101
|
Tickell KD, Atlas HE, Walson JL. Environmental enteric dysfunction: a review of potential mechanisms, consequences and management strategies. BMC Med 2019; 17:181. [PMID: 31760941 PMCID: PMC6876067 DOI: 10.1186/s12916-019-1417-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is an acquired enteropathy of the small intestine, characterized by enteric inflammation, villus blunting and decreased crypt-to-villus ratio. EED has been associated with poor outcomes, including chronic malnutrition (stunting), wasting and reduced vaccine efficacy among children living in low-resource settings. As a result, EED may be a valuable interventional target for programs aiming to reduce childhood morbidity in low and middle-income countries. MAIN TEXT Several highly plausible mechanisms link the proposed pathophysiology underlying EED to adverse outcomes, but causal attribution of these pathways has proved challenging. We provide an overview of recent studies evaluating the causes and consequences of EED. These include studies of the role of subclinical enteric infection as a primary cause of EED, and efforts to understand how EED-associated systemic inflammation and malabsorption may result in long-term morbidity. Finally, we outline recently completed and upcoming clinical trials that test novel interventions to prevent or treat this highly prevalent condition. CONCLUSIONS Significant strides have been made in linking environmental exposure to enteric pathogens and toxins with EED, and in understanding the multifactorial mechanisms underlying this complex condition. Further insights may come from several ongoing and upcoming interventional studies trialing a variety of novel management strategies.
Collapse
Affiliation(s)
- Kirkby D Tickell
- Department of Global Health, University of Washington, 325 9th Avenue (Box 359931), Seattle, WA, 98104, USA. .,Department of Epidemiology, University of Washington, 1959 NE Pacific Street, Health Sciences Bldg, F-262, Box 357236, Seattle, WA, 98195, USA. .,The Childhood Acute Illness and Nutrition Network (CHAIN), Nairobi, Kenya.
| | - Hannah E Atlas
- Department of Global Health, University of Washington, 325 9th Avenue (Box 359931), Seattle, WA, 98104, USA
| | - Judd L Walson
- Department of Global Health, University of Washington, 325 9th Avenue (Box 359931), Seattle, WA, 98104, USA.,Department of Epidemiology, University of Washington, 1959 NE Pacific Street, Health Sciences Bldg, F-262, Box 357236, Seattle, WA, 98195, USA.,The Childhood Acute Illness and Nutrition Network (CHAIN), Nairobi, Kenya.,Department of Allergy and Infectious Disease, University of Washington, 325 9th Avenue (Box 359931), Seattle, WA, 98104, USA.,Department of Pediatrics, University of Washington, 325 9th Avenue (Box 359931), Seattle, WA, 98104, USA
| |
Collapse
|
102
|
Brander RL, Pavlinac PB, Walson JL, John-Stewart GC, Weaver MR, Faruque ASG, Zaidi AKM, Sur D, Sow SO, Hossain MJ, Alonso PL, Breiman RF, Nasrin D, Nataro JP, Levine MM, Kotloff KL. Determinants of linear growth faltering among children with moderate-to-severe diarrhea in the Global Enteric Multicenter Study. BMC Med 2019; 17:214. [PMID: 31767012 PMCID: PMC6878715 DOI: 10.1186/s12916-019-1441-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 10/08/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Moderate-to-severe diarrhea (MSD) in the first 2 years of life can impair linear growth. We sought to determine risk factors for linear growth faltering and to build a clinical prediction tool to identify children most likely to experience growth faltering following an episode of MSD. METHODS Using data from the Global Enteric Multicenter Study of children 0-23 months old presenting with MSD in Africa and Asia, we performed log-binomial regression to determine clinical and sociodemographic factors associated with severe linear growth faltering (loss of ≥ 0.5 length-for-age z-score [LAZ]). Linear regression was used to estimate associations with ΔLAZ. A clinical prediction tool was developed using backward elimination of potential variables, and Akaike Information Criterion to select the best fit model. RESULTS Of the 5902 included children, mean age was 10 months and 43.2% were female. Over the 50-90-day follow-up period, 24.2% of children had severe linear growth faltering and the mean ΔLAZ over follow-up was - 0.17 (standard deviation [SD] 0.54). After adjustment for age, baseline LAZ, and site, several factors were associated with decline in LAZ: young age, acute malnutrition, hospitalization at presentation, non-dysenteric diarrhea, unimproved sanitation, lower wealth, fever, co-morbidity, or an IMCI danger sign. Compared to children 12-23 months old, those 0-6 months were more likely to experience severe linear growth faltering (adjusted prevalence ratio [aPR] 1.97 [95% CI 1.70, 2.28]), as were children 6-12 months of age (aPR 1.72 [95% CI 1.51, 1.95]). A prediction model that included age, wasting, stunting, presentation with fever, and presentation with an IMCI danger sign had an area under the ROC (AUC) of 0.67 (95% CI 0.64, 0.69). Risk scores ranged from 0 to 37, and a cut-off of 21 maximized sensitivity (60.7%) and specificity (63.5%). CONCLUSION Younger age, acute malnutrition, MSD severity, and sociodemographic factors were associated with short-term linear growth deterioration following MSD. Data routinely obtained at MSD may be useful to predict children at risk for growth deterioration who would benefit from interventions.
Collapse
Affiliation(s)
- Rebecca L Brander
- Department of Epidemiology, University of Washington, Seattle, WA, USA.
| | | | - Judd L Walson
- Department of Epidemiology, Global Health, Pediatrics, Medicine, Childhood Acute Illness and Nutrition Network, University of Washington, Seattle, WA, USA
| | - Grace C John-Stewart
- Department of Epidemiology, Global Health, Pediatrics, Medicine, University of Washington, Seattle, WA, USA
| | - Marcia R Weaver
- Department of Global Health, Health Services, Health Metrics Sciences, University of Washington, Seattle, WA, USA
| | - Abu S G Faruque
- International Centre for Diarrhoeal Disease Research, Mohakhali, Dhaka, Bangladesh
| | - Anita K M Zaidi
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan.,Present Address: Enteric and Diarrheal Diseases Program, Bill and Melinda Gates Foundation, Seattle, WA, USA
| | - Dipika Sur
- National Institute of Cholera and Enteric Diseases, Kolkata, India.,Present Address: Translational Health Science and Technology Institute, Faridabad, India
| | - Samba O Sow
- Centre pour le Développement des Vaccines, Bamako, Mali
| | - M Jahangir Hossain
- Medical Research Council Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Pedro L Alonso
- Centro de Investigação em Saúde da Manhiça, Maputo, Mozambique.,Present Address: Global Malaria Programme, World Health Organization, Geneva, Switzerland
| | - Robert F Breiman
- Global Disease Detection Division, Kenya Office of the US Centers for Disease Control and Prevention, Nairobi, Kenya.,Present Address: Global Health Institute Emory University, Atlanta, GA, USA
| | - Dilruba Nasrin
- Center for Vaccine Development, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - James P Nataro
- Center for Vaccine Development, Department of Medicine, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA.,Present Address: Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Myron M Levine
- Center for Vaccine Development and Global Health, Department of Pediatrics and Medicine, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Karen L Kotloff
- Center for Vaccine Development and Global Health, Department of Pediatrics and Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
103
|
Haque MA, Platts-Mills JA, Mduma E, Bodhidatta L, Bessong P, Shakoor S, Kang G, Kosek MN, Lima AAM, Shrestha SK, Alam MA, Havt A, Samie A, Guerrant RL, Lang D, Mahfuz M, Bhutta ZA, Houpt ER, Ahmed T. Determinants of Campylobacter infection and association with growth and enteric inflammation in children under 2 years of age in low-resource settings. Sci Rep 2019; 9:17124. [PMID: 31748573 PMCID: PMC6868199 DOI: 10.1038/s41598-019-53533-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/31/2019] [Indexed: 12/11/2022] Open
Abstract
Campylobacter species infections have been associated with malnutrition and intestinal inflammation among children in low-resource settings. However, it remains unclear whether that association is specific to Campylobacter jejuni/coli. The aim of this study was to assess the association between both all Campylobacter species infections and Campylobacter jejuni/coli infections on growth and enteric inflammation in children aged 1-24 months. We analyzed data from 1715 children followed from birth until 24 months of age in the MAL-ED birth cohort study, including detection of Campylobacter species by enzyme immunoassay and Campylobacter jejuni/coli by quantitative PCR in stool samples. Myeloperoxidase (MPO) concentration in stool, used as a quantitative index of enteric inflammation, was measured. The incidence rate per 100 child-months of infections with Campylobacter jejuni/coli and Campylobacter species during 1-24 month follow up were 17.7 and 29.6 respectively. Female sex of child, shorter duration of exclusive breastfeeding, lower maternal age, mother having less than 3 living children, maternal educational level of <6 years, lack of routine treatment of drinking water, and unimproved sanitation were associated with Campylobacter jejuni/coli infection. The cumulative burden of both Campylobacter jejuni/coli infections and Campylobacter species were associated with poor growth and increased intestinal inflammation.
Collapse
Affiliation(s)
- Md Ahshanul Haque
- Nutrition and Clinical Services Division, icddr,b, Dhaka, Bangladesh.
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| | | | | | | | | | | | - Margaret N Kosek
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
- Asociación Benéfica PRISMA, Iquitos, Peru
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Aldo A M Lima
- Clinical Research Unit and Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | | | - Md Ashraful Alam
- Nutrition and Clinical Services Division, icddr,b, Dhaka, Bangladesh
| | - Alexandre Havt
- Clinical Research Unit and Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | | | - Richard L Guerrant
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| | - Dennis Lang
- Foundation for the National Institutes of Health, Bethesda, MD, USA
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, icddr,b, Dhaka, Bangladesh
| | | | - Eric R Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, icddr,b, Dhaka, Bangladesh
| |
Collapse
|
104
|
Sullivan PB. Environmental enteric dysfunction: a socioeconomic problem looking for a medical solution? Am J Clin Nutr 2019; 110:1051-1052. [PMID: 31667510 DOI: 10.1093/ajcn/nqz225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Peter B Sullivan
- University of Oxford, Department of Pediatrics, Children's Hospital, Oxford, United Kingdom
| |
Collapse
|
105
|
Balamurugan R, Pugazhendhi S, Balachander GM, Dharmalingam T, Mortimer EK, Gopalsamy GL, Woodman RJ, Meng R, Alpers DH, Manary M, Binder HJ, Brown IL, Young GP, Ramakrishna BS. Effect of Native and Acetylated Dietary Resistant Starches on Intestinal Fermentative Capacity of Normal and Stunted Children in Southern India. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E3922. [PMID: 31618992 PMCID: PMC6843365 DOI: 10.3390/ijerph16203922] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/10/2019] [Accepted: 10/12/2019] [Indexed: 12/29/2022]
Abstract
The health benefits of dietary amylase resistant starch (RS) arise from intestinal microbial fermentation and generation of short chain fatty acids (SCFA). We compared the intestinal fermentative capability of stunted and nonstunted ('healthy') children in southern India using two types of RS: high amylose maize starch (HAMS) and acetylated HAMS (HAMSA). Twenty children (10 stunted and 10 healthy) aged 2 to 5 years were fed biscuits containing HAMS (10 g/day) for two weeks followed by a 2-week washout and then HAMSA biscuits (10 g/day) for 2 weeks. Fecal samples were collected at 3-4 day intervals and pH and SCFA analyzed. At entry, stunted children had lower SCFA concentrations compared to healthy children. Both types of RS led to a significant decrease in fecal pH and increase in fecal acetate and propionate in both healthy and stunted children. However, while HAMS increased fecal butyrate in both groups of children, HAMSA increased butyrate in healthy but not stunted children. Furthermore, healthy children showed a significantly greater increase than stunted children in both acetate and butyrate when fed either RS. No adverse effects were reported with either RS. Stunted children have impaired capacity to ferment certain types of RS which has implications for choice of RS in formulations aimed at improving microbial function in stunted children.
