1
|
Meyer R, Arpe L, Kansu A, Kelly V, Lindley K, O'Meara M, del Carmen Rivero M, van Zundert S, Vicente-Santamaría S, Žaja O, Oliveros E, Olivier L, Joosten K. Gastrointestinal changes in paediatric malnutrition that may impact on nutrition choice. Front Pediatr 2025; 13:1523613. [PMID: 40129696 PMCID: PMC11931439 DOI: 10.3389/fped.2025.1523613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/05/2025] [Indexed: 03/26/2025] Open
Abstract
Undernutrition is defined as "a condition resulting from imbalanced nutrition or abnormal utilization of nutrients." In this paper, the term malnutrition is used to refer to undernutrition. Malnutrition may be driven by poor socioeconomic conditions or by disease, and it is estimated that disease-related malnutrition (DRM) impacts up to 28% of hospitalized children in Europe. Malnutrition results in alterations in gastrointestinal function that lead to malabsorption of macro- and micro-nutrients. It can lead to altered gut motility and a deficiency of stomach acid, which can result in intestinal colonization by pathogens, causing diarrhoea and high burdens of intestinal infection. The presence of compromised gastrointestinal function in children with DRM is critical as it negatively impacts the efficacy of nutritional support and recovery. When choosing novel strategies and nutritional therapies for malnourished children, consideration should be given to gut-protective interventions that promote better treatment tolerance. When breastmilk is unavailable, whole protein feeds are currently considered as first-line treatment for malnutrition in children with a normal functioning gastrointestinal tract. However, peptide-based feeds have been associated with improved gastrointestinal tolerance and absorption, reduced diarrhoea, reduced inflammation, improved growth and have restored gut integrity compared with free amino acid and whole-protein feeds. At a recent meeting, experts in this area have identified significant research gaps in the literature on peptide-based feeds in children and possible gaps in clinical practice. Whilst the group acknowledges that further work is needed, this paper provides an overview on this topic to further drive research in this area.
Collapse
Affiliation(s)
- Rosan Meyer
- Department of Medicine, KU Leuven, Leuven, Belgium
| | - Lauren Arpe
- Gastroenterology Department, Great Ormond Street Hospital, London, United Kingdom
| | - Aydan Kansu
- Department of Pediatric Gastroenterology, Hepatology & Nutrition, Ankara University School of Medicine, Ankara, Türkiye
| | - Veronica Kelly
- Department of Pediatrics, Children’s Health Ireland, Dublin, Ireland
| | - Keith Lindley
- Gastroenterology Department, Great Ormond Street Hospital, London, United Kingdom
| | - Mairéad O'Meara
- Department of Pediatrics, Children’s Health Ireland, Dublin, Ireland
| | - Maria del Carmen Rivero
- Department of Pediatric Gastroenterology and Nutrition, Hospital Virgen de la Macarena, Seville, Spain
| | - Suzanne van Zundert
- Department of Nutrition and Dietetics, Amsterdam University Medical Centre, Emma Children’s Hospital, Amsterdam, Netherlands
| | | | - Orjena Žaja
- Sestre Milosrdnice University Hospital Center, University of Zagreb, Zagreb, Croatia
| | | | - Leanne Olivier
- Medical Affairs & Research, Nutrition International, Abbott, Maidenhead, United Kingdom
| | - Koen Joosten
- Department of Intensive Care Neonatology & Pediatrics, Erasmus MC-Sophia Children’s Hospital, Rotterdam, Netherlands
| |
Collapse
|
2
|
Zang P, Chen P, Chen J, Sun J, Lan H, Dong H, Liu W, Xu N, Wang W, Hou L, Sun B, Zhang L, Huang J, Wang P, Ren F, Liu S. Alteration of Gastrointestinal Function and the Ameliorative Effects of Hericium erinaceus Polysaccharides in Tail Suspension Rats. Nutrients 2025; 17:724. [PMID: 40005052 PMCID: PMC11858084 DOI: 10.3390/nu17040724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/13/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Long-term spaceflight in a microgravity environment frequently results in gastrointestinal dysfunction, presenting substantial challenges to astronauts' health. Hericium erinaceus, a plant recognized for its dual use as food and medicine, contains a key functional component called Hericium erinaceus polysaccharide (HEP), which is purported to promote gastrointestinal health. This study aims to investigate the protective effects of HEP against gastrointestinal disturbances induced by simulated weightlessness and to elucidate its regulatory mechanisms. Methods: Sprague Dawley rats subjected to a tail suspension model were administered either a standard diet or a diet supplemented with 0.125% HEP over a period of 4 weeks (the intake of HEP is approximately 157.5 mg/kg bw/d, n = 8), metagenomics and targeted metabolomics to investigate the effects of HEP on gastrointestinal hormone secretion disorders, gut microbiota dysbiosis, and intestinal barrier damage induced by simulated weightlessness. Results: Dietary supplementation with HEP was observed to significantly alleviate weightlessness-induced gastrointestinal hormone disruptions, enhancing motility and intestinal barrier function while reducing inflammation. In addition, HEP improved gut microbiota by boosting beneficial bacteria as Oscillibacter sp.1-3, Firmicutes bacterium ASF500, and Lactobacillus reuteri, while reducing harmful bacteria like Escherichia coli and Mucispirillum schaedleri at the species level. Furthermore, HEP altered the serum metabolic profile of the rats, reducing inflammation by upregulating the tryptophan metabolism pathway and enhancing the production of short-chain fatty acids. Conclusions: HEP effectively protects against gastrointestinal dysfunction induced by simulated weightlessness by regulating hormone secretion and maintaining intestinal homeostasis.
