1
|
Mahfuz M, Coomes D, Abdalla M, Mweetwa M, VanBuskirk K, Iqbal NT, Ali SA, Chandwe K, Das S, Kelly P, Shaikh N, Tarr PI, Denno DM. Biomarker relationships with small bowel histopathology among malnourished children with environmental enteric dysfunction in a multicountry cohort study. Am J Clin Nutr 2024; 120 Suppl 1:S73-S83. [PMID: 39300665 DOI: 10.1016/j.ajcnut.2024.02.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 02/12/2024] [Accepted: 02/22/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Validated biomarkers could catalyze environmental enteric dysfunction (EED) research. OBJECTIVES Leveraging an EED histology scoring system, this multicountry analysis examined biomarker associations with duodenal histology features among children with EED. We also examined differences in 2-h compared with 1-h urine collections in the lactulose rhamnose (LR) dual sugar test. METHODS Three cohorts of undernourished children unresponsive to nutrition intervention underwent esophagogastroduodenoscopy and duodenal biopsies. Histopathology scores were compared to fecal calprotectin (CAL), myeloperoxidase (MPO), neopterin (NEO), and urinary LR ratio and lactulose percentage recovery. Log-transformed biomarkers were used in linear regressions adjusted for age, center, and sample collection-biopsy time interval in multivariable models. RESULTS Data on >1 biomarker were available for 120 Bangladeshi (CAL, MPO, NEO, and LR), 63 Pakistani (MPO, NEO, and LR), and 63 Zambian children (CAL). Median age at endoscopy was similar (19 mo) across centers. Median sample collection prior to endoscopy was consistent with each center's study design: 2 wk in Bangladesh (urine and stool) and Zambia (stool), and 6 (urine) and 11 (stool) mo in Pakistan. In multivariable models, intraepithelial lymphocytes were associated with CAL (exponentiated [exp.] coefficient: 1.19; 95% confidence interval [CI]: 1, 1.41), intramucosal Brunner's glands with MPO (exp. coefficient: 1.33; 95% CI: 1.05, 1.69) and NEO (exp. coefficient: 1.37; 95% CI: 1.1, 1.7), and chronic inflammation with NEO (exp. coefficient: 1.61; 95% CI: 1.17, 2.17). Intraepithelial lymphocytes were associated with lactulose % recovery (exp. coefficient: 1.22; 95% CI: 1.05, 1.41). LR recovery was substantially lower in 1-h collections than in 2-h collections. CONCLUSIONS Four commonly used markers of enteric dysfunction were associated with specific histologic features. One-hour urine collection may be insufficient to reflect small bowel permeability in LR testing. While acknowledging the challenges with obtaining relevant tissue, these findings form the basis for further EED biomarker validation research.
Collapse
Affiliation(s)
- Mustafa Mahfuz
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh.
| | - David Coomes
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, United States
| | - Marwa Abdalla
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, United States
| | - Monica Mweetwa
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Kelley VanBuskirk
- Department of Global Health, University of Washington School of Public Health, Seattle, WA, United States
| | - Najeeha T Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - S Asad Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Kanta Chandwe
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Subhasish Das
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Paul Kelly
- Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Phillip I Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Donna M Denno
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States.
| |
Collapse
|
2
|
Mann ER, Lam YK, Uhlig HH. Short-chain fatty acids: linking diet, the microbiome and immunity. Nat Rev Immunol 2024:10.1038/s41577-024-01014-8. [PMID: 38565643 DOI: 10.1038/s41577-024-01014-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2024] [Indexed: 04/04/2024]
Abstract
The short-chain fatty acids (SCFAs) butyrate, propionate and acetate are microbial metabolites and their availability in the gut and other organs is determined by environmental factors, such as diet and use of antibiotics, that shape the diversity and metabolism of the microbiota. SCFAs regulate epithelial barrier function as well as mucosal and systemic immunity via evolutionary conserved processes that involve G protein-coupled receptor signalling or histone deacetylase activity. Indicatively, the anti-inflammatory role of butyrate is mediated through direct effects on the differentiation of intestinal epithelial cells, phagocytes, B cells and plasma cells, and regulatory and effector T cells. Intestinally derived SCFAs also directly and indirectly affect immunity at extra-intestinal sites, such as the liver, the lungs, the reproductive tract and the brain, and have been implicated in a range of disorders, including infections, intestinal inflammation, autoimmunity, food allergies, asthma and responses to cancer therapies. An ecological understanding of microbial communities and their interrelated metabolic states, as well as the engineering of butyrogenic bacteria may support SCFA-focused interventions for the prevention and treatment of immune-mediated diseases.
Collapse
Affiliation(s)
- Elizabeth R Mann
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Ying Ka Lam
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
- Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
3
|
Ribeiro SA, Braga EL, Queiroga ML, Clementino MA, Fonseca XM, Belém MO, Magalhães LM, de Sousa JK, de Freitas TM, Veras HN, de Aquino CC, Santos AD, de Moura FR, Dos Santos AA, Havt A, Maciel BL, Lima AA. A New Murine Undernutrition Model Based on Complementary Feeding of Undernourished Children Causes Damage to the Morphofunctional Intestinal Epithelium Barrier. J Nutr 2024; 154:1232-1251. [PMID: 38346539 PMCID: PMC11347815 DOI: 10.1016/j.tjnut.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/26/2024] [Accepted: 02/01/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Complementary feeding is critical in establishing undernutrition. However, experimental undernourished diets do not represent the amount of nutrients in the complementary diets of undernourished children. OBJECTIVES To develop, validate, and evaluate the impact of a new murine model of undernutrition on the intestinal epithelium, based on the complementary diet of undernourished children from 7 countries with low-socioeconomic power belonging to the Malnutrition-Enteric Diseases (MAL-ED) cohort study. METHODS We used the difference in the percentage of energy, macronutrients, fiber and zinc in the complementary diet of children without undernutrition compared with stunting (height-for-age Z-score < -2) for the MAL-ED diet formulation. Subsequently, C57BL/6 mice were fed a control diet (AIN-93M diet) or MAL-ED diet for 28 d. Weight was measured daily; body composition was measured every 7 d; lactulose:mannitol ratio (LM) and morphometry were evaluated on days 7 and 28; the cotransport test and analysis of intestinal transporters and tight junctions were performed on day 7. RESULTS The MAL-ED diet presented -8.03% energy, -37.46% protein, -24.20% lipid, -10.83% zinc, +5.93% carbohydrate, and +45.17% fiber compared with the control diet. This diet rapidly reduced weight gain and compromised body growth and energy reserves during the chronic period (P < 0.05). In the intestinal epithelial barrier, this diet caused an increase in the LM (P < 0.001) and reduced (P < 0.001) the villous area associated with an increase in FAT/CD36 in the acute period and increased (P < 0.001) mannitol excretion in the chronic period. CONCLUSIONS The MAL-ED diet induced undernutrition in mice, resulting in acute damage to the integrity of the intestinal epithelial barrier and a subsequent increase in the intestinal area during the chronic period. This study introduces the first murine model of undernutrition for the complementary feeding phase, based on data from undernourished children in 7 different countries.
Collapse
Affiliation(s)
- Samilly A Ribeiro
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil.
| | - Enock Lr Braga
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Marcus L Queiroga
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Marco A Clementino
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Xhaulla Mqc Fonseca
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Mônica O Belém
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Lyvia Mvc Magalhães
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - José K de Sousa
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Thiago M de Freitas
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Herlice N Veras
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Cristiane C de Aquino
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Alan Dc Santos
- Núcleo de Estudos Químicos de Micromoléculas da Amazônia (NEQUIMA), Manaus, Brazil
| | - Flávio Rm de Moura
- Núcleo de Estudos Químicos de Micromoléculas da Amazônia (NEQUIMA), Manaus, Brazil
| | - Armênio A Dos Santos
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Alexandre Havt
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Bruna Ll Maciel
- Nutrition Postgraduation Program, Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Aldo Am Lima
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
4
|
Blaauw J, Chikwana J, Chaima D, Khoswe S, Samikwa L, de Vries I, Voskuijl W. The presence of enteropathy in HIV infected children on antiretroviral therapy in Malawi. PLoS One 2024; 19:e0298310. [PMID: 38330085 PMCID: PMC10852317 DOI: 10.1371/journal.pone.0298310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Undernutrition and malnutrition in children in low- and middle-income countries contribute to high mortality rates. Stunting, a prevalent form of malnutrition, is associated with educational and productivity losses. Environmental enteric dysfunction (EED) and human immunodeficiency virus (HIV) infection worsen these conditions. This study seeks to investigate the presence of enteropathy using EED fecal biomarkers in HIV-infected children who are stable on antiretroviral therapy (ART) across various nutritional statuses. By understanding the interplay between EED, HIV, and nutritional status, this study aims to provide insights that can inform targeted interventions to optimize nutritional outcomes in HIV infected children. METHODS/PRINCIPAL FINDINGS This study evaluated the levels of alpha-1-antitrypsin, calprotectin and myeloperoxidase in frozen fecal samples from 61 HIV infected (mean age 9.16 ±3.08 years) and 31 HIV uninfected (6.65 ±3.41 years) children in Malawi. Anthropometric measurements and clinical data were collected. The height-for-age z-score (-1.66 vs -1.27, p = 0.040) and BMI-for-age z-score (-0.36 vs 0.01, p = 0.037) were lower in HIV infected children. Enzyme-linked immunosorbent assays were used to measure biomarker concentrations. Statistical tests were applied to compare biomarker levels based on HIV status and anthropometric parameters. Myeloperoxidase, alpha-1-antitrypsin, and calprotectin concentrations did not differ between HIV infected and HIV uninfected children of different age groups. In HIV infected children from 5-15 years, there is no difference in biomarker concentration between the stunted and non-stunted groups. CONCLUSION/SIGNIFICANCE Our study found a higher prevalence of stunting in HIV infected children compared to uninfected children, but no significant differences in biomarker concentrations. This suggests no causal relationship between enteropathy and stunting in HIV infected children. These results contribute to the understanding of growth impairment in HIV infected children and emphasize the need for further research, particularly a longitudinal, biopsy-controlled study.