Collapse
Affiliation(s)
- Ramadass Balamurugan
- Wellcome Research Unit (Biochemistry), Christian Medical College, Vellore, Tamil Nadu 632004, India.
| | - Srinivasan Pugazhendhi
- Wellcome Research Unit (Biochemistry), Christian Medical College, Vellore, Tamil Nadu 632004, India.
| | - Gowri M Balachander
- Wellcome Research Unit (Biochemistry), Christian Medical College, Vellore, Tamil Nadu 632004, India.
| | - Tamilselvan Dharmalingam
- Wellcome Research Unit (Biochemistry), Christian Medical College, Vellore, Tamil Nadu 632004, India.
| | - Elissa K Mortimer
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park 5045, South Australia, Australia.
| | - Geetha L Gopalsamy
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park 5045, South Australia, Australia.
| | - Richard J Woodman
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park 5045, South Australia, Australia.
| | - Rosie Meng
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park 5045, South Australia, Australia.
| | - David H Alpers
- Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Mark Manary
- Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Henry J Binder
- Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Ian L Brown
- Australian Cancer Research Foundation, Sydney 2000, Australia.
| | - Graeme P Young
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park 5045, South Australia, Australia.
| | | |
Collapse
|
106
|
Rude KM, Pusceddu MM, Keogh CE, Sladek JA, Rabasa G, Miller EN, Sethi S, Keil KP, Pessah IN, Lein PJ, Gareau MG. Developmental exposure to polychlorinated biphenyls (PCBs) in the maternal diet causes host-microbe defects in weanling offspring mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 253:708-721. [PMID: 31336350 PMCID: PMC6719698 DOI: 10.1016/j.envpol.2019.07.066] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 05/05/2023]
Abstract
The gut microbiota is important for maintaining homeostasis of the host. Gut microbes represent the initial site for toxicant processing following dietary exposures to environmental contaminants. The diet is the primary route of exposure to polychlorinated biphenyls (PCBs), which are absorbed via the gut, and subsequently interfere with neurodevelopment and behavior. Developmental exposures to PCBs have been linked to increased risk of neurodevelopmental disorders (NDD), including autism spectrum disorder (ASD), which are also associated with a high prevalence of gastrointestinal (GI) distress and intestinal dysbiosis. We hypothesized that developmental PCB exposure impacts colonization of the gut microbiota, resulting in GI pathophysiology, in a genetically susceptible host. Mouse dams expressing two heritable human mutations (double mutants [DM]) that result in abnormal Ca2+ dynamics and produce behavioral deficits (gain of function mutation in the ryanodine receptor 1 [T4826I-RYR1] and a human CGG repeat expansion [170-200 CGG repeats] in the fragile X mental retardation gene 1 [FMR1 premutation]). DM and congenic wild type (WT) controls were exposed to PCBs (0-6 mg/kg/d) in the diet starting 2 weeks before gestation and continuing through postnatal day 21 (P21). Intestinal physiology (Ussing chambers), inflammation (qPCR) and gut microbiome (16S sequencing) studies were performed in offspring mice (P28-P30). Developmental exposure to PCBs in the maternal diet caused significant mucosal barrier defects in ileum and colon (increased secretory state and tight junction permeability) of juvenile DM mice. Furthermore, PCB exposure increased the intestinal inflammatory profile (Il6, Il1β, and Il22), and resulted in dysbiosis of the gut microbiota, including altered β-diversity, in juvenile DM mice developmentally exposed to 1 mg/kg/d PCBs when compared to WT controls. Collectively, these findings demonstrate a novel interaction between PCB exposure and the gut microbiota in a genetically susceptible host that provide novel insight into environmental risk factors for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Kavi M Rude
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, United States
| | - Matteo M Pusceddu
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, United States
| | - Ciara E Keogh
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, United States
| | - Jessica A Sladek
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, United States
| | - Gonzalo Rabasa
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, United States
| | - Elaine N Miller
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, United States
| | - Sunjay Sethi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, United States
| | - Kimberly P Keil
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, United States
| | - Isaac N Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, United States
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, United States
| | - Mélanie G Gareau
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, United States.
| |
Collapse
|
107
|
Said-Mohamed R, Stein AD, Pettifor JM, Norris SA. Sanitation and diarrhoea in infancy and CRP level at 18 years: the birth-to-twenty plus cohort. Ann Hum Biol 2019; 46:415-424. [DOI: 10.1080/03014460.2019.1657496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Rihlat Said-Mohamed
- SA MRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Aryeh D. Stein
- SA MRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - John M. Pettifor
- SA MRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Shane A. Norris
- SA MRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| |
Collapse
|
108
|
Hossain M, Nahar B, Haque MA, Mondal D, Mahfuz M, Naila NN, Gazi MA, Hasan MM, Haque NMS, Haque R, Arndt MB, Walson JL, Ahmed T. Serum Adipokines, Growth Factors, and Cytokines Are Independently Associated with Stunting in Bangladeshi Children. Nutrients 2019; 11:nu11081827. [PMID: 31394828 PMCID: PMC6723106 DOI: 10.3390/nu11081827] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/06/2019] [Accepted: 08/06/2019] [Indexed: 02/07/2023] Open
Abstract
Growth in young children is controlled through the release of several hormonal signals, which are affected by diet, infection, and other exposures. Stunting is clearly a growth disorder, yet limited evidence exists documenting the association of different growth biomarkers with child stunting. This study explored the association between different growth biomarkers and stunting in Bangladeshi children. A quasi-experimental study was conducted among 50 stunted (length-for-age Z-score (LAZ) < -2 SD) and 50 control (LAZ ≥ -2 SD) children, aged 12-18 months, residing in a Bangladeshi slum. The enrolled stunted children received an intervention package, which included food supplementation for three months, psychosocial stimulation for six months, and routine clinical care on community nutrition center at the study field site. The controls received routine clinical care only. All children were clinically screened over the study period. Length, weight, fasting blood and fecal biomarkers were measured. All biomarkers levels were similar in both groups except for oxyntomodulin at enrolment. Leptin (adjusted odds ratio, AOR: 4.0, p < 0.01), leptin-adiponectin ratio (AOR 5.07 × 108, p < 0.01), insulin-like growth factor-1 (IGF-1) (AOR 1.02, p < 0.05), and gamma interferon (IFN-γ) (AOR 0.92, p < 0.05) levels were independently associated with stunting at enrolment. Serum leptin, leptin-adiponectin ratio, interleukin-6 (IL-6), IL-10, tumor necrosis factor-alpha (TNF-α), and fecal alpha-1-antitrypsin (AAT) levels increased significantly (p < 0.001), while IFN-γ levels significantly decreased among stunted children after six months of intervention. Leptin, leptin-adiponectin ratio, IGF-1, and IFN-γ are independently associated with stunting in Bangladeshi children. This trial was registered at clinicaltrials.gov as NCT02839148.
Collapse
Affiliation(s)
- Muttaquina Hossain
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh.
| | - Baitun Nahar
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Md Ahshanul Haque
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Dinesh Mondal
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Nurun Nahar Naila
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Md Amran Gazi
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Md Mehedi Hasan
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | | | - Rashidul Haque
- Enteric and Respiratory Infections, icddr,b, Dhaka 1212, Bangladesh
| | - Michael B Arndt
- PATH, Seattle, WA 98109, USA
- Department of Global Health, University of Washington, Seattle, WA 98109, USA
| | - Judd L Walson
- Department of Global Health, University of Washington, Seattle, WA 98109, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
- Childhood Acute Illness and Nutrition Network, Nairobi 00200, Kenya
- Departments of Medicine, University of Washington, Seattle, WA 98109, USA
- Department of Epidemiology, University of Washington, Seattle, WA 98109, USA
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
- Department of Global Health, University of Washington, Seattle, WA 98109, USA
- James P. Grant School of Public Health, BRAC University, Dhaka 1212, Bangladesh
| |
Collapse
|
109
|
Gwela A, Mupere E, Berkley JA, Lancioni C. Undernutrition, Host Immunity and Vulnerability to Infection Among Young Children. Pediatr Infect Dis J 2019; 38:e175-e177. [PMID: 31306401 PMCID: PMC7613497 DOI: 10.1097/inf.0000000000002363] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Agnes Gwela
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi,KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Ezekiel Mupere
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi,Department of Pediatrics, Makerere University, Kampala, Uganda
| | - James A Berkley
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi,KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya,Centre for Tropical Medicine & Global Health, University of Oxford, Oxford, United Kingdom
| | - Christina Lancioni
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi,Department of Pediatrics, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
110
|
Owino VO, Murphy‐Alford AJ, Kerac M, Bahwere P, Friis H, Berkley JA, Jackson AA. Measuring growth and medium- and longer-term outcomes in malnourished children. MATERNAL & CHILD NUTRITION 2019; 15:e12790. [PMID: 30690903 PMCID: PMC7199054 DOI: 10.1111/mcn.12790] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/13/2018] [Accepted: 12/12/2018] [Indexed: 12/26/2022]
Abstract
Severe and moderate acute malnutrition are among the leading causes of mortality among children in low- and middle-income countries. There is strong evidence that growth assessed anthropometrically from conception to 2 years of age marks later risk of ill health. This is central to the concept of the developmental origins of adult disease and is presumed to be related to modification of developmental processes during critical "window(s)" of vulnerability. Interventions to treat acute malnutrition have resulted in dramatic increase in the number of affected children surviving. Ensuring that these children thrive to fulfil their full physical and cognitive potential is a new challenge. Integral to this challenge is the need to be able to measure how earlier insults relate to the ability to survive and thrive to productive adulthood. Despite its obvious value, routine anthropometry does not adequately indicate how earlier adverse exposures affect more refined aspects of growth. Anthropometry is inadequate for predicting how disruption of healthy growth might modulate risk of disease or any subsequent interventions that correct this risk. A clear characterisation of healthy child growth is needed for determining which component best predicts later outcomes. The extent to which postnatal acute malnutrition is a consequence of maternal factors acting preconception or in utero and their relationship to postnatal health and long-term risk of non-communicable diseases is not clear. Body-composition measurement has significant untapped potential allowing us to translate and better understand the relationship between early insults and interventions on early growth in the short-term and long-term health outcomes.
Collapse
Affiliation(s)
- Victor O. Owino
- Nutritional and Health Related Environmental Studies Section, Division of HealthInternational Atomic Energy AgencyViennaAustria
| | - Alexia J. Murphy‐Alford
- Nutritional and Health Related Environmental Studies Section, Division of HealthInternational Atomic Energy AgencyViennaAustria
| | - Marko Kerac
- Department of Population HealthLondon School of Hygiene and Tropical MedicineLondonUK
| | - Paluku Bahwere
- Valid InternationalOxfordUK
- Research Centre in Epidemiology, Biostatistics and Clinical Research, School of Public HealthFree University of BrusselsBrusselsBelgium
| | - Henrik Friis
- Department of Nutrition, Exercise and SportsUniversity of CopenhagenFrederiksbergDenmark
| | - James A. Berkley
- DirectorThe Childhood Acute Illness & Nutrition (CHAIN) NetworkNairobiKenya
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global HealthUniversity of OxfordOxfordUK
| | - Alan A. Jackson
- Human Nutrition, International Malnutrition Task ForceSouthampton General HospitalSouthamptonUK
| |
Collapse
|
111
|
Wang Y, Mortimer EK, Katundu KGH, Kalanga N, Leong LEX, Gopalsamy GL, Christophersen CT, Richard AC, Shivasami A, Abell GCJ, Young GP, Rogers GB. The Capacity of the Fecal Microbiota From Malawian Infants to Ferment Resistant Starch. Front Microbiol 2019; 10:1459. [PMID: 31316490 PMCID: PMC6611432 DOI: 10.3389/fmicb.2019.01459] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/11/2019] [Indexed: 01/10/2023] Open
Abstract
In Low and Middle-Income Countries (LMIC), weaning is associated with environmentally acquired and inflammation-associated enteric disorders. Dietary intake of high amylose maize starch (HAMS) can promote commensal fermentative bacteria and drive the production of short chain fatty acids (SCFAs). By stabilizing commensal gut microbiology, and stimulating the production of anti-inflammatory metabolites, HAMS supplementation might therefore influence enteric health. However, the extent to which the gut microbiota of LMIC infants are capable of fermenting HAMS is unclear. We assessed the capacity of the fecal microbiota from pre-weaning and weaning Malawian infants to ferment HAMS and produce SCFAs using an in vitro fermentation model. Fecal microbiota from both pre-weaning and weaning infants were able to ferment HAMS, as indicated by an increase in bacterial load and total SCFA concentration, and a reduction in pH. All of these changes were more substantial in the weaning group. Acetate production was observed with both pre-weaning and weaning groups, while propionate production was only observed in the weaning group. HAMS fermentation resulted in significant alterations to the fecal microbial community in the weaning group, with significant increases in levels of Prevotella, Veillonella, and Collinsella associated with propionate production. In conclusion, fecal microbiota from Malawian infants before and during weaning has the capacity to produce acetate through HAMS fermentation, with propionate biosynthetic capability appearing only at weaning. Our results suggest that HAMS supplementation might provide benefit to infants during weaning.
