1
|
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
|
2
|
Kawano B, Grisel B, Wischmeyer P, Holsman M, Agarwal S, Fernandez-Moure J, Haines KL. Racial and ethnic demographics in malnutrition related deaths. Clin Nutr ESPEN 2024; 60:135-138. [PMID: 38479901 DOI: 10.1016/j.clnesp.2024.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 01/11/2024] [Accepted: 01/18/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND AND AIMS Currently, 40 million Americans are food insecure. They are forced to skip meals and buy non-nutritious food, leading to health disparities for those of low socioeconomic status. This study aims to investigate relationships between malnutrition deaths and sociodemographic groups. METHODS This cross-sectional study from 2009 to 2018 used aggregate data from the CDC Wide-ranging Online Data for Epidemiologic Research (CDC Wonder). Patients with known race, gender, and Hispanic origin age ≥18 who died from malnutrition (E40-E46) were included. Place of death was grouped into home, inpatient medical facility, hospice facility, nursing facility/long-term care, other (including outpatient, ED, and DOA), and unknown. Crude rates of malnutrition deaths per 100,000 persons for race, gender, and Hispanic origin were calculated using US census estimates. Gross proportions of total deaths were calculated for each place of death. RESULTS Between 2009 and 2018, there were 46,517 malnutrition deaths in the US. Death rates for Black (1.8) and White Americans (2) were twice as high compared to Native Americans (1.1) and Asians or Pacific Islanders (0.7). Death rates among females (2.3) were higher than males (1.5). Death rates among non-Hispanics (2.1) were twice as high compared to Hispanics (0.7). Most people who died of malnutrition died in hospitals (37 %). CONCLUSION Malnutrition deaths occur at greater rates among White, Black, non-Hispanic Americans, and females. Despite reported disparities in food access, Black and White Americans have similar malnutrition mortality rates, raising concerns that malnutrition is under-diagnosed among Black patients. Given the existing nutrition literature, this finding requires further investigation.
Collapse
Affiliation(s)
- Bradley Kawano
- Division of Trauma, Acute, and Critical Care Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| | - Braylee Grisel
- Division of Trauma, Acute, and Critical Care Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| | - Paul Wischmeyer
- Division of Critical Care Surgery, Department of Anesthesia, Duke University Medical Center, Durham, NC, USA.
| | - Maximilian Holsman
- Division of Trauma, Acute, and Critical Care Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| | - Suresh Agarwal
- Division of Trauma, Acute, and Critical Care Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| | - Joseph Fernandez-Moure
- Division of Trauma, Acute, and Critical Care Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| | - Krista L Haines
- Division of Trauma, Acute, and Critical Care Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
3
|
Perlman M, Senger S, Verma S, Carey J, Faherty CS. A foundational approach to culture and analyze malnourished organoids. Gut Microbes 2023; 15:2248713. [PMID: 37724815 PMCID: PMC10512930 DOI: 10.1080/19490976.2023.2248713] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/07/2023] [Indexed: 09/21/2023] Open
Abstract
The gastrointestinal (GI) epithelium plays a major role in nutrient absorption, barrier formation, and innate immunity. The development of organoid-based methodology has significantly impacted the study of the GI epithelium, particularly in the fields of mucosal biology, immunity, and host-microbe interactions. Various effects on the GI epithelium, such as genetics and nutrition, impact patients and alter disease states. Thus, incorporating these effects into organoid-based models will facilitate a better understanding of disease progression and offer opportunities to evaluate therapeutic candidates. One condition that has a significant effect on the GI epithelium is malnutrition, and studying the mechanistic impacts of malnutrition would enhance our understanding of several pathologies. Therefore, the goal of this study was to begin to develop methodology to generate viable malnourished organoids with accessible techniques and resources that can be used for a wide array of mechanistic studies. By selectively limiting distinct macronutrient components of organoid media, we were able to successfully culture and evaluate malnourished organoids. Genetic and protein-based analyses were used to validate the approach and confirm the presence of known biomarkers of malnutrition. Additionally, as proof-of-concept, we utilized malnourished organoid-derived monolayers to evaluate the effect of malnourishment on barrier formation and the ability of the bacterial pathogen Shigella flexneri to infect the GI epithelium. This work serves as the basis for new and exciting techniques to alter the nutritional state of organoids and investigate the related impacts on the GI epithelium.
Collapse
Affiliation(s)
- Meryl Perlman
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stefania Senger
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
| | - Smriti Verma
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - James Carey
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
| | - Christina S. Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Sturgeon JP, Njunge JM, Bourke CD, Gonzales GB, Robertson RC, Bwakura-Dangarembizi M, Berkley JA, Kelly P, Prendergast AJ. Inflammation: the driver of poor outcomes among children with severe acute malnutrition? Nutr Rev 2023; 81:1636-1652. [PMID: 36977352 PMCID: PMC10639108 DOI: 10.1093/nutrit/nuad030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
Severe acute malnutrition (SAM) is the most life-threatening form of undernutrition and underlies at least 10% of all deaths among children younger than 5 years in low-income countries. SAM is a complex, multisystem disease, with physiological perturbations observed in conjunction with the loss of lean mass, including structural and functional changes in many organ systems. Despite the high mortality burden, predominantly due to infections, the underlying pathogenic pathways remain poorly understood. Intestinal and systemic inflammation is heightened in children with SAM. Chronic inflammation and its consequent immunomodulation may explain the increased morbidity and mortality from infections in children with SAM, both during hospitalization and in the longer term after discharge. Recognition of the role of inflammation in SAM is critical in considering new therapeutic targets in this disease, which has not seen a transformational approach to treatment for several decades. This review highlights the central role of inflammation in the wide-ranging pathophysiology of SAM, as well as identifying potential interventions that have biological plausibility based on evidence from other inflammatory syndromes.
Collapse
Affiliation(s)
- Jonathan P Sturgeon
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | - James M Njunge
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Claire D Bourke
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | - Gerard Bryan Gonzales
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, Netherlands
| | - Ruairi C Robertson
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | | | - James A Berkley
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Paul Kelly
- is with the Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| |
Collapse
|
5
|
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: 4.5] [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
|
6
|
Wen B, Farooqui A, Bourdon C, Tarafdar N, Ngari M, Chimwezi E, Thitiri J, Mwalekwa L, Walson JL, Voskuijl W, Berkley JA, Bandsma RHJ. Intestinal disturbances associated with mortality of children with complicated severe malnutrition. COMMUNICATIONS MEDICINE 2023; 3:128. [PMID: 37773543 PMCID: PMC10541881 DOI: 10.1038/s43856-023-00355-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Children admitted to hospital with complicated severe malnutrition (CSM) have high mortality despite compliance with standard WHO management guidelines. Limited data suggests a relationship between intestinal dysfunction and poor prognosis in CSM, but this has not been explicitly studied. This study aimed to evaluate the role of intestinal disturbances in CSM mortality. METHODS A case-control study nested within a randomized control trial was conducted among children hospitalized with CSM in Kenya and Malawi. Children who died (cases, n = 68) were compared with those who were discharged, propensity matched to the cases on age, HIV and nutritional status (controls, n = 68) on fecal metabolomics that targeted about 70 commonly measured metabolites, and enteropathy markers: fecal myeloperoxidase (MPO), fecal calprotectin, and circulating intestinal fatty acid binding protein (I-FABP). RESULTS The fecal metabolomes of cases show specific reductions in amino acids, monosaccharides, and microbial fermentation products, when compared to controls. SCFA levels did not differ between groups. The overall fecal metabolomics signature moderately differentiates cases from controls (AUC = 0.72). Enteropathy markers do not differ between groups overall, although serum I-FABP is elevated in cases in a sensitivity analysis among non-edematous children. Integrative analysis with systemic data suggests an indirect role of intestinal inflammation in the causal path of mortality. CONCLUSIONS Intestinal disturbances appear to have an indirect association with acute mortality. Findings of the study improve our understanding of pathophysiological pathways underlying mortality of children with CSM.
Collapse
Affiliation(s)
- Bijun Wen
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
| | - Amber Farooqui
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
| | - Celine Bourdon
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
| | - Nawar Tarafdar
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
| | - Moses Ngari
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Johnstone Thitiri
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Laura Mwalekwa
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Paediatrics, Coast General Hospital, Mombasa, Kenya
| | - Judd L Walson
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- Departments of Global Health, Medicine, Pediatrics and Epidemiology, University of Washington, Seattle, WA, USA
| | - Wieger Voskuijl
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- Amsterdam Institute for Global Health and Development, Department of Global Health, Amsterdam University Medical Centres, Amsterdam, The Netherlands
- Department of Paediatrics and Child Health, Kamuzu University of Health Sciences (formerly College of Medicine), Blantyre, Malawi
| | - James A Berkley
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Robert H J Bandsma
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada.
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada.
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya.
- Department of Biomedical Sciences, Kamuzu University of Health Sciences (formerly College of Medicine), Blantyre, Malawi.
| |
Collapse
|
7
|
Andres SF, Zhang Y, Kuhn M, Scottoline B. Building better barriers: how nutrition and undernutrition impact pediatric intestinal health. Front Immunol 2023; 14:1192936. [PMID: 37545496 PMCID: PMC10401430 DOI: 10.3389/fimmu.2023.1192936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Chronic undernutrition is a major cause of death for children under five, leaving survivors at risk for adverse long-term consequences. This review focuses on the role of nutrients in normal intestinal development and function, from the intestinal epithelium, to the closely-associated mucosal immune system and intestinal microbiota. We examine what is known about the impacts of undernutrition on intestinal physiology, with focus again on the same systems. We provide a discussion of existing animal models of undernutrition, and review the evidence demonstrating that correcting undernutrition alone does not fully ameliorate effects on intestinal function, the microbiome, or growth. We review efforts to treat undernutrition that incorporate data indicating that improved recovery is possible with interventions focused not only on delivery of sufficient energy, macronutrients, and micronutrients, but also on efforts to correct the abnormal intestinal microbiome that is a consequence of undernutrition. Understanding of the role of the intestinal microbiome in the undernourished state and correction of the phenotype is both complex and a subject that holds great potential to improve recovery. We conclude with critical unanswered questions in the field, including the need for greater mechanistic research, improved models for the impacts of undernourishment, and new interventions that incorporate recent research gains. This review highlights the importance of understanding the mechanistic effects of undernutrition on the intestinal ecosystem to better treat and improve long-term outcomes for survivors.