Collapse
Affiliation(s)
- Julia Blaauw
- Department of Global Child Health, Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jessica Chikwana
- Department of Pediatrics & Child Health, The Kamuzu University of Health Sciences, Blantyre, Malawi
| | - David Chaima
- Department of Pediatrics & Child Health, The Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Stanley Khoswe
- Department of Pediatrics & Child Health, The Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Lyson Samikwa
- Department of Pediatrics & Child Health, The Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Isabelle de Vries
- Department of Global Child Health, Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Wieger Voskuijl
- Department of Global Child Health, Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Sturgeon JP, Njunge JM, Bourke CD, Gonzales GB, Robertson RC, Bwakura-Dangarembizi M, Berkley JA, Kelly P, Prendergast AJ. Inflammation: the driver of poor outcomes among children with severe acute malnutrition? Nutr Rev 2023; 81:1636-1652. [PMID: 36977352 PMCID: PMC10639108 DOI: 10.1093/nutrit/nuad030] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
Severe acute malnutrition (SAM) is the most life-threatening form of undernutrition and underlies at least 10% of all deaths among children younger than 5 years in low-income countries. SAM is a complex, multisystem disease, with physiological perturbations observed in conjunction with the loss of lean mass, including structural and functional changes in many organ systems. Despite the high mortality burden, predominantly due to infections, the underlying pathogenic pathways remain poorly understood. Intestinal and systemic inflammation is heightened in children with SAM. Chronic inflammation and its consequent immunomodulation may explain the increased morbidity and mortality from infections in children with SAM, both during hospitalization and in the longer term after discharge. Recognition of the role of inflammation in SAM is critical in considering new therapeutic targets in this disease, which has not seen a transformational approach to treatment for several decades. This review highlights the central role of inflammation in the wide-ranging pathophysiology of SAM, as well as identifying potential interventions that have biological plausibility based on evidence from other inflammatory syndromes.
Collapse
Affiliation(s)
- Jonathan P Sturgeon
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | - James M Njunge
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Claire D Bourke
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | - Gerard Bryan Gonzales
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, Netherlands
| | - Ruairi C Robertson
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | | | - James A Berkley
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Paul Kelly
- is with the Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| |
Collapse
|
6
|
Ling C, Versloot CJ, Arvidsson Kvissberg ME, Hu G, Swain N, Horcas-Nieto JM, Miraglia E, Thind MK, Farooqui A, Gerding A, van Eunen K, Koster MH, Kloosterhuis NJ, Chi L, ChenMi Y, Langelaar-Makkinje M, Bourdon C, Swann J, Smit M, de Bruin A, Youssef SA, Feenstra M, van Dijk TH, Thedieck K, Jonker JW, Kim PK, Bakker BM, Bandsma RHJ. Rebalancing of mitochondrial homeostasis through an NAD +-SIRT1 pathway preserves intestinal barrier function in severe malnutrition. EBioMedicine 2023; 96:104809. [PMID: 37738832 PMCID: PMC10520344 DOI: 10.1016/j.ebiom.2023.104809] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND The intestine of children with severe malnutrition (SM) shows structural and functional changes that are linked to increased infection and mortality. SM dysregulates the tryptophan-kynurenine pathway, which may impact processes such as SIRT1- and mTORC1-mediated autophagy and mitochondrial homeostasis. Using a mouse and organoid model of SM, we studied the repercussions of these dysregulations on malnutrition enteropathy and the protective capacity of maintaining autophagy activity and mitochondrial health. METHODS SM was induced through feeding male weanling C57BL/6 mice a low protein diet (LPD) for 14-days. Mice were either treated with the NAD+-precursor, nicotinamide; an mTORC1-inhibitor, rapamycin; a SIRT1-activator, resveratrol; or SIRT1-inhibitor, EX-527. Malnutrition enteropathy was induced in enteric organoids through amino-acid deprivation. Features of and pathways to malnutrition enteropathy were examined, including paracellular permeability, nutrient absorption, and autophagic, mitochondrial, and reactive-oxygen-species (ROS) abnormalities. FINDINGS LPD-feeding and ensuing low-tryptophan availability led to villus atrophy, nutrient malabsorption, and intestinal barrier dysfunction. In LPD-fed mice, nicotinamide-supplementation was linked to SIRT1-mediated activation of mitophagy, which reduced damaged mitochondria, and improved intestinal barrier function. Inhibition of mTORC1 reduced intestinal barrier dysfunction and nutrient malabsorption. Findings were validated and extended using an organoid model, demonstrating that resolution of mitochondrial ROS resolved barrier dysfunction. INTERPRETATION Malnutrition enteropathy arises from a dysregulation of the SIRT1 and mTORC1 pathways, leading to disrupted autophagy, mitochondrial homeostasis, and ROS. Whether nicotinamide-supplementation in children with SM could ameliorate malnutrition enteropathy should be explored in clinical trials. FUNDING This work was supported by the Bill and Melinda Gates Foundation, the Sickkids Research Institute, the Canadian Institutes of Health Research, and the University Medical Center Groningen.
Collapse
Affiliation(s)
- Catriona Ling
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Christian J Versloot
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Matilda E Arvidsson Kvissberg
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Guanlan Hu
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nathan Swain
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - José M Horcas-Nieto
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Emily Miraglia
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada; Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mehakpreet K Thind
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Amber Farooqui
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Albert Gerding
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Karen van Eunen
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Mirjam H Koster
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Niels J Kloosterhuis
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Lijun Chi
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - YueYing ChenMi
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Miriam Langelaar-Makkinje
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Celine Bourdon
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jonathan Swann
- Faculty of Medicine, School of Human Development and Health, University of Southampton, United Kingdom; Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom
| | - Marieke Smit
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Alain de Bruin
- Department of Biomolecular Health Sciences, Dutch Molecular Pathology Centre, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Sameh A Youssef
- Department of Biomolecular Health Sciences, Dutch Molecular Pathology Centre, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Janssen Pharmaceutica Research and Development, 2340, Beerse, Belgium
| | - Marjon Feenstra
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Theo H van Dijk
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Kathrin Thedieck
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands; Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria; Freiburg Materials Research Center (FMF), University Freiburg, Freiburg, Germany
| | - Johan W Jonker
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Peter K Kim
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada; Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada.
| | - Barbara M Bakker
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands.
| | - Robert H J Bandsma
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands; Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
7
|
Jakhmola Mani R, Dogra N, Katare DP. The Connection between Chronic Liver Damage and Sporadic Alzheimer's Disease: Evidence and Insights from a Rat Model. Brain Sci 2023; 13:1391. [PMID: 37891760 PMCID: PMC10605053 DOI: 10.3390/brainsci13101391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Junk foods are typically low in essential nutrients, such as vitamins, minerals, and antioxidants. They are also loaded with trans fats and saturated fats, which can increase the level of triglycerides in the blood. High triglyceride levels can contribute to the development of non-alcoholic fatty liver disease (NAFLD), a condition where excess fat accumulates in the liver. A high intake of junk foods can lead to insulin resistance, a condition where the body's cells become less responsive to insulin. A diet lacking in nutrients and loaded with unwanted toxins can impair the liver's ability to detoxify harmful substances and damage its overall function. It is known that the regular consumption of junk food can be linked to memory impairment and cognitive decline. Several studies have shown that diets high in unhealthy fats, sugars, and processed foods can negatively impact brain health, including memory function. In this study, Wistar rats were used to model Late-Onset Alzheimer's Disease (LOAD), which was inspired by knowledge of the liver-brain axis's role in causing dementia. The model mimicked junk-food-induced liver-brain damage, and was developed by using the toxins d-galactosamine, ethanol and d-galactose. To begin with, the model rats demonstrated insulin resistance, a characteristic of LOAD patients. Glucose levels in both the brain and liver tissues were significantly elevated in the model, paralleling clinical findings in LOAD patients. High glucose levels in the brain lead to the increased production of advanced glycation end-products (AGEs), which, along with amyloid beta, harm neighbouring neurons. Histopathological analysis revealed deformed glial nodules, apoptotic neurons, and amyloid plaques in the brain section in the later stages of the disease. Simultaneously, the liver section displayed features of cirrhosis, including an effaced lobular architecture and the extravasation of red blood cells. Liver enzymes ALT, AST and ALP were consistently elevated with disease progression. Furthermore, immunohistochemistry confirmed the presence of amyloid precursor protein (APP) in the diseased brain. The positive expression of Hypoxia-Inducible Factor 3-Alpha (HIF3A) in the brain indicated hypoxic conditions, which is consistent with other LOAD studies. This model also exhibited damaged intestinal villi and excessive bowel and urinary incontinence, indicating malnutrition and a disturbed gut microbiome, which is also consistent with LOAD patients. Bioinformatics analysis on serum protein suggests a few affected molecular pathways, like the amyloid secretase pathway, androgen/oestrogen/progesterone biosynthesis, the apoptosis signalling pathway, the insulin/IGF pathway-protein kinase B signalling cascade, the Metabotropic glutamate receptor group I pathway, the Wnt signalling pathway, etc. Behavioural analysis confirmed memory decline and the loss of muscle strength with disease progression. Overall, this rat model of LOAD sheds valuable light on LOAD pathology and highlights the potential link between liver dysfunction, particularly induced by the excessive consumption of junk food, and LOAD. This study contributes to a deeper understanding of the complex molecular mechanisms involved in LOAD, paving the way for new possibilities in therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Deepshikha Pande Katare
- Proteomics and Translational Research Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida 201301, India; (R.J.M.); (N.D.)