Collapse
Affiliation(s)
- Yanan Wang
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Elissa K. Mortimer
- Flinders University Global GI Health Unit, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Kondwani G. H. Katundu
- Division of Physiology, Biomedical Sciences Department, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Noel Kalanga
- Department of Health Systems and Policy, School of Public Health, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Lex E. X. Leong
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Geetha L. Gopalsamy
- Flinders University Global GI Health Unit, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Claus T. Christophersen
- School of Medical & Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- School of Molecular & Life Sciences, Curtin University, Perth, WA, Australia
| | - Alyson C. Richard
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Aravind Shivasami
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Guy C. J. Abell
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Graeme P. Young
- Flinders University Global GI Health Unit, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Geraint B. Rogers
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
112
|
Sheng X, Sun X, Li F, Wang J, Ma J. Linear growth failure induced by systemic inflammation inhibiting IGF-1/IGFBP axis in rats with asymptomatic colitis. BMC Gastroenterol 2019; 19:96. [PMID: 31221091 PMCID: PMC6585116 DOI: 10.1186/s12876-019-1023-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 06/13/2019] [Indexed: 01/17/2023] Open
Abstract
Background Children in poor areas show significant growth retardation that does not improve with an adequate supply of energy and nutrients, which may be related to asymptomatic intestinal infection caused by poor sanitation. Our objective was to explore the mechanism of intestinal inflammation inhibiting growth in the setting of asymptomatic colitis. Methods Forty-eight 3-week-old Wistar rats were randomly divided into three groups: the control group, colitis group (with asymptomatic colitis induced by 2.5% trinitrobenzenesulphonic acid) and pair-fed group (daily food intake matched to the pair in the colitis group). The linear growth was assessed, and the plasma levels of hormone and systemic cytokines were detected and compared by independent two-sample t-test or one-way ANOVA among groups. Results At d5, the increases in the body length of the control, colitis and pair-fed groups were 1.65 ± 0.34 cm, 1.10 ± 0.30 cm and 1.38 ± 0.26 cm, respectively, and the increase in the body length in the colitis group was significantly less than that in the control group (P < 0.05). There were significant differences in the levels of hormone and cytokines among three groups (P < 0.05). Compared with the control group, rats in the colitis group exhibited linear growth failure, as well as higher expression of calprotectin, tumour necrosis factor-α, interleukin-6 and insulin-like growth factor binding protein 2, lower insulin-like growth factor-1 and insulin-like growth factor binding protein 3, and lower expression of nuclear factor kappa B in hepatocytes. Conclusions In addition to undernutrition, the systemic inflammatory response caused by asymptomatic colitis may inhibit the linear growth of rats by its influence on the insulin-like growth factor/insulin-like growth factor binding protein axis. Electronic supplementary material The online version of this article (10.1186/s12876-019-1023-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoyang Sheng
- Department of Children and Adolescents Health Care, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute for Pediatric Research, MOE-Shanghai Key Laboratory of Children's Environmental Health, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Xueqing Sun
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 S. South Chongqing Road, Shanghai, 200025, China
| | - Feng Li
- Department of Children and Adolescents Health Care, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute for Pediatric Research, MOE-Shanghai Key Laboratory of Children's Environmental Health, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Junli Wang
- Department of Children and Adolescents Health Care, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute for Pediatric Research, MOE-Shanghai Key Laboratory of Children's Environmental Health, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Jingqiu Ma
- Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
113
|
Konyole SO, Omollo SA, Kinyuru JN, Skau JKH, Owuor BO, Estambale BB, Filteau SM, Michaelsen KF, Friis H, Roos N, Owino VO. Effect of locally produced complementary foods on fat-free mass, linear growth, and iron status among Kenyan infants: A randomized controlled trial. MATERNAL AND CHILD NUTRITION 2019; 15:e12836. [PMID: 31045329 DOI: 10.1111/mcn.12836] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/14/2019] [Accepted: 04/24/2019] [Indexed: 12/14/2022]
Abstract
The impact of quality complementary food products on infant growth and body composition has not been adequately investigated. This study evaluated the effect on fat-free mass (FFM) accrual, linear growth, and iron status of locally produced complementary food products comparing to a standard product. In a randomized, double-blind trial, 499 infants at 6 months received nine monthly rations of (a) WinFood Classic (WFC) comprising germinated amaranth (71%), maize (10.4%), small fish (3%), and edible termites (10%); (b) WinFood Lite (WFL) comprising germinated amaranth (82.5%), maize (10.2%), and multimicronutrient premix; or (c) fortified corn-soy blend plus (CSB+). Primary outcomes were changes in FFM, length, and plasma ferritin and transferrin receptors (TfR). FFM was determined using deuterium dilution. Analysis was by intention to treat, based on available cases. Compared with CSB+, there were no differences in change from 6 to 15 months in FFM for WFC 0.0 kg (95% CI [-0.30, 0.29]) and WFL 0.03 kg (95% CI [-0.25, 0.32]) and length change for WFC -0.3 cm (95% CI [-0.9, 0.4]) and WFL -0.3 cm (95% CI [-0.9, 0.3]). TfR increased in WFC group 3.3 mg L-1 (95% CI [1.7, 4.9]) and WFL group 1.7 mg L-1 (95% CI [0.1, 3.4]) compared with CSB+. Compared with the increase in Hb in CSB+ group, there was a reduction in Hb in WFC of -0.9 g dl-1 (95% CI [-1.3, -0.5]) and a lower increase in WFL -0.4 g dl-1 (95% CI [-0.8, 0.0]). In conclusion, the tested WinFoods had the same effect on FFM and length as CSB+, whereas Hb and iron status decreased, suggesting inhibited iron bioavailability from the amaranth-based WinFoods.
Collapse
Affiliation(s)
- Silvenus O Konyole
- Department of Nutritional Sciences, Masinde Muliro University of Science and Technology, Kakamega, Kenya
| | - Selina A Omollo
- Institute of Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
| | - John N Kinyuru
- Department of Food Science and Technology, Jomo Kenyatta University of Agriculture and Technology, Juja, Kenya
| | - Jutta K H Skau
- Department of Paediatrics, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Benson B Estambale
- Division of Research, Innovations and Outreach, Jaramogi Oginga Odinga University of Science and Technology, Bondo, Kenya
| | - Suzanne M Filteau
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Kim F Michaelsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| | - Henrik Friis
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| | - Nanna Roos
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| | - Victor O Owino
- Nutritional and Health Related Environmental Studies Section, International Atomic Energy Agency, Vienna, Austria
| |
Collapse
|
114
|
Mondal P, Long JM, Westcott JE, Islam MM, Ahmed M, Mahfuz M, Ahmed T, Miller LV, Krebs NF. Zinc Absorption and Endogenous Fecal Zinc Losses in Bangladeshi Toddlers at Risk for Environmental Enteric Dysfunction. J Pediatr Gastroenterol Nutr 2019; 68:874-879. [PMID: 31033623 PMCID: PMC6553983 DOI: 10.1097/mpg.0000000000002361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/21/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Environmental enteric dysfunction (EED) impairs zinc absorption from food, and zinc deficiency may contribute to the poor growth associated with EED. We examined zinc absorption from a standardized aqueous zinc dose, and habitual daily endogenous fecal zinc excretion (EFZ) and compared these outcomes between children grouped by the lactulose to mannitol ratio (L:M). METHODS Bangladeshi toddlers (18-24 months) with low (<0.09) and high (≥0.09) L:M were administered isotope-labeled 3 mg aqueous zinc in the fasted state. Fractional absorption of zinc (FAZ) and EFZ were measured by dual stable isotope tracer method and an isotope dilution method, respectively. Secondary aims included examining relationships of biomarkers of systemic and intestinal inflammation and gut function with FAZ and EFZ. RESULTS Forty children completed the study; nearly all had evidence of EED. No differences in zinc homeostasis measurements (mean ± SD) were observed between high and low L:M groups: FAZ was 0.38 ± 0.19 and 0.31 ± 0.19, respectively; both figures were within estimated reference range. Means of EFZ were 0.73 ± 0.27 and 0.76 ± 0.20 mg/day for high and low L:M, respectively, and were 10% to 15% above estimated reference range. Regression analyses indicated that biomarkers of systemic inflammation were directly associated with increasing FAZ, consistent with increased gut permeability. Biomarkers of intestinal inflammation were negatively associated with EFZ, consistent with low-zinc intake and chronic deficiency. CONCLUSIONS In these children at risk of EED, endogenous zinc losses were not markedly increased. Results suggest that efforts to improve zinc status in EED should focus on substantially improving zinc intakes.
Collapse
Affiliation(s)
- Prasenjit Mondal
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Julie M. Long
- University of Colorado School of Medicine, Department of Pediatrics, Section of Nutrition, Aurora, CO
| | - Jamie E. Westcott
- University of Colorado School of Medicine, Department of Pediatrics, Section of Nutrition, Aurora, CO
| | - M. Munirul Islam
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mondar Ahmed
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Leland V. Miller
- University of Colorado School of Medicine, Department of Pediatrics, Section of Nutrition, Aurora, CO
| | - Nancy F. Krebs
- University of Colorado School of Medicine, Department of Pediatrics, Section of Nutrition, Aurora, CO
| |
Collapse
|
115
|
Titaley CR, Ariawan I, Hapsari D, Muasyaroh A, Dibley MJ. Determinants of the Stunting of Children Under Two Years Old in Indonesia: A Multilevel Analysis of the 2013 Indonesia Basic Health Survey. Nutrients 2019; 11:E1106. [PMID: 31109058 PMCID: PMC6567198 DOI: 10.3390/nu11051106] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 11/16/2022] Open
Abstract
Indonesia is ranked fifth among countries with the highest burden of stunting in children under five. This study aims to examine the determinants of stunting in children aged 0-2 years in Indonesia using data derived from the 2013 Indonesia Basic Health Survey. Twenty potential predictors of stunting, categorized into household and housing characteristics; maternal and paternal characteristics; antenatal care services and child characteristics were analyzed. Multilevel analyses were performed to examine the role of cluster/district/provincial differences, as well as individual/household level characteristics and stunting status. Of 24,657 children analyzed, 33.7% (95%CI: 32.8%-34.7%) were stunted. The odds of stunting increased significantly among children living in households with three or more children under five-years-old (aOR = 1.33, 95%CI: 1.03-1.72), households with five to seven household members (aOR =1.11; 95%CI: 1.03-1.20), children whose mothers during pregnancy attended less than four antenatal care services (aOR = 1.22, 95%CI: 1.08-1.39), boys (aOR = 1.33, 95%CI: 1.22-1.45), children aged 12-23 months (aOR = 1.89; 95%CI: 1.54-2.32), and children who weighed <2500 g at birth (aOR = 2.55; 95%CI: 2.05-3.15). The odds also increased significantly with the reduction of household wealth index. Integrated interventions to address environment, an individual level associated with stunting in Indonesia, from the environment- to individual-level factors are important.
Collapse
Affiliation(s)
- Christiana R Titaley
- Faculty of Medicine, Pattimura University, Kampus Poka, Maluku Province, Ambon 97233, Indonesia.
| | - Iwan Ariawan
- Center for Health Research, Faculty of Public Health Universitas Indonesia, West Java Province, Depok 16424, Indonesia.
| | - Dwi Hapsari
- National Institute of Health Research and Development, Ministry of Health Republic of Indonesia, DKI Jakarta 10560, Indonesia.
| | - Anifatun Muasyaroh
- Center for Health Research, Faculty of Public Health Universitas Indonesia, West Java Province, Depok 16424, Indonesia.
| | - Michael J Dibley
- School of Public Health, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
116
|
Abstract
In 2017, an estimated 1 in every 4 (23%) children aged < 5 years were stunted worldwide. With slow progress in stunting reduction in many regions and the realization that a large proportion of stunting is not due to insufficient diet or diarrhea alone, it remains that other factors must explain continued growth faltering. Environmental enteric dysfunction (EED), a subclinical state of intestinal inflammation, can occur in infants across the developing world and is proposed as an immediate causal factor connecting poor sanitation and stunting. A result of chronic pathogen exposure, EED presents multiple causal pathways, and as such the scope and sensitivity of traditional water, sanitation, and hygiene (WASH) interventions have possibly been unsubstantial. Although the definite pathogenesis of EED and the mechanism by which stunting occurs are yet to be defined, this paper reviews the existing literature surrounding the proposed pathology and transmission of EED in infants and considerations for nutrition and WASH interventions to improve linear growth worldwide.