Collapse
Affiliation(s)
- Peng Zang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (P.Z.); (F.R.)
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Pu Chen
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Junli Chen
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Jingchao Sun
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Haiyun Lan
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Haisheng Dong
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Wei Liu
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Nan Xu
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Weiran Wang
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Lingwei Hou
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Bowen Sun
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Lujia Zhang
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (P.C.); (J.C.); (J.S.); (H.L.); (H.D.); (W.L.); (N.X.); (W.W.); (L.H.); (B.S.); (L.Z.)
| | - Jiaqiang Huang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China;
| | - Pengjie Wang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China;
| | - Fazheng Ren
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (P.Z.); (F.R.)
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China;
| | - Siyuan Liu
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China;
| |
Collapse
|
3
|
Cao HH, Molina S, Sumner S, Rushing BR. An untargeted metabolomic analysis of acute AFB1 treatment in liver, breast, and lung cells. PLoS One 2025; 20:e0313159. [PMID: 39883710 PMCID: PMC11781672 DOI: 10.1371/journal.pone.0313159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/18/2024] [Indexed: 02/01/2025] Open
Abstract
Aflatoxin B1 (AFB1) is a class 1 carcinogen and mycotoxin known to contribute to the development of hepatocellular carcinoma (HCC), growth impairment, altered immune system modulation, and malnutrition. AFB1 is synthesized by Aspergillus flavus and is known to widely contaminate foodstuffs, particularly maize, wheat, and groundnuts. The mechanism in which AFB1 causes genetic mutations has been well studied, however its metabolomic effects remained largely unknown. A better understanding of how AFB1 disrupts metabolism would provide insight into how this mycotoxin leads to carcinogenesis, growth impairment, and/or immunomodulation, and may reveal potential targets for pharmacological or nutritional interventions to protect against these effects. The current study evaluated the metabolomic effects of various doses (2.5 μM, 5 μM, 10uM) of AFB1 treatment to HepG2 (liver), MDA-MB-231 (breast), and A549 (lung) cells. Treated and control cells' metabolomic profiles were evaluated via ultra-high performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS). Univariate and multivariate analyses revealed significant alterations in metabolite concentrations from each dose of AFB1 treatment in each cell type. Pathway analysis was then used to understand broader biochemical functions affected by AFB1 treatment in each cell type. HepG2 cell pathway analyses revealed significant pathway perturbations in lipid metabolism, carnitine synthesis, catecholamine biosynthesis, purine metabolism, and spermidine and spermine biosynthesis. Analysis of A549 cells found a greater emphasis of perturbations on various amino acids along with lipid synthesis-related pathways, and catecholamine biosynthesis. Finally, analysis of treated MDA-MB-231 cells found spermidine and spermine biosynthesis, carnitine synthesis, plasma membrane-related pathways (phosphatidylcholine synthesis and alpha linolenic acid and linoleic acid metabolism), and various amino acid metabolism pathways to be most affected. These highlighted pathways should be targeted in future investigations to evaluate their potential in mitigating or preventing the development of negative health effects associated with AFB1 exposure.
