1
|
Carvalho MCDC, Ribeiro SA, de Sousa LS, Lima AÂM, Maciel BLL. Undernutrition and Intestinal Infections in Children: A Narrative Review. Nutrients 2025; 17:1479. [PMID: 40362788 PMCID: PMC12073655 DOI: 10.3390/nu17091479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Undernutrition affects thousands of children under five years old worldwide, and various factors are related to its onset, among which we highlight enteric infections and gastrointestinal barrier dysfunction. The cycle of intestinal infections and undernutrition has long-term consequences, such as cognitive deficits, poor growth, and metabolic diseases in adulthood. This review explores factors linked to childhood undernutrition, focusing on intestinal infections and markers of intestinal permeability that affect child development. This narrative review was conducted using Medline/PubMed, Web of Science, and Scopus, from July 2024 to March 2025. Studies involving children under five years old and addressing undernutrition, intestinal infections, or intestinal permeability markers were included. Exclusion criteria comprised studies without therapeutic focus, and books, case reports, or academic theses. No language restrictions were applied, and registration on global platforms was not required. Overall, the studies reported a close relationship between enteric pathogens, diarrheal and non-diarrheal stools, and undernutrition. Among the pathogens most frequently found in the feces of malnourished children were Shigella, enterotoxigenic Escherichia coli, enteroaggregative E. coli (EAEC), and Cryptosporidium. The studies also showed the relationship between gastrointestinal barrier function and undernutrition, with the deterioration of nutrient absorption and, consequently, repercussions on development, linear growth, and weight in children. Although the studies analyzed had different designs and heterogeneity in the age range of the studied children, it was possible to observe the relationship between the infection/undernutrition cycle. Future studies should optimize personalized nutrient-based therapies, assess long-term effects on gut health and growth, and explore the gut microbiome's role in enteric infection susceptibility and undernutrition.
Collapse
Affiliation(s)
- Maria Clara da Cruz Carvalho
- Graduate Program in Health Science, Center for Health Science, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil;
| | - Samilly Albuquerque Ribeiro
- National Institute of Biomedicine of the Brazilian SemiArid, Faculty of Medicine, Federal University of Ceara, Fortaleza 60430-275, CE, Brazil; (S.A.R.); (A.Â.M.L.)
| | | | - Aldo Ângelo Moreira Lima
- National Institute of Biomedicine of the Brazilian SemiArid, Faculty of Medicine, Federal University of Ceara, Fortaleza 60430-275, CE, Brazil; (S.A.R.); (A.Â.M.L.)
| | - Bruna Leal Lima Maciel
- Department of Nutrition, Center for Health Science, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
| |
Collapse
|
2
|
Cazes C, Stobaugh H, Bahwere P, Binns P, Black RE, Boyd E, Briend A, Isanaka S, Kangas ST, Khara T, Lelijveld N, Mwangome M, Myatt M, Odei Obeng-Amoako G, Trehan I, James PT. Re-thinking "non-response" to wasting treatment: Exploratory analysis from 14 studies. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0003741. [PMID: 39937827 DOI: 10.1371/journal.pgph.0003741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/22/2024] [Indexed: 02/14/2025]
Abstract
Children who receive therapeutic feeding for wasting treatment but do not reach the anthropometric definitions of recovery (usually within 12-16 weeks) are categorised as 'non-responders' and considered as treatment failures. We conducted a pooled analysis to explore the growth trajectories of non-responders and the appropriateness of the definition of 'non-response'. We pooled 14 studies of children aged 6-59 months receiving treatment for wasting. We included children classified by their studies as recovered or as non-responders. Observing the pooled data of non-responders' mid-upper arm circumference (MUAC), weight, weight-for-age z-score, weight-for-height z-score and daily weight gain rate, we found that the first quartile differentiated those who did not grow at all versus those that demonstrated some growth. We therefore defined 'low growth non-responders' as < 25th percentile anthropometric gain between admission and exit using the non-responders' pooled study data, and 'high growth non-responders' as ≥ 25th percentile gain. We plotted the growth trajectories of MUAC-, weight- and height-related indices of the recovered, high growth and low growth non-responder groups over time using mixed effects generalised additive models. We compared age, sex and anthropometric characteristics of the three groups and explored predictors of non-response category using a multivariate multinomial logistic regression model. For all outcomes, the high growth non-responders started with a worse anthropometric status compared to those who recovered, but then tracked along a near-parallel growth trajectory. The low growth non-responders showed limited growth throughout treatment. High growth non-responders are better viewed as 'delayed responders' and may need to be kept longer under treatment to recover and reduce the risks from early discharge. Low growth non-responders are the true treatment failures and should be referred for further investigations as quickly as possible. In conclusion, non-responders are not a homogenous group; ~75% of them respond well to treatment and ~25% are treatment failures.
Collapse
Affiliation(s)
- Cécile Cazes
- Emergency Nutrition Network, Oxford, United Kingdom
- University of Bordeaux, National Institute for Health and Medical Research, Research Institute for Sustainable Development, Bordeaux Population Health Research Centre, Bordeaux, France
| | - Heather Stobaugh
- Action Against Hunger USA, New York City, New York, United States of America
- Friedman School of Nutrition Science and Policy at Tufts University, Boston, Massachusetts, United States of America
| | - Paluku Bahwere
- Center for Epidémiology, Biostatistics and Clinical Research, School of Public Health, Université Libre de Bruxelles, Brussels, Belgium
| | - Paul Binns
- Action Against Hunger UK, London, United Kingdom
| | - Robert E Black
- Johns Hopkins Bloomberg School of Public Health, Institute for International Programs, Baltimore, Maryland, United States of America
| | - Erin Boyd
- Friedman School of Nutrition Science and Policy at Tufts University, Boston, Massachusetts, United States of America
- U.S. Agency for International Development (USAID), Bureau for Global Health, Washington, DC, United States of America
| | - André Briend
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Sheila Isanaka
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Suvi T Kangas
- International Rescue Committee, New York City, New York, United States of America
| | - Tanya Khara
- Emergency Nutrition Network, Oxford, United Kingdom
| | | | - Martha Mwangome
- Kenya Medical Research Institute, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
- Kenya Medical Research Institute, Wellcome Trust Research Programme, Kilifi, Kenya
| | - Mark Myatt
- Brixton Health, Llwyngwril, Gwynedd, Wales, United Kingdom
| | - Gloria Odei Obeng-Amoako
- Department of Nutrition and Food Science, School of Biological Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Ghana
| | - Indi Trehan
- Departments of Paediatrics, Global Health, and Epidemiology, University of Washington, Seattle, Washington, United States of America
| | | |
Collapse
|
3
|
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
|
4
|
Urugo MM, Teka TA, Lema TB, Lusweti JN, Djedjibegovíc J, Lachat C, Tesfamariam K, Mesfin A, Astatkie T, Abdel-Wahhab MA. Dietary aflatoxins exposure, environmental enteropathy, and their relation with childhood stunting. Int J Food Sci Nutr 2024; 75:241-254. [PMID: 38404064 DOI: 10.1080/09637486.2024.2314676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/16/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
Childhood stunting is a global phenomenon affecting more than 149 million children under the age of 5 worldwide. Exposure to aflatoxins (AFs) in utero, during breastfeeding, and consumption of contaminated food affect the gut microbiome, resulting in intestinal dysfunction and potentially contributing to stunting. This review explores the potential relationship between AF exposure, environmental enteropathy and childhood stunting. AFs bind to DNA, disrupt protein synthesis and elicit environmental enteropathy (EE). An EE alters the structure of intestinal epithelial cells, impairs nutrient uptake and leads to malabsorption. This article proposes possible intervention strategies for researchers and policymakers to reduce AF exposure, EE and childhood stunting, such as exposure reduction, the implementation of good agricultural practices, dietary diversification and improving environmental water sanitation and hygiene.
Collapse
Affiliation(s)
- Markos Makiso Urugo
- Department of Food Science and Postharvest Technology, College of Agricultural Sciences, Wachemo University, Hosaina, Ethiopia
- Department of Postharvest Management, College of Agriculture and Veterinary Medicine, Jimma University, Jimma, Ethiopia
| | - Tilahun A Teka
- Department of Postharvest Management, College of Agriculture and Veterinary Medicine, Jimma University, Jimma, Ethiopia
| | - Tefera Belachew Lema
- Department of Nutrition and Dietetics, Faculty of Public Health, Institute of Health, Jimma University, Jimma, Ethiopia
| | | | | | - Carl Lachat
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kokeb Tesfamariam
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
- Department of Public Health, College of Medicine and Health Sciences, Ambo University, Ambo, Ethiopia
| | - Addisalem Mesfin
- Center of Excellence in Mycotoxicology and Public Health, MYTOX-SOUTH, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- Department of Human Nutrition, College of Agriculture, Hawassa University, Hawassa, Ethiopia
| | - Tess Astatkie
- Faculty of Agriculture, Dalhousie University, Truro, NS, Canada
| | - Mosaad A Abdel-Wahhab
- Food Toxicology & Contaminants Department, National Research Center, Dokki, Cairo, Egypt
| |
Collapse
|
5
|
Abtahi S, Sailer A, Roland JT, Haest X, Chanez-Paredes SD, Ahmad K, Sadiq K, Iqbal NT, Ali SA, Turner JR. Intestinal Epithelial Digestive, Transport, and Barrier Protein Expression Is Increased in Environmental Enteric Dysfunction. J Transl Med 2023; 103:100036. [PMID: 36870290 PMCID: PMC10121737 DOI: 10.1016/j.labinv.2022.100036] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 01/11/2023] Open
Abstract
Environmental enteric dysfunction (EED) is characterized by malabsorption and diarrhea that result in irreversible deficits in physical and intellectual growth. We sought to define the expression of transport and tight junction proteins by quantitative analysis of duodenal biopsies from patients with EED. Biopsies from Pakistani children with confirmed EED diagnoses were compared to those from age-matched North American healthy controls, patients with celiac disease, and patients with nonceliac disease with villous atrophy or intraepithelial lymphocytosis. Expression of brush border digestive and transport proteins and paracellular (tight junction) proteins was assessed by quantitative multiplex immunofluorescence microscopy. EED was characterized by partial villous atrophy and marked intraepithelial lymphocytosis. Epithelial proliferation and enteroendocrine, tuft, and Paneth cell numbers were unchanged, but there was significant goblet cell expansion in EED biopsies. Expression of proteins involved in nutrient and water absorption and that of the basolateral Cl- transport protein NKCC1 were also increased in EED. Finally, the barrier-forming tight junction protein claudin-4 (CLDN4) was significantly upregulated in EED, particularly within villous enterocytes. In contrast, expression of CFTR, CLDN2, CLDN15, JAM-A, occludin, ZO-1, and E-cadherin was unchanged. Upregulation of a barrier-forming tight junction protein and brush border and basolateral membrane proteins that support nutrient and water transport in EED is paradoxical, as their increased expression would be expected to be correlated with increased intestinal barrier function and enhanced absorption, respectively. These data suggest that EED activates adaptive intestinal epithelial responses to enhance nutrient absorption but that these changes are insufficient to restore health.
Collapse
Affiliation(s)
- Shabnam Abtahi
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anne Sailer
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Joseph T Roland
- Epithelial Biology Center, Vanderbilt University Medical Center; Nashville, Tennessee
| | - Xenia Haest
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sandra D Chanez-Paredes
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kumail Ahmad
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Kamran Sadiq
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Najeeha Talat 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
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Department of Pathology, University of Chicago, Chicago, Illinois.
| |
Collapse
|
6
|
Kwak JH, Lee SW, Lee JE, Ha EK, Baek HS, Lee E, Kim JH, Han MY. Association of Antibiotic Use during the First 6 Months of Life with Body Mass of Children. Antibiotics (Basel) 2022; 11:antibiotics11040507. [PMID: 35453258 PMCID: PMC9033100 DOI: 10.3390/antibiotics11040507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 01/30/2023] Open
Abstract
In this study, our objective was to assess the association of body mass in preschool children with the use of antibiotics within 6 months after birth. National administrative databases were used to examine all children born between 2008 and 2009 in Korea. Exposure was defined as the use of systemic antibiotics during the first 6 months of age. The observed outcomes were stunting (height for age [HFA] z score < −2.0), short stature (HFA z score < −1.64), overweight (body mass index [BMI] for age z score ≥ 1.04), and obesity (BMI for age z score ≥ 1.64), and the children’s height and body weight were measured from three to six years of age. To balance characteristics between the antibiotic user and non-user groups, propensity score matching was performed. The outcomes were evaluated using a generalized estimation equation with the logit link function. Analysis of antibiotic use by children during the first 6 months of life indicated there were 203,073 users (54.9%) and 166,505 non-users (45.1%). After PS matching, there were 72,983 antibiotic users and 72,983 non-users. Antibiotic use was significantly associated with stunting (aOR = 1.198, 95% CI = 1.056 to 1.360) and short stature (aOR = 1.043, 95% CI = 1.004 to 1.083), and had significant negative association with HFA z score (weighted β = −0.023). The use of an antibiotic for 14 days or more had a marked association with stunting. Antibiotic use was also associated with overweight, obesity, and increased BMI for age z score. Antibiotic use during the first 6 months of life increased the risk of stunting, short stature, overweight, and obesity in preschool children.
Collapse
Affiliation(s)
- Ji Hee Kwak
- Department of Pediatrics, Kangbuk Samsung Hospital, School of Medicine, Sungkyunkwan University, Seoul 03181, Korea;
| | - Seung Won Lee
- Department of Data Science, College of Software Convergence, Sejong University, Seoul 05006, Korea; (S.W.L.); (J.E.L.)
- School of Medicine, Sungkyunkwan University, Suwon 16419, Korea
| | - Jung Eun Lee
- Department of Data Science, College of Software Convergence, Sejong University, Seoul 05006, Korea; (S.W.L.); (J.E.L.)
| | - Eun Kyo Ha
- Department of Pediatrics, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Seoul 07441, Korea;
| | - Hey-Sung Baek
- Department of Pediatrics, Kangdong Sacred Heart Hospital, Hallym University Medical Center, Seoul 05355, Korea;
| | - Eun Lee
- Department of Pediatrics, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Korea;
| | - Ju Hee Kim
- Department of Data Science, College of Software Convergence, Sejong University, Seoul 05006, Korea; (S.W.L.); (J.E.L.)
- Department of Pediatrics, Kangdong Sacred Heart Hospital, Hallym University Medical Center, Seoul 05355, Korea;
- Correspondence: (J.H.K.); (M.Y.H.); Tel.: +82-2-2224-2251 (J.H.K.); +82-31-780-3491 (M.Y.H.); Fax: +82-31-780-3942 (M.Y.H.)
| | - Man Yong Han
- Department of Pediatrics, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam 13496, Korea
- Correspondence: (J.H.K.); (M.Y.H.); Tel.: +82-2-2224-2251 (J.H.K.); +82-31-780-3491 (M.Y.H.); Fax: +82-31-780-3942 (M.Y.H.)
| |
Collapse
|
7
|
Kortekangas E, Fan YM, Chaima D, Lehto KM, Malamba-Banda C, Matchado A, Chingwanda C, Liu Z, Ashorn U, Cheung YB, Dewey KG, Maleta K, Ashorn P. Associations between Gut Microbiota and Intestinal Inflammation, Permeability and Damage in Young Malawian Children. J Trop Pediatr 2022; 68:6527323. [PMID: 35149871 PMCID: PMC8846364 DOI: 10.1093/tropej/fmac012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is common in low- and middle-income countries and associated with childhood undernutrition. The composition of gut microbiota has been implicated in the pathogenesis of EED. Our aim was to assess the associations between gut microbiota and EED biomarkers in rural Malawian children. We hypothesized that there would be an inverse association between microbiota maturity and diversity and fecal concentrations of EED biomarkers. METHODS We used data from fecal samples collected at 6, 18 and 30 months from 611 children who were followed up during a nutrition intervention trial. The primary time point for analysis was 18 months. Microbiota data were obtained through 16S rRNA sequencing and variables included microbiota maturity and diversity, phylogenetic dissimilarity and relative abundances of individual taxa. EED biomarkers included calprotectin (marker of inflammation), alpha-1 antitrypsin (intestinal permeability) and REG1B (intestinal damage). RESULTS There was an inverse association between microbiota maturity and diversity and fecal concentrations of all 3 EED biomarkers at 18 months (p≤0.001). The results were similar at 30 months, while at 6 months inverse associations were found only with calprotectin and alpha-1 antitrypsin concentrations. At 18 months, EED biomarkers were not associated with phylogenetic dissimilarity, but at 6 and 30 months several associations were observed. Individual taxa predicting EED biomarker concentrations at 18 months included several Bifidobacterium and Enterobacteriaceae taxa as well as potentially displaced oral taxa. CONCLUSIONS Our findings support the hypothesis of an inverse association between microbiota maturity and diversity and EED in rural Malawian children.