Collapse
Affiliation(s)
- Sophie Budge
- Cranfield Water Science Institute, Cranfield University, Bedfordshire, United Kingdom
| | - Alison H Parker
- Cranfield Water Science Institute, Cranfield University, Bedfordshire, United Kingdom
| | - Paul T Hutchings
- Cranfield Water Science Institute, Cranfield University, Bedfordshire, United Kingdom
| | | |
Collapse
|
117
|
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
|
118
|
Environmental enteric dysfunction and growth. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2019. [DOI: 10.1016/j.jpedp.2019.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
119
|
Ngari MM, Iversen PO, Thitiri J, Mwalekwa L, Timbwa M, Fegan GW, Berkley JA. Linear growth following complicated severe malnutrition: 1-year follow-up cohort of Kenyan children. Arch Dis Child 2019; 104:229-235. [PMID: 30266874 PMCID: PMC6556974 DOI: 10.1136/archdischild-2018-315641] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/03/2018] [Accepted: 09/05/2018] [Indexed: 12/04/2022]
Abstract
BACKGROUND Stunting is the most common manifestation of childhood undernutrition worldwide. Children presenting with severe acute malnutrition (SAM) are often also severely stunted. We evaluated linear growth and its determinants after medically complicated SAM. METHODS We performed secondary analysis of clinical trial data (NCT00934492) from HIV-uninfected Kenyan children aged 2-59 months hospitalised with SAM. Outcome was change in height/length-for-age z-score (HAZ) between enrolment and 12 months later. Exposures were demographic, clinical, anthropometric characteristics and illness episodes during follow-up. RESULTS Among 1169 children with HAZ values at month 12 (66% of those in original trial), median (IQR) age 11 (7-17) months and mean (SD) HAZ -2.87 (1.6) at enrolment, there was no change in mean HAZ between enrolment and month 12: -0.006Z (95% CI -0.07 to 0.05Z). While 262 (23%) children experienced minimal HAZ change (within ±0.25 HAZ), 472 (40%) lost >0.25 and 435 (37%) gained >0.25 HAZ. After adjusting for regression to the mean, inpatient or outpatient episodes of diarrhoea and inpatient severe pneumonia during follow-up were associated with HAZ loss. Premature birth and not being cared by the biological parent were associated with HAZ gain. Increases in mid-upper arm circumference and weight-for-age were associated with HAZ gain and protected against HAZ loss. Increase in weight-for-height was not associated with HAZ gain but protected against HAZ loss. No threshold of weight gain preceding linear catch-up growth was observed. CONCLUSIONS Interventions to improve dietary quality and prevent illness over a longer period may provide opportunities to improve linear growth.
Collapse
Affiliation(s)
- Moses M Ngari
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya,Childhood Acute Illness and Nutrition (CHAIN) Network, Nairobi, Kenya
| | - Per Ole Iversen
- Department of Nutrition, IBM, University of Oslo, Oslo, Norway,Department of Hematology, Oslo University Hospital, Oslo, Norway,Division of Human Nutrition, Stellenbosch University, Tygerberg, South Africa
| | - Johnstone Thitiri
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya,Childhood Acute Illness and Nutrition (CHAIN) Network, Nairobi, Kenya
| | | | - Molline Timbwa
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya,Childhood Acute Illness and Nutrition (CHAIN) Network, Nairobi, Kenya
| | - Greg W Fegan
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya,Swansea Trials Unit, Swansea University Medical School, Swansea, UK
| | - James Alexander Berkley
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya,Childhood Acute Illness and Nutrition (CHAIN) Network, Nairobi, Kenya,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| |
Collapse
|
120
|
Morais MBD, Silva GAPD. Environmental enteric dysfunction and growth. J Pediatr (Rio J) 2019; 95 Suppl 1:85-94. [PMID: 30629923 DOI: 10.1016/j.jped.2018.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/23/2018] [Accepted: 11/23/2018] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE To describe the current indicators of environmental enteric dysfunction and its association with linear growth deficit and the height-for-age anthropometric indicator. DATA SOURCES Narrative review with articles identified in PubMed and Scopus databases using combinations of the following words: environmental, enteric, dysfunction, enteropathy, and growth, as well as the authors' personal records. DATA SYNTHESIS In the last 15 years, new non-invasive markers have been investigated to characterize environmental enteric dysfunction; however, the best tests to be used have not yet been identified. There is evidence that, in environmental enteric dysfunction, a systemic inflammatory process may also occur as a consequence of increased intestinal permeability, in addition to intestinal mucosa abnormalities. Bacterial overgrowth in the small intestine and changes in fecal microbiota profile have also been identified. There is evidence indicating that environmental enteric dysfunction can impair not only full growth but also the neuropsychomotor development and response to orally administered vaccines. It is important to emphasize that the environmental enteric dysfunction is not a justification for not carrying out vaccination, which must follow the regular schedule. Another aspect to emphasize is the greater risk for those children who had height impairment in early childhood, possibly associated with environmental enteric dysfunction, to present overweight and obesity in adulthood when exposed to a high calorie diet, which has been called "triple burden." CONCLUSIONS According to the analyzed evidence, the control of environmental enteric dysfunction is very important for the full expression of growth, development, and vaccine response in the pediatric age group.
Collapse
Affiliation(s)
- Mauro Batista de Morais
- Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, Disciplina de Gastroenterologia Pediátrica, São Paulo, SP, Brazil.
| | | |
Collapse
|
121
|
Lambrecht NJ, Wilson ML, Jones AD. Assessing the Impact of Animal Husbandry and Capture on Anemia among Women and Children in Low- and Middle-Income Countries: A Systematic Review. Adv Nutr 2019; 10:331-344. [PMID: 30854553 PMCID: PMC6416043 DOI: 10.1093/advances/nmy080] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Animal husbandry and capture (AHC) may mitigate anemia among women and children by supplying a source of micronutrient-rich animal source foods (ASF), yet may concurrently increase exposure to anemia-inducing pathogens such as Plasmodium spp., helminths, and enteropathogens. We conducted a systematic literature review to assess the relation between AHC and anemia among women of reproductive age, school-aged children, and children aged <5 y in low- and middle-income countries (LMICs). We used a 2-stage screening process, in which 1 reviewer searched 4 databases (PubMed, Web of Science, EMBASE, and Global Health) with predetermined search terms for relevant articles. Two reviewers then independently screened studies using a priori exclusion criteria, yielding a total of 23 articles included in the final review. We evaluated evidence from observational studies assessing animal-dependent livelihoods and livestock ownership, and interventions that promoted livestock and fish production. We found little consistency in anemia outcomes across the several AHC exposures and population groups. Poultry production interventions had modest benefits on anemia among women and children, although whether these improvements were a result of increased ASF consumption, or a result of the combined treatment study design could not be determined. Observational studies identified chicken ownership, and no other livestock species, as a risk factor for anemia among young children. However, there was limited evidence to evaluate pathways underlying these associations. Studies tended to rely on self-reported fever and diarrhea to assess illness, and no study directly assessed linkages between AHC, pathogen burden, and anemia. Thus, there is insufficient evidence to conclude whether AHC improves or worsens anemia among women and children in LMICs. Given the current interest in promoting animal production among low-income households, future studies with robust measures of livestock ownership, ASF consumption, pathogen burden, and anemia status are needed to understand the nuances of this complex and potentially contradictory relation.
Collapse
Affiliation(s)
| | - Mark L Wilson
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Andrew D Jones
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI,Address correspondence to ADJ (e-mail: )
| |
Collapse
|
122
|
Kaimila Y, Divala O, Agapova SE, Stephenson KB, Thakwalakwa C, Trehan I, Manary MJ, Maleta KM. Consumption of Animal-Source Protein is Associated with Improved Height-for-Age z Scores in Rural Malawian Children Aged 12⁻36 Months. Nutrients 2019; 11:E480. [PMID: 30823563 PMCID: PMC6413013 DOI: 10.3390/nu11020480] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 01/28/2023] Open
Abstract
Linear growth faltering, caused by insufficient diet, recurrent infections and environmental enteric dysfunction (EED), continues to plague young children in low- and middle-income countries (LMICs). Diets in LMICs are primarily plant based, and thus have poor-quality protein and low levels of essential micronutrients. The aim of this study was to assess the association of the type and protein quality of food consumed with stunting, EED and acute malnutrition in children aged 6⁻36 months in Limera and Masenjere, two rural Southern Malawian communities. This is a secondary analysis of two randomized controlled trials that tested the effects of common bean and cowpea flour on stunting in children aged 6⁻36 months. We used data from two interactive 24-h dietary recalls conducted 12 weeks after enrolment into each trial. Food intakes were compared between the regions using Chi-square and Student's t-test. There were 355 children that participated in the dietary recalls. The diets of children were of poor quality, but the children from Limera consumed more fish (54% vs. 35%, p = 0.009) and more bioavailable protein (26.0 ± 10.3 g/day vs. 23.1 ± 8.1 g/day, p = 0.018, respectively) than children in Masenjere. Food type and protein quality were not associated with any of the outcomes except an association between animal protein consumption and improvement in height-for-age z scores in children aged 12⁻36 months (p = 0.047). These findings support the notion that animal-source food (ASF) consumption in this vulnerable population promotes linear growth.
Collapse
Affiliation(s)
- Yankho Kaimila
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Private Bag 360, Blantyre 3, Malawi.
| | - Oscar Divala
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Private Bag 360, Blantyre 3, Malawi.
| | - Sophia E Agapova
- Washington University School of Medicine, Department of Pediatrics, Campus Box 8116, St. Louis, MO 63110, USA.
| | - Kevin B Stephenson
- Washington University School of Medicine, Department of Pediatrics, Campus Box 8116, St. Louis, MO 63110, USA.
| | - Chrissie Thakwalakwa
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Private Bag 360, Blantyre 3, Malawi.
| | - Indi Trehan
- Washington University School of Medicine, Department of Pediatrics, Campus Box 8116, St. Louis, MO 63110, USA.
- Lao Friends Hospital for Children, P.O. Box 873, Luang Prabang 06000, Lao PDR.
| | - Mark J Manary
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Private Bag 360, Blantyre 3, Malawi.
- Washington University School of Medicine, Department of Pediatrics, Campus Box 8116, St. Louis, MO 63110, USA.
| | - Kenneth M Maleta
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Private Bag 360, Blantyre 3, Malawi.
| |
Collapse
|
123
|
Allen SJ, Adepojou A, Akinyinka OO. Challenges and opportunities for paediatric gastroenterology in low- and middle-income countries: high time for action. Paediatr Int Child Health 2019; 39:4-6. [PMID: 30900525 DOI: 10.1080/20469047.2019.1568022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Stephen J Allen
- a Department of Clinical Sciences , Liverpool School of Tropical Medicine , Liverpool , UK.,b Department of Paediatric Gastroenterology , Alder Hey Children's NHS Foundation Trust , Liverpool , UK
| | - Akinlolu Adepojou
- b Department of Paediatric Gastroenterology , Alder Hey Children's NHS Foundation Trust , Liverpool , UK.,c Department of Paediatrics , University College Hospital; College Of Medicine, University of Ibadan , Ibadan , Nigeria
| | - Olusegun O Akinyinka
- d University of Ibadan , Ibadan , Nigeria.,e University College Hospital , Ibadan , Nigeria
| |
Collapse
|
124
|
Kartini A, Subagio HW, Hadisaputro S, Kartasurya MI, Suhartono S, Budiyono B. Pesticide Exposure and Stunting among Children in Agricultural Areas. THE INTERNATIONAL JOURNAL OF OCCUPATIONAL AND ENVIRONMENTAL MEDICINE 2019; 10:17-29. [PMID: 30685774 PMCID: PMC6522210 DOI: 10.15171/ijoem.2019.1428] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 12/29/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND The prevalence of growth disorders among school-aged children in Indonesia is high (30.7%). Pesticides have been massively used in Indonesian agricultural areas. OBJECTIVE To determine if exposure to pesticides is associated with stunting among children in agricultural areas. METHODS This case-control study included 160 children (48 cases and 112 controls) aged 8-12 years. Exposure to pesticides was measured based on the history of the exposure since perinatal period, infancy, and childhood of the participants. Stunting was determined as a height for age z-score (HAZ) < ‑2 SD. Other variables measured were levels of thyroid stimulating hormone (TSH), insulin-like growth factor-1 (IGF-1), hemoglobin, zinc, albumin, nutrient adequacy level (energy and protein), and history of infection, low-birth weight (LBW), and mother's height. RESULTS There were no significant difference between the cases and controls in terms of in the baseline characteristics, except for the median IGF-1 level; it was significantly (p<0.001) lower in the cases (66.73 ng/mL) than the controls (112.57 ng/mL). High level of pesticide exposure (p=0.029) and low IGF-1 levels (p<0.001) were significantly associated with stunting. After adjusting for confounding variables, these variables were found to be independent risk factors for stunting in children (aOR 3.90, 95% CI 1.15 to 13.26; and aOR 8.35, 95% CI 3.65 to 19.14, respectively). CONCLUSION Pesticide exposure could be a risk factor for the occurrence of growth disorders in children living in agricultural areas. Necessary actions should be taken to protect children living in agricultural areas from exposure to pesticides.