Collapse
Affiliation(s)
- Sarah F. Andres
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Yang Zhang
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Madeline Kuhn
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Brian Scottoline
- Division of Neonatology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
8
|
Connor KL, Bloise E, DeSantis TZ, Lye SJ. Adaptation of the gut holobiont to malnutrition during mouse pregnancy depends on the type of nutritional adversity. J Nutr Biochem 2023; 111:109172. [PMID: 36195213 DOI: 10.1016/j.jnutbio.2022.109172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/09/2022]
Abstract
Malnutrition can influence maternal physiology and programme offspring development. Yet, in pregnancy, little is known about how dietary challenges that influence maternal phenotype affect gut structure and function. Emerging evidence suggests that interactions between the environment, multidrug resistance (MDR) transporters and microbes may influence maternal adaptation to pregnancy and regulate fetoplacental development. We hypothesized that the gut holobiont (host and microbes) during pregnancy adapts differently to suboptimal maternal diets, evidenced by changes in the gut microenvironment, morphology, and expression of key protective MDR transporters during pregnancy. Mice were fed a control diet (CON) during pregnancy, or undernourished (UN) by 30% of control intake from gestational day (GD) 5.5-18.5, or fed 60% high fat diet (HF) for 8 weeks before and during pregnancy. At GD18.5, maternal small intestinal (SI) architecture (H&E), proliferation (Ki67), P-glycoprotein (P-gp - encoded by Abcb1a/b) and breast cancer resistance protein (BCRP/Abcg2) MDR transporter expression and levels of pro-inflammatory biomarkers were assessed. Circulating inflammatory biomarkers and maternal caecal microbiome composition (G3 PhyloChipTM) were measured. MDR transporter expression was also assessed in fetal gut. HF diet increased maternal SI crypt depth and proinflammatory load, and decreased SI expression of Abcb1a mRNA, whilst UN increased SI villi proliferation and Abcb1a, but decreased Abcg2, mRNA expression. There were significant associations between Abcb1a and Abcg2 mRNA levels with relative abundance of specific microbial taxa. Using a systems physiology approach we report that common nutritional adversities provoke adaptations in the pregnancy holobiont in mice, and reveal new mechanisms that could influence reproductive outcomes and fetal development.
Collapse
Affiliation(s)
- Kristin L Connor
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada; Mount Sinai Hospital, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada.
| | - Enrrico Bloise
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Stephen J Lye
- Mount Sinai Hospital, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Izadifar Z, Sontheimer-Phelps A, Lubamba BA, Bai H, Fadel C, Stejskalova A, Ozkan A, Dasgupta Q, Bein A, Junaid A, Gulati A, Mahajan G, Kim S, LoGrande NT, Naziripour A, Ingber DE. Modeling mucus physiology and pathophysiology in human organs-on-chips. Adv Drug Deliv Rev 2022; 191:114542. [PMID: 36179916 DOI: 10.1016/j.addr.2022.114542] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 08/25/2022] [Accepted: 09/13/2022] [Indexed: 01/24/2023]
Abstract
The surfaces of human internal organs are lined by a mucus layer that ensures symbiotic relationships with commensal microbiome while protecting against potentially injurious environmental chemicals, toxins, and pathogens, and disruption of this layer can contribute to disease development. Studying mucus biology has been challenging due to the lack of physiologically relevant human in vitro models. Here we review recent progress that has been made in the development of human organ-on-a-chip microfluidic culture models that reconstitute epithelial tissue barriers and physiologically relevant mucus layers with a focus on lung, colon, small intestine, cervix and vagina. These organ-on-a-chip models that incorporate dynamic fluid flow, air-liquid interfaces, and physiologically relevant mechanical cues can be used to study mucus composition, mechanics, and structure, as well as investigate its contributions to human health and disease with a level of biomimicry not possible in the past.
Collapse
Affiliation(s)
- Zohreh Izadifar
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | | | - Bob A Lubamba
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Haiqing Bai
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Cicely Fadel
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Anna Stejskalova
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Alican Ozkan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Queeny Dasgupta
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Amir Bein
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Abidemi Junaid
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Aakanksha Gulati
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Gautam Mahajan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Seongmin Kim
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Nina T LoGrande
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Arash Naziripour
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States; Vascular Biology Program, Boston Children's Hospital and Department of Pathology, Harvard Medical School, Boston, MA 02115, United States; Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA 02138, United Kingdom.
| |
Collapse
|
10
|
Njunge JM, Tickell K, Diallo AH, Sayeem Bin Shahid ASM, Gazi MA, Saleem A, Kazi Z, Ali S, Tigoi C, Mupere E, Lancioni CL, Yoshioka E, Chisti MJ, Mburu M, Ngari M, Ngao N, Gichuki B, Omer E, Gumbi W, Singa B, Bandsma R, Ahmed T, Voskuijl W, Williams TN, Macharia A, Makale J, Mitchel A, Williams J, Gogain J, Janjic N, Mandal R, Wishart DS, Wu H, Xia L, Routledge M, Gong YY, Espinosa C, Aghaeepour N, Liu J, Houpt E, Lawley TD, Browne H, Shao Y, Rwigi D, Kariuki K, Kaburu T, Uhlig HH, Gartner L, Jones K, Koulman A, Walson J, Berkley J. The Childhood Acute Illness and Nutrition (CHAIN) network nested case-cohort study protocol: a multi-omics approach to understanding mortality among children in sub-Saharan Africa and South Asia. Gates Open Res 2022; 6:77. [PMID: 36415883 PMCID: PMC9646488 DOI: 10.12688/gatesopenres.13635.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2022] [Indexed: 08/10/2023] Open
Abstract
Introduction: Many acutely ill children in low- and middle-income settings have a high risk of mortality both during and after hospitalisation despite guideline-based care. Understanding the biological mechanisms underpinning mortality may suggest optimal pathways to target for interventions to further reduce mortality. The Childhood Acute Illness and Nutrition (CHAIN) Network ( www.chainnnetwork.org) Nested Case-Cohort Study (CNCC) aims to investigate biological mechanisms leading to inpatient and post-discharge mortality through an integrated multi-omic approach. Methods and analysis; The CNCC comprises a subset of participants from the CHAIN cohort (1278/3101 hospitalised participants, including 350 children who died and 658 survivors, and 270/1140 well community children of similar age and household location) from nine sites in six countries across sub-Saharan Africa and South Asia. Systemic proteome, metabolome, lipidome, lipopolysaccharides, haemoglobin variants, toxins, pathogens, intestinal microbiome and biomarkers of enteropathy will be determined. Computational systems biology analysis will include machine learning and multivariate predictive modelling with stacked generalization approaches accounting for the different characteristics of each biological modality. This systems approach is anticipated to yield mechanistic insights, show interactions and behaviours of the components of biological entities, and help develop interventions to reduce mortality among acutely ill children. Ethics and dissemination. The CHAIN Network cohort and CNCC was approved by institutional review boards of all partner sites. Results will be published in open access, peer reviewed scientific journals and presented to academic and policy stakeholders. Data will be made publicly available, including uploading to recognised omics databases. Trial registration NCT03208725.
Collapse
Affiliation(s)
- James M. Njunge
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kirkby Tickell
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - Abdoulaye Hama Diallo
- Department of Public Health, Faculty of Health Sciences, University of Ouagadougou, Ouagadougou, Burkina Faso
| | | | - Md. Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ali Saleem
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Zaubina Kazi
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Syed Ali
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Caroline Tigoi
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Ezekiel Mupere
- Department of Paediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Emily Yoshioka
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - Mohammod Jobayer Chisti
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Moses Mburu
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Moses Ngari
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Narshion Ngao
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Bonface Gichuki
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Elisha Omer
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Wilson Gumbi
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Benson Singa
- Kenya Medical Research Institute, Nairobi, Kenya
| | - Robert Bandsma
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biomedical Sciences, University of Malawi College of Medicine, Blantyre, Malawi
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Wieger Voskuijl
- Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Centre for Global Child Health & Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Thomas N. Williams
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Institute of Global Health Innovation, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Alex Macharia
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | | | | | | | | | - Rupasri Mandal
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - David S. Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Hang Wu
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Lei Xia
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Michael Routledge
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Yun Yun Gong
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Camilo Espinosa
- Departments of Anesthesiology, Pain, and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Nima Aghaeepour
- Departments of Anesthesiology, Pain, and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | | | | | - Yan Shao
- Wellcome Sanger Institute, Hinxton, UK
| | - Doreen Rwigi
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Kevin Kariuki
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Timothy Kaburu
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Holm H. Uhlig
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Paediatrics and Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Lisa Gartner
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kelsey Jones
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Gastroenterology Department, Great Ormond Street Hospital for Children, London, UK
| | - Albert Koulman
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- NIHR BRC Nutritional Biomarker Laboratory, University of Cambridge, Cambridge, UK
| | - Judd Walson
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - James Berkley
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Center for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| |
Collapse
|
11
|
Zhao Y, He L, Peng T, Liu L, Zhou H, Xu Y, Yang X, Huang Y, Chen Z, Xu Y, Li J, Hou X, Tang H, Xu K. Nutritional status and function after high-calorie formula vs. Chinese food intervention in undernourished children with cerebral palsy. Front Nutr 2022; 9:960763. [PMID: 36276835 PMCID: PMC9582948 DOI: 10.3389/fnut.2022.960763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Aim To investigate the efficacy and safety of high-calorie formula vs. Chinese daily food on the nutritional status and motor function of undernourished children with cerebral palsy (CP). Methods In this prospective, assessor-blind, and randomized controlled trial, we recruited children (1-10 years) with CP and undernutrition based on the WHO and the American Society for Parenteral and Enteral Nutrition criteria from the National Children's Medical Center. Participants were randomly allocated (1:1) to a high-calorie formula group or a Chinese daily food diet group (control group) for 6 months. Indirect calorimetry was used to estimate energy requirements. We compared the nutritional status and gross motor function of participants in both groups based on weight, height, z-scores (weight-for-age, height-for-age, weight-for-height, and BMI-for-age), and the Gross Motor Function Measure (GMFM), respectively, at baseline, 3-, and 6-months follow-up. In addition, the effective rate of nutritional intervention, and adverse events were simultaneously assessed. Results From July 2020 to December 2021, a total of 119 participants were enrolled and randomized, and 110 participants completed the study (with 54 children in the high-calorie formula group and 56 children in the control group). After 6 months of treatment, the weight, height, z-scores (weight-for-height, weight-for-age, and BMI-for-age), and GMFM of both groups were significantly improved (p < 0.05). There were significant differences in changes in weight, weight-for-age z-scores, and GMFM between the two groups (p < 0.05). During the study period, 16 children experienced at least one mild adverse event [9 (16.7%) in the formula group and 7 (12.5%) in the control group]. Conclusion Nutritional intervention with a high-calorie formula may be an effective and safe option in children with CP for improving undernutrition and gross motor dysfunction. Clinical trial registration www.chictr.org.cn, identifier: ChiCTR2000033878.