Collapse
Affiliation(s)
- Emma Kortekangas
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere 33014, Finland,Correspondence: Emma Kortekangas, Tampere Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Global Health, Tampere University, Arvo Ylpön katu 34, Arvo building, Tampere 33014, Finland. Tel: +358-3-355-111. Fax +358-3-213-4473. E-mail <>
| | - Yue-Mei Fan
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere 33014, Finland
| | - David Chaima
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Kirsi-Maarit Lehto
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere 33014, Finland
| | - Chikondi Malamba-Banda
- 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,Department of Nutrition and Institute for Global Nutrition, University of California Davis, Davis, CA 95616, USA
| | - Chilungamo Chingwanda
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Zhifei Liu
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere 33014, Finland
| | - Ulla Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere 33014, Finland
| | - Yin Bun Cheung
- Program in Health Services & Systems Research and Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Kathryn G Dewey
- Department of Nutrition and Institute for Global Nutrition, University of California Davis, Davis, CA 95616, USA
| | - Kenneth Maleta
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Per Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere 33014, Finland,Department of Pediatrics, Tampere University Hospital, Tampere 33520, Finland
| |
Collapse
|
8
|
Keddy KH, Saha S, Okeke IN, Kalule JB, Qamar FN, Kariuki S. Combating Childhood Infections in LMICs: evaluating the contribution of Big Data Big data, biomarkers and proteomics: informing childhood diarrhoeal disease management in Low- and Middle-Income Countries. EBioMedicine 2021; 73:103668. [PMID: 34742129 PMCID: PMC8579132 DOI: 10.1016/j.ebiom.2021.103668] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/26/2021] [Accepted: 10/20/2021] [Indexed: 01/20/2023] Open
Abstract
Despite efforts to reduce the global burden of childhood diarrhoea, 50% of all cases globally occur in children under five years in Low–Income and Middle- Income Countries (LMICs) and knowledge gaps remain regarding the aetiological diagnosis, introduction of diarrhoeal vaccines, and the role of environmental enteric dysfunction and severe acute malnutrition. Biomarkers may assist in understanding disease processes, from diagnostics, to management of childhood diarrhoea and the sequelae to vaccine development. Proteomics has the potential to assist in the identification of new biomarkers to understand the processes in the development of childhood diarrhoea and to aid in developing new vaccines. Centralised repositories that enable mining of large data sets to better characterise risk factors, the proteome of both the patient and the different diarrhoeal pathogens, and the environment, could inform patient management and vaccine development, providing a systems biological approach to address the burden of childhood diarrhoea in LMICs.
Collapse
Affiliation(s)
- Karen H Keddy
- Tuberculosis Platform, South African Medical Research Council, 1 Soutpansberg Rd, Pretoria, 0001, South Africa.
| | - Senjuti Saha
- Child Health Research Foundation, 23/2 Khilji Road, Mohammadpur, Dhaka 1207, Bangladesh
| | - Iruka N Okeke
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Oyo State, Nigeria
| | - John Bosco Kalule
- Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Uganda
| | - Farah Naz Qamar
- Department of Pediatrics and Child Health. Aga Khan University, Stadoum road Karachi, Pakistan 74800
| | - Samuel Kariuki
- Centre for Microbiology Research, Kenya Medical Research Institute, Off Mbagathi Road, Nairobi, Kenya
| |
Collapse
|
9
|
Luo X, Huo X, Zhang Y, Cheng Z, Chen S, Xu X. Increased intestinal permeability with elevated peripheral blood endotoxin and inflammatory indices for e-waste lead exposure in children. CHEMOSPHERE 2021; 279:130862. [PMID: 34134434 DOI: 10.1016/j.chemosphere.2021.130862] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/09/2021] [Accepted: 05/09/2021] [Indexed: 02/05/2023]
Abstract
Lead (Pb) entering the body through different channels can damage the function of intestinal mucosal barrier and cause the body stressful inflammatory response to enhance. This study conducted a cross-sectional study to investigate the effects of Pb exposure on intestinal permeability in children by measuring the level of bacterial endotoxin and index of inflammatory cell types in peripheral blood. From November to December 2018, we recruited 187 participants aged 3-6 years by stratified randomization, from an electronic-waste-exposed group (n = 82) and a referent group (n = 105). General demographic information, past history of the digestive system in child, and family situation were informed by children's guardians with questionnaires. Children in the exposed group showed lower weight, height, and body mass index while more diarrhea in a month. Blood Pb and plasma endotoxin were elevated in exposed children than referent children and the positive relationship between them was shown in all children [B (95% CI): 0.072 (0.008, 0.137), P = 0.033]. Peripheral monocyte counts and leukotriene B4 (LTB4) levels were significantly increased in the exposed group. Endotoxin levels were positively correlated with neutrophils, monocytes, and LTB4 [B (95% CI): 0.054 (0.015, 0.093), 0.018 (0.005, 0.031), and 0.049 (0.011, 0.087), respectively, P < 0.05]. To sum up, the exposed children showed lower physical growth levels, poorer gut health, and increased intestinal permeability, which was related to high blood Pb and peripheral inflammatory indices. These results suggest the possible adverse impact of environmental Pb exposure on the intestinal health of children.
Collapse
Affiliation(s)
- Xiuli Luo
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China
| | - Yuling Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Zhiheng Cheng
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Shuqin Chen
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
10
|
Budge S, Hutchings P, Parker A, Tyrrel S, Norton S, Garbutt C, Woldemedhin F, Jemal MY, Moges M, Hussen S, Beyene H. A randomised controlled feasibility trial of a BabyWASH household playspace: The CAMPI study. PLoS Negl Trop Dis 2021; 15:e0009514. [PMID: 34260591 PMCID: PMC8312948 DOI: 10.1371/journal.pntd.0009514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 07/26/2021] [Accepted: 05/28/2021] [Indexed: 11/26/2022] Open
Abstract
Background Water, sanitation and hygiene (WASH) interventions should support infant growth but trial results are inconsistent. Frequently, interventions do not consider behaviours or transmission pathways specific to age. A household playspace (HPS) is one intervention component which may block faecal-oral transmission. This study was a two-armed, parallel-group, randomised, controlled feasibility trial of a HPS in rural Ethiopia. It aimed to recommend proceeding to a definitive trial. Secondary outcomes included effects on infant health, injury prevention and women’s time. Methods November 2019−January 2020 106 households were identified and assessed for eligibility. Recruited households (N = 100) were randomised (blinded prior to the trial start) to intervention or control (both n = 50). Outcomes included recruitment, attrition, adherence, and acceptability. Data were collected at baseline, two and four weeks. Findings Recruitment met a priori criteria (≥80%). There was no loss to follow-up, and no non-use, meeting adherence criteria (both ≤10%). Further, 48.0% (95% CI 33.7−62.6; n = 24) of households appropriately used and 56.0% (41.3−70.0; n = 28) cleaned the HPS over four weeks, partly meeting adherence criteria (≥50%). For acceptability, 41.0% (31.3−51.3; n = 41) of infants were in the HPS during random visits, failing criteria (≥50%). Further, the proportion of HPS use decreased during some activities, failing criteria (no decrease in use). A modified Barrier Analysis described good acceptability and multiple secondary benefits, including on women’s time burden and infant injury prevention. Interpretation Despite failing some a priori criteria, the trial demonstrated mixed adherence and good acceptability among intervention households. A definitive trial to determine efficacy is warranted if recommended adjustments are made. Funding People In Need; Czech Development Agency. Trial registration RIDIE-ID-5de0b6938afb8. This research tested a new way to protect infants and young children from infections that are caused by pathogens in human and animal faeces. It tested the feasibility of using a household playspace to reduce infection by creating a hygienic environment for children to play-in in rural Ethiopia. The results show that the household playspace was well accepted, used regularly and cleaned well by participants in the study. The study also suggests a potential positive impact in reducing diarrhoea. Based on these results, we suggest that a larger scale trial be conducted to conclusively assess whether a household playspace can protect young children and infants from infection in rural Ethiopia or similar contexts.
Collapse
Affiliation(s)
- Sophie Budge
- Cranfield Water Science Institute, Cranfield University, Cranfield, United Kingdom
| | - Paul Hutchings
- Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, United Kingdom
- * E-mail:
| | - Alison Parker
- Cranfield Water Science Institute, Cranfield University, Cranfield, United Kingdom
| | - Sean Tyrrel
- Cranfield Water Science Institute, Cranfield University, Cranfield, United Kingdom
| | - Sam Norton
- Psychology Department, Institute of Psychiatry Psychology & Neuroscience, King’s College London, London, United Kingdom
| | | | | | | | - Mathewos Moges
- Hawassa University College of Medicine and Health Sciences, Hawassa, Ethiopia
| | - Siraj Hussen
- Hawassa University College of Medicine and Health Sciences, Hawassa, Ethiopia
| | - Hunachew Beyene
- Hawassa University College of Medicine and Health Sciences, Hawassa, Ethiopia
| |
Collapse
|
11
|
Pesu H, Mutumba R, Mbabazi J, Olsen MF, Mølgaard C, Michaelsen KF, Ritz C, Filteau S, Briend A, Mupere E, Friis H, Grenov B. The Role of Milk Protein and Whey Permeate in Lipid-based Nutrient Supplements on the Growth and Development of Stunted Children in Uganda: A Randomized Trial Protocol (MAGNUS). Curr Dev Nutr 2021; 5:nzab067. [PMID: 34027295 PMCID: PMC8128719 DOI: 10.1093/cdn/nzab067] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/25/2021] [Accepted: 04/16/2021] [Indexed: 11/13/2022] Open
Abstract
Stunting is associated with cognitive impairment and later chronic disease. Previous trials to prevent stunting have had little effect, and no trials seem to have provided larger amounts of energy and high-quality proteins to already stunted children. We aimed to assess the effects of milk protein (MP) and whey permeate (WP) in large-quantity lipid-based nutrient supplements (LNS-LQ), among stunted children, on linear growth and child development. This was a randomized, double-blind, 2-by-2 factorial trial. Stunted children aged 12-59 mo from eastern Uganda (n = 750) were randomly assigned to receive 100 g LNS-LQ with or without MP and WP (n = 4 × 150) or no supplement (n = 150) for 3 mo. The primary outcomes were change in knee-heel and total length. Secondary outcomes included child development, body composition, anthropometry, and hemoglobin. Micronutrient status, intestinal function, and microbiota were also assessed. Our findings will contribute to an understanding of the role of milk ingredients and LNS in linear catch-up growth. This trial was registered at www.isrctn.com as ISRCTN13093195.
Collapse
Affiliation(s)
- Hannah Pesu
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Rolland Mutumba
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
- Department of Paediatrics and Child Health, School of Medicine College of Health Sciences, Makerere University, Kampala, Uganda
| | - Joseph Mbabazi
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
- Department of Paediatrics and Child Health, School of Medicine College of Health Sciences, Makerere University, Kampala, Uganda
| | - Mette F Olsen
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Christian Mølgaard
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Kim F Michaelsen
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Christian Ritz
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Suzanne Filteau
- Department of Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - André Briend
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
- Tampere Centre for Child Health Research, Tampere University, Tampere, Finland
| | - Ezekiel Mupere
- Department of Paediatrics and Child Health, School of Medicine College of Health Sciences, Makerere University, Kampala, Uganda
| | - Henrik Friis
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Benedikte Grenov
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Njunge JM, Gonzales GB, Ngari MM, Thitiri J, Bandsma RH, Berkley JA. Systemic inflammation is negatively associated with early post discharge growth following acute illness among severely malnourished children - a pilot study. Wellcome Open Res 2021; 5:248. [PMID: 33969227 PMCID: PMC8080977 DOI: 10.12688/wellcomeopenres.16330.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Rapid growth should occur among children with severe malnutrition (SM) with medical and nutritional management. Systemic inflammation (SI) is associated with death among children with SM and is negatively associated with linear growth. However, the relationship between SI and weight gain during therapeutic feeding following acute illness is unknown. We hypothesised that growth post-hospital discharge is associated with SI among children with SM. Methods: We conducted secondary analysis of data from HIV-uninfected children with SM (n=98) who survived and were not readmitted to hospital during one year of follow-up. We examined the relationship between changes in absolute deficits in weight and mid-upper-arm circumference (MUAC) from enrolment at stabilisation to 60 days and one year later, and untargeted plasma proteome, targeted cytokines/chemokines, leptin, and soluble CD14 using multivariate regularized linear regression. Results: The mean change in absolute deficit in weight and MUAC was -0.50kg (standard deviation; SD±0.69) and -1.20cm (SD±0.89), respectively, from enrolment to 60 days later. During the same period, mean weight and MUAC gain was 3.3g/kg/day (SD±2.4) and 0.22mm/day (SD±0.2), respectively. Enrolment interleukins; IL17-alpha and IL-2, and serum amyloid P were negatively associated with weight and MUAC gain during 60 days. Lipopolysaccharide binding protein and complement component 2 were negatively associated with weight gain only. Leptin was positively associated with weight gain. Soluble CD14, beta-2 microglobulin, and macrophage inflammatory protein 1 beta were negatively associated with MUAC gain only. Glutathione peroxidase 3 was positively associated with weight and MUAC gain during one year. Conclusions: Early post-hospital discharge weight and MUAC gain were rapid and comparable to children with uncomplicated SM treated in the community. Higher concentrations of SI markers were associated with less weight and MUAC gain, suggesting inflammation negatively impacts recovery from wasting. This finding warrants further research on reducing inflammation on growth among children with SM.
Collapse
Affiliation(s)
- James M. Njunge
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Gerard Bryan Gonzales
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Moses M. Ngari
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Johnstone Thitiri
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Robert H.J. Bandsma
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - James A. Berkley
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
13
|
Budge S, Parker A, Hutchings P, Garbutt C, Rosenbaum J, Tulu T, Woldemedhin F, Jemal M, Engineer B, Williams L. Multi-Sectoral Participatory Design of a BabyWASH Playspace for Rural Ethiopian Households. Am J Trop Med Hyg 2021; 104:884-897. [PMID: 33534743 PMCID: PMC7941829 DOI: 10.4269/ajtmh.20-0945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/21/2020] [Indexed: 12/21/2022] Open
Abstract
Growing evidence suggests current water, sanitation, and hygiene interventions do not improve domestic hygiene sufficiently to improve infant health, nor consider the age-specific behaviors which increase infection risk. A household playspace (HPS) is described as one critical intervention to reduce direct fecal-oral transmission within formative growth periods. This article details both the design and development (materials and methods), and testing (results) of a HPS for rural Ethiopian households. Design and testing followed a multi-sectoral, multistep participatory process. This included a focus group discussion (FGD), two user-centered and participatory design workshops in the United Kingdom and Ethiopia, discussions with local manufacturers, and a Trials by Improved Practices (TIPs) leading to a final prototype design. Testing included the FGD and TIPs study and a subsequent randomized controlled feasibility trial in Ethiopian households. This multi-sectoral, multistage development process demonstrated a HPS is an acceptable and feasible intervention in these low-income, rural subsistence Ethiopian households. A HPS may help reduce fecal-oral transmission and infection-particularly in settings where free-range domestic livestock present an increased risk. With the need to better tailor interventions to improve infant health, this article also provides a framework for future groups developing similar material inputs and highlights the value of participatory design in this field.
Collapse
Affiliation(s)
- Sophie Budge
- Cranfield Water Science Institute, Cranfield University, Cranfield, United Kingdom
| | - Alison Parker
- Cranfield Water Science Institute, Cranfield University, Cranfield, United Kingdom
| | - Paul Hutchings
- Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, United Kingdom
| | | | - Julia Rosenbaum
- FHI360/United States Agency for International Development WASHPaLS Project, Washington, District of Columbia
| | | | | | | | | | - Leon Williams
- Centre for Competitive Creative Design, Cranfield University, Cranfield, United Kingdom
| |
Collapse
|
14
|
Chen D, McKune SL, Singh N, Yousuf Hassen J, Gebreyes W, Manary MJ, Bardosh K, Yang Y, Diaz N, Mohammed A, Terefe Y, Roba KT, Ketema M, Ameha N, Assefa N, Rajashekara G, Deblais L, Ghanem M, Yimer G, Havelaar AH. Campylobacter Colonization, Environmental Enteric Dysfunction, Stunting, and Associated Risk Factors Among Young Children in Rural Ethiopia: A Cross-Sectional Study From the Campylobacter Genomics and Environmental Enteric Dysfunction (CAGED) Project. Front Public Health 2021; 8:615793. [PMID: 33553097 PMCID: PMC7862945 DOI: 10.3389/fpubh.2020.615793] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/17/2020] [Indexed: 11/19/2022] Open
Abstract
Livestock farming provides a possible mechanism by which smallholder farmers can meet their household need for animal source foods (ASF), which may reduce the risk of stunting. However, direct/indirect contacts with domestic animals may increase colonization by Campylobacter spp., which has been associated with Environmental Enteric Dysfunction (EED) and stunting. A cross-sectional study involving 102 randomly selected children between 12 and 16 months of age was conducted in rural eastern Ethiopia to establish prevalence rates of Campylobacter colonization, EED, and stunting, and evaluate potential risk factors. Data were collected between September and December 2018. The prevalence of EED and stunting was 50% (95% CI: 40-60%) and 41% (95% CI: 32-51%), respectively. Among enrolled children, 56% had consumed some ASF in the previous 24 h; 47% had diarrhea and 50% had fever in the past 15 days. 54, 63, 71 or 43% of households owned at least one chicken, cow/bull, goat, or sheep; 54 (53%) households kept chickens indoors overnight and only half of these confined the animals. Sanitation was poor, with high levels of unimproved latrines and open defecation. Most households had access to an improved source of drinking water. The prevalence of Campylobacter colonization was 50% (95% CI: 41-60%) by PCR. In addition to the thermotolerant species Campylobacter jejuni, Campylobacter coli and Campylobacter upsaliensis, non-thermotolerant species related to Campylobacter hyointestinalis and Campylobacter fetus were frequently detected by Meta-total RNA sequencing (MeTRS). Current breastfeeding and ASF consumption increased the odds of Campylobacter detection by PCR, while improved drinking water supply decreased the odds of EED. No risk factors were significantly associated with stunting. Further studies are necessary to better understand reservoirs and transmission pathways of Campylobacter spp. and their potential impact on child health.