| |
Collapse
|
8
|
Abtahi S, Sailer A, Roland JT, Haest X, Chanez-Paredes SD, Ahmad K, Sadiq K, Iqbal NT, Ali SA, Turner JR. Intestinal Epithelial Digestive, Transport, and Barrier Protein Expression Is Increased in Environmental Enteric Dysfunction. J Transl Med 2023; 103:100036. [PMID: 36870290 PMCID: PMC10121737 DOI: 10.1016/j.labinv.2022.100036] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 01/11/2023] Open
Abstract
Environmental enteric dysfunction (EED) is characterized by malabsorption and diarrhea that result in irreversible deficits in physical and intellectual growth. We sought to define the expression of transport and tight junction proteins by quantitative analysis of duodenal biopsies from patients with EED. Biopsies from Pakistani children with confirmed EED diagnoses were compared to those from age-matched North American healthy controls, patients with celiac disease, and patients with nonceliac disease with villous atrophy or intraepithelial lymphocytosis. Expression of brush border digestive and transport proteins and paracellular (tight junction) proteins was assessed by quantitative multiplex immunofluorescence microscopy. EED was characterized by partial villous atrophy and marked intraepithelial lymphocytosis. Epithelial proliferation and enteroendocrine, tuft, and Paneth cell numbers were unchanged, but there was significant goblet cell expansion in EED biopsies. Expression of proteins involved in nutrient and water absorption and that of the basolateral Cl- transport protein NKCC1 were also increased in EED. Finally, the barrier-forming tight junction protein claudin-4 (CLDN4) was significantly upregulated in EED, particularly within villous enterocytes. In contrast, expression of CFTR, CLDN2, CLDN15, JAM-A, occludin, ZO-1, and E-cadherin was unchanged. Upregulation of a barrier-forming tight junction protein and brush border and basolateral membrane proteins that support nutrient and water transport in EED is paradoxical, as their increased expression would be expected to be correlated with increased intestinal barrier function and enhanced absorption, respectively. These data suggest that EED activates adaptive intestinal epithelial responses to enhance nutrient absorption but that these changes are insufficient to restore health.
Collapse
Affiliation(s)
- Shabnam Abtahi
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anne Sailer
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Joseph T Roland
- Epithelial Biology Center, Vanderbilt University Medical Center; Nashville, Tennessee
| | - Xenia Haest
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sandra D Chanez-Paredes
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kumail Ahmad
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Kamran Sadiq
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Najeeha Talat Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - S Asad Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Department of Pathology, University of Chicago, Chicago, Illinois.
| |
Collapse
|
9
|
Horcas-Nieto JM, Versloot CJ, Langelaar-Makkinje M, Gerding A, Blokzijl T, Koster MH, Baanstra M, Martini IA, Coppes RP, Bourdon C, van Ijzendoorn SCD, Kim P, Bandsma RHJ, Bakker BM. Organoids as a model to study intestinal and liver dysfunction in severe malnutrition. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166635. [PMID: 36581145 DOI: 10.1016/j.bbadis.2022.166635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/02/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Affiliation(s)
- José M Horcas-Nieto
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Christian J Versloot
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Miriam Langelaar-Makkinje
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Albert Gerding
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Tjasso Blokzijl
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mirjam H Koster
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Mirjam Baanstra
- Department of Biomedical Sciences of Cell & Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ingrid A Martini
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Robert P Coppes
- Department of Biomedical Sciences of Cell & Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Céline Bourdon
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sven C D van Ijzendoorn
- Department of Biomedical Sciences of Cell & Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Peter Kim
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Robert H J Bandsma
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands; Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, ON, Canada.
| | - Barbara M Bakker
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands.
| |
Collapse
|
10
|
Hossain MS, Begum SMKN, Rahman MM, Parvez M, Mazumder RN, Sarker SA, Hasan MM, Fahim SM, Gazi MA, Das S, Mahfuz M, Ahmed T. Environmental enteric dysfunction and small intestinal histomorphology of stunted children in Bangladesh. PLoS Negl Trop Dis 2023; 17:e0010472. [PMID: 36656867 PMCID: PMC9888706 DOI: 10.1371/journal.pntd.0010472] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 01/31/2023] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
There is lack of information on the histological characteristics of the intestinal mucosa in Bangladeshi children. Collection of intestinal biopsy samples and assessment of the histomorphological features is considered to be the traditional gold standard for diagnosis of environmental enteric dysfunction (EED). The purpose of the study was to evaluate the intestinal histological characteristics of stunted children aged between 12-18 months with possible EED. 110 children with chronic malnutrition (52 stunted with length-for-age Z score, LAZ<-2 and 58 at risk of stunting with LAZ <-1 to -2) from the Bangladesh Environmental Enteric Dysfunction (BEED) study protocol who underwent upper gastrointestinal (GI) endoscopy were selected for this study. To explore the association of EED with childhood stunting, upper GI endoscopy was done and the biopsy specimens were studied for histopathology. Villous height and crypt depth were measured and the presence and intensity of inflammatory infiltrates in the lamina propria was investigated. Bivariate analysis was performed to examine the relationship between stunting and histologic morphology. More than 90% children irrespective of nutritional status were diagnosed to have chronic non-specific duodenitis on histopathology. Half of the children from both groups had villous atrophy as well as crypt hyperplasia and lymphocytic infiltration was present in more than 90% children, irrespective of groups. However, no statistically significant difference was observed when compared between the groups. The prevalence of chronic non-specific duodenitis in Bangladeshi children, irrespective of nutritional status, was high. A significant number of these children had abnormal findings in intestinal histomorphology. Trial registration number: ClinicalTrials.gov ID: NCT02812615 Date of first registration: 24/06/2016. https://clinicaltrials.gov/ct2/results?cond=NCT02812615&term=&cntry=&state=&city=&dist.
Collapse
Affiliation(s)
- Md. Shabab Hossain
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- * E-mail:
| | | | - M Masudur Rahman
- Department of Gastroenterology, Sheikh Russel National Gastroliver Institute and Hospital, Dhaka, Bangladesh
| | - Mashud Parvez
- Department of Pathology, Bangladesh Specialized Hospital, Dhaka, Bangladesh
| | - Ramendra Nath Mazumder
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Shafiqul Alam Sarker
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Mehedi Hasan
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Shah Mohammad Fahim
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Subhasish Das
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Public Health Nutrition, James P Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| |
Collapse
|
11
|
Diarrheal disease and gut microbiome. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 192:149-177. [DOI: 10.1016/bs.pmbts.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
12
|
Keddy KH, Saha S, Okeke IN, Kalule JB, Qamar FN, Kariuki S. Combating Childhood Infections in LMICs: evaluating the contribution of Big Data Big data, biomarkers and proteomics: informing childhood diarrhoeal disease management in Low- and Middle-Income Countries. EBioMedicine 2021; 73:103668. [PMID: 34742129 PMCID: PMC8579132 DOI: 10.1016/j.ebiom.2021.103668] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/26/2021] [Accepted: 10/20/2021] [Indexed: 01/20/2023] Open
Abstract
Despite efforts to reduce the global burden of childhood diarrhoea, 50% of all cases globally occur in children under five years in Low–Income and Middle- Income Countries (LMICs) and knowledge gaps remain regarding the aetiological diagnosis, introduction of diarrhoeal vaccines, and the role of environmental enteric dysfunction and severe acute malnutrition. Biomarkers may assist in understanding disease processes, from diagnostics, to management of childhood diarrhoea and the sequelae to vaccine development. Proteomics has the potential to assist in the identification of new biomarkers to understand the processes in the development of childhood diarrhoea and to aid in developing new vaccines. Centralised repositories that enable mining of large data sets to better characterise risk factors, the proteome of both the patient and the different diarrhoeal pathogens, and the environment, could inform patient management and vaccine development, providing a systems biological approach to address the burden of childhood diarrhoea in LMICs.