Collapse
Affiliation(s)
- Yiting Zhao
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China,Department of Sports and Health, Guangzhou Sport University, Guangzhou, China
| | - Lu He
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Tingting Peng
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Liru Liu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Hongyu Zhou
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China,Department of Sports and Health, Guangzhou Sport University, Guangzhou, China
| | - Yunxian Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xubo Yang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yuan Huang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China,School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhaofang Chen
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yi Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China,Department of Sports and Health, Guangzhou Sport University, Guangzhou, China
| | - Jinling Li
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaohui Hou
- Department of Sports and Health, Guangzhou Sport University, Guangzhou, China,Xiaohui Hou
| | - Hongmei Tang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China,Hongmei Tang
| | - Kaishou Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China,*Correspondence: Kaishou Xu
| |
Collapse
|
12
|
Diaz JN, Dulience SJL, Wolthausen N, Jiang X, Gyimah E, Marhône Pierre FJ, Kuhlmann FM, Iannotti LL. Choline, DHA, and Diarrheal Disease Associated with Growth Faltering in a Case-Control Study. Curr Dev Nutr 2022; 6:nzac140. [PMID: 36204326 PMCID: PMC9529221 DOI: 10.1093/cdn/nzac140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/17/2022] [Accepted: 09/01/2022] [Indexed: 12/03/2022] Open
Abstract
Background Children with recurrent infectious diarrhea are susceptible to growth faltering. DHA and choline may play a role in this relationship due to their involvement in lipid metabolism, gut immunity, and inflammatory pathways. Objectives This study aimed to characterize the contributions made by DHA and choline status and enteric damage in young children in the association between diarrheal illness and child growth. Methods A longitudinal case-control study was conducted among children aged 6-36 mo (N = 195) in Cap-Haitien, Haiti. Mother-child dyads were recruited from community health posts and outpatient clinics. Cases were defined as children experiencing acute diarrhea within the last 3 d and matched to healthy controls. Child anthropometry, dietary intake, and blood and stool samples were collected at baseline and follow-up. Plasma DHA, choline, and betaine were determined by LC-MS/MS methods (n = 49) and intestinal fatty acid-binding protein (I-FABP) by ELISA (n = 183). Multivariate regression models were applied with mediation analyses to examine associations and adjust for confounding factors. Results At baseline, mean plasma DHA concentrations (1.03 µg/mL; 95% CI: 0.91, 1.15) were not significantly different between cases and controls, nor was there a difference in mean plasma choline concentrations (4.5 µg/mL; 95% CI: 3.8, 5.1). Mean plasma I-FABP concentrations were significantly higher at follow-up in cases (3.34; 95% CI: 3.28, 3.40) than controls (3.20; 95% CI: 3.13, 3.27; P = 0.002). In adjusted multilinear regression models, higher plasma DHA concentrations at follow-up were associated with a negative change in weight-age z score (P = 0.016), and follow-up I-FABP was inversely associated with height-age z score (P = 0.035). No interaction or mediation effects were found. Conclusions I-FABP concentrations were significantly higher in cases as compared with controls at follow-up, suggesting ongoing enteric damage and increased risk for malnutrition. Plasma DHA and I-FABP may have a role in childhood growth outcomes.
Collapse
Affiliation(s)
- Jenna N Diaz
- Department of Pediatrics, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | | | - Noah Wolthausen
- Brown School, Institute for Public Health, Washington University in St Louis, St Louis, Missouri, USA
| | - Xuntian Jiang
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Emmanuel Gyimah
- Brown School, Institute for Public Health, Washington University in St Louis, St Louis, Missouri, USA
| | - Francesca J Marhône Pierre
- Unité de Coordination du Programme National d'Alimentation et de Nutrition, Ministère de la Santé Publique et de la Population, Port-au-Prince, Haiti
| | - F Matthew Kuhlmann
- Department of Medicine, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Lora L Iannotti
- Brown School, Institute for Public Health, Washington University in St Louis, St Louis, Missouri, USA
| |
Collapse
|
13
|
Pinho RM, Garas LC, Huang BC, Weimer BC, Maga EA. Malnourishment affects gene expression along the length of the small intestine. Front Nutr 2022; 9:894640. [PMID: 36118759 PMCID: PMC9478944 DOI: 10.3389/fnut.2022.894640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Malnourishment is a risk factor for childhood mortality, jeopardizing the health of children by aggravating pneumonia/acute respiratory infections and diarrheal diseases. Malnourishment causes morphophysiological changes resulting in stunting and wasting that have long-lasting consequences such as cognitive deficit and metabolic dysfunction. Using a pig model of malnutrition, the interplay between the phenotypic data displayed by the malnourished animals, the gene expression pattern along the intestinal tract, microbiota composition of the intestinal contents, and hepatic metabolite concentrations from the same animals were correlated using a multi-omics approach. Samples from the duodenum, jejunum, and ileum of malnourished (protein and calorie-restricted diet) and full-fed (no dietary restrictions) piglets were subjected to RNA-seq. Gene co-expression analysis and phenotypic correlations were made with WGCNA, while the integration of transcriptome with microbiota composition and the hepatic metabolite profile was done using mixOmics. Malnourishment caused changes in tissue gene expression that influenced energetic balance, cell proliferation, nutrient absorption, and response to stress. Repression of antioxidant genes, including glutathione peroxidase, in coordination with induction of metal ion transporters corresponded to the hepatic metabolite changes. These data indicate oxidative stress in the intestine of malnourished animals. Furthermore, several of the phenotypes displayed by these animals could be explained by changes in gene expression.
Collapse
Affiliation(s)
- Raquel M. Pinho
- Department of Animal Science, University of California, Davis, Davis, CA, United States
- *Correspondence: Raquel M. Pinho
| | - Lydia C. Garas
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - B. Carol Huang
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Bart C. Weimer
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Elizabeth A. Maga
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
14
|
Wetherill MS, Bakhsh C, Caywood L, Williams MB, Hartwell ML, Wheeler DL, Hubach RD, Teague TK, Köhler G, Hebert JR, Weiser SD. Unpacking determinants and consequences of food insecurity for insulin resistance among people living with HIV: Conceptual framework and protocol for the NOURISH-OK study. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2022; 3:947552. [PMID: 36225538 PMCID: PMC9552993 DOI: 10.3389/fcdhc.2022.947552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/01/2022] [Indexed: 11/22/2022]
Abstract
Background Over the past four decades, advances in HIV treatment have contributed to a longer life expectancy for people living with HIV (PLWH). With these gains, the prevention and management of chronic co-morbidities, such as diabetes, are now central medical care goals for this population. In the United States, food insecurity disproportionately impacts PLWH and may play a role in the development of insulin resistance through direct and indirect pathways. The Nutrition to Optimize, Understand, and Restore Insulin Sensitivity in HIV for Oklahoma (NOURISH-OK) will use a novel, multi-level, integrated framework to explore how food insecurity contributes to insulin resistance among PLWH. Specifically, it will explore how food insecurity may operate as an intermediary risk factor for insulin resistance, including potential linkages between upstream determinants of health and downstream consequences of poor diet, other behavioral risk factors, and chronic inflammation. Methods/design This paper summarizes the protocol for the first aim of the NOURISH-OK study, which involves purposeful cross-sectional sampling of PLWH (n=500) across four levels of food insecurity to test our conceptual framework. Developed in collaboration with community stakeholders, this initial phase involves the collection of anthropometrics, fasting blood samples, non-blood biomarkers, 24-hour food recall to estimate the Dietary Inflammatory Index (DII®) score, and survey data. A 1-month, prospective observational sub-study (total n=100; n=25 for each food security group) involves weekly 24-hour food recalls and stool samples to identify temporal associations between food insecurity, diet, and gut microbiome composition. Using structural equation modeling, we will explore how upstream risk factors, including early life events, current discrimination, and community food access, may influence food insecurity and its potential downstream impacts, including diet, other lifestyle risk behaviors, and chronic inflammation, with insulin resistance as the ultimate outcome variable. Findings from these analyses of observational data will inform the subsequent study aims, which involve qualitative exploration of significant pathways, followed by development and testing of a low-DII® food as medicine intervention to reverse insulin resistance among PLWH (ClinicalTrials.gov Identifier: NCT05208671). Discussion The NOURISH-OK study will address important research gaps to inform the development of food as medicine interventions to support healthy aging for PLWH.