Collapse
Affiliation(s)
- Dehao Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Sarah L. McKune
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
- Center for African Studies, University of Florida, Gainesville, FL, United States
| | - Nitya Singh
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Animal Sciences, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Jemal Yousuf Hassen
- Department of Rural Development and Agricultural Extension, Haramaya University, Dire Dawa, Ethiopia
| | - Wondwossen Gebreyes
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, United States
| | - Mark J. Manary
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - Kevin Bardosh
- Department of Anthropology, University of Florida, Gainesville, FL, United States
| | - Yang Yang
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Biostatistics, College of Public Health and Health Professions & College of Medicine, University of Florida, Gainesville, FL, United States
| | - Nicholas Diaz
- Center for African Studies, University of Florida, Gainesville, FL, United States
| | | | - Yitagele Terefe
- College of Veterinary Medicine, Haramaya University, Dire Dawa, Ethiopia
| | - Kedir Teji Roba
- College of Health and Medical Sciences, Haramaya University, Dire Dawa, Ethiopia
| | - Mengistu Ketema
- School of Agricultural Economics and Agribusiness, Haramaya University, Dire Dawa, Ethiopia
| | - Negassi Ameha
- School of Animal and Range Science, Haramaya University, Dire Dawa, Ethiopia
| | - Nega Assefa
- College of Health and Medical Sciences, Haramaya University, Dire Dawa, Ethiopia
| | - Gireesh Rajashekara
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, United States
| | - Loïc Deblais
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, United States
| | - Mostafa Ghanem
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, United States
| | - Getnet Yimer
- Global One Health initiative, Office of International Affairs, The Ohio State University, Eastern Africa Regional Office, Addis Ababa, Ethiopia
| | - Arie H. Havelaar
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Animal Sciences, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
- Institute for Sustainable Food Systems, University of Florida, Gainesville, FL, United States
| |
Collapse
|
15
|
George CM, Cirhuza LB, Kuhl J, Williams C, Coglianese N, Thomas E, Bauler S, François R, Saxton R, Presence AS, Birindwa A, Jean Claude BR, Perin J, Mirindi P. Child Mouthing of Feces and Fomites and Animal Contact are Associated with Diarrhea and Impaired Growth Among Young Children in the Democratic Republic of the Congo: A Prospective Cohort Study (REDUCE Program). J Pediatr 2021; 228:110-116.e1. [PMID: 32918918 DOI: 10.1016/j.jpeds.2020.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/04/2020] [Accepted: 09/04/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To identify exposure pathways to fecal pathogens that are significant contributors to diarrheal diseases and impaired growth in young children, and to evaluate scalable interventions to reduce fecal contamination from these pathways. STUDY DESIGN Reducing Enteropathy, Undernutrition, and Contamination in the Environment (REDUCE) was a prospective cohort study of 370 children <5 years of age was conducted in Walungu Territory, South Kivu, Democratic Republic of the Congo. Child mouthing behaviors were assessed through caregiver reports and 5-hour structured observations. Caregiver reports of child contact with animals and child diarrhea were also obtained. Anthropometric measurements were collected at baseline and at a 6-month follow-up. RESULTS Children observed putting soil in their mouth during structured observation at baseline had a significantly higher odds of diarrhea at the 6-month follow-up (OR, 1.79; 95% CI, 1.04 to 3.07). Children observed mouthing feces during structured observation had a significant reduction in height-for-age z-score (HAZ) from baseline to the 6-month follow-up (ΔHAZ, -0.69; 95% CI, -1.34 to -0.04). A significant reduction in HAZ was also observed for children with caregiver reports of touching guinea pigs (-0.33; 95% CI, -0.58 to -0.08) and rabbits (-0.34; 95% CI, -0.64 to -0.04) and children with feces in their sleeping space during unannounced spot checks (-0.41; 95% CI, -0.74 to -0.09). CONCLUSIONS These findings emphasize the urgent need for infant water, sanitation, and hygiene interventions targeting child mouthing behaviors, fecal contamination in child living spaces, and child contact with domestic animals to reduce exposure to fecal pathogens among susceptible populations.
Collapse
Affiliation(s)
- Christine Marie George
- Department of International Health, Johns Hopkins School Bloomberg of Public Health, Baltimore, MD.
| | - Lucien Bisimwa Cirhuza
- Food for the Hungry, Phoenix, AZ; Unit for Research and Training in Ecology and Control of Infectious Diseases, Microbiology Department, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Jennifer Kuhl
- Department of International Health, Johns Hopkins School Bloomberg of Public Health, Baltimore, MD
| | - Camille Williams
- Department of International Health, Johns Hopkins School Bloomberg of Public Health, Baltimore, MD
| | | | - Elizabeth Thomas
- Department of International Health, Johns Hopkins School Bloomberg of Public Health, Baltimore, MD
| | | | - Ruthly François
- Department of International Health, Johns Hopkins School Bloomberg of Public Health, Baltimore, MD
| | - Ronald Saxton
- Department of International Health, Johns Hopkins School Bloomberg of Public Health, Baltimore, MD
| | | | | | | | - Jamie Perin
- Department of International Health, Johns Hopkins School Bloomberg of Public Health, Baltimore, MD
| | | |
Collapse
|
16
|
Hart JD, Kalua K, Keenan JD, Lietman TM, Bailey RL. Effect of Mass Treatment with Azithromycin on Causes of Death in Children in Malawi: Secondary Analysis from the MORDOR Trial. Am J Trop Med Hyg 2020; 103:1319-1328. [PMID: 32342837 PMCID: PMC7470551 DOI: 10.4269/ajtmh.19-0613] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Recent evidence indicates mass drug administration with azithromycin may reduce child mortality. This study uses verbal autopsy (VA) to investigate the causes of individual deaths during the Macrolides Oraux pour Réduire les Décès avec un Oeil sur la Résistance (MORDOR) trial in Malawi. Cluster randomization was performed as part of MORDOR. Biannual household visits were conducted to distribute azithromycin or placebo to children aged 1–59 months and update the census to identify deaths for VA. MORDOR was not powered to investigate mortality effects at individual sites, but the available evidence is presented here for hypothesis generation regarding the mechanism through which azithromycin may reduce child mortality. Automated VA analysis was performed to infer the likely cause of death using two major analysis programs, InterVA and SmartVA. A total of 334 communities were randomized to azithromycin or placebo, with more than 130,000 person-years of follow-up. During the study, there were 1,184 deaths, of which 1,131 were followed up with VA. Mortality was 9% lower in azithromycin-treated communities than in placebo communities (rate ratio 0.91 [95% CI: 0.79–1.05]; P = 0.20). The intention-to-treat analysis by cause using InterVA suggested fewer HIV/AIDS deaths in azithromycin-treated communities (rate ratio 0.70 [95% CI: 0.50–0.97]; P = 0.03) and fewer pneumonia deaths (rate ratio 0.82 [95% CI: 0.60–1.12]; P = 0.22). The use of the SmartVA algorithm suggested fewer diarrhea deaths (rate ratio 0.71 [95% CI: 0.51–1.00]; P = 0.05) and fewer pneumonia deaths (rate ratio 0.58 [95% CI: 0.33–1.00]; P = 0.05). Although this study is not able to provide strong evidence, the data suggest that the mortality reduction during MORDOR in Malawi may have been due to effects on pneumonia and diarrhea or HIV/AIDS mortality.
Collapse
Affiliation(s)
- John D Hart
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Khumbo Kalua
- Blantyre Institute for Community Outreach and College of Medicine, University of Malawi, Blantyre, Malawi
| | - Jeremy D Keenan
- Department of Ophthalmology, Francis I. Proctor Foundation, University of California, San Francisco, San Francisco, California
| | - Thomas M Lietman
- Department of Ophthalmology, Francis I. Proctor Foundation, University of California, San Francisco, San Francisco, California
| | - Robin L Bailey
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
17
|
Njunge JM, Gonzales GB, Ngari MM, Thitiri J, Bandsma RH, Berkley JA. Systemic inflammation is negatively associated with early post discharge growth following acute illness among severely malnourished children - a pilot study. Wellcome Open Res 2020; 5:248. [PMID: 33969227 PMCID: PMC8080977 DOI: 10.12688/wellcomeopenres.16330.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2020] [Indexed: 11/03/2023] Open
Abstract
Background: Rapid growth should occur among children with severe malnutrition (SM) when medically and nutritionally treated. Systemic inflammation (SI) is associated with death among children with SM and is negatively associated with linear growth. However, the relationship between SI and weight gain during therapeutic feeding following acute illness is unknown. We hypothesised that growth in the first 60 days post-hospital discharge is associated with SI among children with SM. Methods: We conducted secondary analysis of data from HIV-uninfected children with SM (n=98) who survived and were not readmitted to hospital during one year of follow up. We examined the relationship between changes in absolute deficits in weight and mid-upper-arm circumference (MUAC) from enrolment at stabilisation to 60 days later and untargeted plasma proteome, targeted cytokines/chemokines, leptin, and soluble CD14 (sCD14) using multivariate regularized linear regression. Results: The mean change in absolute deficit in weight and MUAC was -0.50kg (standard deviation; SD±0.69) and -1.20cm (SD±0.89), respectively, from enrolment to 60 days later. During the same period, mean weight and MUAC gain was 3.3g/kg/day (SD±2.4) and 0.22mm/day (SD±0.2), respectively. Enrolment inflammatory cytokines interleukin 17 alpha (IL17α), interleukin 2 (IL2), and serum amyloid P (SAP) were negatively associated with weight and MUAC gain. Lipopolysaccharide binding protein (LBP) and complement component 2 were negatively associated with weight gain only. Leptin was positively associated with weight gain. sCD14, beta-2 microglobulin (β2M), and macrophage inflammatory protein 1 beta (MIP1β) were negatively associated with MUAC gain only. Conclusions: Early post-hospital discharge weight and MUAC gain were rapid and comparable to children with uncomplicated SM treated with similar diet in the community. Higher concentrations of SI markers were associated with less weight and MUAC gain, suggesting inflammation negatively impacts recovery from wasting. This finding warrants further research on the role of inflammation on growth among children with SM.
Collapse
Affiliation(s)
- James M. Njunge
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Gerard Bryan Gonzales
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Moses M. Ngari
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Johnstone Thitiri
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Robert H.J. Bandsma
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - James A. Berkley
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Robertson LJ, Johansen ØH, Kifleyohannes T, Efunshile AM, Terefe G. Cryptosporidium Infections in Africa-How Important Is Zoonotic Transmission? A Review of the Evidence. Front Vet Sci 2020; 7:575881. [PMID: 33195574 PMCID: PMC7580383 DOI: 10.3389/fvets.2020.575881] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/27/2020] [Indexed: 01/25/2023] Open
Abstract
Cryptosporidium, a protozoan parasite in the phylum Apicomplexa, is the etiological agent of cryptosporidiosis, an intestinal infection characterized by profuse watery diarrhea. Over 30 species of Cryptosporidium are recognized, some host specific whereas others infect a broader host range. Cryptosporidium hominis and Cryptosporidium parvum are the species most commonly associated with human infection; C. hominis is largely associated only with human infections, but C. parvum is also associated with infection in animals, especially young ruminants. In some regions, cryptosporidiosis is a serious veterinary problem, particularly for calves, and lambs. Many outbreaks of human cryptosporidiosis have been associated with zoonotic transmission following contact with infected animals. In Africa, where cryptosporidiosis is a major contributor to pediatric morbidity and mortality, evidence suggests transmission is principally anthroponotic. Given the frequent close contact between humans and animals in Africa, the apparent predominance of human-to-human transmission is both interesting and puzzling. In this article, after a brief "text book" introduction to the parasite, we consider in separate sections the different aspects of relevance to Cryptosporidium transmission in African countries, describing different aspects of the various species and subtypes in human and animal infections, considering livestock management practices in different African countries, and looking for any characteristic "hot spots" where zoonotic transmission has apparently occurred. Studies where transmission networks have been investigated are particularly relevant. Finally, in a separate section, we try to gather these different strands of evidence together in order to assess the reasons behind the apparent predominance of anthroponotic transmission in Africa. Reviewing the available evidence provides an opportunity to re-think transmission pathways, not only in Africa but also elsewhere, and also to pose questions. Does the predominance of human-to-human transmission in Africa reflect a relative absence of zoonotic C. parvum in African livestock? Are Africans less susceptible to zoonotic Cryptosporidium infection, perhaps resulting from early immunostimulation by C. hominis or due to inherent genetic traits? Is the African environment-in all its variety-simply more detrimental to oocyst survival? Will the so-called hypertransmissible subtypes, currently relatively rare in Africa, be introduced from Europe or elsewhere, and, if so, will they fade out or establish and spread? Our intention with this manuscript is not only to summarize and consolidate diverse data, thereby providing an overview of data gaps, but also to provide food for thought regarding transmission of a parasite that continues to have a considerable impact on both human and animal health.
Collapse
Affiliation(s)
- Lucy J. Robertson
- Parasitology Laboratory, Department of Paraclinical Science, Faculty of Veterinary Medicine Norwegian University of Life Sciences, Oslo, Norway
| | - Øystein Haarklau Johansen
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Microbiology, Vestfold Hospital Trust, Tønsberg, Norway
| | - Tsegabirhan Kifleyohannes
- Parasitology Laboratory, Department of Paraclinical Science, Faculty of Veterinary Medicine Norwegian University of Life Sciences, Oslo, Norway
- Department of Veterinary Basic and Diagnostic Sciences, College of Veterinary Medicine, Mekelle University, Mekelle, Ethiopia
| | - Akinwale Michael Efunshile
- Department of Medical Microbiology, Alex Ekwueme Federal University Teaching Hospital, Abakaliki, Nigeria
- Department of Medical Microbiology, Ebonyi State University, Abakaliki, Nigeria
| | - Getachew Terefe
- College of Veterinary Medicine and Agriculture, Department of Pathology and Parasitology, Addis Ababa University, Bishoftu, Ethiopia
| |
Collapse
|
19
|
Lauer JM, Ghosh S, Ausman LM, Webb P, Bashaasha B, Agaba E, Turyashemererwa FM, Tran HQ, Gewirtz AT, Erhardt J, Duggan CP. Markers of Environmental Enteric Dysfunction Are Associated with Poor Growth and Iron Status in Rural Ugandan Infants. J Nutr 2020; 150:2175-2182. [PMID: 32455424 PMCID: PMC7398767 DOI: 10.1093/jn/nxaa141] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/25/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED), characterized by altered intestinal permeability/inflammation, microbial translocation, and systemic inflammation (SI), may be a significant contributor to micronutrient deficiencies and poor growth in infants from low-resource settings. OBJECTIVE We examined associations among EED, SI, growth, and iron status at 6 mo of age. METHODS We performed a cross-sectional analysis of 6-mo-old infants (n = 548) enrolled in a Ugandan birth-cohort study (NCT04233944). EED was assessed via serum concentrations of anti-flagellin and anti- LPS immunoglobulins (Igs); SI was assessed via serum concentrations of ɑ1-acid glycoprotein (AGP) and C-reactive protein (CRP); iron status was assessed via serum concentrations of hemoglobin (Hb), soluble transferrin receptor (sTfR), and ferritin. Associations were assessed using adjusted linear regression analysis. RESULTS At 6 mo, ∼35% of infants were stunted [length-for-age z score (LAZ) < -2] and ∼53% were anemic [hemoglobin (Hb) <11.0 g/dL]. Nearly half (∼46%) had elevated AGP (>1 g/L) and ∼30% had elevated CRP (>5 mg/L). EED and SI biomarkers were significantly correlated (r = 0.142-0.193, P < 0.001 for all). In adjusted linear regression models, which included adjustments for SI, higher anti-flagellin IgA, anti-LPS IgA, and anti-LPS IgG concentrations were each significantly associated with lower LAZ [β (95% CI): -0.21 (-0.41, 0.00), -0.23 (-0.44, -0.03), and -0.33 (-0.58, -0.09)]. Furthermore, higher anti-flagellin IgA, anti-flagellin IgG, and anti-LPS IgA concentrations were significantly associated with lower Hb [β (95% CI): -0.24 (-0.45, -0.02), -0.58 (-1.13, 0.00), and -0.26 (-0.51, 0.00)] and higher anti-flagellin IgG and anti-LPS IgG concentrations were significantly associated with higher sTfR [β (95% CI): 2.31 (0.34, 4.28) and 3.13 (0.75, 5.51)]. CONCLUSIONS EED is associated with both low LAZ and iron status in 6-mo-old infants. Further research on the mechanisms by which EED affects growth and micronutrient status is warranted.