Collapse
Affiliation(s)
- Apoina Kartini
- Department of Public Health Nutrition, Faculty of Public Health, Diponegoro University, Semarang, Indonesia
| | - Hertanto W Subagio
- Department of Nutrition, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Suharyo Hadisaputro
- Department of Internal Medicine, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Martha I Kartasurya
- Department of Public Health Nutrition, Faculty of Public Health, Diponegoro University, Semarang, Indonesia
| | - Suhartono Suhartono
- Department of Environmental Health, Faculty of Public Health, Diponegoro University, Semarang, Indonesia.
| | - Budiyono Budiyono
- Department of Environmental Health, Faculty of Public Health, Diponegoro University, Semarang, Indonesia
| |
Collapse
|
125
|
Freedman SB, Williamson-Urquhart S, Farion KJ, Gouin S, Willan AR, Poonai N, Hurley K, Sherman PM, Finkelstein Y, Lee BE, Pang XL, Chui L, Schnadower D, Xie J, Gorelick M, Schuh S. Multicenter Trial of a Combination Probiotic for Children with Gastroenteritis. N Engl J Med 2018; 379:2015-2026. [PMID: 30462939 DOI: 10.1056/nejmoa1802597] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Gastroenteritis accounts for approximately 1.7 million visits to the emergency department (ED) by children in the United States every year. Data to determine whether the use of probiotics improves outcomes in these children are lacking. METHODS We conducted a randomized, double-blind trial involving 886 children 3 to 48 months of age with gastroenteritis who presented to six pediatric EDs in Canada. Participants received a 5-day course of a combination probiotic product containing Lactobacillus rhamnosus R0011 and L. helveticus R0052, at a dose of 4.0×109 colony-forming units twice daily or placebo. The primary outcome was moderate-to-severe gastroenteritis, which was defined according to a post-enrollment modified Vesikari scale symptom score of 9 or higher (scores range from 0 to 20, with higher scores indicating more severe disease). Secondary outcomes included the duration of diarrhea and vomiting, the percentage of children who had unscheduled physician visits, and the presence or absence of adverse events. RESULTS Moderate-to-severe gastroenteritis within 14 days after enrollment occurred in 108 of 414 participants (26.1%) who were assigned to probiotics and 102 of 413 participants (24.7%) who were assigned to placebo (odds ratio, 1.06; 95% confidence interval [CI], 0.77 to 1.46; P=0.72). After adjustment for trial site, age, detection of rotavirus in stool, and frequency of diarrhea and vomiting before enrollment, trial-group assignment did not predict moderate-to-severe gastroenteritis (odds ratio, 1.06; 95% CI, 0.76 to 1.49; P=0.74). There were no significant differences between the probiotic group and the placebo group in the median duration of diarrhea (52.5 hours [interquartile range, 18.3 to 95.8] and 55.5 hours [interquartile range, 20.2 to 102.3], respectively; P=0.31) or vomiting (17.7 hours [interquartile range, 0 to 58.6] and 18.7 hours [interquartile range, 0 to 51.6], P=0.18), the percentages of participants with unscheduled visits to a health care provider (30.2% and 26.6%; odds ratio, 1.19; 95% CI, 0.87 to 1.62; P=0.27), and the percentage of participants who reported an adverse event (34.8% and 38.7%; odds ratio, 0.83; 95% CI, 0.62 to 1.11; P=0.21). CONCLUSIONS In children who presented to the emergency department with gastroenteritis, twice-daily administration of a combined L. rhamnosus-L. helveticus probiotic did not prevent the development of moderate-to-severe gastroenteritis within 14 days after enrollment. (Funded by the Canadian Institutes of Health Research and others; PROGUT ClinicalTrials.gov number, NCT01853124 .).
Collapse
Affiliation(s)
- Stephen B Freedman
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Sarah Williamson-Urquhart
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Ken J Farion
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Serge Gouin
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Andrew R Willan
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Naveen Poonai
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Katrina Hurley
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Philip M Sherman
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Yaron Finkelstein
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Bonita E Lee
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Xiao-Li Pang
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Linda Chui
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - David Schnadower
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Jianling Xie
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Marc Gorelick
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| | - Suzanne Schuh
- From the Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute (S.B.F.), and the Section of Pediatric Emergency Medicine, Alberta Children's Hospital (S.W.-U., J.X.), Cumming School of Medicine, University of Calgary, Calgary, the Departments of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa (K.J.F.), the Division of Pediatric Emergency Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal (S.G.), Ontario Child Health Support Unit, SickKids Research Institute, the Division of Biostatistics, Dalla Lana School of Public Health (A.R.W.), the Division of Gastroenterology, Hepatology, and Nutrition (P.M.S.), the Divisions of Pediatric Emergency Medicine and Clinical Pharmacology and Toxicology (Y.F.), and the Division of Pediatric Emergency Medicine and Research Institute (S.S.), Hospital for Sick Children, University of Toronto, Toronto, the Division of Emergency Medicine, London Health Sciences Centre, Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON (N.P.), the Division of Pediatric Emergency Medicine, IWK Health Centre, Halifax, NS (K.H.), the Department of Pediatrics, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute (B.E.L.) and the Provincial Laboratory for Public Health-Alberta Public Laboratories and Department of Laboratory Medicine and Pathology (X.-L.P., L.C.), University of Alberta, Edmonton - all in Canada; the Division of Pediatric Emergency Medicine, Washington University School of Medicine, St. Louis (D.S.); and Children's Minnesota and Department of Pediatrics, University of Minnesota Medical School, Minneapolis (M.G.)
| |
Collapse
|
126
|
Knee J, Sumner T, Adriano Z, Berendes D, de Bruijn E, Schmidt WP, Nalá R, Cumming O, Brown J. Risk factors for childhood enteric infection in urban Maputo, Mozambique: A cross-sectional study. PLoS Negl Trop Dis 2018; 12:e0006956. [PMID: 30419034 PMCID: PMC6258421 DOI: 10.1371/journal.pntd.0006956] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/26/2018] [Accepted: 10/29/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Enteric infections are common where public health infrastructure is lacking. This study assesses risk factors for a range of enteric infections among children living in low-income, unplanned communities of urban Maputo, Mozambique. METHODS & FINDINGS We conducted a cross-sectional survey in 17 neighborhoods of Maputo to assess the prevalence of reported diarrheal illness and laboratory-confirmed enteric infections in children. We collected stool from children aged 1-48 months, independent of reported symptoms, for molecular detection of 15 common enteric pathogens by multiplex RT-PCR. We also collected survey and observational data related to water, sanitation, and hygiene (WASH) characteristics; other environmental factors; and social, economic, and demographic covariates. We analyzed stool from 759 children living in 425 household clusters (compounds) representing a range of environmental conditions. We detected ≥1 enteric pathogens in stool from most children (86%, 95% confidence interval (CI): 84-89%) though diarrheal symptoms were only reported for 16% (95% CI: 13-19%) of children with enteric infections and 13% (95% CI: 11-15%) of all children. Prevalence of any enteric infection was positively associated with age and ranged from 71% (95% CI: 64-77%) in children 1-11 months to 96% (95% CI: 93-98%) in children 24-48 months. We found poor sanitary conditions, such as presence of feces or soiled diapers around the compound, to be associated with higher risk of protozoan infections. Certain latrine features, including drop-hole covers and latrine walls, and presence of a water tap on the compound grounds were associated with a lower risk of bacterial and protozoan infections. Any breastfeeding was also associated with reduced risk of infection. CONCLUSIONS We found a high prevalence of enteric infections, primarily among children without diarrhea, and weak associations between bacterial and protozoan infections and environmental risk factors including WASH. Findings suggest that environmental health interventions to limit infections would need to be transformative given the high prevalence of enteric pathogen shedding and poor sanitary conditions observed. TRIAL REGISTRATION ClinicalTrials.gov NCT02362932.
Collapse
Affiliation(s)
- Jackie Knee
- School of Civil and Environmental Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Trent Sumner
- School of Civil and Environmental Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Zaida Adriano
- We Consult, Maputo, Mozambique
- Departamento de Geografia, Universidade Eduardo Mondlane, Maputo, Mozambique
| | - David Berendes
- Waterborne Disease Prevention Branch, Division of Foodborne, Waterborne, and Environmental Diseases, National Center for Emerging Zoonotic and Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | | | - Wolf-Peter Schmidt
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Rassul Nalá
- Ministério da Saúde, Instituto Nacional de Saúde Maputo, Maputo, Republic of Mozambique
| | - Oliver Cumming
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Joe Brown
- School of Civil and Environmental Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
127
|
Justine M, Yeconia A, Nicodemu I, Augustino D, Gratz J, Mduma E, Heysell SK, Kivuyo S, Mfinanga S, Peloquin CA, Zagurski T, Kibiki GS, Mmbaga B, Houpt ER, Thomas TA. Pharmacokinetics of First-Line Drugs Among Children With Tuberculosis in Rural Tanzania. J Pediatric Infect Dis Soc 2018; 9:14-20. [PMID: 30395239 PMCID: PMC7317157 DOI: 10.1093/jpids/piy106] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 10/17/2018] [Indexed: 11/13/2022]
Abstract
BACKGROUND Dosing recommendations for treating childhood tuberculosis (TB) were revised by the World Health Organization, yet so far, pharmacokinetic studies that have evaluated these changes are relatively limited. We evaluated plasma drug concentrations of rifampicin (RIF), isoniazid (INH), pyrazinamide (PZA), and ethambutol (EMB) among children undergoing TB treatment in Tanzania when these dosing recommendations were being implemented. METHODS At the end of intensive-phase TB therapy, blood was obtained 2 hours after witnessed medication administration to estimate the peak drug concentration (C2h), measured using high-performance liquid chromatography or liquid chromatography-tandem mass spectrometry methods. Differences in median drug concentrations were compared on the basis of the weight-based dosing strategy using the Mann-Whitney U test. Risk factors for low drug concentrations were analyzed using multivariate regression analysis. RESULTS We enrolled 51 human immunodeficiency virus-negative children (median age, 5.3 years [range, 0.75-14 years]). The median C2hs were below the target range for each TB drug studied. Compared with children who received the "old" dosages, those who received the "revised" WHO dosages had a higher median C2h for RIF (P = .049) and PZA (P = .015) but not for INH (P = .624) or EMB (P = .143); however, these revised dosages did not result in the target range for RIF, INH, and EMB being achieved. A low starting dose was associated with a low C2h for RIF (P = .005) and PZA (P = .005). Malnutrition was associated with a low C2h for RIF (P = .001) and INH (P = .001). CONCLUSIONS Among this cohort of human immunodeficiency virus-negative Tanzanian children, use of the revised dosing strategy for treating childhood TB did not result in the target drug concentration for RIF, INH, or EMB being reached.
Collapse
Affiliation(s)
- Museveni Justine
- Center for Global Health Research, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Anita Yeconia
- Center for Global Health Research, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Ingi Nicodemu
- Center for Global Health Research, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Domitila Augustino
- Center for Global Health Research, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Jean Gratz
- Center for Global Health Research, Haydom Lutheran Hospital, Haydom, Tanzania,Division of Infectious Diseases and International Health, University of Virginia, Charlottesville
| | - Estomih Mduma
- Center for Global Health Research, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Scott K Heysell
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville
| | - Sokoine Kivuyo
- National Institute of Medical Research Muhimbili, Dar es Salaam, Tanzania
| | - Sayoki Mfinanga
- National Institute of Medical Research Muhimbili, Dar es Salaam, Tanzania
| | - Charles A Peloquin
- Infectious Disease Pharmacokinetic Laboratory, University of Florida, Gainesville
| | - Theodore Zagurski
- Infectious Disease Pharmacokinetic Laboratory, University of Florida, Gainesville
| | - Gibson S Kibiki
- East African Health Research Commission, East African Community, Arusha, Tanzania
| | - Blandina Mmbaga
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical College, Moshi, Tanzania
| | - Eric R Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville
| | - Tania A Thomas
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville,Correspondence: T. A. Thomas, MD, MPH, University of Virginia, Division of Infectious Diseases and International Health, PO Box 801340, Charlottesville, VA 22908 ()
| |
Collapse
|
128
|
Affiliation(s)
- Michael Freemark
- From the Division of Pediatric Endocrinology and Diabetes, Duke University Medical Center, Durham, NC
| |
Collapse
|
129
|
Nutrient intake and environmental enteric dysfunction among Nepalese children 9-24 months old-the MAL-ED birth cohort study. Pediatr Res 2018; 84:509-515. [PMID: 30030503 DOI: 10.1038/s41390-018-0108-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/04/2018] [Accepted: 06/23/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Nutrient deficiencies limit the growth and turnover of intestinal mucosa, but studies assessing whether specific nutrients protect against or improve environmental enteric dysfunction (EED) are scarce. We aimed to investigate associations between nutrient intake and EED assessed by lactulose:mannitol (L:M) ratio, anti-1-antitrypsin, myeloperoxidase (MPO), and neopterin (NEO) among children 9-24 months in Bhaktapur, Nepal. METHODS Among 231 included children, nutrient intake was assessed monthly by 24 h recalls, and 3-month usual intake was estimated using Multiple Source Method. Associations between nutrient intake and L:M ratio (measured at 15 months) were assessed using multiple linear regression, while associations between nutrient intake and fecal markers (measured quarterly) were assessed using Generalized Estimating Equations (GEE) models. RESULTS We found that associations between nutrient intake from complementary food and L:M ratio, alpha-1-antitrypsin (AAT), MPO and NEO were generally negative but weak. The only significant associations between nutrient intake (potassium, magnesium, phosphorous, folate, and vitamin C) and markers for intestinal inflammation were found for MPO. CONCLUSION Negative but weak associations between nutrient intake and markers of intestinal inflammation were found. Significant associations between several nutrients and MPO might merit further investigation.