Collapse
Affiliation(s)
- Marianna S. Wetherill
- Department of Health Promotion Sciences, Hudson College of Public Health, University of Oklahoma Tulsa Schusterman Center, Tulsa, OK, United States
- Department of Family and Community Medicine, University of Oklahoma School of Community Medicine, Tulsa, OK, United States
| | | | - Lacey Caywood
- Department of Health Promotion Sciences, Hudson College of Public Health, University of Oklahoma Tulsa Schusterman Center, Tulsa, OK, United States
| | - Mary B. Williams
- Department of Family and Community Medicine, University of Oklahoma School of Community Medicine, Tulsa, OK, United States
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Tulsa Schusterman Center, Tulsa, OK, United States
| | - Micah L. Hartwell
- Department of Psychiatry, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Denna L. Wheeler
- Center for Rural Health, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Randolph D. Hubach
- Department of Public Health, Purdue University, West Lafayette, IN, United States
| | - T. Kent Teague
- Department of Surgery, University of Oklahoma School of Community Medicine, Tulsa, OK, United States
- Department of Psychiatry, University of Oklahoma School of Community Medicine, Tulsa, OK, United States
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Gerwald Köhler
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - James R. Hebert
- Department of Epidemiology and Biostatistics, University of South Carolina, Columbia, SC, United States
| | - Sheri D. Weiser
- Division of HIV, Infectious Disease and Global Medicine, Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, United States
| |
Collapse
|
15
|
Njunge JM, Tickell K, Diallo AH, Sayeem Bin Shahid ASM, Gazi MA, Saleem A, Kazi Z, Ali S, Tigoi C, Mupere E, Lancioni CL, Yoshioka E, Chisti MJ, Mburu M, Ngari M, Ngao N, Gichuki B, Omer E, Gumbi W, Singa B, Bandsma R, Ahmed T, Voskuijl W, Williams TN, Macharia A, Makale J, Mitchel A, Williams J, Gogain J, Janjic N, Mandal R, Wishart DS, Wu H, Xia L, Routledge M, Gong YY, Espinosa C, Aghaeepour N, Liu J, Houpt E, Lawley TD, Browne H, Shao Y, Rwigi D, Kariuki K, Kaburu T, Uhlig HH, Gartner L, Jones K, Koulman A, Walson J, Berkley J. The Childhood Acute Illness and Nutrition (CHAIN) network nested case-cohort study protocol: a multi-omics approach to understanding mortality among children in sub-Saharan Africa and South Asia. Gates Open Res 2022; 6:77. [PMID: 36415883 PMCID: PMC9646488 DOI: 10.12688/gatesopenres.13635.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 02/15/2024] Open
Abstract
Introduction: Many acutely ill children in low- and middle-income settings have a high risk of mortality both during and after hospitalisation despite guideline-based care. Understanding the biological mechanisms underpinning mortality may suggest optimal pathways to target for interventions to further reduce mortality. The Childhood Acute Illness and Nutrition (CHAIN) Network ( www.chainnnetwork.org) Nested Case-Cohort Study (CNCC) aims to investigate biological mechanisms leading to inpatient and post-discharge mortality through an integrated multi-omic approach. Methods and analysis; The CNCC comprises a subset of participants from the CHAIN cohort (1278/3101 hospitalised participants, including 350 children who died and 658 survivors, and 270/1140 well community children of similar age and household location) from nine sites in six countries across sub-Saharan Africa and South Asia. Systemic proteome, metabolome, lipidome, lipopolysaccharides, haemoglobin variants, toxins, pathogens, intestinal microbiome and biomarkers of enteropathy will be determined. Computational systems biology analysis will include machine learning and multivariate predictive modelling with stacked generalization approaches accounting for the different characteristics of each biological modality. This systems approach is anticipated to yield mechanistic insights, show interactions and behaviours of the components of biological entities, and help develop interventions to reduce mortality among acutely ill children. Ethics and dissemination. The CHAIN Network cohort and CNCC was approved by institutional review boards of all partner sites. Results will be published in open access, peer reviewed scientific journals and presented to academic and policy stakeholders. Data will be made publicly available, including uploading to recognised omics databases. Trial registration NCT03208725.
Collapse
Affiliation(s)
- James M. Njunge
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kirkby Tickell
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - Abdoulaye Hama Diallo
- Department of Public Health, Faculty of Health Sciences, University of Ouagadougou, Ouagadougou, Burkina Faso
| | | | - Md. Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ali Saleem
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Zaubina Kazi
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Syed Ali
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Caroline Tigoi
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Ezekiel Mupere
- Department of Paediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Emily Yoshioka
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - Mohammod Jobayer Chisti
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Moses Mburu
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Moses Ngari
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Narshion Ngao
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Bonface Gichuki
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Elisha Omer
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Wilson Gumbi
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Benson Singa
- Kenya Medical Research Institute, Nairobi, Kenya
| | - Robert Bandsma
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biomedical Sciences, University of Malawi College of Medicine, Blantyre, Malawi
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Wieger Voskuijl
- Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Centre for Global Child Health & Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Thomas N. Williams
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Institute of Global Health Innovation, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Alex Macharia
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | | | | | | | | | - Rupasri Mandal
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - David S. Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Hang Wu
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Lei Xia
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Michael Routledge
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Yun Yun Gong
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Camilo Espinosa
- Departments of Anesthesiology, Pain, and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Nima Aghaeepour
- Departments of Anesthesiology, Pain, and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | | | | | - Yan Shao
- Wellcome Sanger Institute, Hinxton, UK
| | - Doreen Rwigi
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Kevin Kariuki
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Timothy Kaburu
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Holm H. Uhlig
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Paediatrics and Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Lisa Gartner
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kelsey Jones
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Gastroenterology Department, Great Ormond Street Hospital for Children, London, UK
| | - Albert Koulman
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- NIHR BRC Nutritional Biomarker Laboratory, University of Cambridge, Cambridge, UK
| | - Judd Walson
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - James Berkley
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Center for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| |
Collapse
|
16
|
Patterson GT, Osorio EY, Peniche A, Dann SM, Cordova E, Preidis GA, Suh JH, Ito I, Saldarriaga OA, Loeffelholz M, Ajami NJ, Travi BL, Melby PC. Pathologic Inflammation in Malnutrition Is Driven by Proinflammatory Intestinal Microbiota, Large Intestine Barrier Dysfunction, and Translocation of Bacterial Lipopolysaccharide. Front Immunol 2022; 13:846155. [PMID: 35720380 PMCID: PMC9204284 DOI: 10.3389/fimmu.2022.846155] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
Acute malnutrition, or wasting, is implicated in over half of all deaths in children under five and increases risk of infectious disease. Studies in humans and preclinical models have demonstrated that malnutrition is linked to an immature intestinal microbiota characterized by increased prevalence of Enterobacteriaceae. Observational studies in children with moderate acute malnutrition (MAM) have also observed heightened systemic inflammation and increased circulating bacterial lipopolysaccharides (LPS; endotoxin). However, the mechanisms that underpin the systemic inflammatory state and endotoxemia, and their pathophysiological consequences, remain uncertain. Understanding these pathophysiological mechanisms is necessary to design targeted treatments that will improve the unacceptable rate of failure or relapse that plague current approaches. Here we use a mouse model of MAM to investigate the mechanisms that promote inflammation in the malnourished host. We found that mice with MAM exhibited increased systemic inflammation at baseline, increased translocation of bacteria and bacterial LPS, and an exaggerated response to inflammatory stimuli. An exaggerated response to bacterial LPS was associated with increased acute weight loss. Remarkably, intestinal inflammation and barrier dysfunction was found in the cecum and colon. The cecum showed a dysbiotic microbiota with expansion of Gammaproteobacteria and some Firmicutes, and contraction of Bacteroidetes. These changes were paralleled by an increase in fecal LPS bioactivity. The inflammatory phenotype and weight loss was modulated by oral administration of non-absorbable antibiotics that altered the proportion of cecal Gammaproteobacteria. We propose that the heightened inflammation of acute malnutrition is the result of changes in the intestinal microbiota, intestinal barrier dysfunction in the cecum and colon, and increased systemic exposure to LPS.
Collapse
Affiliation(s)
- Grace T Patterson
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Elvia Y Osorio
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Alex Peniche
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Sara M Dann
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States.,Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX, United States
| | - Erika Cordova
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Geoffrey A Preidis
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Ji Ho Suh
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Ichiaki Ito
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Omar A Saldarriaga
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Michael Loeffelholz
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Nadim J Ajami
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Bruno L Travi
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States.,Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX, United States
| | - Peter C Melby
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States.,Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
17
|
Fan Y, Yao Q, Liu Y, Jia T, Zhang J, Jiang E. Underlying Causes and Co-existence of Malnutrition and Infections: An Exceedingly Common Death Risk in Cancer. Front Nutr 2022; 9:814095. [PMID: 35284454 PMCID: PMC8906403 DOI: 10.3389/fnut.2022.814095] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/24/2022] [Indexed: 12/16/2022] Open
Abstract
In nutrition science, malnutrition is a state of imbalance between intake and the needs of the organism, leading to metabolic changes, impaired physiological functions, and weight loss. Regardless of the countless efforts being taken and researched for years, the burden of malnutrition is still alarming and considered a significant agent of mortality across the globe. Around 45% of 12 million children deaths (0–5 years old) annually are due to malnutrition, mostly from developing countries. Malnutrition develops associations with other infections and leads to substantial clinical outcomes, such as mortality, more visits to hospitals, poor quality of life and physical frailty, and socioeconomic issues. Here, in this review, we intend to provide an overview of the current burden, underlying risk factors, and co-existence of malnutrition and other infections, such as cancer. Following the rising concern of the vicious interplay of malnutrition and other medical illnesses, we believed that this narrative review would highlight the need to re-make and re-define the future strategies by giving comprehensive and sustainable programs to alleviate poverty and combat the rampant infectious diseases and those nutrition-related health problems. Furthermore, the study also raises the concern for hospitalized malnourished cancer patients as it is crucially important to knowledge the caregiver healthcare staff for early interventions of providing nutritional support to delay or prevent the onset of malnutrition.