Collapse
Affiliation(s)
- Jacqueline M Lauer
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA,USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA, USA,Address correspondence to JML (e-mail: )
| | - Shibani Ghosh
- USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA, USA,Gerald J and Dorothy R Friedman School of Nutrition Science and Policy at Tufts University, Boston, MA, USA
| | - Lynne M Ausman
- USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA, USA,Gerald J and Dorothy R Friedman School of Nutrition Science and Policy at Tufts University, Boston, MA, USA
| | - Patrick Webb
- USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA, USA,Gerald J and Dorothy R Friedman School of Nutrition Science and Policy at Tufts University, Boston, MA, USA
| | - Bernard Bashaasha
- Department of Agribusiness and Natural Resource Economics, Makerere University, Kampala, Uganda
| | - Edgar Agaba
- USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA, USA
| | | | - Hao Q Tran
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Andrew T Gewirtz
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | | | - Christopher P Duggan
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA,USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA, USA,Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
20
|
Monira S, Bhuyian MSI, Parvin T, Uddin IM, Zohura F, Hasan MT, Biswas SK, Hasan K, Masud J, Rashid M, Rahman Z, Papri N, Rafique R, Islam A, Barman I, Jubyda FT, Johura F, Sultana M, Sanin KI, Sack DA, Perin J, Alam M, George C. Child mouthing of soil and presence of animals in child sleeping spaces are associated with growth faltering among young children in Dhaka, Bangladesh (CHoBI7 Program). Trop Med Int Health 2020; 25:1016-1023. [DOI: 10.1111/tmi.13417] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shirajum Monira
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | | | - Tahmina Parvin
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Ismat Minhaj Uddin
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Fatema Zohura
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - M. Tasdik Hasan
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Shwapon K. Biswas
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Khaled Hasan
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Jahed Masud
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Mahamud‐ur Rashid
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Zillur Rahman
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Nowshin Papri
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Raisa Rafique
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Aminul Islam
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Indrajeet Barman
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Fatema Tuz Jubyda
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Fatema‐Tuz Johura
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Marzia Sultana
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Kazi Istiaque Sanin
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - David A. Sack
- Department of International Health Johns Hopkins Bloomberg School of Public Health Baltimore MD USA
| | - Jamie Perin
- Department of International Health Johns Hopkins Bloomberg School of Public Health Baltimore MD USA
| | - Munirul Alam
- International Centre for Diarrhoeal Disease Research, Bangladesh Dhaka Bangladesh
| | - Christine George
- Department of International Health Johns Hopkins Bloomberg School of Public Health Baltimore MD USA
| |
Collapse
|
21
|
Parvin T, Minhaj Uddin I, Islam Bhuyian MS, Saxton R, Zohura F, Sultana M, Johura FT, Monira S, Hasan MT, Papri N, Haque MA, Biswas SK, Sack DA, Perin J, Alam M, George CM. Prospective cohort study of child mouthing of faeces and fomites in Dhaka, Bangladesh (CHoBI7 Program). Trop Med Int Health 2020; 25:976-984. [PMID: 32406964 DOI: 10.1111/tmi.13413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To characterise childhood mouthing and handling behaviours and to assess the association between hand-to-object and object-to-mouth contacts and diarrhoea prevalence in young children in urban Dhaka, Bangladesh. METHODS A prospective cohort study was conducted among 494 children under 5 years of age in Dhaka, Bangladesh. This study was nested within the randomised controlled trial of the Cholera Hospital-Based Intervention for 7 Days (CHoBI7) mobile health (mHealth) program. The CHoBI7 mHealth program focuses on promoting handwashing with soap and water treatment to diarrhoea patients and their household members through mobile messages and a single in person visit. Mouthing and handling of faeces and fomites among young children was measured by five-hour structured observation and caregiver reports. Diarrhoea surveillance data was collected monthly for 12 months. RESULTS Fifty five percent of caregivers reported that their child put a visibly dirty fomite (object or soil) in their mouth in the past week. Caregivers reported that 50% of children had mouthed visibly dirty objects, 26% had mouthed dirt, and 2% had mouthed faeces. Forty five percent of children were observed mouthing a visibly dirty fomite during structured observation, 40% of children were observed mouthing a visibly dirty object, 10% were observed mouthing soil, and one child (0.2%) was observed mouthing faeces. Mouthing of visibly dirty fomites was highest for children 12-18 months of age with 69% of these children having caregiver reports and 54% having observed events. Children with caregiver reports of mouthing faeces had a significantly higher odds of diarrhoea over the subsequent month (Odds Ratio: 4.54; 95% Confidence Interval: 1.06, 19.48). CONCLUSION These findings demonstrate that mouthing of contaminated fomites among young children is frequent in urban environments in Bangladesh, and that mouthing faeces is associated with a significantly higher odds of diarrhoea. Interventions are urgently needed to protect young children from faecal pathogens in their play spaces.
Collapse
Affiliation(s)
- Tahmina Parvin
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ismat Minhaj Uddin
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | | | - Ronald Saxton
- Department of International Health, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Fatema Zohura
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Marzia Sultana
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Fatema-Tuz Johura
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Shirajum Monira
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md Tasdik Hasan
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh.,University of Liverpool, Liverpool, UK
| | - Nowshin Papri
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md Ahshanul Haque
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Shwapon K Biswas
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh.,Rangpur Medical College and Hospital, Rangpur, Bangladesh
| | - David A Sack
- Department of International Health, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Jamie Perin
- Department of International Health, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Munirul Alam
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Christine Marie George
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| |
Collapse
|
22
|
Hinnouho GM, Wessells KR, Barffour MA, Sayasone S, Arnold CD, Kounnavong S, Hess SY. Impact of Different Strategies for Delivering Supplemental Zinc on Selected Fecal Markers of Environmental Enteric Dysfunction among Young Laotian Children: A Randomized Controlled Trial. Am J Trop Med Hyg 2020; 103:1416-1426. [PMID: 32618258 PMCID: PMC7543857 DOI: 10.4269/ajtmh.20-0106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The objective of this study was to assess the impact of different strategies for delivering supplemental zinc on fecal myeloperoxidase (MPO), neopterin (NEO), and calprotectin (CAL) among young Laotian children. In a double-blind controlled trial, children aged 6-23 months were randomized to receive either daily preventive zinc (PZ) tablets (7 mg/day), daily micronutrient powder (MNP; containing 10 mg zinc and 14 other micronutrients), therapeutic zinc (TZ) supplements for diarrhea treatment (20 mg/day for 10 days), or daily placebo powder and followed for ∼36 weeks. Stool samples were collected at baseline and endline. Fecal MPO, NEO, and CAL concentrations were determined in a randomly selected subsample of 720 children using commercially available ELISA kits. At baseline, the mean age was 14.1 ± 4.9 months and prevalence of stunting was 39%. The endline prevalence of stunting was 43%; there was no overall treatment effect on physical growth in the parent trial. At endline, the mean (95% CI) MPO in the PZ group was 1,590 [1,396; 1,811] ng/mL and did not differ from that in the MNP (1,633 [1,434; 1,859] ng/mL), TZ (1,749 [1,535; 1,992] ng/mL), and control (1,612 [1,415; 1,836] ng/mL) groups (P = 0.749). Similarly, there was no overall treatment effect on NEO and CAL concentrations (P = 0.226 and 0.229, respectively). In this population, the provision of PZ or TZ supplements or MNP had no impact on growth or environmental enteric dysfunction (EED) as assessed by fecal MPO, NEO, and CAL. Additional research is needed to better understand the etiology and proposed mechanisms of EED pathogenesis.
Collapse
Affiliation(s)
- Guy-Marino Hinnouho
- Helen Keller International, Washington, District of Columbia.,Department of Nutrition, Institute for Global Nutrition, University of California, Davis, Davis, California
| | - K Ryan Wessells
- Department of Nutrition, Institute for Global Nutrition, University of California, Davis, Davis, California
| | - Maxwell A Barffour
- Public Health Program, College of Health and Human Services, Missouri State University, Springfield, Missouri.,Department of Nutrition, Institute for Global Nutrition, University of California, Davis, Davis, California
| | - Somphou Sayasone
- Lao Tropical and Public Health Institute, Vientiane, Lao People's Democratic Republic
| | - Charles D Arnold
- Department of Nutrition, Institute for Global Nutrition, University of California, Davis, Davis, California
| | - Sengchanh Kounnavong
- Lao Tropical and Public Health Institute, Vientiane, Lao People's Democratic Republic
| | - Sonja Y Hess
- Department of Nutrition, Institute for Global Nutrition, University of California, Davis, Davis, California
| |
Collapse
|
23
|
Desai C, Handley SA, Rodgers R, Rodriguez C, Ordiz MI, Manary MJ, Holtz LR. Growth velocity in children with Environmental Enteric Dysfunction is associated with specific bacterial and viral taxa of the gastrointestinal tract in Malawian children. PLoS Negl Trop Dis 2020; 14:e0008387. [PMID: 32574158 PMCID: PMC7310680 DOI: 10.1371/journal.pntd.0008387] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 05/13/2020] [Indexed: 12/22/2022] Open
Abstract
Environmental enteric dysfunction (EED) is characterized by diffuse villous atrophy of the small bowel. EED is strongly associated with stunting, a major public health problem linked to increased childhood morbidity and mortality. EED and subsequent stunting of linear growth are surmised to have microbial origins. To interrogate this relationship, we defined the comprehensive virome (eukaryotic virus and bacteriophage) and bacterial microbiome of a longitudinal cohort of rural Malawian children with extensive metadata and intestinal permeability testing at each time point. We found thirty bacterial taxa differentially associated with linear growth. We detected many eukaryotic viruses. Neither the total number of eukaryotic families nor a specific viral family was statistically associated with improved linear growth. We identified 3 differentially abundant bacteriophage among growth velocities. Interestingly, there was a positive correlation between bacteria and bacteriophage richness in children with subsequent adequate/moderate growth which children with subsequent poor growth lacked. This suggests that a disruption in the equilibrium between bacteria and bacteriophage communities might be associated with subsequent poor growth. Future studies of EED and stunting should include the evaluation of viral communities in addition to bacterial microbiota to understand the complete microbial ecology of these poorly understood entities.
Collapse
Affiliation(s)
- Chandni Desai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Scott A. Handley
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Rachel Rodgers
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Cynthia Rodriguez
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Maria I. Ordiz
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Mark J. Manary
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Lori R. Holtz
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
24
|
Costa PN, Soares AM, Filho JQ, Junior FS, Ambikapathi R, Rogawski McQuade ET, Guerrant RL, Caulfield LE, Lima AAM, Maciel BLL. Dietary intake from complementary feeding is associated with intestinal barrier function and environmental enteropathy in Brazilian children from the MAL-ED cohort study. Br J Nutr 2020; 123:1003-1012. [PMID: 31964426 PMCID: PMC7282865 DOI: 10.1017/s0007114520000215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/26/2019] [Accepted: 01/07/2020] [Indexed: 12/19/2022]
Abstract
A child's diet contains nutrients and other substances that influence intestinal health. The present study aimed to evaluate the relations between complementary feeding, intestinal barrier function and environmental enteropathy (EE) in infants. Data from 233 children were obtained from the Brazilian site of the Etiology, Risk Factors, and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health and Development Project cohort study. Habitual dietary intake from complementary feeding was estimated using seven 24-h dietary recalls, from 9 to 15 months of age. Intestinal barrier function was assessed using the lactulose-mannitol test (L-M), and EE was determined as a composite measure using faecal biomarkers concentrations - α-1-antitrypsin, myeloperoxidase (MPO) and neopterin (NEO) at 15 months of age. The nutrient adequacies explored the associations between dietary intake and the intestinal biomarkers. Children showed adequate nutrient intakes (with the exception of fibre), impaired intestinal barrier function and intestinal inflammation. There was a negative correlation between energy adequacy and L-M (ρ = -0·19, P < 0·05) and between folate adequacy and NEO concentrations (ρ = -0·21, P < 0·01). In addition, there was a positive correlation between thiamine adequacy and MPO concentration (ρ = 0·22, P < 0·01) and between Ca adequacy and NEO concentration (ρ = 0·23; P < 0·01). Multiple linear regression models showed that energy intakes were inversely associated with intestinal barrier function (β = -0·19, P = 0·02), and fibre intake was inversely associated with the EE scores (β = -0·20, P = 0·04). Findings suggest that dietary intake from complementary feeding is associated with decreased intestinal barrier function and EE in children.
Collapse
Affiliation(s)
- P. N. Costa
- Nutrition Post-Graduation Program, Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - A. M. Soares
- Department of Physiology and Pharmacology, INCT – Instituto de Biomedicina do Semiárido Brasileiro (IBISAB), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - J. Q. Filho
- Department of Physiology and Pharmacology, INCT – Instituto de Biomedicina do Semiárido Brasileiro (IBISAB), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - F. S. Junior
- Department of Physiology and Pharmacology, INCT – Instituto de Biomedicina do Semiárido Brasileiro (IBISAB), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - R. Ambikapathi
- Department of Public Health, Purdue University, West Lafayette, IN, USA
| | | | - R. L. Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - L. E. Caulfield
- Center for Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - A. A. M. Lima
- Department of Public Health, Purdue University, West Lafayette, IN, USA
| | - B. L. L. Maciel
- Nutrition Post-Graduation Program, Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| |
Collapse
|
25
|
Perin J, Burrowes V, Almeida M, Ahmed S, Haque R, Parvin T, Biswas S, Azmi IJ, Bhuyian SI, Talukder KA, Faruque AG, Stine OC, George CM. A Retrospective Case-Control Study of the Relationship between the Gut Microbiota, Enteropathy, and Child Growth. Am J Trop Med Hyg 2020; 103:520-527. [PMID: 32431271 DOI: 10.4269/ajtmh.19-0761] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The microbial communities residing in the child gut are thought to play an important role in child growth, although the relationship is not well understood. We examined a cohort of young children from Mirzapur, Bangladesh, prospectively over 18 months. Four fecal markers of environmental enteropathy (EE) (high levels of alpha-1-antitrypsin, calprotectin, myeloperoxidase, and neopterin) were examined and anthropometric measures obtained from a cohort of 68 children. The 16S rRNA gene of bacterial DNA was sequenced from stool samples and used to estimate amplicon sequence variants (ASVs). We age-matched children with poor growth to children with normal growth within 1 month and compared the change in abundance and diversity of ASVs over time. Elevated EE markers and poor linear growth in children were associated with changes in microbial communities in the gut. There were increased amounts of Escherichia/Shigella and Proteobacteria and decreased amounts of Prevotella associated with poorly growing children consistent with the mounting evidence supporting the relationship between intestinal inflammation, child growth, and changes in gut microbiota composition. Future research is needed to investigate this association among young children in low- and middle-income countries.