Collapse
|
130
|
Fahim SM, Das S, Gazi MA, Mahfuz M, Ahmed T. Association of intestinal pathogens with faecal markers of environmental enteric dysfunction among slum-dwelling children in the first 2 years of life in Bangladesh. Trop Med Int Health 2018; 23:1242-1250. [PMID: 30133067 PMCID: PMC6282798 DOI: 10.1111/tmi.13141] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Environmental Enteric Dysfunction (EED) can be assessed by faecal biomarkers such as Myeloperoxidase (MPO), Neopterin (NEO) and Alpha-1 anti-trypsin (AAT). We aimed to test the association of intestinal pathogens with faecal markers of EED among slum-dwelling children in first 2 years of life. METHODS The MAL-ED birth cohort data of Bangladesh site were used to conduct this analysis. Multivariable analyses using Generalized Estimating Equations (GEE) were performed to test the association between intestinal pathogens and faecal markers of EED. RESULTS Giardiasis, ascariasis and trichuriasis were the most frequent parasitic infections and Campylobacter spp., Enteroaggregative Escherichia coli (EAEC) and Enterotoxigenic Escherichia coli (ETEC) were the common bacterial pathogens observed in stool samples of the children. Overall, 71%, 97% and 58% of stool samples were above values considered normal in non-tropical settings for MPO, NEO and AAT respectively. Giardiasis was found to be significantly associated with MPO (Coefficient = 0.55; 95% CI = 0.15, 0.95; P-value = 0.008) and AAT concentrations (Coefficient = 0.34; 95% CI = 0.04, 0.63; P-value = 0.03). A significant association was found between trichuriasis and NEO (Coefficient = 0.90; 95% CI = 0.19, 1.61; P-value = 0.01). Trichuriasis (Coefficient = 1.71; 95% CI = 0.32, 3.11; P-value = 0.02) and giardiasis (Coefficient = 1.51; 95% CI = 0.79, 2.23; P-value <0.001) were significantly associated with EED score. Children with EAEC had significantly higher MPO concentrations (Coefficient = 0.33; 95% CI = 0.06, 0.61; P-value = 0.02). CONCLUSION The study results imply the importance of intestinal pathogens in contributing to intestinal inflammation and increased intestinal permeability in young children.
Collapse
Affiliation(s)
| | - Subhasish Das
- Nutrition and Clinical Services Division, icddr,b, Dhaka, Bangladesh
| | - Md Amran Gazi
- Nutrition and Clinical Services Division, icddr,b, Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, icddr,b, Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, icddr,b, Dhaka, Bangladesh
| |
Collapse
|
131
|
Marie C, Ali A, Chandwe K, Petri WA, Kelly P. Pathophysiology of environmental enteric dysfunction and its impact on oral vaccine efficacy. Mucosal Immunol 2018; 11:1290-1298. [PMID: 29988114 DOI: 10.1038/s41385-018-0036-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/03/2018] [Accepted: 04/11/2018] [Indexed: 02/08/2023]
Abstract
Environmental enteric dysfunction (EED) refers to a subclinical disorder of intestinal function common in tropical countries and in settings of poverty and economic disadvantage. The enteropathy that underlies this syndrome is characterized by mucosal inflammation and villus blunting mediated by T cell activation. Epithelial cell disruption and microbial translocation drive systemic inflammation. EED in young children is associated geographically with growth failure, malnutrition, and greatly impaired responses to oral vaccines, notably rotavirus and poliovirus vaccines. In this review, we describe the pathophysiology of EED and examine the evidence linking EED and oral vaccine failure. This evidence is far from conclusive. Although our understanding of EED is still sketchy, there is limited evidence of disturbed innate immunity, B cell disturbances including aggregation into lymphoid follicles, and autoantibody generation. Pathways of T cell activation and the possibility of dendritic cell anergy, which could help explain oral vaccine failure, require further work.
Collapse
Affiliation(s)
- Chelsea Marie
- The University of Virginia, Charlottesville, VA, USA
| | - Asad Ali
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Kanta Chandwe
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine, Lusaka, Zambia
| | | | - Paul Kelly
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine, Lusaka, Zambia. .,Barts & The London School of Medicine, Queen Mary University of London, London, E1 4AT, UK.
| |
Collapse
|
132
|
Mello CS, Rodrigues MSDC, Filho HBDA, Melli LCFL, Tahan S, Pignatari ACC, de Morais MB. Fecal microbiota analysis of children with small intestinal bacterial overgrowth among residents of an urban slum in Brazil. J Pediatr (Rio J) 2018; 94:483-490. [PMID: 29049893 DOI: 10.1016/j.jped.2017.09.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 09/08/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To analyze the fecal microbiota composition of children living in an urban slum in Brazil, with or without small intestinal bacterial overgrowth, and to investigate the occurrence of stunting and anemia. METHODS A total of 100 children were studied, aged 5-11 years, from the municipality of Osasco, São Paulo. Small intestinal bacterial overgrowth was screened through hydrogen and methane breath test with lactulose. Weight and height were measured, and the height-for-age and body mass-for-age anthropometric indexes were calculated. The occurrence of anemia was investigated by capillary hemoglobin. Analysis of bacterial phylum, genus, and species was performed by real-time polymerase chain reaction in fecal samples. RESULTS Small intestinal bacterial overgrowth was identified in 61.0% of the children. A lower mean of height-for-age Z-score ([-0.48±0.90] vs. [-0.11±0.97]; p=0.027), as well as capillary hemoglobin ([12.61±1.03g/dL] vs. [13.44±1.19g/dL]; p<0.001) was demonstrated in children with SIBO when compared with children without small intestinal bacterial overgrowth. Children with small intestinal bacterial overgrowth presented a higher frequency of Salmonella spp., when compared to those without small intestinal bacterial overgrowth (37.7% vs. 10.3%; p=0.002). Higher counts of total Eubacteria (p=0.014) and Firmicutes (p=0.038) were observed in children without small intestinal bacterial overgrowth; however, a higher count of Salmonella (p=0.002) was found in children with small intestinal bacterial overgrowth. CONCLUSION Children who lived in a slum and were diagnosed with small intestinal bacterial overgrowth showed lower H/A Z-scores and hemoglobin levels. Furthermore, differences were observed in the fecal microbiota of children with small intestinal bacterial overgrowth, when compared to those without it; specifically, a higher frequency and count of Salmonella, and lower counts of Firmicutes and total Eubacteria.
Collapse
Affiliation(s)
- Carolina Santos Mello
- Universidade Federal de São Paulo (UNIFESP), Departamento de Pediatria, Disciplina de Gastroenterologia Pediátrica, São Paulo, SP, Brazil
| | - Mirian Silva do Carmo Rodrigues
- Universidade Federal de São Paulo (UNIFESP), Departamento de Pediatria, Disciplina de Gastroenterologia Pediátrica, São Paulo, SP, Brazil
| | - Humberto Bezerra de Araújo Filho
- Universidade Federal de São Paulo (UNIFESP), Departamento de Pediatria, Disciplina de Gastroenterologia Pediátrica, São Paulo, SP, Brazil
| | - Lígia Cristina Fonseca Lahoz Melli
- Universidade Federal de São Paulo (UNIFESP), Departamento de Pediatria, Disciplina de Gastroenterologia Pediátrica, São Paulo, SP, Brazil
| | - Soraia Tahan
- Universidade Federal de São Paulo (UNIFESP), Departamento de Pediatria, Disciplina de Gastroenterologia Pediátrica, São Paulo, SP, Brazil
| | - Antônio Carlos Campos Pignatari
- Universidade Federal de São Paulo (UNIFESP), Departamento de Pediatria, Disciplina de Gastroenterologia Pediátrica, São Paulo, SP, Brazil
| | - Mauro Batista de Morais
- Universidade Federal de São Paulo (UNIFESP), Departamento de Pediatria, Disciplina de Gastroenterologia Pediátrica, São Paulo, SP, Brazil.
| |
Collapse
|
133
|
Fecal microbiota analysis of children with small intestinal bacterial overgrowth among residents of an urban slum in Brazil. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2018. [DOI: 10.1016/j.jpedp.2017.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
134
|
Environmental Enteropathy, Micronutrient Adequacy, and Length Velocity in Nepalese Children: the MAL-ED Birth Cohort Study. J Pediatr Gastroenterol Nutr 2018; 67:242-249. [PMID: 29620600 DOI: 10.1097/mpg.0000000000001990] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Environmental enteropathy (EE) is likely associated with growth retardation in children, but the association between EE and length velocity z score (LVZ) has not been investigated. The objective of the study was to assess associations between fecal markers for intestinal inflammation and LVZ and whether these associations were influenced by micronutrient adequacy among 9 to 24 months old children in Bhaktapur, Nepal. METHODS Data were divided into 5 time slots (9-12, 12-15, 15-18, 18-21, and 21-24 months). Anthropometric measurement and dietary assessment (by 24 hour recall) were performed monthly. Mean nutrient density adequacy was calculated based on nutrient density adequacy of 10 micronutrients (thiamin, riboflavin, niacin, vitamin B6, folate, vitamin C, vitamin A, calcium, iron, and zinc). Anti-1-antitrypsin (AAT), myeloperoxidase (MPO), and neopterin (NEO) were measured in stool samples collected at the beginning of each time slot. An EE score was calculated based on all 3 fecal markers. Associations between AAT, MPO, NEO and EE score and LVZ were assessed by multiple linear regression analyses and Generalized Estimating Equations models. RESULTS Associations between fecal markers and EE score and LVZ were generally weak. EE score and MPO for 3-month and MPO for 6-month growth periods were significantly associated with LVZ from 9 to 24 months. These associations were slightly modified by mean nutrient density adequacy. CONCLUSIONS EE score and MPO were significantly associated with LVZ in 9 to 24 months old Nepali children. Further studies to establish the usefulness of AAT, MPO, and NEO in assessing EE and growth retardation are warranted.
Collapse
|
135
|
Fahim SM, Das S, Sanin KI, Gazi MA, Mahfuz M, Islam MM, Ahmed T. Association of Fecal Markers of Environmental Enteric Dysfunction with Zinc and Iron Status among Children at First Two Years of Life in Bangladesh. Am J Trop Med Hyg 2018; 99:489-494. [PMID: 29893201 PMCID: PMC6090336 DOI: 10.4269/ajtmh.17-0985] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 04/10/2018] [Indexed: 02/01/2023] Open
Abstract
Environmental enteric dysfunction (EED) causes gut inflammation and increased intestinal permeability leading to deficiencies in micronutrients such as zinc and iron. Fecal markers such as myeloperoxidase (MPO), neopterin (NEO), and alpha-1-anti-trypsin (AAT) can predict EED. The aim of this study was to examine the association between fecal markers of EED with zinc and iron status among children at first 2 years of life. Malnutrition and Enteric Disease Study Bangladeshi birth cohort data were used to conduct this analysis. Multivariable analyses using generalized estimating equations were performed to test the association between individual fecal markers with zinc or iron status of the children. A total of 265 children were enrolled in the study (male:female = 1:1). Of the 627 stool samples collected (N = 222 children), 535, 511, and 577 were accompanied by zinc, ferritin, and soluble transferrin receptor values, respectively. Median (interquartile range [IQR]) values of AAT, MPO, and NEO were 0.33 (0.18-0.62) mg/g, 3,895.42 (1,563.76-8,432.82) ng/mL, and 890.81 (331.57-2,089.04) nmol/L, respectively. Overall, 60%, 71%, and 97% of samples were above the values considered normal in nontropical settings for AAT, MPO, and NEO, respectively. High AAT levels were significantly associated with low ferritin values after adjusting for age and gender (coefficient = -5.85; 95% confidence interval = -11.23 to -0.47; P value = 0.03). No such association was found between AAT and plasma zinc status. Myeloperoxidase and NEO were not associated with plasma zinc or iron status. The study results imply the importance of enteric protein loss in contributing to reduced ferritin levels at first 2 years of life.