Collapse
Affiliation(s)
- Yuanyuan Fan
- School of Life Sciences, Henan University, Kaifeng, China
| | - Qianqian Yao
- Institute of Nursing and Health, Henan University, Kaifeng, China
| | - Yufeng Liu
- Institute of Nursing and Health, Henan University, Kaifeng, China
| | - Tiantian Jia
- Institute of Nursing and Health, Henan University, Kaifeng, China
- DeDepartment of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, China
| | - Junjuan Zhang
- DeDepartment of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, China
- Junjuan Zhang
| | - Enshe Jiang
- Institute of Nursing and Health, Henan University, Kaifeng, China
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
- *Correspondence: Enshe Jiang
| |
Collapse
|
18
|
Wen B, Njunge JM, Bourdon C, Gonzales GB, Gichuki BM, Lee D, Wishart DS, Ngari M, Chimwezi E, Thitiri J, Mwalekwa L, Voskuijl W, Berkley JA, Bandsma RHJ. Systemic inflammation and metabolic disturbances underlie inpatient mortality among ill children with severe malnutrition. SCIENCE ADVANCES 2022; 8:eabj6779. [PMID: 35171682 PMCID: PMC8849276 DOI: 10.1126/sciadv.abj6779] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/23/2021] [Indexed: 06/14/2023]
Abstract
Children admitted to hospital with an acute illness and concurrent severe malnutrition [complicated severe malnutrition (CSM)] have a high risk of dying. The biological processes underlying their mortality are poorly understood. In this case-control study nested within a multicenter randomized controlled trial among children with CSM in Kenya and Malawi, we found that blood metabolomic and proteomic profiles robustly differentiated children who died (n = 92) from those who survived (n = 92). Fatalities were characterized by increased energetic substrates (tricarboxylic acid cycle metabolites), microbial metabolites (e.g., propionate and isobutyrate), acute phase proteins (e.g., calprotectin and C-reactive protein), and inflammatory markers (e.g., interleukin-8 and tumor necrosis factor-α). These perturbations indicated disruptions in mitochondria-related bioenergetic pathways and sepsis-like responses. This study identified specific biomolecular disturbances associated with CSM mortality, revealing that systemic inflammation and bioenergetic deficits are targetable pathophysiological processes for improving survival of this vulnerable population.
Collapse
Affiliation(s)
- Bijun Wen
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
| | - James M. Njunge
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Celine Bourdon
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
| | - Gerard Bryan Gonzales
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, Netherlands
| | - Bonface M. Gichuki
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Dorothy Lee
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
| | | | - Moses Ngari
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Johnstone Thitiri
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Laura Mwalekwa
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Paediatrics, Coast General Hospital, Mombasa, Kenya
| | - Wieger Voskuijl
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- Department of Global Health, Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centres, Amsterdam, Netherlands
- Department of Pediatrics, the College of Medicine, University of Malawi, Blantyre, Malawi
| | - James A. Berkley
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Robert HJ Bandsma
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- Department of Pediatrics, the College of Medicine, University of Malawi, Blantyre, Malawi
- Department of Biomedical Sciences, the College of Medicine, University of Malawi, Blantyre, Malawi
| |
Collapse
|
19
|
Daniel AI, Bwanali M, Tenthani JC, Gladstone M, Voskuijl W, Potani I, Ziwoya F, Chidzalo K, Mbale E, Heath A, Bourdon C, Njirammadzi J, van den Heuvel M, Bandsma RHJ. A Mixed-Methods Cluster-Randomized Controlled Trial of a Hospital-Based Psychosocial Stimulation and Counseling Program for Caregivers and Children with Severe Acute Malnutrition. Curr Dev Nutr 2021; 5:nzab100. [PMID: 34447897 PMCID: PMC8382273 DOI: 10.1093/cdn/nzab100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/17/2021] [Accepted: 07/16/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Children with severe acute malnutrition (SAM) who require nutritional rehabilitation unit (NRU) treatment often have poor developmental and nutritional outcomes following discharge. The Kusamala Program is a 4-d hospital-based counseling program for caregivers of children with SAM that integrates nutrition, water, sanitation, and hygiene and psychosocial stimulation, aimed at improving these outcomes. OBJECTIVES The aim was to evaluate the effects of the Kusamala Program on child development and nutritional status in children with SAM 6 mo after NRU discharge. The other aim was to qualitatively understand perceptions and experiences of caregivers who participated in the intervention. METHODS A cluster-randomized controlled trial was conducted with caregivers and their children 6-59 mo of age with SAM admitted to the Moyo NRU in Blantyre, Malawi. The primary outcome of the trial was child development according to Malawi Developmental Assessment Tool (MDAT) composite z-scores of gross motor, fine motor, language, and social domains. A qualitative component with focus group discussions and in-depth interviews was also completed with a subset of caregivers who participated in the trial. RESULTS Sixty-eight caregivers and children were enrolled to clusters by week and randomly assigned to the comparison arm and 104 to the intervention arm. There were no differences in child development, with mean MDAT composite z-scores in the comparison arm of -1.2 (95% CI: -2.1, -0.22) and in the intervention arm of -1.1 (95% CI: -1.9, -0.40) (P = 0.93). The qualitative evaluation with 20 caregivers indicated that the 3 modules of the Kusamala Program were appropriate and that they applied many of the lessons learned at home as much as possible. CONCLUSIONS The Kusamala Program did not result in improved developmental or nutritional outcomes, yet it was viewed positively by caregivers according to qualitative results. Future research should evaluate more intensive interventions for caregivers and children with SAM. This trial was registered at www.clinicaltrials.gov as NCT03072433.
Collapse
Affiliation(s)
- Allison I Daniel
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mike Bwanali
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Blantyre, Malawi
| | | | - Melissa Gladstone
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Wieger Voskuijl
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Blantyre, Malawi
- Department of Pediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
- Amsterdam Centre for Global Child Health, Emma Children's Hospital, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
| | - Isabel Potani
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Blantyre, Malawi
| | - Frank Ziwoya
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Blantyre, Malawi
| | - Kate Chidzalo
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Blantyre, Malawi
| | - Emmie Mbale
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Blantyre, Malawi
- Department of Pediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Anna Heath
- Child Health Evaluative Sciences, Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Department of Statistical Science, University College London, London, United Kingdom
| | - Celine Bourdon
- Translational Medicine Program, Hospital for Sick Children, Toronto, Ontario, Canada
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Blantyre, Malawi
| | - Jenala Njirammadzi
- Department of Pediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Meta van den Heuvel
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Blantyre, Malawi
- Division of Pediatric Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Robert H J Bandsma
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Blantyre, Malawi
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Biomedical Sciences, College of Medicine, University of Malawi, Blantyre, Malawi
| |
Collapse
|
20
|
Krishinchand H, Naidoo K, Mahabeer P, Archary M. Paediatric Enterobacteriaceae infections in hospitalised children in Durban, KwaZulu-Natal. S Afr J Infect Dis 2021; 36:279. [PMID: 39376941 PMCID: PMC11457515 DOI: 10.4102/sajid.v36i1.279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/10/2021] [Indexed: 10/09/2024] Open
Abstract
Background Community-acquired Gram-negative Enterobacteriaceae infections in malnourished and HIV-infected hospitalised children are not well documented and are of concern because of increasing antibiotic resistance and limited available treatment options. This study describes the clinical characteristics and outcomes of hospitalised children with positive Enterobacteriaceae cultures. Method A retrospective chart review of children with Gram-negative Enterobacteriaceae infections was performed in King Edward VIII Hospital, a referral hospital in Durban, KwaZulu-Natal. Standard descriptive and analytical statistics, including regression analysis, were performed to determine the clinical characteristics associated with Enterobacteriaceae infections in children hospitalised in the study period. Results Of all hospitalised children in the study period, 207 (3.5%) had positive cultures for Enterobacteriaceae isolates, with Escherichia coli 109 (44.5%) and Klebsiella spp. 59 (24.1%) making up most of the infections. Urine (126; 58%) followed by stool (34; 14.8%) and blood (35; 14.0%) were the commonest samples that yielded positive cultures. Diarrhoeal hospitalisations especially posed a higher risk for Enterobacteriaceae infections. Severe acutely malnourished and HIV-infected children were at higher risk. These comorbidities were independently associated with an increased risk of Enterobacteriaceae infection. Prolonged hospitalisation and increased risk of death were also associated with Enterobacteriaceae infection. Conclusion Enterobacteriaceae infections were common in hospitalised children and posed an increased risk, especially in malnourished and HIV-infected children. Further studies investigating the relationships between diarrhoea, urinary tract infections and Enterobacteriaceae infections are needed.