Collapse
Affiliation(s)
- Karen H Keddy
- Tuberculosis Platform, South African Medical Research Council, 1 Soutpansberg Rd, Pretoria, 0001, South Africa.
| | - Senjuti Saha
- Child Health Research Foundation, 23/2 Khilji Road, Mohammadpur, Dhaka 1207, Bangladesh
| | - Iruka N Okeke
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Oyo State, Nigeria
| | - John Bosco Kalule
- Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Uganda
| | - Farah Naz Qamar
- Department of Pediatrics and Child Health. Aga Khan University, Stadoum road Karachi, Pakistan 74800
| | - Samuel Kariuki
- Centre for Microbiology Research, Kenya Medical Research Institute, Off Mbagathi Road, Nairobi, Kenya
| |
Collapse
|
13
|
Coley EJL, Hsiao EY. Malnutrition and the microbiome as modifiers of early neurodevelopment. Trends Neurosci 2021; 44:753-764. [PMID: 34303552 DOI: 10.1016/j.tins.2021.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/23/2021] [Accepted: 06/11/2021] [Indexed: 01/16/2023]
Abstract
Malnutrition refers to a dearth, excess, or altered differential ratios of calories, macronutrients, or micronutrients. Malnutrition, particularly during early life, is a pressing global health and socioeconomic burden that is increasingly associated with neurodevelopmental impairments. Understanding how perinatal malnutrition influences brain development is crucial to uncovering fundamental mechanisms for establishing behavioral neurocircuits, with the potential to inform public policy and clinical interventions for neurodevelopmental conditions. Recent studies reveal that the gut microbiome can mediate dietary effects on host physiology and that the microbiome modulates the development and function of the nervous system. This review discusses evidence that perinatal malnutrition alters brain development and examines the maternal and neonatal microbiome as a potential contributing factor.
Collapse
Affiliation(s)
- Elena J L Coley
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Elaine Y Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
14
|
Patterson GT, Manthi D, Osuna F, Muia A, Olack B, Mbuchi M, Saldarriaga OA, Ouma L, Inziani M, Yu X, Otieno P, Melby PC. Environmental, Metabolic, and Inflammatory Factors Converge in the Pathogenesis of Moderate Acute Malnutrition in Children: An Observational Cohort Study. Am J Trop Med Hyg 2021; 104:1877-1888. [PMID: 33755580 PMCID: PMC8103470 DOI: 10.4269/ajtmh.20-0963] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/07/2021] [Indexed: 01/04/2023] Open
Abstract
Acute malnutrition affects more than 50 million children worldwide. These children are at an increased risk of morbidity and mortality from infectious disease. However, the pathogenesis of acute malnutrition and mechanisms underlying the increased risk and poor outcomes from infection are not well understood. Our objective was to identify differences in inflammation and inflammatory responses between children with moderate acute malnutrition (MAM) and healthy controls (HCs), and search for environmental, pathophysiological, and metabolic factors that may influence this response. Sixteen children with MAM and 16 HCs aged 18-36 months were studied in Nairobi, Kenya. None of the children had symptoms of an infectious disease (fever, diarrhea, or cough) in the 2 weeks before enrollment and sample collection. Demographic and health data were provided by their primary caregivers. Blood samples were collected to measure various biomarkers and the response to an inflammatory stimulus. Children with MAM were more frequently from households with contaminated water, crowding, and unstable income sources. They also had increases in basal inflammation, circulating bacterial lipopolysaccharide (LPS), markers of intestinal damage, and an exaggerated whole blood inflammatory response to LPS. Metabolic changes in children with MAM led to increased plasma levels of long-chain fatty acids, which were found to contribute to the pro-inflammatory state. These exploratory findings suggest convergence of multiple factors to promote dysregulated inflammatory responses and prompt several mechanistic hypotheses that can be pursued to better understand the pathogenesis of MAM.
Collapse
Affiliation(s)
- Grace T. Patterson
- Department of Internal Medicine and Infectious Disease, University of Texas Medical Branch, Galveston, Texas
| | - Dennis Manthi
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Finnley Osuna
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Alfred Muia
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Beatrice Olack
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Margaret Mbuchi
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Omar A. Saldarriaga
- Department of Internal Medicine and Infectious Disease, University of Texas Medical Branch, Galveston, Texas
| | - Linet Ouma
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Mary Inziani
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Xiaoying Yu
- Department of Preventative Medicine and Population Health, University of Texas Medical Branch, Galveston, Texas
| | - Phelgona Otieno
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya;,Address correspondence to Phelgona Otieno, Kenya Medical Research Institute, Mbagathi Road, Nairobi, Kenya, E-mail: or Peter C. Melby, Department of Internal Medicine and Infectious Disease, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, E-mail:
| | - Peter C. Melby
- Department of Internal Medicine and Infectious Disease, University of Texas Medical Branch, Galveston, Texas;,Address correspondence to Phelgona Otieno, Kenya Medical Research Institute, Mbagathi Road, Nairobi, Kenya, E-mail: or Peter C. Melby, Department of Internal Medicine and Infectious Disease, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, E-mail:
| |
Collapse
|
15
|
Diversifying the Utilization of Maize at Household Level in Zambia: Quality and Consumer Preferences of Maize-Based Snacks. Foods 2021; 10:foods10040750. [PMID: 33916052 PMCID: PMC8067053 DOI: 10.3390/foods10040750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 11/21/2022] Open
Abstract
This study evaluated the nutritional, antinutritional properties, and consumer preferences of five maize-based snacks at the household level. The physical, nutritional, and antinutritional properties were analyzed with standard laboratory methods, while a structured questionnaire was used for the data collection on consumer preferences of the maize products. There were significant (p < 0.05) differences in the proximate parameters of the maize snack samples. Antinutritional properties among maize snacks all fell within the permissible range. Respondents from all districts showed no significant (p > 0.05) differences in maize chin-chin variants’ and maize finger variants’ except for Serenje and Mkushi districts where maize chin-chin and maize finger showed significant (p < 0.05) differences in their sensory ratings. However, across districts, the most rated maize finger variant was the spiced 100% maize finger. In conclusion, maize-based snacks enriched with soybean flour have proven nutritious with a reasonable acceptability level.
Collapse
|
16
|
Singh A, Ghosh S, Ward H, Manary MJ, Rogers BL, Rosenberg IH. Biomarkers of environmental enteric dysfunction are differently associated with recovery and growth among children with moderate acute malnutrition in Sierra Leone. Am J Clin Nutr 2021; 113:1556-1564. [PMID: 33668048 PMCID: PMC8243924 DOI: 10.1093/ajcn/nqaa434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) may influence growth during and recovery from moderate acute malnutrition (MAM), however, biomarkers to assess these relations have yet to be identified. OBJECTIVES The objectives of this study were to: 1) develop a score for EED based on host fecal mRNA transcripts, 2) compare biomarkers of EED with each other, and 3) examine associations between the EED biomarkers and recovery from MAM and growth outcomes. METHODS In a cohort of 520 Sierra Leonean MAM children, biomarkers of EED included the lactulose: mannitol (L: M) test, 15 host fecal mRNA transcripts, and host fecal proteins [α-1-antitrypsin (AAT), myeloperoxidase (MPO), neopterin (NEO)]. Anthropometry data were also collected and z scores were computed for length-for-age (LAZ) and weight-for-length (WLZ). Recovery from MAM was defined as midupper arm circumference ≥12.5 cm. Factor analysis was used to identify EED scores using the mRNA transcripts, and mixed effects regression was conducted to test for associations. RESULTS The 15 host fecal mRNA transcripts were clustered into 3 scores: gut inflammation (GI) score, gut structure (GS) score, and gut defense (GD) score. We found agreement between certain inflammation markers (GI score and MPO), and permeability markers (GS score and AAT; AAT and the L: M excretion ratio). Antimicrobial gut defense (GD score) was inversely associated with percent lactulose excreted, a measure of intestinal permeability. LAZ (β: -0.08; 95% CI: -0.14, -0.02) and WLZ (β: -0.03; 95% CI: -0.06, -0.01) were negatively associated with GI score. A high GD score (β: 0.36; 95% CI: 0.08, 0.64) and low AAT (β: -1.35; 95% CI: -2.35, -0.36) were associated with recovery from MAM. CONCLUSIONS Scores derived from host fecal mRNA transcript variably correlated with the L: M test and host fecal proteins. Markers of intestinal inflammation, permeability, and defense were associated with growth outcomes and recovery from MAM.