Collapse
Affiliation(s)
- Shah Mohammad Fahim
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Subhasish Das
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Kazi Istiaque Sanin
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Amran Gazi
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - M. Munirul Islam
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| |
Collapse
|
136
|
Sié A, Dah C, Ouermi L, Tapsoba C, Zabre P, Bärnighausen T, Lebas E, Arzika AM, Snyder BM, Porco TC, Lietman TM, Keenan JD, Oldenburg CE. Effect of Antibiotics on Short-Term Growth among Children in Burkina Faso: A Randomized Trial. Am J Trop Med Hyg 2018; 99:789-796. [PMID: 30014828 DOI: 10.4269/ajtmh.18-0342] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Antibiotics improve both weight and height gain in randomized trials of preschool children with preexisting morbidity. Here, we assess the effect of a short course of three different antibiotics (amoxicillin, azithromycin, and cotrimoxazole) on short-term linear and ponderal growth in a population-based sample of preschool children in rural Burkina Faso. We randomized households with at least two children in the Nouna district, Burkina Faso, to a 5-day course of amoxicillin, azithromycin, cotrimoxazole, or placebo. Within each antibiotic-randomized household, one child was randomly assigned to receive the antibiotic and the other to receive the placebo. Weight and height measurements were taken at baseline and 30 days following the last study medication dose. Weight-for-height Z (WHZ), height-for-age Z (HAZ), and weight-for-age Z (WAZ) scoreswere calculated based on the 2006 World Health Organization standards. Of the 124 households and 248 children enrolled, 229 had anthropometry measurements at 1 month and were analyzed. Children randomized to amoxicillin gained significantly more weight compared with both the placebo household (mean difference 317 g, 95% confidence interval [CI]: 115-519 g) and placebo sibling (mean difference 315 g, 95% CI: 147-482 g) controls. Growth velocity in g/kg/day, and WHZ and WAZ scores were higher in amoxicillin-treated children compared with placebo households and siblings. There were no differences in weight gain in children randomized to azithromycin or cotrimoxazole compared with placebo households or placebo siblings. There were no differences in height gain or HAZ across any of the study arms. Amoxicillin may have short-term growth-promoting effects in healthy children.
Collapse
Affiliation(s)
- Ali Sié
- Centre de Recherche en Santé de Nouna, Nouna, Burkina Faso
| | - Clarisse Dah
- Centre de Recherche en Santé de Nouna, Nouna, Burkina Faso
| | | | | | - Pascal Zabre
- Centre de Recherche en Santé de Nouna, Nouna, Burkina Faso
| | - Till Bärnighausen
- Africa Health Research Institute, KwaZulu-Natal, South Africa.,Department of Global Health and Population, Harvard T. H. Chan School of Public Health, Boston, Massachusetts.,Heidelberg Institute of Public Health, University of Heidelberg, Heidelberg, Germany
| | - Elodie Lebas
- Francis I. Proctor Foundation, University of California, San Francisco, California
| | | | - Blake M Snyder
- Francis I. Proctor Foundation, University of California, San Francisco, California
| | - Travis C Porco
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California.,Department of Ophthalmology, University of California, San Francisco, San Francisco, California.,Francis I. Proctor Foundation, University of California, San Francisco, California
| | - Thomas M Lietman
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California.,Department of Ophthalmology, University of California, San Francisco, San Francisco, California.,Francis I. Proctor Foundation, University of California, San Francisco, California
| | - Jeremy D Keenan
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California.,Department of Ophthalmology, University of California, San Francisco, San Francisco, California.,Francis I. Proctor Foundation, University of California, San Francisco, California
| | - Catherine E Oldenburg
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California.,Department of Ophthalmology, University of California, San Francisco, San Francisco, California.,Francis I. Proctor Foundation, University of California, San Francisco, California
| |
Collapse
|
137
|
Ngoma TN, Chimimba UK, Mwangwela AM, Thakwalakwa C, Maleta KM, Manary MJ, Trehan I. Effect of cowpea flour processing on the chemical properties and acceptability of a novel cowpea blended maize porridge. PLoS One 2018; 13:e0200418. [PMID: 29990380 PMCID: PMC6039016 DOI: 10.1371/journal.pone.0200418] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 06/26/2018] [Indexed: 11/17/2022] Open
Abstract
Childhood growth stunting is a pervasive problem in Malawi and is in large part due to low quality complementary foods and chronic gut inflammation. Introducing legumes such as cowpea (Vigna unguiculata) into the complementary diet has the potential to improve childhood growth by improving diet quality through improvements in macro- and micronutrients and also by reducing gut inflammation. However, cowpea is relatively underutilized in complementary feeding in Malawi due to its strong taste, long processing time, and high energy requirements for processing. Effective utilization of cowpea in complementary feeding requires processing which may affect chemical composition as well as sensory quality. The present study evaluated the effect of processing on the retention of zinc, crude fibre, and flavonoid in roasted, boiled, and dehulled cowpea flours, and assessed the acceptability of maize porridge (70%) enriched with one of the three cowpea flours (30%). Roasting, dehulling, and boiling did not have any effect on zinc content. Crude fibre content increased after processing by all methods. Processing had no effect on measurable flavonoids. Roasted, boiled, and dehulled cowpea blended maize porridges were acceptable to children with mean quantities of leftover food of less than 3g from the given 100g. Caregivers also rated the blended flours to be highly acceptable to them as well, with maize porridge blended with dehulled cowpea flour the most acceptable to both children and caregivers. These results demonstrate that cowpea flour, processed by any of these three different methods, could serve as a useful addition to maize porridge for complementary feeding of children in sub-Saharan Africa.
Collapse
Affiliation(s)
- Theresa N Ngoma
- Department of Food Science and Technology, Lilongwe University of Agriculture and Natural Resources, Lilongwe, Malawi
| | - Ulemu K Chimimba
- Department of Food Science and Technology, Lilongwe University of Agriculture and Natural Resources, Lilongwe, Malawi
| | - Agnes M Mwangwela
- Department of Food Science and Technology, Lilongwe University of Agriculture and Natural Resources, Lilongwe, Malawi
| | - Chrissie Thakwalakwa
- School of Public Health and Family Medicine, University of Malawi College of Medicine, Blantyre, Malawi
| | - Kenneth M Maleta
- School of Public Health and Family Medicine, University of Malawi College of Medicine, Blantyre, Malawi
| | - Mark J Manary
- School of Public Health and Family Medicine, University of Malawi College of Medicine, Blantyre, Malawi.,Department of Pediatrics, Washington University in St. Louis, St Louis, Missouri, United States of America
| | - Indi Trehan
- Department of Pediatrics, Washington University in St. Louis, St Louis, Missouri, United States of America.,Department of Paediatrics and Child Health, University of Malawi College of Medicine, Blantyre, Malawi
| |
Collapse
|
138
|
Church JA, Parker EPK, Kosek MN, Kang G, Grassly NC, Kelly P, Prendergast AJ. Exploring the relationship between environmental enteric dysfunction and oral vaccine responses. Future Microbiol 2018; 13:1055-1070. [PMID: 29926747 PMCID: PMC6136084 DOI: 10.2217/fmb-2018-0016] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/26/2018] [Indexed: 12/11/2022] Open
Abstract
Oral vaccines significantly underperform in low-income countries. One possible contributory factor is environmental enteric dysfunction (EED), a subclinical disorder of small intestinal structure and function among children living in poverty. Here, we review studies describing oral vaccine responses and EED. We identified eight studies evaluating EED and oral vaccine responses. There was substantial heterogeneity in study design and few consistent trends emerged. Four studies reported a negative association between EED and oral vaccine responses; two showed no significant association; and two described a positive correlation. Current evidence is therefore insufficient to determine whether EED contributes to oral vaccine underperformance. We identify roadblocks in the field and future research needs, including carefully designed studies those can investigate this hypothesis further.
Collapse
Affiliation(s)
- James A Church
- Zvitambo Institute for Maternal & Child Health Research, Harare, Zimbabwe
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, UK
| | - Edward PK Parker
- Department of Infectious Disease Epidemiology, St Mary's Campus, Imperial College London, London, UK
| | - Margaret N Kosek
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Gagandeep Kang
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Nicholas C Grassly
- Department of Infectious Disease Epidemiology, St Mary's Campus, Imperial College London, London, UK
| | - Paul Kelly
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, UK
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal & Child Health Research, Harare, Zimbabwe
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, UK
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
139
|
Owino VO, Cornelius C, Loechl CU. Elucidating Adverse Nutritional Implications of Exposure to Endocrine-Disrupting Chemicals and Mycotoxins through Stable Isotope Techniques. Nutrients 2018; 10:nu10040401. [PMID: 29570653 PMCID: PMC5946186 DOI: 10.3390/nu10040401] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
Multiple drivers of the double burden of malnutrition (DBM) include a rapid shift from predominantly plant-based diets to energy-dense foods based on meats, milk, animal fats and vegetable oils. The shift to overweight and obesity is driven by increased exposure to mass media, urbanization, technological advances in food processing, rising income and increased population density associated with increased access to cheap foods. At the same time, undernutrition persists mainly due to food insecurity and lack of access to safe water, sanitation and adequate health care. All known nutrition interventions result in only one third reduction in stunting. Little consideration has been given to hazardous exposure to endocrine disrupting chemicals (EDCs) and microbial toxins as major components of the malnutrition-causal framework. These hazards include microbial toxins, for example, mycotoxins, and environmental pollutants such as persistent organic pollutants (POPs), some of which are known to disrupt the endocrine system. These hazards sit at the cross road of undernutrition and overweight and obesity since the exposure cuts across the critical window of opportunity (the first 1000 days). In this review, we update on the role of food and environmental contaminants, especially EDCs and aflatoxins, in child growth and on the implications for metabolic dysfunction and disease risk in later life, and discuss potential applications of nuclear and isotopic techniques to elucidate the underlying biological mechanisms, outcome indicators, as well as occurrence levels.
Collapse
Affiliation(s)
- Victor O Owino
- Nutrition and Health Related Environmental Studies Section, Division of Health, International Atomic Energy Agency, Vienna International Centre P.O. Box 100, A-1400 Vienna, Austria.
| | - Carolin Cornelius
- Nutrition and Health Related Environmental Studies Section, Division of Health, International Atomic Energy Agency, Vienna International Centre P.O. Box 100, A-1400 Vienna, Austria.
| | - Cornelia U Loechl
- Nutrition and Health Related Environmental Studies Section, Division of Health, International Atomic Energy Agency, Vienna International Centre P.O. Box 100, A-1400 Vienna, Austria.
| |
Collapse
|
140
|
Promising Biomarkers of Environmental Enteric Dysfunction: A Prospective Cohort study in Pakistani Children. Sci Rep 2018; 8:2966. [PMID: 29445110 PMCID: PMC5813024 DOI: 10.1038/s41598-018-21319-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/22/2018] [Indexed: 12/13/2022] Open
Abstract
Environmental Enteric Dysfunction (EED), a syndrome characterized by chronic gut inflammation, contributes towards stunting and poor response to enteric vaccines in children in developing countries. In this study, we evaluated major putative biomarkers of EED using growth faltering as its clinical proxy. Newborns (n = 380) were enrolled and followed till 18 months with monthly anthropometry. Biomarkers associated with gut and systemic inflammation were assessed at 6 and 9 months. Linear mixed effects model was used to determine the associations of these biomarkers with growth faltering between birth and 18 months. Fecal myeloperoxidase (neutrophil activation marker) at 6 months [β = −0.207, p = 0.005], and serum GLP 2 (enterocyte proliferation marker) at 6 and 9 months [6M: β = −0.271, p = 0.035; 9M: β = −0.267, p = 0.045] were associated with decreasing LAZ score. Ferritin at 6 and 9 months was associated with decreasing LAZ score [6M: β = −0.882, p < 0.0001; 9M: β = −0.714, p < 0.0001] and so was CRP [β = −0.451, p = 0.039] and AGP [β = −0.443, p = 0.012] at 9 months. Both gut specific and systemic biomarkers correlated negatively with IGF-1, but only weakly correlated, if at all with each other. We therefore conclude that EED may be contributing directly towards growth faltering, and this pathway is not entirely through the pathway of systemic inflammation.