Collapse
Affiliation(s)
- Harshna Krishinchand
- Department of Paediatrics and Child Health, School of
Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South
Africa
| | - Kimesh Naidoo
- Department of Paediatrics and Child Health, School of
Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South
Africa
- Department of Paediatrics, King Edward VIII Hospital,
Durban, South Africa
| | - Prasha Mahabeer
- Department of Microbiology, King Edward VIII Hospital,
Durban, South Africa
- Department of Medical Microbiology, School of Laboratory
Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal,
Durban, South Africa
| | - Moherndran Archary
- Department of Paediatrics and Child Health, School of
Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South
Africa
- Department of Paediatrics and Child Health, King Edward
VIII Hospital, Durban, South Africa
| |
Collapse
|
21
|
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: 2.8] [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
|
22
|
Extracellular vesicles isolated from milk can improve gut barrier dysfunction induced by malnutrition. Sci Rep 2021; 11:7635. [PMID: 33828139 PMCID: PMC8026962 DOI: 10.1038/s41598-021-86920-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 03/18/2021] [Indexed: 02/01/2023] Open
Abstract
Malnutrition impacts approximately 50 million children worldwide and is linked to 45% of global mortality in children below the age of five. Severe acute malnutrition (SAM) is associated with intestinal barrier breakdown and epithelial atrophy. Extracellular vesicles including exosomes (EVs; 30-150 nm) can travel to distant target cells through biofluids including milk. Since milk-derived EVs are known to induce intestinal stem cell proliferation, this study aimed to examine their potential efficacy in improving malnutrition-induced atrophy of intestinal mucosa and barrier dysfunction. Mice were fed either a control (18%) or a low protein (1%) diet for 14 days to induce malnutrition. From day 10 to 14, they received either bovine milk EVs or control gavage and were sacrificed on day 15, 4 h after a Fluorescein Isothiocyanate (FITC) dose. Tissue and blood were collected for histological and epithelial barrier function analyses. Mice fed low protein diet developed intestinal villus atrophy and barrier dysfunction. Despite continued low protein diet feeding, milk EV treatment improved intestinal permeability, intestinal architecture and cellular proliferation. Our results suggest that EVs enriched from milk should be further explored as a valuable adjuvant therapy to standard clinical management of malnourished children with high risk of morbidity and mortality.
Collapse
|
23
|
Lezo A, Aidala E, Deorsola L, Cascarano MT, Rizzo A, Iannandrea S, Peruzzi L, Runfola F, Pace Napoleone C. Malnutrition and chyle leakage: A life-threatening duo in heart transplantation post-Fontan procedure. Clin Case Rep 2020; 8:2055-2059. [PMID: 33088551 PMCID: PMC7562867 DOI: 10.1002/ccr3.3060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/25/2020] [Accepted: 05/31/2020] [Indexed: 11/08/2022] Open
Abstract
Protein‐losing enteropathy and chyle leakage may lead to severe malnutrition in heart transplantation for failing Fontan. Nutritional management may be challenging from defining nutrient needs to diagnosis of malnutrition enteropathy, and expertise is necessary. Body composition and hematological nutritional indices may help define malnutrition severity and guide nutritional strategy.
Collapse
Affiliation(s)
- Antonella Lezo
- Clinical Nutrition Unit Children's Hospital Regina Margherita Città della Salute e della Scienza Turin Italy
| | - Enrico Aidala
- Cardiac Surgery Department Regina Margherita Children's Hospital, Città della Salute e della Scienza Turin Italy
| | - Luca Deorsola
- Cardiac Surgery Department Regina Margherita Children's Hospital, Città della Salute e della Scienza Turin Italy
| | - Maria Teresa Cascarano
- Cardiac Surgery Department Regina Margherita Children's Hospital, Città della Salute e della Scienza Turin Italy
| | - Alberta Rizzo
- Cardiac Intensive Care Unit Regina Margherita Children's Hospital, Città della Salute e della Scienza Turin Italy
| | - Stefania Iannandrea
- Cardiac Intensive Care Unit Regina Margherita Children's Hospital, Città della Salute e della Scienza Turin Italy
| | - Licia Peruzzi
- Nephrology Department Regina Margherita Children's Hospital, Città della Salute e della Scienza Turin Italy
| | - Federica Runfola
- University of Turin Postgraduate School of Specialization in Paediatrics Turin Italy
| | - Carlo Pace Napoleone
- Cardiac Surgery Department Regina Margherita Children's Hospital, Città della Salute e della Scienza Turin Italy
| |
Collapse
|
24
|
Song H, Sui H, Zhang Q, Wang P, Wang F. Cucurbitacin E Induces Autophagy-Involved Apoptosis in Intestinal Epithelial Cells. Front Physiol 2020; 11:1020. [PMID: 32982778 PMCID: PMC7479753 DOI: 10.3389/fphys.2020.01020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/27/2020] [Indexed: 12/17/2022] Open
Abstract
Apoptosis plays a crucial role in maintaining the structural and functional integrity of the intestinal epithelial barrier. Autophagy mediates injury to and repair of the intestinal epithelial barrier through multiple pathways in pathophysiological conditions. Our earlier study has found that cucurbitacin E (CuE) regulates the proliferation, migration, and permeability of human intestinal epithelial cells (IECs); however, its effects and mechanisms on apoptosis and autophagy are still unclear. This study reported CuE induced apoptosis and promoted autophagy of IECs in a concentration-dependent manner. The results showed that CuE could inhibit the expression of apoptosis-related protein Bcl-2 and drove activation of caspase-3 and cleavage of its substrate poly (ADP-ribose) polymerase. CuE also facilitated the expression of endoplasmic reticulum stress-related proteins, CHOP and Grp78, and autophagy-related proteins, Beclin1 and LC3, while inhibiting the phosphorylation of AKT and mammalian target of rapamycin (mTOR). An autophagy inhibitor, 3-methyladenine, reduced CuE-induced apoptosis. These results suggest that CuE may induce apoptosis and autophagy in IECs via the PI3K/AKT/mTOR signaling pathway and that autophagy following endoplasmic reticulum stress participates in the pro-apoptotic process induced by CuE.
Collapse
Affiliation(s)
- Huapei Song
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hehuan Sui
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Pharmacy, Central Hospital of Nanchong, The Second Clinical School of North Sichuan Medical College, Nanchong, China.,Nanchong Key Laboratory of Individualized Drug Therapy, Nanchong, China
| | - Qiong Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Pei Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fengjun Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
25
|
Liu TC, VanBuskirk K, Ali SA, Kelly MP, Holtz LR, Yilmaz OH, Sadiq K, Iqbal N, Amadi B, Syed S, Ahmed T, Moore S, Ndao IM, Isaacs MH, Pfeifer JD, Atlas H, Tarr PI, Denno DM, Moskaluk CA. A novel histological index for evaluation of environmental enteric dysfunction identifies geographic-specific features of enteropathy among children with suboptimal growth. PLoS Negl Trop Dis 2020; 14:e0007975. [PMID: 31929525 PMCID: PMC6980693 DOI: 10.1371/journal.pntd.0007975] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 01/24/2020] [Accepted: 12/06/2019] [Indexed: 02/06/2023] Open
Abstract
Background A major limitation to understanding the etiopathogenesis of environmental enteric dysfunction (EED) is the lack of a comprehensive, reproducible histologic framework for characterizing the small bowel lesions. We hypothesized that the development of such a system will identify unique histology features for EED, and that some features might correlate with clinical severity. Methods Duodenal endoscopic biopsies from two cohorts where EED is prevalent (Pakistan, Zambia) and North American children with and without gluten sensitive enteropathy (GSE) were processed for routine hematoxylin & eosin (H&E) staining, and scanned to produce whole slide images (WSIs) which we shared among study pathologists via a secure web browser-based platform. A semi-quantitative scoring index composed of 11 parameters encompassing tissue injury and response patterns commonly observed in routine clinical practice was constructed by three gastrointestinal pathologists, with input from EED experts. The pathologists then read the WSIs using the EED histology index, and inter-observer reliability was assessed. The histology index was further used to identify within- and between-child variations as well as features common across and unique to each cohort, and those that correlated with host phenotype. Results Eight of the 11 histologic scoring parameters showed useful degrees of variation. The overall concordance across all parameters was 96% weighted agreement, kappa 0.70, and Gwet’s AC 0.93. Zambian and Pakistani tissues shared some histologic features with GSE, but most features were distinct, particularly abundance of intraepithelial lymphocytes in the Pakistani cohort, and marked villous destruction and loss of secretory cell lineages in the Zambian cohort. Conclusions We propose the first EED histology index for interpreting duodenal biopsies. This index should be useful in future clinical and translational studies of this widespread, poorly understood, and highly consequential disorder, which might be caused by multiple contributing processes, in different regions of the world. The study of EED has been limited by the lack of a rigorously tested, reproducible histology index that can provide insight to the pathogenesis of this entity. In this study we report the first duodenal histology index that was developed using an unbiased approach, with excellent inter-observer reproducibility, for the study of EED. The EED histology index readily identified histologic features that are common or unique to cohorts of distinct geographic locations. Incorporating the histology index into future clinical studies will provide useful insight into the pathogenesis and for intervention strategy development.
Collapse
Affiliation(s)
- Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Kelley VanBuskirk
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Syed A. Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - M. Paul Kelly
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine, Lusaka, Zambia
- Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Lori R. Holtz
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Omer H. Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, Department of Pathology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Kamran Sadiq
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Najeeha Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Beatrice Amadi
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Sana Syed
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
- Department of Pediatrics, University of Virginia, Charlottesville, VA, United States of America
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- James P. Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| | - Sean Moore
- Department of Pediatrics, University of Virginia, Charlottesville, VA, United States of America
| | - I. Malick Ndao
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Michael H. Isaacs
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - John D. Pfeifer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Hannah Atlas
- Departments of Pediatrics and Global Health, University of Washington, Seattle, WA, United States of America
| | - Phillip I. Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Donna M. Denno
- Departments of Pediatrics and Global Health, University of Washington, Seattle, WA, United States of America
| | - Christopher A. Moskaluk
- Department of Pathology, University of Virginia, Charlottesville, VA, United States of America
- * E-mail:
| |
Collapse
|
26
|
Schulze KV, Swaminathan S, Howell S, Jajoo A, Lie NC, Brown O, Sadat R, Hall N, Zhao L, Marshall K, May T, Reid ME, Taylor-Bryan C, Wang X, Belmont JW, Guan Y, Manary MJ, Trehan I, McKenzie CA, Hanchard NA. Edematous severe acute malnutrition is characterized by hypomethylation of DNA. Nat Commun 2019; 10:5791. [PMID: 31857576 PMCID: PMC6923441 DOI: 10.1038/s41467-019-13433-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
Edematous severe acute childhood malnutrition (edematous SAM or ESAM), which includes kwashiorkor, presents with more overt multi-organ dysfunction than non-edematous SAM (NESAM). Reduced concentrations and methyl-flux of methionine in 1-carbon metabolism have been reported in acute, but not recovered, ESAM, suggesting downstream DNA methylation changes could be relevant to differences in SAM pathogenesis. Here, we assess genome-wide DNA methylation in buccal cells of 309 SAM children using the 450 K microarray. Relative to NESAM, ESAM is characterized by multiple significantly hypomethylated loci, which is not observed among SAM-recovered adults. Gene expression and methylation show both positive and negative correlation, suggesting a complex transcriptional response to SAM. Hypomethylated loci link to disorders of nutrition and metabolism, including fatty liver and diabetes, and appear to be influenced by genetic variation. Our epigenetic findings provide a potential molecular link to reported aberrant 1-carbon metabolism in ESAM and support consideration of methyl-group supplementation in ESAM.