Collapse
Affiliation(s)
| | - Shibani Ghosh
- Friedman School of Nutrition Science and Policy, Tufts
University, Boston, MA, USA
| | - Honorine Ward
- Tufts Medical Center and Tufts University School of
Medicine, Boston, MA, USA
| | - Mark J Manary
- Department of Pediatrics, Washington University,
St Louis, MO, USA
| | - Beatrice L Rogers
- Friedman School of Nutrition Science and Policy, Tufts
University, Boston, MA, USA
| | - Irwin H Rosenberg
- Friedman School of Nutrition Science and Policy, Tufts
University, Boston, MA, USA
| |
Collapse
|
17
|
Hossain MS, Begum SMKN, Rahman MM, Mazumder RN, Parvez M, Gazi MA, Hasan MM, Fahim SM, Das S, Mahfuz M, Sarker SA, Ahmed T. Alterations in the histological features of the intestinal mucosa in malnourished adults of Bangladesh. Sci Rep 2021; 11:2355. [PMID: 33504937 PMCID: PMC7840936 DOI: 10.1038/s41598-021-82079-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
There is paucity of knowledge on the histological features of the intestinal mucosa in malnourished adults of Bangladesh. The purpose of the study was to explore the histological features of the intestinal mucosa in malnourished adults of Bangladesh and to compare the findings with their well-nourished counterparts. 64 adults (37 malnourished with body mass index, BMI < 18.5 kg/m2 and 27 controls with BMI > 18.5 kg/m2) from the Bangladesh Environmental Enteric Dysfunction (BEED) study, who underwent upper-gastrointestinal endoscopy, were selected for this study. With a view to address the association of environmental enteric dysfunction (EED) with malnutrition, upper-gastrointestinal endoscopy was performed and mucosal biopsies from the distal duodenum were studied for histopathology. Villous height, crypt depth, and presence of inflammatory infiltrates in lamina propria were investigated. Bivariate analysis was performed to quantify the relation between malnutrition and the histological features. About 95% adults, irrespective of nutritional status, were diagnosed to have chronic non-specific duodenitis on histopathology. Malnourished adults suffered significantly more from chronic active duodenitis compared to their well-nourished counterparts (p = 0.003). Malnourished adults also had significantly higher frequency of subtotal villous atrophy, crypt hyperplasia and marked cellular infiltration in the lamina propria than the healthy controls (p < 0.05).
Collapse
Affiliation(s)
- Md Shabab Hossain
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh.
| | | | - M Masudur Rahman
- Department of Gastroenterology, Sheikh Russel National Gastroliver Institute and Hospital, Dhaka, 1212, Bangladesh
| | - Ramendra Nath Mazumder
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Mashud Parvez
- Department of Pathology, Bangladesh Specialized Hospital, Dhaka, 1207, Bangladesh
| | - Md Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Mehedi Hasan
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Shah Mohammad Fahim
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Subhasish Das
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Shafiqul Alam Sarker
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| |
Collapse
|
18
|
Selimoglu MA, Kansu A, Aydogdu S, Sarioglu AA, Erdogan S, Dalgic B, Yuce A, Cullu Cokugras F. Nutritional Support in Malnourished Children With Compromised Gastrointestinal Function: Utility of Peptide-Based Enteral Therapy. Front Pediatr 2021; 9:610275. [PMID: 34164352 PMCID: PMC8215107 DOI: 10.3389/fped.2021.610275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 05/07/2021] [Indexed: 12/20/2022] Open
Abstract
This review focuses on nutritional support in malnourished children with compromised gastrointestinal function addressing the interplay between malnutrition and gastrointestinal dysfunction, and the specific role of peptide-based enteral therapy in pediatric malnutrition. Malnutrition is associated with impaired gut functions such as increased intestinal permeability, malabsorption, and diarrhea, while pre-existing functional gastrointestinal disorders may also lead to malnutrition. Presence of compromised gastrointestinal function in malnourished children is critical given that alterations such as malabsorption and increased intestinal permeability directly interfere with efficacy of nutritional support and recovery from malnutrition. Appropriate nutritional intervention is the key step in the management of malnutrition, while alterations in gastrointestinal functions in malnourished children are likely even in those with mild degree malnutrition. Therefore, nutritional therapy in children with compromised gastrointestinal function is considered to involve gut-protective interventions that address the overlapping and interacting effects of diarrhea, enteropathy and malnutrition to improve child survival and developmental potential in the long-term. Peptide-based enteral formulas seem to have clinical applications in malnourished children with compromised gastrointestinal function, given their association with improved gastrointestinal tolerance and absorption, better nitrogen retention/ balance, reduced diarrhea and bacterial translocation, enhanced fat absorption, and maintained/restored gut integrity as compared with free amino acid or whole-protein formulas.
Collapse
Affiliation(s)
- Mukadder Ayse Selimoglu
- Department of Pediatric Gastroenterology, Atasehir and Bahcelievler Memorial Hospitals, Istanbul, Turkey
| | - Aydan Kansu
- Department of Pediatric Gastroenterology, Ankara University School of Medicine, Ankara, Turkey
| | - Sema Aydogdu
- Department of Pediatric Gastroenterology, Ege University Faculty of Medicine, Izmir, Turkey
| | | | | | - Buket Dalgic
- Department of Pediatric Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Aysel Yuce
- Department of Pediatric Gastroenterology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Fugen Cullu Cokugras
- Department of Pediatric Gastroenterology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
19
|
Som SV, Van Der Hoeven M, Laillou A, Poirot E, Chan T, Polman K, Ponce MC, Wieringa FT. Adherence to Child Feeding Practices and Child Growth: A Retrospective Cohort Analysis in Cambodia. Nutrients 2020; 13:nu13010137. [PMID: 33396559 PMCID: PMC7823716 DOI: 10.3390/nu13010137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022] Open
Abstract
Appropriate feeding in the first 1000 days of a child’s life is critical for their health and growth. We determined associations between adherence to age-appropriate feeding practices and child growth in Cambodia. Children (n = 1079) were included in the first follow-up (FU) data analyses and followed for 30 months (six FUs). Data were analyzed by generalized linear mixed-effect models. Children who adhered to feeding practices on at least three FUs, with an adequate minimal dietary diversity (MDD), a minimal acceptable diet (MAD), and age-appropriate daily feeding (ADF) were less stunted (14.8%, 12.3%, and 6.4%, respectively) than children who never adhered to these indicators (25.2%, 30.1%, and 24.8%, respectively). A higher adherence to MDD and ADF was associated with a higher height-for-age Z-score (HAZ) (β: 0.13, 95% CI: 0.01–0.25 and β: 0.36, 95% CI: 0.22–0.50), while a higher adherence to the MDD and MAD was associated with a higher weight-for-height Z-score (WHZ) (β: 0.19, 95% CI: 0.08–0.30; and β: 0.16, 95% CI: 0.05–0.27). A higher adherence to a minimum meal frequency (MMF) was associated with a lower HAZ (β: −0.99, 95% CI: −1.28–−0.70). Our findings showed that to reduce wasting and stunting in Cambodia, interventions should focus on improving both the quality and quantity of food intake of children under two while targeting the whole complementary feeding period.
Collapse
Affiliation(s)
- Somphos Vicheth Som
- Section of Infectious Diseases, Department of Health Sciences, Vrije Universiteit Amsterdam, de Boelelaan 1091, 1081 HV Amsterdam, The Netherlands; (M.V.D.H.); (K.P.); (M.C.P.)
- Correspondence: ; Tel.: +31-20-59-84395
| | - Marinka Van Der Hoeven
- Section of Infectious Diseases, Department of Health Sciences, Vrije Universiteit Amsterdam, de Boelelaan 1091, 1081 HV Amsterdam, The Netherlands; (M.V.D.H.); (K.P.); (M.C.P.)
| | - Arnaud Laillou
- Department of Child Survival and Development, United Nations Children’s Fund Cambodia, Exchange Square Building, Phnom Penh 12101, Cambodia; (A.L.); (E.P.)
| | - Etienne Poirot
- Department of Child Survival and Development, United Nations Children’s Fund Cambodia, Exchange Square Building, Phnom Penh 12101, Cambodia; (A.L.); (E.P.)
| | - Theary Chan
- Reproductive and Child Health Alliance, Phnom Penh 12100, Cambodia;
| | - Katja Polman
- Section of Infectious Diseases, Department of Health Sciences, Vrije Universiteit Amsterdam, de Boelelaan 1091, 1081 HV Amsterdam, The Netherlands; (M.V.D.H.); (K.P.); (M.C.P.)