Collapse
Affiliation(s)
- Jamie Perin
- Johns Hopkins School of Public Health, Baltimore, Maryland
| | | | | | - Shahnawaz Ahmed
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Rashidul Haque
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tahmina Parvin
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Shwapon Biswas
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ishrat J Azmi
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Sazzadul Islam Bhuyian
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Kaisar A Talukder
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Abu G Faruque
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | | | | |
Collapse
|
26
|
Devi S, Varkey A, Dharmar M, Holt RR, Allen LH, Sheshshayee MS, Preston T, Keen CL, Kurpad AV. Amino Acid Digestibility of Extruded Chickpea and Yellow Pea Protein is High and Comparable in Moderately Stunted South Indian Children with Use of a Dual Stable Isotope Tracer Method. J Nutr 2020; 150:1178-1185. [PMID: 32006007 PMCID: PMC7198288 DOI: 10.1093/jn/nxaa004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/19/2019] [Accepted: 01/07/2020] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Legumes are an excellent plant source of the limiting indispensable amino acid (IAA) lysine in vegetarian, cereal-based diets. However, their digestibility is poor largely because of their antiprotease content. Extrusion can enhance digestibility by inactivating trypsin inhibitors and thus potentially improve the protein quality of legumes. OBJECTIVE We measured the digestibility of extruded chickpea and yellow pea protein with use of a dual stable isotope method in moderately stunted South Indian primary school children. METHODS Twenty-eight moderately stunted children (height-for-age z scores <-2.0 SD and >-3.0 SD) aged 6-11 y from low to middle socioeconomic status were randomly assigned to receive a test protein (extruded intrinsically [2H]-labeled chickpea or yellow pea) along with a standard of U-[13C]-spirulina protein to measure amino acid (AA) digestibility with use of a dual stable isotope method. Individual AA digestibility in the test protein was calculated by the ratios of AA enrichments in the test protein to the standard protein in the food and their appearance in blood plasma collected at 6 and 6.5 h during the experiment, representing a plateau state. RESULTS The mean AA digestibility of extruded chickpea and yellow pea protein in moderately stunted children (HAZ; -2.86 to -1.2) was high and similar in both extruded test proteins (89.0% and 88.0%, respectively, P = 0.83). However, lysine and proline digestibilities were higher in extruded chickpea than yellow pea (79.2% compared with 76.5% and 75.0% compared with 72.0%, respectively, P < 0.02). CONCLUSION Extruded chickpea and yellow pea protein had good IAA digestibility in moderately stunted children, which was 20% higher than an earlier report of their digestibility when pressure-cooked, measured by the same method in adults. Higher digestibility of lysine and proline highlights better retention of these AA in chickpea during extrusion-based processing. Extrusion might be useful for developing high-quality protein foods from legumes. This trial was registered at www.ctri.nic.in as CTRI/2018/03/012439.
Collapse
Affiliation(s)
- Sarita Devi
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - Aneesia Varkey
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - Madan Dharmar
- Betty Irene Moore School of Nursing and the Department of Pediatrics, University of California, Davis School of Medicine, Sacramento, CA, USA
| | - Roberta R Holt
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Lindsay H Allen
- United States Department of Agriculture, Agricultural Research Service Western Human Nutrition Research Center, Davis, CA, USA
| | - M S Sheshshayee
- Department of Crop Physiology, University of Agricultural Sciences, Bangalore, India
| | - Thomas Preston
- Scottish Universities Environmental Research Centre, East Kilbride, UK
| | - Carl L Keen
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Anura V Kurpad
- Department of Physiology, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| |
Collapse
|
27
|
Zambruni M, Ochoa TJ, Somasunderam A, Cabada MM, Morales ML, Mitreva M, Rosa BA, Acosta GJ, Vigo NI, Riveros M, Arango S, Durand D, Berends MN, Melby P, Utay NS. Stunting Is Preceded by Intestinal Mucosal Damage and Microbiome Changes and Is Associated with Systemic Inflammation in a Cohort of Peruvian Infants. Am J Trop Med Hyg 2020; 101:1009-1017. [PMID: 31482782 DOI: 10.4269/ajtmh.18-0975] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Stunting, defined as height-for-age Z score equal to or lower than -2, is associated with increased childhood mortality, cognitive impairment, and chronic diseases. The aim of the study was to investigate the relationship between linear growth, intestinal damage, and systemic inflammation in infants at risk of stunting. We followed up 78 infants aged 5-12 months living in rural areas of Peru for 6 months. Blood samples for biomarkers of intestinal damage (intestinal fatty-acid-binding protein [I-FABP] and zonulin) and systemic inflammation (interleukin-1β, interleukin-6, tumor necrosis factor α [TNF-α], soluble CD14, and lipopolysaccharide-binding protein [LBP]) and fecal samples for microbiome analysis were collected at baseline and closure of the study. The children's growth and health status were monitored through biweekly home visits by trained staff. Twenty-one percent of the children became stunted: compared with non-stunted children, they had worse nutritional parameters and higher levels of serum I-FABP at baseline. The likelihood of becoming stunted was strongly associated with an increase in sCD14 over time; LBP and TNF-α showed a trend toward increase in stunted children but not in controls. The fecal microbiota composition of stunted children had an increased beta diversity compared with that of healthy controls throughout the study. The relative abundance of Ruminococcus 1 and 2, Clostridium sensu stricto, and Collinsella increased in children becoming stunted but not in controls, whereas Providencia abundance decreased. In conclusion, stunting in our population was preceded by an increase in markers of enterocyte turnover and differences in the fecal microbiota and was associated with increasing levels of systemic inflammation markers.
Collapse
Affiliation(s)
- Mara Zambruni
- Department of Pediatrics, The University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - Theresa J Ochoa
- Instituto de Medicina Tropical "Alexander von Humboldt," Universidad Peruana Cayetano Heredia, Lima, Peru.,Department of Pediatrics, The University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - Anoma Somasunderam
- Division of Infectious Diseases, The University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - Miguel M Cabada
- Infectious Diseases Division, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas.,Universidad Peruana Cayetano Heredia-University of Texas Medical Branch Collaborative Research Center Cusco, Universidad Peruana Cayetano Heredia, Cusco, Peru
| | - Maria L Morales
- Universidad Peruana Cayetano Heredia-University of Texas Medical Branch Collaborative Research Center Cusco, Universidad Peruana Cayetano Heredia, Cusco, Peru
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.,The McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Bruce A Rosa
- The McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Gonzalo J Acosta
- Instituto de Medicina Tropical "Alexander von Humboldt," Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Natalia I Vigo
- Instituto de Medicina Tropical "Alexander von Humboldt," Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Maribel Riveros
- Instituto de Medicina Tropical "Alexander von Humboldt," Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Sara Arango
- Instituto de Medicina Tropical "Alexander von Humboldt," Universidad Peruana Cayetano Heredia, Lima, Peru
| | - David Durand
- Instituto de Medicina Tropical "Alexander von Humboldt," Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Maitreyee N Berends
- Infectious Diseases Division, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Peter Melby
- Infectious Diseases Division, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Netanya S Utay
- Division of Infectious Diseases, The University of Texas Health Science Center at Houston Medical School, Houston, Texas
| |
Collapse
|
28
|
Lauer JM, Duggan CP, Ausman LM, Griffiths JK, Webb P, Bashaasha B, Agaba E, Turyashemererwa FM, Ghosh S. Unsafe Drinking Water Is Associated with Environmental Enteric Dysfunction and Poor Growth Outcomes in Young Children in Rural Southwestern Uganda. Am J Trop Med Hyg 2019; 99:1606-1612. [PMID: 30350765 PMCID: PMC6283503 DOI: 10.4269/ajtmh.18-0143] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Environmental enteric dysfunction (EED), a subclinical disorder of the small intestine, and poor growth are associated with living in poor water, sanitation, and hygiene (WASH) conditions, but specific risk factors remain unclear. Nested within a birth cohort study, this study investigates relationships among water quality, EED, and growth in 385 children living in southwestern Uganda. Water quality was assessed using a portable water quality test when children were 6 months, and safe water was defined as lacking Escherichia coli contamination. Environmental enteric dysfunction was assessed using the lactulose:mannitol (L:M) test at 12-16 months. Anthropometry and covariate data were extracted from the cohort study, and associations were assessed using linear and logistic regression models. Less than half of the households (43.8%) had safe water, and safe versus unsafe water did not correlate with improved versus unimproved water source. In adjusted linear regression models, children from households with safe water had significantly lower log-transformed (ln) L:M ratios (β: -0.22, 95% confidence interval (CI): -0.44, -0.00) and significantly higher length-for-age (β: 0.29, 95% CI: 0.00, 0.58) and weight-for-age (β: 0.20, 95% CI: 0.05, 0.34) Z-scores at 12-16 months. Furthermore, in adjusted linear regression models, ln L:M ratios at 12-16 months significantly decreased with increasing length-for-age Z-scores at birth, 6 months, and 9 months (β: -0.05, 95% CI: -0.10, -0.004; β: -0.06, 95% CI: -0.11, -0.006; and β: -0.05, 95% CI: -0.09, -0.005, respectively). Overall, our data suggest that programs seeking to improve nutrition should address poor WASH conditions simultaneously, particularly related to household drinking water quality.
Collapse
Affiliation(s)
- Jacqueline M Lauer
- United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts.,Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts.,Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| | - Christopher P Duggan
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts.,United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts
| | - Lynne M Ausman
- United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts.,Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| | - Jeffrey K Griffiths
- Tufts University Cummings School of Veterinary Medicine, Tufts University School of Engineering, Medford, Massachusetts.,Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts
| | - Patrick Webb
- United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts.,Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| | - Bernard Bashaasha
- Department of Agribusiness and Natural Resource Economics, Makerere University, Kampala, Uganda
| | - Edgar Agaba
- United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts
| | | | - Shibani Ghosh
- United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts.,Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| |
Collapse
|
29
|
Kastl AJ, Terry NA, Wu GD, Albenberg LG. The Structure and Function of the Human Small Intestinal Microbiota: Current Understanding and Future Directions. Cell Mol Gastroenterol Hepatol 2019; 9:33-45. [PMID: 31344510 PMCID: PMC6881639 DOI: 10.1016/j.jcmgh.2019.07.006] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/17/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023]
Abstract
Despite growing literature characterizing the fecal microbiome and its association with health and disease, few studies have analyzed the microbiome of the small intestine. Here, we examine what is known about the human small intestinal microbiota in terms of community structure and functional properties. We examine temporal dynamics of select bacterial populations in the small intestine, and the effects of dietary carbohydrates and fats on shaping these populations. We then evaluate dysbiosis in the small intestine in several human disease models, including small intestinal bacterial overgrowth, short-bowel syndrome, pouchitis, environmental enteric dysfunction, and irritable bowel syndrome. What is clear is that the bacterial biology, and mechanisms of bacteria-induced pathophysiology, are enormously broad and elegant in the small intestine. Studying the small intestinal microbiota is challenged by rapidly fluctuating environmental conditions in these intestinal segments, as well as the complexity of sample collection and bioinformatic analysis. Because the functionality of the digestive tract is determined primarily by the small intestine, efforts must be made to better characterize this unique and important microbial ecosystem.
Collapse
Affiliation(s)
- Arthur J. Kastl
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Correspondence Address correspondence to: Arthur J. Kastl Jr, MD, Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, 7NW, Philadelphia, Pennsylvania 19104. fax: (215) 590-3606.
| | - Natalie A. Terry
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Gary D Wu
- Division of Gastroenterology, Hepatology, and Nutrition, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lindsey G. Albenberg
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
30
|
Iqbal NT, Syed S, Sadiq K, Khan MN, Iqbal J, Ma JZ, Umrani F, Ahmed S, Maier EA, Denson LA, Haberman Y, McNeal MM, Setchell KDR, Zhao X, Qureshi S, Shen L, Moskaluk CA, Liu TC, Yilmaz O, Brown DE, Barratt MJ, Kung VL, Gordon JI, Moore SR, Ali SA. Study of Environmental Enteropathy and Malnutrition (SEEM) in Pakistan: protocols for biopsy based biomarker discovery and validation. BMC Pediatr 2019; 19:247. [PMID: 31331393 PMCID: PMC6643315 DOI: 10.1186/s12887-019-1564-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 05/31/2019] [Indexed: 12/26/2022] Open
Abstract
Background Environmental Enteropathy (EE), characterized by alterations in intestinal structure, function, and immune activation, is believed to be an important contributor to childhood undernutrition and its associated morbidities, including stunting. Half of all global deaths in children < 5 years are attributable to under-nutrition, making the study of EE an area of critical priority. Methods Community based intervention study, divided into two sub-studies, 1) Longitudinal analyses and 2) Biopsy studies for identification of EE features via omics analyses. Birth cohorts in Matiari, Pakistan established: moderately or severely malnourished (weight for height Z score (WHZ) < − 2) children, and well-nourished (WHZ > 0) children. Blood, urine, and fecal samples, for evaluation of potential biomarkers, will be collected at various time points from all participants (longitudinal analyses). Participants will receive appropriate educational and nutritional interventions; non-responders will undergo further evaluation to determine eligibility for further workup, including upper gastrointestinal endoscopy. Histopathological changes in duodenal biopsies will be compared with duodenal biopsies obtained from USA controls who have celiac disease, Crohn’s disease, or who were found to have normal histopathology. RNA-Seq will be employed to characterize mucosal gene expression across groups. Duodenal biopsies, luminal aspirates from the duodenum, and fecal samples will be analyzed to define microbial community composition (omic analyses). The relationship between histopathology, mucosal gene expression, and community configuration will be assessed using a variety of bioinformatic tools to gain better understanding of disease pathogenesis and to identify mechanism-based biomarkers. Ethical review committees at all collaborating institutions have approved this study. All results will be made available to the scientific community. Discussion Operational and ethical constraints for safely obtaining intestinal biopsies from children in resource-poor settings have led to a paucity of human tissue-based investigations to understand and reverse EE in vulnerable populations. Furthermore, EE biomarkers have rarely been correlated with gold standard histopathological confirmation. The Study of Environmental Enteropathy and Malnutrition (SEEM) is designed to better understand the pathophysiology, predictors, biomarkers, and potential management strategies of EE to inform strategies to eradicate this debilitating pathology and accelerate progress towards the 2030 Sustainable Development Goals. Trial registration Retrospectively registered; clinicaltrials.gov ID NCT03588013. Electronic supplementary material The online version of this article (10.1186/s12887-019-1564-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Najeeha T Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan.,Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Sana Syed
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan.,Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | - Kamran Sadiq
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Marium N Khan
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | - Junaid Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan.,Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Jennie Z Ma
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Fayaz Umrani
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sheraz Ahmed
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Elizabeth A Maier
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lee A Denson
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yael Haberman
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Monica M McNeal
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kenneth D R Setchell
- Clinical Mass Spectrometry, Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xueheng Zhao
- Clinical Mass Spectrometry, Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Shahida Qureshi
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Lanlan Shen
- Department of Pediatrics, Baylor College of Medicine, USDA/ARS Children's Nutrition Research Center, Houston, TX, USA
| | | | - Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Omer Yilmaz
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Koch Institute for Integrative Cancer Research at MIT and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Donald E Brown
- Data Science Institute, University of Virginia, Charlottesville, VA, USA
| | - Michael J Barratt
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vanderlene L Kung
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeffrey I Gordon
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sean R Moore
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA.
| | - S Asad Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan.
| |
Collapse
|
31
|
Lauer JM, McDonald CM, Kisenge R, Aboud S, Fawzi WW, Liu E, Tran HQ, Gewirtz AT, Manji KP, Duggan CP. Markers of Systemic Inflammation and Environmental Enteric Dysfunction Are Not Reduced by Zinc or Multivitamins in Tanzanian Infants: A Randomized, Placebo-Controlled Trial. J Pediatr 2019; 210:34-40.e1. [PMID: 30952509 PMCID: PMC6590867 DOI: 10.1016/j.jpeds.2019.02.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/30/2019] [Accepted: 02/13/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To examine whether daily zinc and/or multivitamin supplementation reduce biomarkers of environmental enteric dysfunction (EED), systemic inflammation, or markers of growth in a sample of infants from Dar es Salaam, Tanzania. STUDY DESIGN Subgroup analysis of infants participating in a randomized, double-blind, placebo-controlled trial received daily oral supplementation of zinc, multivitamins, zinc + multivitamins, or placebo for 18 months starting at 6 weeks of age. EED (anti-flagellin and anti-lipopolysaccharide immunoglobulins), systemic inflammation (C-reactive protein and alpha-1-acid glycoprotein), and growth biomarkers (insulin-like growth factor-1 and insulin-like growth factor binding protein-3) were measured via enzyme-linked immunosorbent assay in a subsample of 590 infants at 6 weeks and 6 months of age. EED biomarkers also were measured in 162 infants at 12 months of age. RESULTS With the exception of anti-lipopolysaccharide IgG concentrations, which were significantly greater in infants who received multivitamins compared with those who did not (1.41 ± 0.61 vs 1.26 ± 0.65, P = .006), and insulin-like growth factor binding protein-3 concentrations, which were significantly lower in children who received zinc compared with those who did not (981.13 ± 297.59 vs 1019.10 ± 333.01, P = .03), at 6 months of age, we did not observe any significant treatment effects of zinc or multivitamins on EED, systemic inflammation, or growth biomarkers. CONCLUSIONS Neither zinc nor multivitamin supplementation ameliorated markers of EED or systemic inflammation during infancy. Other interventions should be prioritized for future trials. TRIAL REGISTRATION Clinicaltrials.gov: NCT00421668.