Collapse
Affiliation(s)
- Katharina V Schulze
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- USDA/ARS/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Shanker Swaminathan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- USDA/ARS/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Sharon Howell
- Tropical Metabolism Research Unit, Caribbean Institute for Health Research, University of the West Indies, Mona, Jamaica
| | - Aarti Jajoo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- USDA/ARS/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Natasha C Lie
- USDA/ARS/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Orgen Brown
- Tropical Metabolism Research Unit, Caribbean Institute for Health Research, University of the West Indies, Mona, Jamaica
| | - Roa Sadat
- USDA/ARS/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Nancy Hall
- USDA/ARS/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Liang Zhao
- Precision Medicine Research Center, Taihe Hospital, Shiyan City, China
| | - Kwesi Marshall
- Tropical Metabolism Research Unit, Caribbean Institute for Health Research, University of the West Indies, Mona, Jamaica
| | - Thaddaeus May
- USDA/ARS/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Marvin E Reid
- Tropical Metabolism Research Unit, Caribbean Institute for Health Research, University of the West Indies, Mona, Jamaica
| | - Carolyn Taylor-Bryan
- Tropical Metabolism Research Unit, Caribbean Institute for Health Research, University of the West Indies, Mona, Jamaica
| | - Xueqing Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- USDA/ARS/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - John W Belmont
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- USDA/ARS/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Yongtao Guan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- USDA/ARS/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Mark J Manary
- Departments of Paediatrics and Child Health and Community Health, University of Malawi, Blantyre, Malawi
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Indi Trehan
- Departments of Paediatrics and Child Health and Community Health, University of Malawi, Blantyre, Malawi
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
- Departments of Pediatrics and Global Health, University of Washington, Seattle, WA, USA
| | - Colin A McKenzie
- Tropical Metabolism Research Unit, Caribbean Institute for Health Research, University of the West Indies, Mona, Jamaica
| | - Neil A Hanchard
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- USDA/ARS/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
27
|
Kastl AJ, Terry NA, Wu GD, Albenberg LG. The Structure and Function of the Human Small Intestinal Microbiota: Current Understanding and Future Directions. Cell Mol Gastroenterol Hepatol 2019; 9:33-45. [PMID: 31344510 PMCID: PMC6881639 DOI: 10.1016/j.jcmgh.2019.07.006] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/17/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023]
Abstract
Despite growing literature characterizing the fecal microbiome and its association with health and disease, few studies have analyzed the microbiome of the small intestine. Here, we examine what is known about the human small intestinal microbiota in terms of community structure and functional properties. We examine temporal dynamics of select bacterial populations in the small intestine, and the effects of dietary carbohydrates and fats on shaping these populations. We then evaluate dysbiosis in the small intestine in several human disease models, including small intestinal bacterial overgrowth, short-bowel syndrome, pouchitis, environmental enteric dysfunction, and irritable bowel syndrome. What is clear is that the bacterial biology, and mechanisms of bacteria-induced pathophysiology, are enormously broad and elegant in the small intestine. Studying the small intestinal microbiota is challenged by rapidly fluctuating environmental conditions in these intestinal segments, as well as the complexity of sample collection and bioinformatic analysis. Because the functionality of the digestive tract is determined primarily by the small intestine, efforts must be made to better characterize this unique and important microbial ecosystem.
Collapse
Affiliation(s)
- Arthur J. Kastl
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Correspondence Address correspondence to: Arthur J. Kastl Jr, MD, Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, 7NW, Philadelphia, Pennsylvania 19104. fax: (215) 590-3606.
| | - Natalie A. Terry
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Gary D Wu
- Division of Gastroenterology, Hepatology, and Nutrition, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lindsey G. Albenberg
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
28
|
Abstract
Purpose of review Almost half of all childhood deaths worldwide occur in children with malnutrition, predominantly in sub-Saharan Africa and South Asia. This review summarizes the mechanisms by which malnutrition and serious infections interact with each other and with children's environments. Recent findings It has become clear that whilst malnutrition results in increased incidence, severity and case fatality of common infections, risks continue beyond acute episodes resulting in significant postdischarge mortality. A well established concept of a ‘vicious-cycle’ between nutrition and infection has now evolving to encompass dysbiosis and pathogen colonization as precursors to infection; enteric dysfunction constituting malabsorption, dysregulation of nutrients and metabolism, inflammation and bacterial translocation. All of these interact with a child's diet and environment. Published trials aiming to break this cycle using antimicrobial prophylaxis or water, sanitation and hygiene interventions have not demonstrated public health benefit so far. Summary As further trials are planned, key gaps in knowledge can be filled by applying new tools to re-examine old questions relating to immune competence during and after infection events and changes in nutritional status; and how to characterize overt and subclinical infection, intestinal permeability to bacteria and the role of antimicrobial resistance using specific biomarkers.
Collapse
|
29
|
L'Huillier C, Jarbeau M, Achamrah N, Belmonte L, Amamou A, Nobis S, Goichon A, Salameh E, Bahlouli W, do Rego JL, Déchelotte P, Coëffier M. Glutamine, but not Branched-Chain Amino Acids, Restores Intestinal Barrier Function during Activity-Based Anorexia. Nutrients 2019; 11:nu11061348. [PMID: 31208031 PMCID: PMC6628073 DOI: 10.3390/nu11061348] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/21/2022] Open
Abstract
Background: During activity-based anorexia (ABA) in mice, enhanced paracellular permeability and reduced protein synthesis have been shown in the colon while the gut–brain axis has received increasing attention in the regulation of intestinal and mood disorders that frequently occur during anorexia nervosa, a severe eating disorder for which there is no specific treatment. In the present study, we assessed the effects of oral glutamine (Gln) or branched-chain amino acids (BCAA) supplementation during ABA to target intestinal functions, body composition and feeding behavior. Methods: C57BL/6 male mice were randomized in Control (CTRL) and ABA groups. After ABA induction, mice received, or not, either 1% Gln or 2.5% BCAA (Leu, Ile, Val) for one week in drinking water. Results: Neither Gln nor BCAA supplementation affected body weight and body composition, while only Gln supplementation slightly increased food intake. ABA mice exhibited increased paracellular permeability and reduced protein synthesis in the colonic mucosa. Oral Gln restored colonic paracellular permeability and protein synthesis and increased the mucin-2 mRNA level, whereas BCAA did not affect colonic parameters. Conclusion: In conclusion, oral Gln specifically improves colonic response during ABA. These data should be further confirmed in AN patients.
Collapse
Affiliation(s)
- Clément L'Huillier
- UNIROUEN, INSERM UMR 1073 "Nutrition, Inflammation and Gut-Brain Axis", Normandie University, 76183 Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
| | - Marine Jarbeau
- UNIROUEN, INSERM UMR 1073 "Nutrition, Inflammation and Gut-Brain Axis", Normandie University, 76183 Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
| | - Najate Achamrah
- UNIROUEN, INSERM UMR 1073 "Nutrition, Inflammation and Gut-Brain Axis", Normandie University, 76183 Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
- Department of Nutrition, Rouen University Hospital, 76183 Rouen, France.
| | - Liliana Belmonte
- UNIROUEN, INSERM UMR 1073 "Nutrition, Inflammation and Gut-Brain Axis", Normandie University, 76183 Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
- Department of Nutrition, Rouen University Hospital, 76183 Rouen, France.
| | - Asma Amamou
- UNIROUEN, INSERM UMR 1073 "Nutrition, Inflammation and Gut-Brain Axis", Normandie University, 76183 Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
| | - Séverine Nobis
- UNIROUEN, INSERM UMR 1073 "Nutrition, Inflammation and Gut-Brain Axis", Normandie University, 76183 Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
| | - Alexis Goichon
- UNIROUEN, INSERM UMR 1073 "Nutrition, Inflammation and Gut-Brain Axis", Normandie University, 76183 Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
| | - Emmeline Salameh
- UNIROUEN, INSERM UMR 1073 "Nutrition, Inflammation and Gut-Brain Axis", Normandie University, 76183 Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
| | - Wafa Bahlouli
- UNIROUEN, INSERM UMR 1073 "Nutrition, Inflammation and Gut-Brain Axis", Normandie University, 76183 Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
| | - Jean-Luc do Rego
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
- Animal Behavior Facility, SCAC, UNIROUEN, 76183 Rouen, France.
| | - Pierre Déchelotte
- UNIROUEN, INSERM UMR 1073 "Nutrition, Inflammation and Gut-Brain Axis", Normandie University, 76183 Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
- Department of Nutrition, Rouen University Hospital, 76183 Rouen, France.
| | - Moïse Coëffier
- UNIROUEN, INSERM UMR 1073 "Nutrition, Inflammation and Gut-Brain Axis", Normandie University, 76183 Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), UNIROUEN, Normandie University, 76183 Rouen, France.