- Department of Biomedical Sciences, Institute of Tropical Medicine, Nationalestraat 155, 2000 Antwerp, Belgium
| | - Maiza Campos Ponce
- Section of Infectious Diseases, Department of Health Sciences, Vrije Universiteit Amsterdam, de Boelelaan 1091, 1081 HV Amsterdam, The Netherlands; (M.V.D.H.); (K.P.); (M.C.P.)
| | - Frank T. Wieringa
- UMR Qualisud, French National Research Institute for Sustainable Development (IRD), IRD/CIRAD/Université de Montpellier/SupAgro, 911 avenue d’Agropolis, 34394 CEDEX 5 Montpellier, France;
| |
Collapse
|
20
|
Martinez EE, Zurakowski D, Pereira L, Freire R, Emans JB, Nurko S, Duggan CP, Fasano A, Mehta NM. Interleukin-10 and Zonulin Are Associated With Postoperative Delayed Gastric Emptying in Critically Ill Surgical Pediatric Patients: A Prospective Pilot Study. JPEN J Parenter Enteral Nutr 2020; 44:1407-1416. [PMID: 32386238 PMCID: PMC7754495 DOI: 10.1002/jpen.1874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND Impaired gastric emptying (GE) is associated with morbidity in surgical critically ill children. The relationship between inflammation, gut barrier integrity (lipopolysaccharide binding protein [LBP]; zonulin), and GE has not been described in this cohort. METHODS Children ≥2 years of age and requiring critical care after surgery were enrolled. Preoperative and postoperative levels of serum cytokines, LBP, and zonulin, and GE by the acetaminophen absorption test, were measured, allowing patients to serve as their own controls. Postoperative delayed GE was defined as a decrease in GE by ≥20% compared with preoperative GE. The following were examined : comparison between postoperative andpreoperative values, correlations between fold change (postoperative/preoperative) in study variables, and fold change in study variables between patients with and without postoperative delayed GE. RESULTS Twenty patients, median age 14 years (12.25, 18), 12 female, were included. Eight of 20 patients had postoperative delayed GE. Postoperative interleukin-6 (IL-6), IL-8, IL-10, and LBP were increased, and zonulin was decreased (P-values < .05). Fold change in IL-10 and zonulin were inversely correlated (ρ -0.618, P = .004). Patients with postoperative delayed GE had greater fold increase in IL-10 (P = .0159) and fold decrease in zonulin (P = .0160). Five of 7 (71%) patients with both fold increase in IL-10 and decrease in zonulin had delayed GE. CONCLUSION Postoperative changes in IL-10 and zonulin were associated with delayed GE in surgical critically ill children, which might suggest a mechanism to for delayed GE in postoperative inflammation and gut barrier dysregulation after surgery.
Collapse
Affiliation(s)
- Enid E. Martinez
- Department of Anesthesiology, Critical Care and Pain MedicineBoston Children's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - David Zurakowski
- Department of Anesthesiology, Critical Care and Pain MedicineBoston Children's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Luis Pereira
- Department of Anesthesiology, Critical Care and Pain MedicineBoston Children's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Rachel Freire
- Division of Pediatric Gastroenterology and NutritionDepartment of PediatricsMassachusetts General Hospital for ChildrenBostonMassachusettsUSA
| | - John B. Emans
- Harvard Medical SchoolBostonMassachusettsUSA
- Orthopedic CenterBoston Children's HospitalBostonMassachusettsUSA
| | - Samuel Nurko
- Harvard Medical SchoolBostonMassachusettsUSA
- Division of Gastroenterology, Hepatology and NutritionBoston Children's HospitalBostonMassachusettsUSA
| | - Christopher P. Duggan
- Harvard Medical SchoolBostonMassachusettsUSA
- Division of Gastroenterology, Hepatology and NutritionBoston Children's HospitalBostonMassachusettsUSA
- Center for NutritionBoston Children's HospitalBostonMassachusettsUSA
| | - Alessio Fasano
- Harvard Medical SchoolBostonMassachusettsUSA
- Division of Pediatric Gastroenterology and NutritionDepartment of PediatricsMassachusetts General Hospital for ChildrenBostonMassachusettsUSA
| | - Nilesh M. Mehta
- Department of Anesthesiology, Critical Care and Pain MedicineBoston Children's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Center for NutritionBoston Children's HospitalBostonMassachusettsUSA
| |
Collapse
|
21
|
Giallourou N, Fardus-Reid F, Panic G, Veselkov K, McCormick BJJ, Olortegui MP, Ahmed T, Mduma E, Yori PP, Mahfuz M, Svensen E, Ahmed MMM, Colston JM, Kosek MN, Swann JR. Metabolic maturation in the first 2 years of life in resource-constrained settings and its association with postnatal growths. SCIENCE ADVANCES 2020; 6:eaay5969. [PMID: 32284996 PMCID: PMC7141821 DOI: 10.1126/sciadv.aay5969] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 01/14/2020] [Indexed: 05/10/2023]
Abstract
Malnutrition continues to affect the growth and development of millions of children worldwide, and chronic undernutrition has proven to be largely refractory to interventions. Improved understanding of metabolic development in infancy and how it differs in growth-constrained children may provide insights to inform more timely, targeted, and effective interventions. Here, the metabolome of healthy infants was compared to that of growth-constrained infants from three continents over the first 2 years of life to identify metabolic signatures of aging. Predictive models demonstrated that growth-constrained children lag in their metabolic maturity relative to their healthier peers and that metabolic maturity can predict growth 6 months into the future. Our results provide a metabolic framework from which future nutritional programs may be more precisely constructed and evaluated.
Collapse
Affiliation(s)
- N. Giallourou
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, London, UK
| | - F. Fardus-Reid
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, London, UK
| | - G. Panic
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, London, UK
| | - K. Veselkov
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, London, UK
| | | | - M. P. Olortegui
- Asociación Benéfica PRISMA, Unidad de Investigación Biomedica, Iquitos, Peru
| | - T. Ahmed
- International Center for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - E. Mduma
- Haydom Global Health Institute, Haydom, Tanzania
| | - P. P. Yori
- Asociación Benéfica PRISMA, Unidad de Investigación Biomedica, Iquitos, Peru
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - M. Mahfuz
- International Center for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - E. Svensen
- Haydom Global Health Institute, Haydom, Tanzania
- Haukeland University Hospital, Bergen, Norway
| | - M. M. M. Ahmed
- International Center for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - J. M. Colston
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - M. N. Kosek
- Asociación Benéfica PRISMA, Unidad de Investigación Biomedica, Iquitos, Peru
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
- Corresponding author.
| | - J. R. Swann
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, London, UK
- School of Human Development and Health, Faculty of Medicine, University of Southampton, UK
| |
Collapse
|
22
|
Tran H, Lee D, Petnic SE, Bozzini JA, Lu S. Peanut butter-based formulations of amoxicillin for pediatric applications. Int Health 2020; 12:43-49. [PMID: 31365083 PMCID: PMC6964218 DOI: 10.1093/inthealth/ihz031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/08/2019] [Accepted: 04/09/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Child mortality is a major global health challenge, especially in regions of limited resources. Accessibility to lifesaving medicine and adequate nutrition is essential to reduce child mortality and improve the health and well-being of the world's most vulnerable children. METHODS We have developed NutMox, a novel pediatric formulation of the β-lactam antibiotic amoxicillin in a matrix of peanut-based ready-to-use therapeutic food (RUTF) consisting of peanut butter, sugar, vegetable oil, dry milk and vitamins. NutMox is ready to use and thermostable, requires no chewing or pill swallowing and provides both an antibiotic and nutrition. RESULTS Amoxicillin in NutMox formulations was stable for at least 12 months at storage temperatures of 4°C, 25°C and 37°C. Amoxicillin formulated in NutMox displayed similar pharmacokinetics in mice to that in suspension. CONCLUSIONS Our results demonstrated the feasibility of a peanut butter-based matrix for pediatric formulations of amoxicillin, suggesting that such a matrix can serve as a base for delivering medications in addition to its current use as an RUTF.
Collapse
Affiliation(s)
- Helen Tran
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA, USA
| | - Danny Lee
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA, USA
| | - Sarah E Petnic
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA, USA
| | - Julianne A Bozzini
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA, USA
| | - Sangwei Lu
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA, USA
| |
Collapse
|
23
|
Salameh E, Morel FB, Zeilani M, Déchelotte P, Marion-Letellier R. Animal Models of Undernutrition and Enteropathy as Tools for Assessment of Nutritional Intervention. Nutrients 2019; 11:nu11092233. [PMID: 31527523 PMCID: PMC6770013 DOI: 10.3390/nu11092233] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/24/2019] [Accepted: 09/08/2019] [Indexed: 02/06/2023] Open
Abstract
: Undernutrition is a major public health problem leading to 1 in 5 of all deaths in children under 5 years. Undernutrition leads to growth stunting and/or wasting and is often associated with environmental enteric dysfunction (EED). EED mechanisms leading to growth failure include intestinal hyperpermeability, villus blunting, malabsorption and gut inflammation. As non-invasive methods for investigating gut function in undernourished children are limited, pre-clinical models are relevant to elucidating the pathophysiological processes involved in undernutrition and EED, and to identifying novel therapeutic strategies. In many published models, undernutrition was induced using protein or micronutrient deficient diets, but these experimental models were not associated with EED. Enteropathy models mainly used gastrointestinal injury triggers. These models are presented in this review. We found only a few studies investigating the combination of undernutrition and enteropathy. This highlights the need for further developments to establish an experimental model reproducing the impact of undernutrition and enteropathy on growth, intestinal hyperpermeability and inflammation, that could be suitable for preclinical evaluation of innovative therapeutic intervention.
Collapse
Affiliation(s)
- Emmeline Salameh
- UniRouen, Inserm UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France.
- Nutriset SAS, 76770 Malaunay, France.
| | | | | | - Pierre Déchelotte
- UniRouen, Inserm UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France.