Collapse
Affiliation(s)
- Jacqueline M Lauer
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA.
| | | | - Rodrick Kisenge
- Department of Pediatrics and Child Health, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Said Aboud
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Wafaie W Fawzi
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Enju Liu
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA; Institutional Centers of Clinical and Translational Research, Boston Children's Hospital, Boston, MA
| | - Hao Q Tran
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA
| | - Andrew T Gewirtz
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA
| | - 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, MA; Department of Pediatrics and Child Health, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
32
|
Khalil IA, Troeger C, Rao PC, Blacker BF, Brown A, Brewer TG, Colombara DV, De Hostos EL, Engmann C, Guerrant RL, Haque R, Houpt ER, Kang G, Korpe PS, Kotloff KL, Lima AAM, Petri WA, Platts-Mills JA, Shoultz DA, Forouzanfar MH, Hay SI, Reiner RC, Mokdad AH. Morbidity, mortality, and long-term consequences associated with diarrhoea from Cryptosporidium infection in children younger than 5 years: a meta-analyses study. LANCET GLOBAL HEALTH 2019; 6:e758-e768. [PMID: 29903377 PMCID: PMC6005120 DOI: 10.1016/s2214-109x(18)30283-3] [Citation(s) in RCA: 280] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 03/08/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022]
Abstract
Background The protozoan Cryptosporidium is a leading cause of diarrhoea morbidity and mortality in children younger than 5 years. However, the true global burden of Cryptosporidium infection in children younger than 5 years might have been underestimated in previous quantifications because it only took account of the acute effects of diarrhoea. We aimed to demonstrate whether there is a causal relation between Cryptosporidium and childhood growth and, if so, to quantify the associated additional burden. Methods The Global Burden of Diseases, Injuries, and Risk Factors study (GBD) 2016 was a systematic and scientific effort to quantify the morbidity and mortality associated with more than 300 causes of death and disability, including diarrhoea caused by Cryptosporidium infection. We supplemented estimates on the burden of Cryptosporidium in GBD 2016 with findings from a systematic review of published and unpublished cohort studies and a meta-analysis of the effect of childhood diarrhoea caused by Cryptosporidium infection on physical growth. Findings In 2016, Cryptosporidium infection was the fifth leading diarrhoeal aetiology in children younger than 5 years, and acute infection caused more than 48 000 deaths (95% uncertainty interval [UI] 24 600–81 900) and more than 4·2 million disability-adjusted life-years lost (95% UI 2·2 million–7·2 million). We identified seven data sources from the scientific literature and six individual-level data sources describing the relation between Cryptosporidium and childhood growth. Each episode of diarrhoea caused by Cryptosporidium infection was associated with a decrease in height-for-age Z score (0·049, 95% CI 0·014–0·080), weight-for-age Z score (0·095, 0·055–0·134), and weight-for-height Z score (0·126, 0·057–0·194). We estimated that diarrhoea from Cryptosporidium infection caused an additional 7·85 million disability-adjusted life-years (95% UI 5·42 million–10·11 million) after we accounted for its effect on growth faltering—153% more than that estimated from acute effects alone. Interpretation Our findings show that the substantial short-term burden of diarrhoea from Cryptosporidium infection on childhood growth and wellbeing is an underestimate of the true burden. Interventions designed to prevent and effectively treat infection in children younger than 5 years will have enormous public health and social development impacts. Funding The Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Ibrahim A Khalil
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Christopher Troeger
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Puja C Rao
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Brigette F Blacker
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Alexandria Brown
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Thomas G Brewer
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Danny V Colombara
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | | | - Cyril Engmann
- PATH, Seattle, WA, USA; School of Public Health, University of Washington, Seattle, WA, USA
| | - Richard L Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | | | - Eric R Houpt
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Gagandeep Kang
- Translational Health Science and Technology Institute, Faridabad, India
| | - Poonum S Korpe
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Karen L Kotloff
- Departments of Pediatrics and Medicine, Center for Vaccine Development, Institute for Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aldo A M Lima
- Center for Global Health, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - William A Petri
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - James A Platts-Mills
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | | | | | - Simon I Hay
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA; Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Robert C Reiner
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Ali H Mokdad
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA.
| |
Collapse
|
33
|
Supplementation With Lactoferrin and Lysozyme Ameliorates Environmental Enteric Dysfunction: A Double-Blind, Randomized, Placebo-Controlled Trial. Am J Gastroenterol 2019; 114:671-678. [PMID: 30829679 DOI: 10.14309/ajg.0000000000000170] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Environmental enteric dysfunction (EED) predisposes children throughout the developing world to high rates of systemic exposure to enteric pathogens and stunting. Effective interventions that treat or prevent EED may help children achieve their full physical and cognitive potential. The objective of this study is to test whether 2 components of breast milk would improve a biomarker of EED and linear growth during the second year of life. METHODS A prospective, randomized, double-blind, placebo-controlled clinical trial among children aged 12-23 months was conducted in rural Malawi. The experimental group received a daily supplement of 1.5 g of lactoferrin and 0.2 g of lysozyme for 16 weeks. The primary outcome was an improvement in EED, as measured by the change in the percentage of ingested lactulose excreted into the urine (Δ%L). RESULTS Among 214 children who completed the study, there was a significant difference in Δ%L between the control and experimental groups over 8 weeks (an increase of 0.23% vs 0.14%, respectively; P = 0.04). However, this relative improvement was not as strongly sustained over the full 16 weeks of the study (an increase of 0.16% vs 0.11%, respectively; P = 0.17). No difference in linear growth over this short period was observed. The experimental intervention group had significantly lower rates of hospitalization and the development of acute malnutrition during the course of the study (2.5% vs 10.3%, relative risk 0.25; P < 0.02). DISCUSSION Supplementation with lactoferrin and lysozyme in a population of agrarian children during the second year of life has a beneficial effect on gut health. This intervention also protected against hospitalization and the development of acute malnutrition, a finding with a significant clinical and public health importance. This finding should be pursued in larger studies with longer follow-up and optimized dosing.
Collapse
|
34
|
Bartelt LA, Bolick DT, Guerrant RL. Disentangling Microbial Mediators of Malnutrition: Modeling Environmental Enteric Dysfunction. Cell Mol Gastroenterol Hepatol 2019; 7:692-707. [PMID: 30630118 PMCID: PMC6477186 DOI: 10.1016/j.jcmgh.2018.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022]
Abstract
Environmental enteric dysfunction (EED) (also referred to as environmental enteropathy) is a subclinical chronic intestinal disorder that is an emerging contributor to early childhood malnutrition. EED is common in resource-limited settings, and is postulated to consist of small intestinal injury, dysfunctional nutrient absorption, and chronic inflammation that results in impaired early child growth attainment. Although there is emerging interest in the hypothetical potential for chemical toxins in the environmental exposome to contribute to EED, the propensity of published data, and hence the focus of this review, implicates a critical role of environmental microbes. Early childhood malnutrition and EED are most prevalent in resource-limited settings where food is limited, and inadequate access to clean water and sanitation results in frequent gastrointestinal pathogen exposures. Even as overt diarrhea rates in these settings decline, silent enteric infections and faltering growth persist. Furthermore, beyond restricted physical growth, EED and/or enteric pathogens also associate with impaired oral vaccine responses, impaired cognitive development, and may even accelerate metabolic syndrome and its cardiovascular consequences. As these potentially costly long-term consequences of early childhood enteric infections increasingly are appreciated, novel therapeutic strategies that reverse damage resulting from nutritional deficiencies and microbial insults in the developing small intestine are needed. Given the inherent limitations in investigating how specific intestinal pathogens directly injure the small intestine in children, animal models provide an affordable and controlled opportunity to elucidate causal sequelae of specific enteric infections, to differentiate consequences of defined nutrient deprivation alone from co-incident enteropathogen insults, and to correlate the resulting gut pathologies with their functional impact during vulnerable early life windows.
Collapse
Affiliation(s)
- Luther A Bartelt
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Gastrointestinal Biology and Disease, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - David T Bolick
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Richard L Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
35
|
Stunted childhood growth is associated with decompartmentalization of the gastrointestinal tract and overgrowth of oropharyngeal taxa. Proc Natl Acad Sci U S A 2018; 115:E8489-E8498. [PMID: 30126990 DOI: 10.1073/pnas.1806573115] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Linear growth delay (stunting) affects roughly 155 million children under the age of 5 years worldwide. Treatment has been limited by a lack of understanding of the underlying pathophysiological mechanisms. Stunting is most likely associated with changes in the microbial community of the small intestine, a compartment vital for digestion and nutrient absorption. Efforts to better understand the pathophysiology have been hampered by difficulty of access to small intestinal fluids. Here, we describe the microbial community found in the upper gastrointestinal tract of stunted children aged 2-5 y living in sub-Saharan Africa. We studied 46 duodenal and 57 gastric samples from stunted children, as well as 404 fecal samples from stunted and nonstunted children living in Bangui, Central African Republic, and in Antananarivo, Madagascar, using 16S Illumina Amplicon sequencing and semiquantitative culture methods. The vast majority of the stunted children showed small intestinal bacterial overgrowth dominated by bacteria that normally reside in the oropharyngeal cavity. There was an overrepresentation of oral bacteria in fecal samples of stunted children, opening the way for developing noninvasive diagnostic markers. In addition, Escherichia coli/Shigella sp. and Campylobacter sp. were found to be more prevalent in stunted children, while Clostridia, well-known butyrate producers, were reduced. Our data suggest that stunting is associated with a microbiome "decompartmentalization" of the gastrointestinal tract characterized by an increased presence of oropharyngeal bacteria from the stomach to the colon, hence challenging the current view of stunting arising solely as a consequence of small intestine overstimulation through recurrent infections by enteric pathogens.
Collapse
|
36
|
Ordiz MI, Wold K, Kaimila Y, Divala O, Gilstrap M, Lu HZ, Manary MJ. Detection and interpretation of fecal host mRNA in rural Malawian infants aged 6-12 months at risk for environmental enteric dysfunction. Exp Biol Med (Maywood) 2018; 243:985-989. [PMID: 30099958 DOI: 10.1177/1535370218794418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Recent studies have suggested that environmental enteric dysfunction can be assessed in rural African children by measuring levels of fecal mRNA transcripts. The field collection of fecal samples is less invasive and cumbersome than administration of the lactulose:mannitol test, which is typically used to assess environmental enteric dysfunction. This study sought to determine if, as in children aged 12-60 months, an array of seven fecal host transcripts (CD53, CDX1, HLA-DRA, TNF, S100A8, MUC12, and REG1A) could predict environmental enteric dysfunction in rural African infants. Host fecal transcript abundance was correlated to the percentage of lactulose (%L) excreted in the urine for 340 samples from Malawian children aged 6-12 months. Permeability was categorized as not severe (%L < 0.45) and severe (%L ≥ 0.45). This study found the prevalence of severe environmental enteric dysfunction to be 114/834 (14%), lower than what was previously reported for 12-60 months old children, 595/1521 (39%, P = 0.001). In linear regression analysis with the seven host transcripts, two were associated with %L: β coefficients of -1.843 ( P = 0.035) and 0.215 ( P = 0.006) for CDX1 and REG1A, respectively. The seven fecal host transcripts in a random forest model did not predict severe environmental enteric dysfunction. Future models utilizing different transcripts identified from an untargeted, agnostic assessment of all potential host transcripts could provide accurate predictions of environmental enteric dysfunction in infants. Impact statement Environmental enteric dysfunction (EED) is associated with reduced linear growth. The dual sugar absorption test has been used as a non-invasive method to determine the gut health of individuals. Alternative methods using fecal host mRNAs as predictors of the gut health are promising. In older children, we have determined that seven transcripts can predict the gut health in a random forest model. Our current study determined that the host fecal mRNA is abundant in infants and toddlers alike. Severe EED in rural Malawian children is less prevalent in infants than in young children. REG1A and CDX1 are associated with gut health. Fecal host mRNA may well be a means to assess gut health in African infants, but the panel of transcripts used to do this will differ from that in older children.
Collapse
Affiliation(s)
- M Isabel Ordiz
- 1 Department of Pediatrics, Washington University at Saint Louis, St. Louis, MO 63110, USA
| | - Karl Wold
- 1 Department of Pediatrics, Washington University at Saint Louis, St. Louis, MO 63110, USA
| | - Yankho Kaimila
- 2 School of Public Health and Family Medicine, University of Malawi, Blantyre 3, Malawi
| | - Oscar Divala
- 2 School of Public Health and Family Medicine, University of Malawi, Blantyre 3, Malawi
| | - Madeline Gilstrap
- 1 Department of Pediatrics, Washington University at Saint Louis, St. Louis, MO 63110, USA
| | - Henry Z Lu
- 1 Department of Pediatrics, Washington University at Saint Louis, St. Louis, MO 63110, USA
| | - Mark J Manary
- 1 Department of Pediatrics, Washington University at Saint Louis, St. Louis, MO 63110, USA.,2 School of Public Health and Family Medicine, University of Malawi, Blantyre 3, Malawi.,3 Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
37
|
Vonaesch P, Randremanana R, Gody JC, Collard JM, Giles-Vernick T, Doria M, Vigan-Womas I, Rubbo PA, Etienne A, Andriatahirintsoa EJ, Kapel N, Brown E, Huus KE, Duffy D, Finlay B, Hasan M, Hunald FA, Robinson A, Manirakiza A, Wegener-Parfrey L, Vray M, Sansonetti PJ. Identifying the etiology and pathophysiology underlying stunting and environmental enteropathy: study protocol of the AFRIBIOTA project. BMC Pediatr 2018; 18:236. [PMID: 30025542 PMCID: PMC6053792 DOI: 10.1186/s12887-018-1189-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 06/21/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Globally one out of four children under 5 years is affected by linear growth delay (stunting). This syndrome has severe long-term sequelae including increased risk of illness and mortality and delayed psychomotor development. Stunting is a syndrome that is linked to poor nutrition and repeated infections. To date, the treatment of stunted children is challenging as the underlying etiology and pathophysiological mechanisms remain elusive. We hypothesize that pediatric environmental enteropathy (PEE), a chronic inflammation of the small intestine, plays a major role in the pathophysiology of stunting, failure of nutritional interventions and diminished response to oral vaccines, potentially via changes in the composition of the pro- and eukaryotic intestinal communities. The main objective of AFRIBIOTA is to describe the intestinal dysbiosis observed in the context of stunting and to link it to PEE. Secondary objectives include the identification of the broader socio-economic environment and biological and environmental risk factors for stunting and PEE as well as the testing of a set of easy-to-use candidate biomarkers for PEE. We also assess host outcomes including mucosal and systemic immunity and psychomotor development. This article describes the rationale and study protocol of the AFRIBIOTA project. METHODS AFRIBIOTA is a case-control study for stunting recruiting children in Bangui, Central African Republic and in Antananarivo, Madagascar. In each country, 460 children aged 2-5 years with no overt signs of gastrointestinal disease are recruited (260 with no growth delay, 100 moderately stunted and 100 severely stunted). We compare the intestinal microbiota composition (gastric and small intestinal aspirates; feces), the mucosal and systemic immune status and the psychomotor development of children with stunting and/or PEE compared to non-stunted controls. We also perform anthropological and epidemiological investigations of the children's broader living conditions and assess risk factors using a standardized questionnaire. DISCUSSION To date, the pathophysiology and risk factors of stunting and PEE have been insufficiently investigated. AFRIBIOTA will add new insights into the pathophysiology underlying stunting and PEE and in doing so will enable implementation of new biomarkers and design of evidence-based treatment strategies for these two syndromes.