- Department of Nutrition, Rouen University Hospital, 76183 Rouen, France.
| |
Collapse
|
30
|
Environmental enteric dysfunction and growth. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2019. [DOI: 10.1016/j.jpedp.2019.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
31
|
Morais MBD, Silva GAPD. Environmental enteric dysfunction and growth. J Pediatr (Rio J) 2019; 95 Suppl 1:85-94. [PMID: 30629923 DOI: 10.1016/j.jped.2018.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/23/2018] [Accepted: 11/23/2018] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE To describe the current indicators of environmental enteric dysfunction and its association with linear growth deficit and the height-for-age anthropometric indicator. DATA SOURCES Narrative review with articles identified in PubMed and Scopus databases using combinations of the following words: environmental, enteric, dysfunction, enteropathy, and growth, as well as the authors' personal records. DATA SYNTHESIS In the last 15 years, new non-invasive markers have been investigated to characterize environmental enteric dysfunction; however, the best tests to be used have not yet been identified. There is evidence that, in environmental enteric dysfunction, a systemic inflammatory process may also occur as a consequence of increased intestinal permeability, in addition to intestinal mucosa abnormalities. Bacterial overgrowth in the small intestine and changes in fecal microbiota profile have also been identified. There is evidence indicating that environmental enteric dysfunction can impair not only full growth but also the neuropsychomotor development and response to orally administered vaccines. It is important to emphasize that the environmental enteric dysfunction is not a justification for not carrying out vaccination, which must follow the regular schedule. Another aspect to emphasize is the greater risk for those children who had height impairment in early childhood, possibly associated with environmental enteric dysfunction, to present overweight and obesity in adulthood when exposed to a high calorie diet, which has been called "triple burden." CONCLUSIONS According to the analyzed evidence, the control of environmental enteric dysfunction is very important for the full expression of growth, development, and vaccine response in the pediatric age group.
Collapse
Affiliation(s)
- Mauro Batista de Morais
- Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, Disciplina de Gastroenterologia Pediátrica, São Paulo, SP, Brazil.
| | | |
Collapse
|
32
|
Abstract
BACKGROUND Persistent diarrhoea (PD), defined as diarrhoeal symptoms for longer than 2 weeks, still forms a substantial disease burden in children under 5 years of age. This article provides an overview of the current knowledge of PD and discusses novel concepts. METHODS A literature search on PD was performed which focused on evidence on epidemiology, pathophysiology and management of the disease. RESULTS The prevalence of PD has potentially decreased over the last decades. Debate remains around the role of specific bacterial, viral and parasitic infections with PD. PD is associated with malnutrition and a compromised immune system, including that caused by HIV infection. Management includes fluid resuscitation and improving nutritional status. There is a lack of evidence on the use of antibiotic therapy for PD. There is increasing interest in nutrient-based interventions, including pre- and/or probiotics that can modify the microbiome and thereby potentially prevent or improve the outcome of PD in children. CONCLUSION As PD remains a significant health burden, multicentre clinical trials are needed to inform future treatment guidelines. ABBREVIATIONS PD, persistent diarrhoea; EED, environmental enteric dysfunction; IBD, inflammatory bowel disease; WHO, World Health Organization.
Collapse
Affiliation(s)
- Robert H J Bandsma
- a Division of Gastroenterology, Hepatology and Nutrition , Hospital for Sick Children , Toronto , Canada.,b Translational Medicine Program, Hospital for Sick Children , Toronto , Canada.,c Centre for Global Child Health, Hospital for Sick Children , Toronto , Canada.,d Department of Nutrition Sciences , University of Toronto , Toronto , Canada
| | - Kamran Sadiq
- e Department of Paediatrics and Child Health , Aga Khan University , Karachi , Pakistan
| | - Zulfiqar A Bhutta
- c Centre for Global Child Health, Hospital for Sick Children , Toronto , Canada.,d Department of Nutrition Sciences , University of Toronto , Toronto , Canada.,e Department of Paediatrics and Child Health , Aga Khan University , Karachi , Pakistan
| |
Collapse
|
33
|
Bartelt LA, Bolick DT, Guerrant RL. Disentangling Microbial Mediators of Malnutrition: Modeling Environmental Enteric Dysfunction. Cell Mol Gastroenterol Hepatol 2019; 7:692-707. [PMID: 30630118 PMCID: PMC6477186 DOI: 10.1016/j.jcmgh.2018.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022]
Abstract
Environmental enteric dysfunction (EED) (also referred to as environmental enteropathy) is a subclinical chronic intestinal disorder that is an emerging contributor to early childhood malnutrition. EED is common in resource-limited settings, and is postulated to consist of small intestinal injury, dysfunctional nutrient absorption, and chronic inflammation that results in impaired early child growth attainment. Although there is emerging interest in the hypothetical potential for chemical toxins in the environmental exposome to contribute to EED, the propensity of published data, and hence the focus of this review, implicates a critical role of environmental microbes. Early childhood malnutrition and EED are most prevalent in resource-limited settings where food is limited, and inadequate access to clean water and sanitation results in frequent gastrointestinal pathogen exposures. Even as overt diarrhea rates in these settings decline, silent enteric infections and faltering growth persist. Furthermore, beyond restricted physical growth, EED and/or enteric pathogens also associate with impaired oral vaccine responses, impaired cognitive development, and may even accelerate metabolic syndrome and its cardiovascular consequences. As these potentially costly long-term consequences of early childhood enteric infections increasingly are appreciated, novel therapeutic strategies that reverse damage resulting from nutritional deficiencies and microbial insults in the developing small intestine are needed. Given the inherent limitations in investigating how specific intestinal pathogens directly injure the small intestine in children, animal models provide an affordable and controlled opportunity to elucidate causal sequelae of specific enteric infections, to differentiate consequences of defined nutrient deprivation alone from co-incident enteropathogen insults, and to correlate the resulting gut pathologies with their functional impact during vulnerable early life windows.
Collapse
Affiliation(s)
- Luther A Bartelt
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Gastrointestinal Biology and Disease, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - David T Bolick
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Richard L Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
34
|
Sîrbe C, Grama A, Pop TL. Severe acute malnutrition, a consequence of low socioeconomic status – clinical case series. PEDIATRU.RO 2019; 2:18. [DOI: 10.26416/pedi.54.2.2019.2434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
35
|
Abstract
The volume of research into the pathogenesis and treatment of malnutrition has increased markedly over the past ten years, providing mechanistic insights that can be leveraged into more effective treatment options. These discoveries have been driven by several landmark studies employing metabolomics, metagenomics, and new preclinical models. This review highlights some of the most important recent findings, focusing in particular on the emerging roles of prenatal and perinatal factors, protein deficiency, impaired gut barrier function, immune deficiency, and the intestinal microbiome.
Collapse
|
36
|
Ngari MM, Mwalekwa L, Timbwa M, Hamid F, Ali R, Iversen PO, Fegan GW, Berkley JA. Changes in susceptibility to life-threatening infections after treatment for complicated severe malnutrition in Kenya. Am J Clin Nutr 2018; 107:626-634. [PMID: 29635501 PMCID: PMC6134064 DOI: 10.1093/ajcn/nqy007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/08/2018] [Indexed: 12/15/2022] Open
Abstract
Background Goals of treating childhood severe acute malnutrition (SAM), in addition to anthropometric recovery and preventing short-term mortality, include reducing the risks of subsequent serious infections. How quickly and how much the risk of serious illness changes during rehabilitation are unknown but could inform improving the design and scope of interventions. Objective The aim of this study was to investigate changes in the risk of life-threatening events (LTEs) in relation to anthropometric recovery from SAM. Design This was a secondary analysis of a clinical trial including 1778 HIV-uninfected Kenyan children aged 2-59 mo with complicated SAM, enrolled after the inpatient stabilization phase of treatment, and followed for 12 mo. The main outcome was LTEs, defined as infections requiring rehospitalization or causing death. We examined anthropometric variables measured at months 1, 3, and 6 after enrollment in relation to LTEs occurring during the 6 mo after each of these time points. Results Over 12 mo, there were 823 LTEs (257 fatal), predominantly severe pneumonia and diarrhea. At months 1, 3, and 6, 557 (34%), 764 (49%), and 842 (56%) children had a weight-for-height or -length z score (WHZ) ≥-2, respectively, which, compared with a WHZ <-3, was associated with lower risks of subsequent LTEs [adjusted HRs (95% CIs): 0.50 (0.40, 0.64), 0.30 (0.23, 0.39), and 0.23 (0.16, 0.32), respectively]. However, children with a WHZ ≥-2 at 1, 3, and 6 mo still had 39 (95% CI: 32, 47), 26 (95% CI: 22, 32), and 15 (95% CI: 12, 20) LTEs/100 child-years of observation during the following 6 mo. WHZ at study enrollment predicted subsequent WHZ but not the risk of LTEs. Changes in height-for-age z score did not predict LTEs. Conclusions Anthropometric response was associated with a rapid and substantial reduction in risk of LTEs. However, reduction in susceptibility lagged behind anthropometric improvement. Disease events, together with anthropometric assessment, may provide a clearer picture of the effectiveness of interventions. Robust protocols for detecting and treating poor anthropometric recovery and addressing broader vulnerabilities that complicated SAM indicates may save lives. This trial was registered at www.clinicaltrials.gov as NCT00934492.
Collapse
Affiliation(s)
- Moses M Ngari
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- The Childhood Acute Illness and Nutrition (CHAIN) Network, Nairobi, Kenya
| | | | - Molline Timbwa
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- The Childhood Acute Illness and Nutrition (CHAIN) Network, Nairobi, Kenya
| | - Fauzat Hamid
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Rehema Ali
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Per Ole Iversen
- Department of Nutrition, IBM, University of Oslo, Norway
- Department of Hematology, Oslo University Hospital, Oslo, Norway
| | - Greg W Fegan
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- Swansea Trials Unit, Swansea University Medical School, Swansea, United Kingdom
| | - James A Berkley
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- The Childhood Acute Illness and Nutrition (CHAIN) Network, Nairobi, Kenya
- Center for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|