- Department of Nutrition, Rouen University Hospital, 76183 Rouen, France.
| | - Rachel Marion-Letellier
- UniRouen, Inserm UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France.
| |
Collapse
|
24
|
Hossain MS, Das S, Gazi MA, Alam MA, Haque NMS, Mahfuz M, Ahmed T, Damman CJ. Association of faecal pH with childhood stunting: Results from a cross-sectional study. BMJ Paediatr Open 2019; 3:e000549. [PMID: 31646200 PMCID: PMC6782033 DOI: 10.1136/bmjpo-2019-000549] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Gut microbiota plays an important role in the growth of children. The gut of children with optimum growth is enriched in certain species, especially Bifidobacteria and Clostridia. Bifidobacteria and commensal Clostridia both contribute to formation of acidic stool, and an elevated faecal pH indicates reduction of these species in the gut. The purpose of the study was to investigate the association of faecal pH with childhood stunting. METHODS In this cross-sectional study, 100 children with length-for-age Z score (LAZ) <-1 aged between 12 and 18 months were enrolled from the ongoing Bangladesh Environmental Enteric Dysfunction study conducted in Dhaka, Bangladesh. LAZ was measured by anthropometry and data on factors affecting linear growth were recorded. Faecal pH measurement was done using pH metre on freshly collected non-diarrhoeal faecal samples following standard procedure. Multiple quantile regression was done to quantify the relation between faecal pH and LAZ scores. RESULTS The mean LAZ and faecal pH of the children were -2.12±0.80 and 5.84±1.11, respectively. Pearson correlation analysis showed a statistically significant negative correlation between stool pH and the LAZ scores (p<0.01). After inclusion of other factors affecting linear growth into the regression model, a statistically significant inverse association was observed between faecal pH and LAZ score (p<0.01). CONCLUSION Elevated faecal pH was found to have a significant association with stunted growth. As an indicator of gut microbiota status, faecal pH might have emerged as a possible indirect determinant of childhood stunting. TRIAL REGISTRATION NUMBER NCT02812615.
Collapse
Affiliation(s)
- Md. Shabab Hossain
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Subhasish Das
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Md. Amran Gazi
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Md. Ashraful Alam
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Nur Muhammad Shahedul Haque
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Chris J Damman
- Division of Gastroenterology, University of Washington, Seattle, Washington, USA
- Bill and Melinda Gates Foundation, Seattle, Washington, USA
| |
Collapse
|
25
|
Bourke CD, Jones KDJ, Prendergast AJ. Current Understanding of Innate Immune Cell Dysfunction in Childhood Undernutrition. Front Immunol 2019; 10:1728. [PMID: 31417545 PMCID: PMC6681674 DOI: 10.3389/fimmu.2019.01728] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022] Open
Abstract
Undernutrition affects millions of children in low- and middle-income countries (LMIC) and underlies almost half of all deaths among children under 5 years old. The growth deficits that characterize childhood undernutrition (stunting and wasting) result from simultaneous underlying defects in multiple physiological processes, and current treatment regimens do not completely normalize these pathways. Most deaths among undernourished children are due to infections, indicating that their anti-pathogen immune responses are impaired. Defects in the body's first-line-of-defense against pathogens, the innate immune system, is a plausible yet understudied pathway that could contribute to this increased infection risk. In this review, we discuss the evidence for innate immune cell dysfunction from cohort studies of childhood undernutrition in LMIC, highlighting knowledge gaps in almost all innate immune cell types. We supplement these gaps with insights from relevant experimental models and make recommendations for how human and animal studies could be improved. A better understanding of innate immune function could inform future tractable immune-targeted interventions for childhood undernutrition to reduce mortality and improve long-term health, growth and development.
Collapse
Affiliation(s)
- Claire D Bourke
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, London, United Kingdom.,Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Kelsey D J Jones
- Kennedy Institute for Rheumatology, University of Oxford, Oxford, United Kingdom.,Department of Paediatric Gastroenterology & Nutrition, University of Oxford NHS Foundation Trust, Oxford, United Kingdom
| | - Andrew J Prendergast
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, London, United Kingdom.,Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| |
Collapse
|
26
|
Daniel AI, Kvissberg MEA, Senga E, Versloot CJ, Harawa PP, Voskuijl W, Wishart D, Mandal R, Bandsma R, Bourdon C. Urinary Organic Acids Increase After Clinical Stabilization of Hospitalized Children With Severe Acute Malnutrition. Food Nutr Bull 2019; 40:532-543. [PMID: 31303023 DOI: 10.1177/0379572119853930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Despite a reduction of child mortality in low-income countries, acutely ill undernourished children still have an elevated risk of death. Those at highest risk are children with severe acute malnutrition (SAM) who often show metabolic dysregulation that remains poorly understood. OBJECTIVE We performed a pilot study to examine changes in urinary organic acids during nutritional rehabilitation of children with SAM, and to identify metabolites associated with the presence of edema or with mortality. METHODS This study included 76 children aged between 6 and 60 months, hospitalized for SAM at the Moyo Nutritional Rehabilitation and Research Unit in Blantyre, Malawi. Urine was collected at admission and 3 days after clinical stabilization and metabolomics were performed using gas chromatography-mass spectrometry. Metabolite concentrations were evaluated with both uni- and multivariate approaches. RESULTS Most metabolites increased 3 days after clinical stabilization, and total urinary concentration changed from 1.2 mM (interquartile range [IQR], 0.78-1.7) at admission to 3.8 mM (IQR, 2.1-6.6) after stabilization (P < .0001). In particular, 6 metabolites showed increases: 3-hydroxybutyric, 4-hydroxyhippuric, p-hydroxyphenylacetic, oxoglutaric, succinic, and lactic acids. Urinary creatinine was low at both time points, but levels did increase from 0.63 mM (IQR, 0.2-1.2) to 2.6 mM (IQR,1.6-4.4; P < .0001). No differences in urinary profiles were found between children who died versus those who survived, nor between children with severe wasting or edematous SAM. CONCLUSIONS Total urinary metabolites and creatinine increase after stabilization and may reflect partial recovery of overall metabolism linked to refeeding. The use of urinary metabolites for risk assessment should be furthered explored. TRIAL REGISTRATION TranSAM study (ISRCTN13916953).
Collapse
Affiliation(s)
- Allison I Daniel
- Hospital for Sick Children, Toronto, Canada.,Faculty of Medicine, University of Toronto, Toronto, Canada.,Allison I. Daniel and Matilda E. Arvidsson Kvissberg are co-first authors
| | - Matilda E Arvidsson Kvissberg
- Hospital for Sick Children, Toronto, Canada.,University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Allison I. Daniel and Matilda E. Arvidsson Kvissberg are co-first authors
| | - Edward Senga
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Christian J Versloot
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Wieger Voskuijl
- College of Medicine, University of Malawi, Blantyre, Malawi.,The Childhood Acute Illness and Nutrition Network, Narobi, Kenya.,Academic Medical Centre, University of Amsterdam, The Netherlands
| | | | | | - Robert Bandsma
- Hospital for Sick Children, Toronto, Canada.,Faculty of Medicine, University of Toronto, Toronto, Canada.,University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,College of Medicine, University of Malawi, Blantyre, Malawi.,The Childhood Acute Illness and Nutrition Network, Narobi, Kenya
| | - Céline Bourdon
- Hospital for Sick Children, Toronto, Canada.,The Childhood Acute Illness and Nutrition Network, Narobi, Kenya
| |
Collapse
|
27
|
A review of GI conditions critical to oral drug absorption in malnourished children. Eur J Pharm Biopharm 2019; 137:9-22. [DOI: 10.1016/j.ejpb.2019.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/30/2019] [Accepted: 02/03/2019] [Indexed: 02/06/2023]
|
28
|
Bwakura-Dangarembizi M, Amadi B, Bourke CD, Robertson RC, Mwapenya B, Chandwe K, Kapoma C, Chifunda K, Majo F, Ngosa D, Chakara P, Chulu N, Masimba F, Mapurisa I, Besa E, Mutasa K, Mwakamui S, Runodamoto T, Humphrey JH, Ntozini R, Wells JCK, Manges AR, Swann JR, Walker AS, Nathoo KJ, Kelly P, Prendergast AJ. Health Outcomes, Pathogenesis and Epidemiology of Severe Acute Malnutrition (HOPE-SAM): rationale and methods of a longitudinal observational study. BMJ Open 2019; 9:e023077. [PMID: 30782694 PMCID: PMC6361330 DOI: 10.1136/bmjopen-2018-023077] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 10/25/2018] [Accepted: 11/07/2018] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Mortality among children hospitalised for complicated severe acute malnutrition (SAM) remains high despite the implementation of WHO guidelines, particularly in settings of high HIV prevalence. Children continue to be at high risk of morbidity, mortality and relapse after discharge from hospital although long-term outcomes are not well documented. Better understanding the pathogenesis of SAM and the factors associated with poor outcomes may inform new therapeutic interventions. METHODS AND ANALYSIS The Health Outcomes, Pathogenesis and Epidemiology of Severe Acute Malnutrition (HOPE-SAM) study is a longitudinal observational cohort that aims to evaluate the short-term and long-term clinical outcomes of HIV-positive and HIV-negative children with complicated SAM, and to identify the risk factors at admission and discharge from hospital that independently predict poor outcomes. Children aged 0-59 months hospitalised for SAM are being enrolled at three tertiary hospitals in Harare, Zimbabwe and Lusaka, Zambia. Longitudinal mortality, morbidity and nutritional data are being collected at admission, discharge and for 48 weeks post discharge. Nested laboratory substudies are exploring the role of enteropathy, gut microbiota, metabolomics and cellular immune function in the pathogenesis of SAM using stool, urine and blood collected from participants and from well-nourished controls. ETHICS AND DISSEMINATION The study is approved by the local and international institutional review boards in the participating countries (the Joint Research Ethics Committee of the University of Zimbabwe, Medical Research Council of Zimbabwe and University of Zambia Biomedical Research Ethics Committee) and the study sponsor (Queen Mary University of London). Caregivers provide written informed consent for each participant. Findings will be disseminated through peer-reviewed journals, conference presentations and to caregivers at face-to-face meetings.