Collapse
Affiliation(s)
- Pascale Vonaesch
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Rindra Randremanana
- Unité d’Epidémiologie et de Recherche Clinique, Institut Pasteur de Madagascar, BP 1274 Ambatofotsikely, Avaradoha, 101 Antananarivo, Madagascar
| | - Jean-Chrysostome Gody
- Centre Pédiatrique de Bangui, Avenue de l’Indépendance, Bangui, Central African Republic
| | - Jean-Marc Collard
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, BP 1274 Ambatofotsikely, Avaradoha, 101 Antananarivo, Madagascar
| | - Tamara Giles-Vernick
- Unité d’Epidémiologie des Maladies Emergentes, Institut Pasteur, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Maria Doria
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Inès Vigan-Womas
- Unité d’Immunologie des Maladies Infectieuses, Institut Pasteur de Madagascar, BP 1274 Ambatofotsikely, Avaradoha, 101 Antananarivo, Madagascar
| | - Pierre-Alain Rubbo
- Laboratoire d’Analyses Médicales, Institut Pasteur de Bangui, Avenue de l’Indépendance, Bangui, Central African Republic
| | - Aurélie Etienne
- Unité d’Epidémiologie et de Recherche Clinique, Institut Pasteur de Madagascar, BP 1274 Ambatofotsikely, Avaradoha, 101 Antananarivo, Madagascar
| | - Emilson Jean Andriatahirintsoa
- Centre Hospitalier Universitaire Mère-Enfant de Tsaralalàna (CHUMET), rue Patrice Lumumba, Tsaralalàna, 101 Antananarivo, Madagascar
| | - Nathalie Kapel
- Laboratoire de Coprologie Fonctionnelle, Hôpital Pitié-Salpêtrière, 47-83 Bd de l’Hôpital, 75013 Paris, France
| | - Eric Brown
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, V6T1Z4 Canada
| | - Kelsey E. Huus
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, V6T1Z4 Canada
| | - Darragh Duffy
- Unité de la Biologie des Cellules Dendritiques, Institut Pasteur, 25 Rue du Dr. Roux, 75015 Paris, France
| | - B.Brett Finlay
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, V6T1Z4 Canada
| | - Milena Hasan
- Centre de Recherche Translationnelle, Institut Pasteur, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Francis Allen Hunald
- Centre Hospitalier Universitaire Joseph Ravoahangy Andrianavalona (CHUJRA), Antananarivo, Madagascar
| | - Annick Robinson
- Centre Hospitalier Universitaire Mère Enfant de Tsaralalana, Antananarivo, Madagascar
| | - Alexandre Manirakiza
- Unité d’Epidémiologie, Institut Pasteur de Bangui, Avenue de l’Indépendance, Bangui, Central African Republic
| | - Laura Wegener-Parfrey
- Departments of Botany and Zoology, and Biodiversity Research Centre, University of British Columbia, 3200-6270 University Boulevard, Vancouver, V6T1Z4 Canada
| | - Muriel Vray
- Unité d’Epidémiologie des Maladies Emergentes, Institut Pasteur, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Philippe J. Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 28 Rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
38
|
Reid B, Seu R, Orgle J, Roy K, Pongolani C, Chileshe M, Fundira D, Stoltzfus R. A Community-Designed Play-Yard Intervention to Prevent Microbial Ingestion: A Baby Water, Sanitation, and Hygiene Pilot Study in Rural Zambia. Am J Trop Med Hyg 2018; 99:513-525. [PMID: 29869596 DOI: 10.4269/ajtmh.17-0780] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Malnourished children in low-income contexts usually suffer from environmental enteric dysfunction, which is damage to the intestines caused by chronic exposure to bacterial pathogens from feces hypothesized to contribute to stunting. Many intervention studies are piloting "Baby water, sanitation, and hygiene (WASH)" to help rural farming families reduce infant and young children's (IYC's) exposure to human and free-range livestock feces. One proposed Baby WASH intervention is a play-yard, which consists of a baby-proofed structure (i.e., playpen) that caregivers can place IYC into while doing chores around the household yard. This article describes the pilot development and assessment of a community-built Baby WASH play-yard and a plastic play-yard intervention with 21 caregivers of 6- to 24-month-old IYC in rural Zambia. A modified Trials of Improved Practices approach was used to conduct three visits in each household: an introductory visit during which play-yard use was explained, a second visit consisting of a semi-structured interview and a session of behavioral counseling, and a final visit which included a 2-hour observation of play-yard use. The second and final visits also included 24-hour recalls, and all three visits included spot observations of play-yard use. Reports from caregivers suggest that the community-built play-yard protected IYC from ingesting soil and livestock feces. Barriers to intervention use included caregivers' WASH beliefs and practices, community reactions, and play-yard maintenance. More work is needed to examine the role of women's time use in their home environment, community reactions to the intervention, and the biological efficacy to reduce microbial ingestion.
Collapse
Affiliation(s)
- Brie Reid
- Institute of Child Development, University of Minnesota, Minneapolis, Minnesota
| | - Rie Seu
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Jennifer Orgle
- Cooperative for Assistance and Relief Everywhere (CARE) USA, Atlanta, Georgia
| | - Khrist Roy
- Cooperative for Assistance and Relief Everywhere (CARE) USA, Atlanta, Georgia
| | - Catherine Pongolani
- Cooperative for Assistance and Relief Everywhere (CARE) International Zambia, Lusaka, Zambia
| | | | - Dadirai Fundira
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Rebecca Stoltzfus
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| |
Collapse
|
39
|
Jones AD, Colecraft EK, Awuah RB, Boatemaa S, Lambrecht NJ, Adjorlolo LK, Wilson ML. Livestock ownership is associated with higher odds of anaemia among preschool-aged children, but not women of reproductive age in Ghana. MATERNAL AND CHILD NUTRITION 2018; 14:e12604. [PMID: 29608248 PMCID: PMC6055803 DOI: 10.1111/mcn.12604] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/21/2017] [Accepted: 02/07/2018] [Indexed: 12/21/2022]
Abstract
Livestock ownership may influence anaemia through complex and possibly contradictory mechanisms. In this study, we aimed to determine the association of household livestock ownership with anaemia among women aged 15–49 years and children aged 6–59 months in Ghana and to examine the contribution of animal source foods (ASFs) to consumption patterns as a potential mechanism mediating this association. We analysed data on 4,441 women and 2,735 children from the 2014 Ghana Demographic and Health Survey and 16,772 households from the Ghana Living Standards Survey Round 6. Haemoglobin measurements were used to define anaemia (non‐pregnant women: <120 g/L; children: <110 g/L). Child‐ and household‐level ASF consumption data were collected from 24‐hour food group intake and food consumption and expenditure surveys, respectively. In multiple logistic regression models, household livestock ownership was associated with anaemia among children (OR, 95% CI: 1.5 [1.1, 2.0]), but not women (1.0 [0.83, 1.2]). Household ownership of chickens was associated with higher odds of anaemia among children (1.6 [1.2, 2.2]), but ownership of other animal species was not associated with anaemia among women or children. In path analyses, we observed no evidence of mediation of the association of household livestock ownership with child anaemia by ASF consumption. Ownership of livestock likely has limited importance for consumption of ASFs among young children in Ghana and may in fact place children at an increased risk of anaemia. Further research is needed to elucidate if and how pathogen exposure associated with livestock rearing may underlie this increased risk of anaemia.
Collapse
Affiliation(s)
- Andrew D Jones
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Esi K Colecraft
- Nutrition and Food Science Department, University of Ghana, Accra, Ghana
| | - Raphael B Awuah
- Regional Institute for Population Studies, University of Ghana, Accra, Ghana
| | - Sandra Boatemaa
- Regional Institute for Population Studies, University of Ghana, Accra, Ghana
| | - Nathalie J Lambrecht
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Mark L Wilson
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
40
|
Ordiz MI, Davitt C, Stephenson K, Agapova S, Divala O, Shaikh N, Manary MJ. EB 2017 Article: Interpretation of the lactulose:mannitol test in rural Malawian children at risk for perturbations in intestinal permeability. Exp Biol Med (Maywood) 2018; 243:677-683. [PMID: 29597877 DOI: 10.1177/1535370218768508] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The dual sugar absorption test, specifically the lactulose:mannitol test, is used to assess gut health. Lactulose absorption is said to represent gut damage and mannitol absorption is used as a measure of normal small bowel function and serves as normalizing factor for lactulose. A underappreciated limitation of this common understanding of the lactulose:mannitol test is that mannitol is not absorbed to any substantial extent by a transcellular process. Additionally, this interpretation of lactulose:mannitol is not consistent with current understanding of paracellular pathways, where three pathway types exist: pore, leak, and unrestricted. Pore and leak pathways are regulated biological constructions of the small bowel barrier, and unrestricted pathways represent micropathological damage. We analyzed 2334 lactulose:mannitol measurements rigorously collected from 622 young rural Malawian children at high risk for poor gut health in light of the pathway model. An alternative method of normalizing for gut length utilizing autopsy data is described. In our population, absorbed lactulose and mannitol are strongly correlated, r = 0.68 P <0.0001, suggesting lactulose and mannitol are traversing the gut barrier via the same pathways. Considering measurements where pore pathways predominate, mannitol flux is about 14 times that of lactulose. As more leak pathways are present, this differential flux mannitol:lactulose falls to 8:1 and when increased numbers of unrestricted pathways are present, the differential flux of mannitol:lactulose is 6:1. There was no substantial correlation between the lactulose:mannitol and linear growth. Given that mannitol will always pass through a given pathway at a rate at least equal to that of lactulose, and lactulose absorption is a composite measure of flux through both physiologic and pathologic pathways, we question the utility of the lactulose:mannitol test. We suggest using lactulose alone is as informative as lactulose:mannitol in a sugar absorption testing in subclinical gut inflammation. Impact statement Our work integrates the standard interpretation of the lactulose:mannitol test (L:M), with mechanistic insight of intestinal permeability. There are three paracellular pathways in the gut epithelium; pore, leak, and unrestricted. Using thousands of L:M measurements from rural Malawian children at risk for increased intestinal permeability, we predict the differential flux of L and M through the pathways. Our findings challenge the traditional notions that little L is absorbed through a normal epithelial barrier and that M is a normalizing factor for L. Our observations are consistent with pore pathways allowing only M to pass. And that substantial amounts of L and M pass through leak pathways which are normal, regulated, cell-junctional adaptations. So M is a composite measure of all pathways, and L is not a measure solely of pathologic gut damage. Using L alone as a probe will yield more information about gut health than L:M.
Collapse
Affiliation(s)
- M Isabel Ordiz
- 1 Department of Pediatrics, Washington University at Saint Louis, St. Louis, MO 63110, USA
| | - Caroline Davitt
- 1 Department of Pediatrics, Washington University at Saint Louis, St. Louis, MO 63110, USA
| | - Kevin Stephenson
- 1 Department of Pediatrics, Washington University at Saint Louis, St. Louis, MO 63110, USA
| | - Sophia Agapova
- 1 Department of Pediatrics, Washington University at Saint Louis, St. Louis, MO 63110, USA
| | - Oscar Divala
- 2 School of Public Health and Family Medicine, University of Malawi, Blantyre 3, Malawi
| | - Nurmohammad Shaikh
- 1 Department of Pediatrics, Washington University at Saint Louis, St. Louis, MO 63110, USA
| | - Mark J Manary
- 1 Department of Pediatrics, Washington University at Saint Louis, St. Louis, MO 63110, USA.,2 School of Public Health and Family Medicine, University of Malawi, Blantyre 3, Malawi.,3 Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
41
|
Environmental enteric dysfunction and systemic inflammation predict reduced weight but not length gain in rural Bangladeshi children. Br J Nutr 2018; 119:407-414. [DOI: 10.1017/s0007114517003683] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AbstractEnvironmental enteric dysfunction (EED) and systemic inflammation (SI) are common in developing countries and may cause stunting. In Bangladesh, >40 % of preschool children are stunted, but EED and SI contributions are unknown. We aimed to determine the impact of EED and SI (assessed with multiple indicators) on growth in children (n 539) enrolled in a community-based randomised food supplementation trial in rural Bangladesh. EED was defined with faecal myeloperoxidase, α-1 antitrypsin and neopterin and serum endotoxin core antibody and glucagon-like peptide-2, consolidated into gut inflammation (GI) and permeability (GP) scores, and urinary lactulose:mannitol α-1 acid glycoprotein (AGP) characterised SI. Biomarker associations with anthropometry (15-, 18- and 24-month length-for-age (LAZ), weight-for-length (WLZ) and weight-for-age (WAZ) z scores) were examined in pairwise correlations and adjusted mixed-effects regressions. Stunting, wasting and underweight prevalence at 18 months were 45, 15 and 37 %, respectively, with elevated EED and SI markers common. EED and SI were not associated with 15–24-month length trajectory. Elevated (worse) GI and GP scores predicted reduced 18–24-month WLZ change (β −0·01 (se 0·00) z score/month for both). Elevated GP was also associated with reduced 15–18-month WLZ change (β −0·03 (se 0·01) z score/month) and greater 15-month WLZ (β 0·16 (se 0·05)). Higher AGP was associated with reduced prior and increased subsequent WLZ change (β −0·04 (se 0·01) and β 0·02 (se 0·00) z score/month for 15–18 and 18–24 months). The hypothesised link from EED to stunting was not observed in this sample of Bangladeshi 18-month-olds, but the effects of EED on constrained weight gain may have consequences for later linear growth or for other health and development outcomes.
Collapse
|
42
|
Agapova SE, Stephenson KB, Divala O, Kaimila Y, Maleta KM, Thakwalakwa C, Ordiz MI, Trehan I, Manary MJ. Additional Common Bean in the Diet of Malawian Children Does Not Affect Linear Growth, but Reduces Intestinal Permeability. J Nutr 2018; 148:267-274. [PMID: 29490090 DOI: 10.1093/jn/nxx013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/18/2017] [Indexed: 01/26/2023] Open
Abstract
Background Chronic malnutrition, as manifested by linear growth faltering, is pervasive among rural African children. Improvements in complementary feeding may decrease the burden of environmental enteric dysfunction (EED) and thus improve growth in children during the critical first 1000 d of development. Objective We tested the hypothesis that systematically including common bean or cowpea into complementary feeding would reduce EED and growth faltering among children in rural Malawi. Methods This was a double-blind clinical trial in which children 12-23 mo of age were randomly assigned to receive complementary feeding with 1 of 3 foods: roasted cowpea or common bean flour, or an isoenergetic amount of corn-soy blend as a control food for 48 wk. Children aged 12-23 mo received 155 kcal/d and thereafter until 35 mo received 200 kcal/d. The primary outcomes were change in length-for-age z score (LAZ) and improvements in a biomarker of EED, the percentage of lactulose (%L) excreted as part of the lactulose:mannitol dual-sugar absorption test. Anthropometric measurements and urinary %L excretion were compared between the 2 intervention groups and the control group separately with the use of linear mixed model analyses for repeated measures. Results A total of 331 children completed the clinical trial. Compliance with the study interventions was excellent, with >90% of the intervention flour consumed as intended. No significant effects on LAZ, change in LAZ, or weight-for-length z score were observed due to either intervention legume, compared to the control. %L was reduced with common bean consumption (effect estimate was -0.07 percentage points of lactulose, P = 0.0007). The lactulose:mannitol test was not affected by the legume intervention. Conclusion The addition of common bean to complementary feeding of rural Malawian children during the second year of life led to an improvement in a biomarker of gut health, although this did not directly translate into improved linear growth. This trial was registered at clinicaltrials.gov as NCT02472301.
Collapse
Affiliation(s)
| | | | - Oscar Divala
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Yankho Kaimila
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Kenneth M Maleta
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Chrissie Thakwalakwa
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - M Isabel Ordiz
- Department of Pediatrics, Washington University, St Louis, MO
| | - Indi Trehan
- Department of Pediatrics, Washington University, St Louis, MO.,Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Mark J Manary
- Department of Pediatrics, Washington University, St Louis, MO.,School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi.,Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
43
|
Harper KM, Mutasa M, Prendergast AJ, Humphrey J, Manges AR. Environmental enteric dysfunction pathways and child stunting: A systematic review. PLoS Negl Trop Dis 2018; 12:e0006205. [PMID: 29351288 PMCID: PMC5792022 DOI: 10.1371/journal.pntd.0006205] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/31/2018] [Accepted: 01/03/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is commonly defined as an acquired subclinical disorder of the small intestine, characterized by villous atrophy and crypt hyperplasia. EED has been proposed to underlie stunted growth among children in developing countries. A collection of biomarkers, organized into distinct domains, has been used to measure different aspects of EED. Here, we examine whether these hypothesized relationships, among EED domains and between each domain and stunting, are supported by data from recent studies. METHODOLOGY A systematic literature search was conducted using PubMed, MEDLINE, EMBASE, Web of Science, and CINAHL between January 1, 2010 and April 20, 2017. Information on study objective, design, population, location, biomarkers, and results were recorded, as well as qualitative and quantitative definitions of EED. Biomarkers were organized into five EED domains, and the number of studies that support or do not support relationships among domains and between each domain with stunting were summarized. RESULTS There was little evidence to support the pathway from intestinal permeability to microbial translocation and from microbial translocation to stunting, but stronger support existed for the link between intestinal inflammation and systemic inflammation and for intestinal inflammation and stunting. There was conflicting evidence for the pathways from intestinal damage to intestinal permeability and intestinal damage to stunting. CONCLUSIONS These results suggest that certain EED biomarkers may require reconsideration, particularly those most difficult to measure, such as microbial translocation and intestinal permeability. We discuss several issues with currently used biomarkers and recommend further analysis of pathogen-induced changes to the intestinal microbiota as a pathway leading to stunting.