Collapse
|
141
|
Syed S, Dinallo V, Iqbal NT, Di Iorio L, Di Fusco D, Guleria S, Amadi BC, Sadiq K, Moskaluk C, Ali SA, Kelly P, Monteleone G. High SMAD7 and p-SMAD2,3 expression is associated with environmental enteropathy in children. PLoS Negl Trop Dis 2018; 12:e0006224. [PMID: 29415065 PMCID: PMC5819826 DOI: 10.1371/journal.pntd.0006224] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/20/2018] [Accepted: 01/09/2018] [Indexed: 01/10/2023] Open
Abstract
Enteropathies such as Crohn's disease are associated with enteric inflammation characterized by impaired TGF-β signaling, decreased expression of phosphorylated (p)-SMAD2,3 and increased expression of SMAD7 (an inhibitor of SMAD3 phosphorylation). Environmental enteropathy (EE) is an acquired inflammatory disease of the small intestine (SI), which is associated with linear growth disruption, cognitive deficits, and reduced oral vaccine responsiveness in children <5 y in resource-poor countries. We aimed to characterize EE inflammatory pathways by determining SMAD7 and p-SMAD2,3 levels (using Western blotting) in EE duodenal biopsies (N = 19 children, 7 from Pakistan, 12 from Zambia) and comparing these with healthy controls (Ctl) and celiac disease (CD) patients from Italy. Densitometric analysis of immunoblots showed that EE SI biopsies expressed higher levels of both SMAD7 (mean±SD in arbitrary units [a.u.], Ctl = 0.47±0.20 a.u., EE = 1.13±0.25 a.u., p-value = 0.03) and p-SMAD2,3 (mean±SD, Ctl = 0.38±0.14 a.u., EE = 0.60±0.10 a.u., p-value = 0.03). Immunohistochemistry showed that, in EE, SMAD7 is expressed in both the epithelium and in mononuclear cells of the lamina propria (LP). In contrast, p-SMAD3 in EE is expressed much more prominently in epithelial cells than in the LP. The high SMAD7 immunoreactivity and lack of p-SMAD3 expression in the LP suggests defective TGF-β signaling in the LP in EE similar to a previously reported SMAD7-mediated inflammatory pathway in refractory CD and Crohn's disease. However, Western blot densitometry showed elevated p-SMAD2,3 levels in EE, possibly suggesting a different inflammatory pathway than Crohn's disease but more likely reflecting cumulative protein expression from across all compartments of the mucosa as opposed to the LP alone. Further studies are needed to substantiate these preliminary results and to illustrate the relationship between SMAD proteins, TGF-β signaling, and inflammatory cytokine production, all of which may be potential therapeutic targets.
Collapse
Affiliation(s)
- Sana Syed
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
- Department of Pediatrics, Division of Gastroenterology, Hepatology, & Nutrition, University of Virginia, Charlottesville, United States of America
| | - Vincenzo Dinallo
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Najeeha T. Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Laura Di Iorio
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Davide Di Fusco
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Shan Guleria
- Department of Pediatrics, Division of Gastroenterology, Hepatology, & Nutrition, University of Virginia, Charlottesville, United States of America
| | - Beatrice C. Amadi
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Kamran Sadiq
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Christopher Moskaluk
- 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
| | - Paul Kelly
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine, Lusaka, Zambia
- Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
- * E-mail:
| |
Collapse
|
142
|
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
|
143
|
Gómez-Duarte O. Contaminación de agua en países de bajos y medianos recursos es un problema de salud pública global. REVISTA DE LA FACULTAD DE MEDICINA 2018. [DOI: 10.15446/revfacmed.v66n1.70775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
El agua es un elemento esencial para la vida de todos los seres vivientes del planeta, además es un derecho humano fundamental (1). El 97.2% del agua en el planeta Tierra es salina y solo el 2.5% corresponde a agua fresca; de ese 2.5%, 30% es subterránea, 68% está en los glaciares y otras capas de nieve y solo el 1.2% es superficial y se encuentra en ríos, lagos y otras formas de agua de superficie (2).El agua potable está amenazada por la continua contaminación que genera la actividad humana y por la disminución de los recursos hídricos como consecuencia del calentamiento global (3). Según la Organización Mundial de la Salud, desde el año 2015 solo el 89% de la población mundial tiene acceso a agua apta para consumo y se anticipa que este porcentaje continuará disminuyendo (4), aunque se estima que la situación ya es crítica para 260 millones de personas que carecen de agua apta para el consumo (5).
Collapse
|
144
|
Position paper: The potential role of optical biopsy in the study and diagnosis of environmental enteric dysfunction. Nat Rev Gastroenterol Hepatol 2017; 14:727-738. [PMID: 29139480 DOI: 10.1038/nrgastro.2017.147] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Environmental enteric dysfunction (EED) is a disease of the small intestine affecting children and adults in low and middle income countries. Arising as a consequence of repeated infections, gut inflammation results in impaired intestinal absorptive and barrier function, leading to poor nutrient uptake and ultimately to stunting and other developmental limitations. Progress towards new biomarkers and interventions for EED is hampered by the practical and ethical difficulties of cross-validation with the gold standard of biopsy and histology. Optical biopsy techniques - which can provide minimally invasive or noninvasive alternatives to biopsy - could offer other routes to validation and could potentially be used as point-of-care tests among the general population. This Consensus Statement identifies and reviews the most promising candidate optical biopsy technologies for applications in EED, critically assesses them against criteria identified for successful deployment in developing world settings, and proposes further lines of enquiry. Importantly, many of the techniques discussed could also be adapted to monitor the impaired intestinal barrier in other settings such as IBD, autoimmune enteropathies, coeliac disease, graft-versus-host disease, small intestinal transplantation or critical care.
Collapse
|
145
|
Yu EA, Huey SL, Peña‐Rosas JP, Mehta S. The effects of oral vitamin D supplementation on linear growth and non‐communicable diseases among infants and children younger than five years of age. Cochrane Database Syst Rev 2017; 2017:CD012875. [PMCID: PMC6486017 DOI: 10.1002/14651858.cd012875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
This is a protocol for a Cochrane Review (Intervention). The objectives are as follows: To assess effects of oral vitamin D supplementation on preventing and treating stunting and non‐communicable diseases among infants and children younger than five years of age.
Collapse
Affiliation(s)
- Elaine A Yu
- Cornell UniversityDivision of Nutritional Sciences316 Savage HallIthacaUSA14853
| | - Samantha L Huey
- Cornell UniversityDivision of Nutritional Sciences316 Savage HallIthacaUSA14853
| | - Juan Pablo Peña‐Rosas
- World Health OrganizationEvidence and Programme Guidance, Department of Nutrition for Health and Development20 Avenue AppiaGenevaSwitzerland1211
| | - Saurabh Mehta
- Cornell UniversityDivision of Nutritional Sciences316 Savage HallIthacaUSA14853
| |
Collapse
|
146
|
Lee D, Lee SH, Song J, Jee HJ, Cha SH, Chang GT. Effects of Astragalus Extract Mixture HT042 on Height Growth in Children with Mild Short Stature: A Multicenter Randomized Controlled Trial. Phytother Res 2017; 32:49-57. [PMID: 29130588 DOI: 10.1002/ptr.5886] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 07/10/2017] [Accepted: 07/16/2017] [Indexed: 11/09/2022]
Abstract
HT042 is a standardized functional food ingredient approved by Korean Food and Drug Administration with a claim 'HT042 can help height growth of children'. We aimed to evaluate the safety and efficacy of HT042 on height growth in children with mild short stature. A multicenter, randomized, double-blind, placebo-controlled parallel study was performed on children aged 6-8 years with height ranked below the 25th percentile. In 129 children, height gain was significantly higher in HT042 group than placebo group after 24 weeks (mean difference, 0.29 cm; 95% CI, 0.01 to 0.57 cm; p = 0.027). The difference was elevated when the efficacy analysis was restricted to children below the 10th percentile (mean difference, 0.45 cm; 95% CI, 0.04 to 0.87 cm; p = 0.031). Because bone age advancement was lower in HT042 group, height standard deviation score gain for bone age was higher in HT042 group and the difference was significant in children below the 10th percentile (mean difference, 0.20 score; 95% CI, 0.00 to 0.39 points; p = 0.045). Serum IGF-1 and IGFBP-3 levels were significantly increased compared with baseline within HT042 group, but group difference was not significant. HT042 supplementation helped to increase height growth in children without skeletal maturation and was more effective in much shorter children. The effects might be mediated by increases in serum IGF-1 and IGFBP-3 levels. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Donghun Lee
- Department of Herbal Pharmacology, Kyung Hee University College of Korean Medicine, Seoul, South Korea
| | - Sun Haeng Lee
- Department of Pediatrics, Kyung Hee University College of Korean Medicine, Seoul, South Korea
| | - Jungbin Song
- Department of Herbal Pharmacology, Kyung Hee University College of Korean Medicine, Seoul, South Korea
| | - Hee-Jung Jee
- Department of Biostatistics, Korea University College of Medicine, Seoul, South Korea
| | - Sung Ho Cha
- Department of Pediatrics, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Gyu Tae Chang
- Pediatrics of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
| |
Collapse
|
147
|
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
|
148
|
Abstract
Malnutrition contributes significantly to death and illness worldwide and especially to the deaths of children younger than 5 years. The relation between intestinal changes in malnutrition and morbidity and mortality has not been well characterized; however, recent research indicates that the functional and morphologic changes of the intestine secondary to malnutrition itself contribute significantly to these negative clinical outcomes and may be potent targets of intervention. The aim of this review was to summarize current knowledge of experimental and clinically observed changes in the intestine from malnutrition preclinical models and human studies. Limited clinical studies have shown villous blunting, intestinal inflammation, and changes in the intestinal microbiome of malnourished children. In addition to these findings, experimental data using various animal models of malnutrition have found evidence of increased intestinal permeability, upregulated intestinal inflammation, and loss of goblet cells. More mechanistic studies are urgently needed to improve our understanding of malnutrition-related intestinal dysfunction and to identify potential novel targets for intervention.
Collapse
|
149
|
Abstract
The main forms of childhood malnutrition occur predominantly in children <5 years of age living in low-income and middle-income countries and include stunting, wasting and kwashiorkor, of which severe wasting and kwashiorkor are commonly referred to as severe acute malnutrition. Here, we use the term 'severe malnutrition' to describe these conditions to better reflect the contributions of chronic poverty, poor living conditions with pervasive deficits in sanitation and hygiene, a high prevalence of infectious diseases and environmental insults, food insecurity, poor maternal and fetal nutritional status and suboptimal nutritional intake in infancy and early childhood. Children with severe malnutrition have an increased risk of serious illness and death, primarily from acute infectious diseases. International growth standards are used for the diagnosis of severe malnutrition and provide therapeutic end points. The early detection of severe wasting and kwashiorkor and outpatient therapy for these conditions using ready-to-use therapeutic foods form the cornerstone of modern therapy, and only a small percentage of children require inpatient care. However, the normalization of physiological and metabolic functions in children with malnutrition is challenging, and children remain at high risk of relapse and death. Further research is urgently needed to improve our understanding of the pathophysiology of severe malnutrition, especially the mechanisms causing kwashiorkor, and to develop new interventions for prevention and treatment.
Collapse
Affiliation(s)
- Zulfiqar A Bhutta
- Centre for Global Child Health, Hospital for Sick Children, Peter Gilgan Centre for Research &Learning, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
- Center of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - James A Berkley
- Clinical Research Department, KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- The Childhood Acute Illness &Nutrition (CHAIN) Network, Nairobi, Kenya
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Robert H J Bandsma
- Centre for Global Child Health, Hospital for Sick Children, Peter Gilgan Centre for Research &Learning, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
- The Childhood Acute Illness &Nutrition (CHAIN) Network, Nairobi, Kenya
- Department of Biomedical Sciences, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Marko Kerac
- Department of Population Health, London School of Hygiene &Tropical Medicine, London, UK
| | - Indi Trehan
- Lao Friends Hospital for Children, Luang Prabang, Laos
- Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Paediatrics and Child Health, University of Malawi, Blantyre, Malawi
| | - André Briend
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
150
|
Abstract
PURPOSE OF REVIEW The term 'tropical enteropathy' originated in observations in the 1960s that small intestinal morphology and function differed in the tropics from the norms found in temperate climates. It was subsequently shown that this enteropathy is more closely related to environmental conditions than latitude, and it was re-labelled 'environmental enteropathy'. It is now recognised that environmental enteropathy (also now called environmental enteric dysfunction) has implications for the health and linear growth of children in low- and middle-income countries, and it may underlie poor responses to oral vaccination in these countries. The purpose of this review is to define and clarify this enteropathy despite the confusing terminology it has attracted and to contrast it with other enteropathic states. RECENT FINDINGS Recent work has begun to demonstrate the nature of the mucosal lesion and the relationship with microbial translocation which is currently thought to link a failure of mucosal barrier function and the cascade of systemic inflammation which inhibits growth. The evidence is still correlative rather than definitive, but derives some additional support from animal models. There are some common features between environmental enteropathy and other enteropathies, but there are important differences also. The mechanism of the link between enteropathy and vaccine failure is not understood, and neither is it clear how the more severe form of enteropathy, which we refer to as malnutrition enteropathy, is driven by nutrient depletion and intestinal infection. Tropical enteropathies form a group of disorders which include environmental and nutritional enteropathies. The long-term health implications of these disorders for health in low-income countries are just being explored, but the scale of their effects is very large, with millions of people affected.
Collapse
Affiliation(s)
- John Louis-Auguste
- Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, Newark Street, London, E1 2AT, UK
| | - Paul Kelly
- Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, Newark Street, London, E1 2AT, UK.
- TROPGAN, University of Zambia School of Medicine, University Teaching Hospital, Lusaka, Zambia.
| |
Collapse
|