Collapse
Affiliation(s)
- Mutsa Bwakura-Dangarembizi
- Department of Paediatrics and Child Health, University of Zimbabwe College of Health Sciences, Harare, Zimbabwe
| | - Beatrice Amadi
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Claire D Bourke
- Blizard Institute, Queen Mary University of London, London, UK
| | | | - Benjamin Mwapenya
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Kanta Chandwe
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Chanda Kapoma
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Kapula Chifunda
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Florence Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Deophine Ngosa
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Pamela Chakara
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Nivea Chulu
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Faithfull Masimba
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Idah Mapurisa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Ellen Besa
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Simutanyi Mwakamui
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | | | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | - Amee R Manges
- University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - Kusum J Nathoo
- Department of Paediatrics and Child Health, University of Zimbabwe College of Health Sciences, Harare, Zimbabwe
| | - Paul Kelly
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
- Blizard Institute, Queen Mary University of London, London, UK
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London, UK
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | |
Collapse
|
29
|
Bartelt LA, Bolick DT, Guerrant RL. Disentangling Microbial Mediators of Malnutrition: Modeling Environmental Enteric Dysfunction. Cell Mol Gastroenterol Hepatol 2019; 7:692-707. [PMID: 30630118 PMCID: PMC6477186 DOI: 10.1016/j.jcmgh.2018.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022]
Abstract
Environmental enteric dysfunction (EED) (also referred to as environmental enteropathy) is a subclinical chronic intestinal disorder that is an emerging contributor to early childhood malnutrition. EED is common in resource-limited settings, and is postulated to consist of small intestinal injury, dysfunctional nutrient absorption, and chronic inflammation that results in impaired early child growth attainment. Although there is emerging interest in the hypothetical potential for chemical toxins in the environmental exposome to contribute to EED, the propensity of published data, and hence the focus of this review, implicates a critical role of environmental microbes. Early childhood malnutrition and EED are most prevalent in resource-limited settings where food is limited, and inadequate access to clean water and sanitation results in frequent gastrointestinal pathogen exposures. Even as overt diarrhea rates in these settings decline, silent enteric infections and faltering growth persist. Furthermore, beyond restricted physical growth, EED and/or enteric pathogens also associate with impaired oral vaccine responses, impaired cognitive development, and may even accelerate metabolic syndrome and its cardiovascular consequences. As these potentially costly long-term consequences of early childhood enteric infections increasingly are appreciated, novel therapeutic strategies that reverse damage resulting from nutritional deficiencies and microbial insults in the developing small intestine are needed. Given the inherent limitations in investigating how specific intestinal pathogens directly injure the small intestine in children, animal models provide an affordable and controlled opportunity to elucidate causal sequelae of specific enteric infections, to differentiate consequences of defined nutrient deprivation alone from co-incident enteropathogen insults, and to correlate the resulting gut pathologies with their functional impact during vulnerable early life windows.
Collapse
Affiliation(s)
- Luther A Bartelt
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Gastrointestinal Biology and Disease, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - David T Bolick
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Richard L Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
30
|
Oriá RB, Malva JO, Foley PL, Freitas RS, Bolick DT, Guerrant RL. Revisiting Inbred Mouse Models to Study the Developing Brain: The Potential Role of Intestinal Microbiota. Front Hum Neurosci 2018; 12:358. [PMID: 30283311 PMCID: PMC6156437 DOI: 10.3389/fnhum.2018.00358] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/20/2018] [Indexed: 12/23/2022] Open
Affiliation(s)
- Reinaldo B Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - João O Malva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Patricia L Foley
- Division of Comparative Medicine, Department of Microbiology and Immunology, Georgetown University, Washington, DC, United States
| | - Raul S Freitas
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - David T Bolick
- Division of Infectious Diseases and International Health, Center for Global Health, University of Virginia, Charlottesville, VA, United States
| | - Richard L Guerrant
- Division of Infectious Diseases and International Health, Center for Global Health, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
31
|
Panic G, Coulibaly JT, Harvey N, Keiser J, Swann J. Characterizing the Biochemical Response to Schistosoma mansoni Infection and Treatment with Praziquantel in Preschool and School Aged Children. J Proteome Res 2018; 17:2028-2033. [PMID: 29701975 DOI: 10.1021/acs.jproteome.7b00910] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Schistosomiasis is a widespread chronic neglected tropical disease prevalent mostly in children in under-resourced rural areas. Its pathological effects have been clinically characterized, yet the molecular-level effects are understudied. In this study, the biochemical effects of Schistosoma mansoni infection and praziquantel treatment were studied in 130 preschool aged and 159 school aged infected children and 11 noninfected children in Azaguié, Côte d'Ivoire. Urine samples were collected prior to receiving 20, 40, or 60 mg/kg of praziquantel or a placebo, as well as 24 h post-treatment, and at the 3-week follow up. Urinary metabolic phenotypes were measured using 1H NMR spectroscopy, and metabolic variation associated with S. mansoni infection and praziquantel administration was identified using multivariate statistical techniques. Discriminatory metabolic signatures were detected between heavily infected and noninfected children at baseline as well as according to the dose of praziquantel administered 24 h post treatment. These signatures were primarily associated with the metabolic activity of the gut microbiota, gut health and growth biomarkers and energy and liver metabolism. These analyses provide insights into the metabolic phenotype of schistosomiasis and treatment with praziquantel in two important demographics.
Collapse
Affiliation(s)
- Gordana Panic
- Department of Medical Parasitology and Infection Biology , Swiss Tropical and Public Health Institute , CH-4002 Basel , Switzerland.,University of Basel , CH-4003 Basel , Switzerland
| | - Jean T Coulibaly
- Department of Medical Parasitology and Infection Biology , Swiss Tropical and Public Health Institute , CH-4002 Basel , Switzerland.,University of Basel , CH-4003 Basel , Switzerland.,Unité de Formation et de Recherche Biosciences , Université Félix Houphouët-Boigny , 01 BP V34 , Abidjan 01 , Côte d'Ivoire
| | - Nikita Harvey
- Division of Integrative Systems Medicine and Digestive Diseases , Department of Surgery and Cancer, Imperial College London , London SW7 2AZ , United Kingdom
| | - Jennifer Keiser
- Department of Medical Parasitology and Infection Biology , Swiss Tropical and Public Health Institute , CH-4002 Basel , Switzerland.,University of Basel , CH-4003 Basel , Switzerland
| | - Jonathan Swann
- Division of Integrative Systems Medicine and Digestive Diseases , Department of Surgery and Cancer, Imperial College London , London SW7 2AZ , United Kingdom
| |
Collapse
|
32
|
Correction: Characterizing the metabolic phenotype of intestinal villus blunting in Zambian children with severe acute malnutrition and persistent diarrhea. PLoS One 2018; 13:e0196934. [PMID: 29715324 PMCID: PMC5929521 DOI: 10.1371/journal.pone.0196934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
33
|
Metabolic phenotyping of malnutrition during the first 1000 days of life. Eur J Nutr 2018; 58:909-930. [PMID: 29644395 PMCID: PMC6499750 DOI: 10.1007/s00394-018-1679-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/26/2018] [Indexed: 02/07/2023]
Abstract
Nutritional restrictions during the first 1000 days of life can impair or delay the physical and cognitive development of the individual and have long-term consequences for their health. Metabolic phenotyping (metabolomics/metabonomics) simultaneously measures a diverse range of low molecular weight metabolites in a sample providing a comprehensive assessment of the individual's biochemical status. There are a growing number of studies applying such approaches to characterize the metabolic derangements induced by various forms of early-life malnutrition. This includes acute and chronic undernutrition and specific micronutrient deficiencies. Collectively, these studies highlight the diverse and dynamic metabolic disruptions resulting from various forms of nutritional deficiencies. Perturbations were observed in many pathways including those involved in energy, amino acid, and bile acid metabolism, the metabolic interactions between the gut microbiota and the host, and changes in metabolites associated with gut health. The information gleaned from such studies provides novel insights into the mechanisms linking malnutrition with developmental impairments and assists in the elucidation of candidate biomarkers to identify individuals at risk of developmental shortfalls. As the metabolic profile represents a snapshot of the biochemical status of an individual at a given time, there is great potential to use this information to tailor interventional strategies specifically to the metabolic needs of the individual.
Collapse
|