Collapse
Affiliation(s)
- Kaitlyn M. Harper
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Maxine Mutasa
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Andrew J. Prendergast
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jean Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Amee R. Manges
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
- British Columbia Centre for Disease Control, Vancouver, Canada
| |
Collapse
|
44
|
George CM, Burrowes V, Perin J, Oldja L, Biswas S, Sack D, Ahmed S, Haque R, Bhuiyan NA, Parvin T, Bhuyian SI, Akter M, Li S, Natarajan G, Shahnaij M, Faruque AG, Stine OC. Enteric Infections in Young Children are Associated with Environmental Enteropathy and Impaired Growth. Trop Med Int Health 2017; 23:26-33. [PMID: 29121442 DOI: 10.1111/tmi.13002] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To investigate the relationship between faecal contamination in child play spaces, enteric infections, environmental enteropathy (EE) and impaired growth among young children. METHODS A prospective cohort study was conducted of 203 children 6-30 months of age in rural Bangladesh. Stool samples were analysed by quantitative PCR for Shigella, Enterotoxigenic Escherichia coli (ETEC), Campylobacter jejuni, Giardia intestinalis and Cryptosporidium spp. Four faecal markers of intestinal inflammation were also measured: alpha-1-antitrypsin, myeloperoxidase, neopterin and calprotectin. Child growth was measured at baseline and 9 months after enrolment. E. coli was measured in soil in child play spaces. RESULTS Forty-seven percent of study children had three or more enteric pathogens in their stool. Thirty five percent (71/203) of children had Shigella, 30% (61/203) had ETEC, 73% (148/203) had C. jejuni, 79% (160/203) had Giardia intestinalis and none had Cryptosporidium. Children with ETEC had significantly higher calprotectin concentrations (Coefficient: 1.35, 95% Confidence Interval [CI]: 1.005, 1.82). Children with Shigella had a significantly higher odds of being stunted at our 9-month follow-up (OR: 2.01, 95% CI: 1.02, 3.93). Children with Giardia intestinalis had significantly higher E.coli counts in the soil collected from their play spaces (OR: 1.23, 95% CI: 1.02, 1.48). CONCLUSION Enteric infections were significantly associated with EE and impaired growth in rural Bangladesh. These findings provide further evidence to support the hypothesis that contaminated soil in child play spaces can lead to enteric infections, many of which are likely subclinical, resulting in EE and impaired growth in young children.
Collapse
Affiliation(s)
- Christine Marie George
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Vanessa Burrowes
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jamie Perin
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Lauren Oldja
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shwapon Biswas
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh.,Department of Internal Medicine, Rangpur Medical College Hospital, Rangpur, Bangladesh
| | - David Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shahnawaz Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Nurul Amin Bhuiyan
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Tahmina Parvin
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | | | - Mahmuda Akter
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Shan Li
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gayathri Natarajan
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Mohammad Shahnaij
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Abu G Faruque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - O Colin Stine
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
45
|
Stephenson KB, Agapova SE, Divala O, Kaimila Y, Maleta KM, Thakwalakwa C, Ordiz MI, Trehan I, Manary MJ. Complementary feeding with cowpea reduces growth faltering in rural Malawian infants: a blind, randomized controlled clinical trial. Am J Clin Nutr 2017; 106:1500-1507. [PMID: 29092882 PMCID: PMC6482976 DOI: 10.3945/ajcn.117.160986] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/04/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Growth faltering is common in rural African children and is attributed to inadequate dietary intake and environmental enteric dysfunction (EED).Objective: We tested the hypothesis that complementary feeding with cowpea or common bean flour would reduce growth faltering and EED in 6-mo-old rural Malawians compared with the control group receiving a corn-soy blend.Design: A prospective, double-blind, randomized controlled clinical trial was conducted in which children received daily feeding for 6 mo (200 kcal/d when 6-9 mo old and 300 kcal/d when 10-12 mo old). The primary outcomes were change in length-for-age z score (LAZ) and improvements in EED, as measured by percentage of lactulose excretion (%L). %L <0.2% was considered normal. Anthropometric measurements and %L through urine were compared between each legume group and the control group with Student's t test.Results: Of the 355 infants enrolled, 291 infants completed the trial, and 288 were breastfed throughout the duration of the study. Cowpea and common bean added 4.6-5.2 g protein/d and 4-5 g indigestible carbohydrate/d to the diet. LAZ and weight-for-height z score were reduced in all 3 groups from 6 to 12 mo of age. The changes in LAZ [mean (95% CI)] for the cowpea, common bean, and control groups from 6 to 9 mo were -0.14 (-0.24, -0.04), -0.27 (-0.38, -0.16), and -0.27 (-0.35, -0.19), respectively. LAZ was reduced less in infants receiving cowpea than in those receiving control food from 6 to 9 mo (P = 0.048). The absolute value of %L did not differ between the dietary groups at 9 mo of age (mean ± SD: 0.30 ± 0.43, 0.23 ± 0.21, and 0.26 ± 0.31 for cowpea, common bean, and control, respectively), nor did the change in %L from 6 to 9 mo.Conclusion: Addition of cowpea to complementary feeding in Malawian infants resulted in less linear growth faltering. This trial was registered at clinicaltrials.gov as NCT02472262.
Collapse
Affiliation(s)
| | | | - Oscar Divala
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - Yankho Kaimila
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - Kenneth M Maleta
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - Chrissie Thakwalakwa
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - M Isabel Ordiz
- Department of Pediatrics, Washington University, St. Louis, MO
| | - Indi Trehan
- Department of Pediatrics, Washington University, St. Louis, MO,Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Mark J Manary
- Department of Pediatrics, Washington University, St. Louis, MO; .,School of Public Health and Family Medicine and.,Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
46
|
Abstract
The global impact of childhood malnutrition is staggering. The synergism between malnutrition and infection contributes substantially to childhood morbidity and mortality. Anthropometric indicators of malnutrition are associated with the increased risk and severity of infections caused by many pathogens, including viruses, bacteria, protozoa, and helminths. Since childhood malnutrition commonly involves the inadequate intake of protein and calories, with superimposed micronutrient deficiencies, the causal factors involved in impaired host defense are usually not defined. This review focuses on literature related to impaired host defense and the risk of infection in primary childhood malnutrition. Particular attention is given to longitudinal and prospective cohort human studies and studies of experimental animal models that address causal, mechanistic relationships between malnutrition and host defense. Protein and micronutrient deficiencies impact the hematopoietic and lymphoid organs and compromise both innate and adaptive immune functions. Malnutrition-related changes in intestinal microbiota contribute to growth faltering and dysregulated inflammation and immune function. Although substantial progress has been made in understanding the malnutrition-infection synergism, critical gaps in our understanding remain. We highlight the need for mechanistic studies that can lead to targeted interventions to improve host defense and reduce the morbidity and mortality of infectious diseases in this vulnerable population.
Collapse
|
47
|
Cheng WD, Wold KJ, Benzoni NS, Thakwalakwa C, Maleta KM, Manary MJ, Trehan I. Lactoferrin and lysozyme to reduce environmental enteric dysfunction and stunting in Malawian children: study protocol for a randomized controlled trial. Trials 2017; 18:523. [PMID: 29110675 PMCID: PMC5674751 DOI: 10.1186/s13063-017-2278-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/25/2017] [Indexed: 12/11/2022] Open
Abstract
Background Chronic childhood malnutrition, as manifested by stunted linear growth, remains a persistent barrier to optimal child growth and societal development. Environmental enteric dysfunction (EED) is a significant underlying factor in the causal pathway to stunting, delayed cognitive development, and ultimately morbidity and mortality. Effective therapies against EED and stunting are lacking and further clinical trials are warranted to effectively identify and operationalize interventions. Methods/design A prospective randomized placebo-controlled parallel-group randomized controlled trial will be conducted to determine if a daily supplement of lactoferrin and lysozyme, two important proteins found in breast milk, can decrease the burden of EED and stunting in rural Malawian children aged 12–23 months old. The intervention and control groups will have a sample size of 86 subjects each. All field and laboratory researchers will be blinded to the assigned intervention group, as will the subjects and their caregivers. The percentage of ingested lactulose excreted in the urine (Δ%L) after 4 h will be used as the biomarker for EED and linear growth as the measure of chronic malnutrition (stunting). The primary outcomes of interest will be change in Δ%L from baseline to 8 weeks and to 16 weeks. Intention-to-treat analyses will be used. Discussion A rigorous clinical trial design will be used to assess the biologically plausible use of lactoferrin and lysozyme as dietary supplements for children at high risk for EED. If proven effective, these safe proteins may serve to markedly reduce the burden of childhood malnutrition and improve survival. Trial Registration Clinicaltrials.gov, NCT02925026. Registered on 4 October 2016. Electronic supplementary material The online version of this article (doi:10.1186/s13063-017-2278-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- William D Cheng
- Department of Pediatrics, Washington University in St. Louis, One Children's Place, Campus Box 8116, Saint Louis, MO, 63110, USA
| | - Karl J Wold
- Department of Pediatrics, Washington University in St. Louis, One Children's Place, Campus Box 8116, Saint Louis, MO, 63110, USA
| | - Nicole S Benzoni
- Department of Pediatrics, Washington University in St. Louis, One Children's Place, Campus Box 8116, Saint Louis, MO, 63110, USA
| | - Chrissie Thakwalakwa
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - Kenneth M Maleta
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - Mark J Manary
- Department of Pediatrics, Washington University in St. Louis, One Children's Place, Campus Box 8116, Saint Louis, MO, 63110, USA.,School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi.,Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Indi Trehan
- Department of Pediatrics, Washington University in St. Louis, One Children's Place, Campus Box 8116, Saint Louis, MO, 63110, USA. .,Department of Paediatrics and Child Health, University of Malawi, Blantyre, Malawi. .,Lao Friends Hospital for Children, Luang Prabang, Lao PDR.
| |
Collapse
|
48
|
Dorshow RB, Hall-Moore C, Shaikh N, Talcott MR, Faubion WA, Rogers TE, Shieh JJ, Debreczeny MP, Johnson JR, Dyer RB, Singh RJ, Tarr PI. Measurement of gut permeability using fluorescent tracer agent technology. Sci Rep 2017; 7:10888. [PMID: 28883476 PMCID: PMC5589723 DOI: 10.1038/s41598-017-09971-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 07/31/2017] [Indexed: 12/27/2022] Open
Abstract
The healthy gut restricts macromolecular and bacterial movement across tight junctions, while increased intestinal permeability accompanies many intestinal disorders. Dual sugar absorption tests, which measure intestinal permeability in humans, present challenges. Therefore, we asked if enterally administered fluorescent tracers could ascertain mucosal integrity, because transcutaneous measurement of differentially absorbed molecules could enable specimen-free evaluation of permeability. We induced small bowel injury in rats using high- (15 mg/kg), intermediate- (10 mg/kg), and low- (5 mg/kg) dose indomethacin. Then, we compared urinary ratios of enterally administered fluorescent tracers MB-402 and MB-301 to urinary ratios of sugar tracers lactulose and rhamnose. We also tested the ability of transcutaneous sensors to measure the ratios of absorbed fluorophores. Urinary fluorophore and sugar ratios reflect gut injury in an indomethacin dose dependent manner. The fluorophores generated smooth curvilinear ratio trajectories with wide dynamic ranges. The more chaotic sugar ratios had narrower dynamic ranges. Fluorophore ratios measured through the skin distinguished indomethacin-challenged from same day control rats. Enterally administered fluorophores can identify intestinal injury in a rat model. Fluorophore ratios are measureable through the skin, obviating drawbacks of dual sugar absorption tests. Pending validation, this technology should be considered for human use.
Collapse
Affiliation(s)
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Michael R Talcott
- Division of Comparative Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - William A Faubion
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Roy B Dyer
- Immunochemical Core Laboratory, Mayo Clinic, Rochester, MN, USA
| | | | - Phillip I Tarr
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
49
|
Conan A, O’Reilly CE, Ogola E, Ochieng JB, Blackstock AJ, Omore R, Ochieng L, Moke F, Parsons MB, Xiao L, Roellig D, Farag TH, Nataro JP, Kotloff KL, Levine MM, Mintz ED, Breiman RF, Cleaveland S, Knobel DL. Animal-related factors associated with moderate-to-severe diarrhea in children younger than five years in western Kenya: A matched case-control study. PLoS Negl Trop Dis 2017; 11:e0005795. [PMID: 28783751 PMCID: PMC5559092 DOI: 10.1371/journal.pntd.0005795] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/16/2017] [Accepted: 07/11/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Diarrheal disease remains among the leading causes of global mortality in children younger than 5 years. Exposure to domestic animals may be a risk factor for diarrheal disease. The objectives of this study were to identify animal-related exposures associated with cases of moderate-to-severe diarrhea (MSD) in children in rural western Kenya, and to identify the major zoonotic enteric pathogens present in domestic animals residing in the homesteads of case and control children. METHODOLOGY/PRINCIPAL FINDINGS We characterized animal-related exposures in a subset of case and control children (n = 73 pairs matched on age, sex and location) with reported animal presence at home enrolled in the Global Enteric Multicenter Study in western Kenya, and analysed these for an association with MSD. We identified potentially zoonotic enteric pathogens in pooled fecal specimens collected from domestic animals resident at children's homesteads. Variables that were associated with decreased risk of MSD were washing hands after animal contact (matched odds ratio [MOR] = 0.2; 95% CI 0.08-0.7), and presence of adult sheep that were not confined in a pen overnight (MOR = 0.1; 0.02-0.5). Variables that were associated with increased risk of MSD were increasing number of sheep owned (MOR = 1.2; 1.0-1.5), frequent observation of fresh rodent excreta (feces/urine) outside the house (MOR = 7.5; 1.5-37.2), and participation of the child in providing water to chickens (MOR = 3.8; 1.2-12.2). Of 691 pooled specimens collected from 2,174 domestic animals, 159 pools (23%) tested positive for one or more potentially zoonotic enteric pathogens (Campylobacter jejuni, C. coli, non-typhoidal Salmonella, diarrheagenic E. coli, Giardia, Cryptosporidium, or rotavirus). We did not find any association between the presence of particular pathogens in household animals, and MSD in children. CONCLUSIONS AND SIGNIFICANCE Public health agencies should continue to promote frequent hand washing, including after animal contact, to reduce the risk of MSD. Future studies should address specific causal relations of MSD with sheep and chicken husbandry practices, and with the presence of rodents.
Collapse
Affiliation(s)
- Anne Conan
- Ross University School of Veterinary Medicine, Basseterre, St Kitts and Nevis
| | - Ciara E. O’Reilly
- Division of Foodborne, Waterborne, and Environmental Diseases, US Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Eric Ogola
- School of Health Sciences, Jaramogi Oginga Odinga University of Science and Technology, Bondo, Kenya
| | - J. Benjamin Ochieng
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Anna J. Blackstock
- Division of Foodborne, Waterborne, and Environmental Diseases, US Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Richard Omore
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Linus Ochieng
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Fenny Moke
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Michele B. Parsons
- Division of Global Health and Protection, Center for Global Health, US Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Lihua Xiao
- Division of Foodborne, Waterborne, and Environmental Diseases, US Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Dawn Roellig
- Division of Foodborne, Waterborne, and Environmental Diseases, US Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Tamer H. Farag
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - James P. Nataro
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Karen L. Kotloff
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Myron M. Levine
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Eric D. Mintz
- Division of Foodborne, Waterborne, and Environmental Diseases, US Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Robert F. Breiman
- International Emerging Infections Program, Centers for Disease Control and Prevention, Nairobi, Kenya
| | - Sarah Cleaveland
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Darryn L. Knobel
- Ross University School of Veterinary Medicine, Basseterre, St Kitts and Nevis
- * E-mail:
| |
Collapse
|
50
|
Mosites E, Dawson-Hahn E, Walson J, Rowhani-Rahbar A, Neuhouser ML. Piecing together the stunting puzzle: a framework for attributable factors of child stunting. Paediatr Int Child Health 2017; 37:158-165. [PMID: 27680199 DOI: 10.1080/20469047.2016.1230952] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Reducing the burden of stunting in childhood is critical to improving health in low- and middle-income settings. However, because many aetiologies underlie linear growth failure, stunting has proved difficult to prevent and reverse. Understanding the contributions these aetiologies make to the burden of stunting can help the development of targeted, effective interventions. To begin to frame these causes, a qualitative and a quantitative framework of the primary drivers of stunting in low-resource settings were developed. Population attributable fractions (PAF) were estimated to inform the quantitative framework. According to these estimates, infectious diseases were responsible for large attributable fractions in all settings, and a combination of dietary indicators also comprised a large fraction in Africa. However, the PAF calculation was found to have several limitations, including a requirement for a binary outcome and sensitivity to confounding, which necessitate broad interpretation of the results. More robust tools to model complex causality are needed in order to understand the causal aetiology of stunting.
Collapse
Affiliation(s)
- Emily Mosites
- a Department of Epidemiology , University of Washington , Seattle , USA.,b Paul G. Allen School for Global Animal Health , Washington State University , Pullman , USA
| | | | - Judd Walson
- a Department of Epidemiology , University of Washington , Seattle , USA.,c Department of Pediatrics , University of Washington , Seattle , USA.,d Department of Global Health , University of Washington , Seattle , USA.,e Department of Allergy and Infectious Disease , University of Washington , Seattle , USA
| | | | - Marian L Neuhouser
- a Department of Epidemiology , University of Washington , Seattle , USA.,f Division of Public Health Sciences , Fred Hutchinson Cancer Research Center , Seattle , USA
| |
Collapse
|