Collapse
Affiliation(s)
- Heidi H. Cao
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Sabrina Molina
- Nutrition Research Institute, University of North Carolina Chapel Hill, Kannapolis, NC, United States of America
| | - Susan Sumner
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Nutrition Research Institute, University of North Carolina Chapel Hill, Kannapolis, NC, United States of America
| | - Blake R. Rushing
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Nutrition Research Institute, University of North Carolina Chapel Hill, Kannapolis, NC, United States of America
| |
Collapse
|
4
|
Mostafa I, Sthity RA, Lamiya UH, Tariqujjaman M, Mahfuz M, Hasan SMT, Ahmed T. Effect of Gut Microbiota-Directed Complementary Food Supplementation on Fecal and Plasma Biomarkers of Gut Health and Environmental Enteric Dysfunction in Slum-Dwelling Children with Moderate Acute Malnutrition. CHILDREN (BASEL, SWITZERLAND) 2024; 11:69. [PMID: 38255381 PMCID: PMC10814735 DOI: 10.3390/children11010069] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 12/31/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024]
Abstract
Dietary supplementation with a gut microbiota-directed complementary food (MDCF-2) significantly improved weight gain and repaired gut microbiota, as reported in a recent randomized controlled trial on Bangladeshi children with moderate acute malnutrition (MAM). Environmental enteric dysfunction (EED) is a small bowel disorder, and recent evidence shows that it is linked to growth failure in children. Therefore, we intended to investigate whether supplementation with MDCF-2 has any role in modifying gut health by changing the levels of biomarkers of EED and gut inflammation in children with MAM. We randomly assigned 124 children aged 12-18 months to one of two intervention diets, either MDCF-2 or ready-to-use supplementary food (RUSF). Approximately 50 g of the diet was administered in two feeding sessions daily for 12 weeks. Stool and plasma biomarkers were assessed to evaluate intestinal health. Results showed that the average change in citrulline concentration (µmol/L) significantly increased among children who consumed MDCF-2 compared to those who consumed RUSF (mean difference-in-differences: 123.10; 95% CI: 3.60, 242.61; p = 0.044). The research findings demonstrated that MDCF-2 might have a beneficial effect on improving the gastrointestinal health of malnourished children.
Collapse
Affiliation(s)
- Ishita Mostafa
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212, Bangladesh; (R.A.S.); (U.H.L.); (M.T.); (M.M.); (S.M.T.H.); (T.A.)
- Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland
| | - Rahvia Alam Sthity
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212, Bangladesh; (R.A.S.); (U.H.L.); (M.T.); (M.M.); (S.M.T.H.); (T.A.)
| | - Umme Habiba Lamiya
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212, Bangladesh; (R.A.S.); (U.H.L.); (M.T.); (M.M.); (S.M.T.H.); (T.A.)
| | - Md. Tariqujjaman
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212, Bangladesh; (R.A.S.); (U.H.L.); (M.T.); (M.M.); (S.M.T.H.); (T.A.)
| | - Mustafa Mahfuz
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212, Bangladesh; (R.A.S.); (U.H.L.); (M.T.); (M.M.); (S.M.T.H.); (T.A.)
- Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland
| | - S. M. Tafsir Hasan
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212, Bangladesh; (R.A.S.); (U.H.L.); (M.T.); (M.M.); (S.M.T.H.); (T.A.)
| | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212, Bangladesh; (R.A.S.); (U.H.L.); (M.T.); (M.M.); (S.M.T.H.); (T.A.)
- Department of Public Health Nutrition, James P Grant School of Public Health, BRAC University, Dhaka 1212, Bangladesh
| |
Collapse
|
5
|
Horowitz A, Chanez-Paredes SD, Haest X, Turner JR. Paracellular permeability and tight junction regulation in gut health and disease. Nat Rev Gastroenterol Hepatol 2023:10.1038/s41575-023-00766-3. [PMID: 37186118 PMCID: PMC10127193 DOI: 10.1038/s41575-023-00766-3] [Citation(s) in RCA: 265] [Impact Index Per Article: 132.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2023] [Indexed: 05/17/2023]
Abstract
Epithelial tight junctions define the paracellular permeability of the intestinal barrier. Molecules can cross the tight junctions via two distinct size-selective and charge-selective paracellular pathways: the pore pathway and the leak pathway. These can be distinguished by their selectivities and differential regulation by immune cells. However, permeability increases measured in most studies are secondary to epithelial damage, which allows non-selective flux via the unrestricted pathway. Restoration of increased unrestricted pathway permeability requires mucosal healing. By contrast, tight junction barrier loss can be reversed by targeted interventions. Specific approaches are needed to restore pore pathway or leak pathway permeability increases. Recent studies have used preclinical disease models to demonstrate the potential of pore pathway or leak pathway barrier restoration in disease. In this Review, we focus on the two paracellular flux pathways that are dependent on the tight junction. We discuss the latest evidence that highlights tight junction components, structures and regulatory mechanisms, their impact on gut health and disease, and opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Arie Horowitz
- UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, Normandie University, Rouen, France
| | - Sandra D Chanez-Paredes
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xenia Haest
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Abtahi S, Sailer A, Roland JT, Haest X, Chanez-Paredes SD, Ahmad K, Sadiq K, Iqbal NT, Ali SA, Turner JR. Intestinal Epithelial Digestive, Transport, and Barrier Protein Expression Is Increased in Environmental Enteric Dysfunction. J Transl Med 2023; 103:100036. [PMID: 36870290 PMCID: PMC10121737 DOI: 10.1016/j.labinv.2022.100036] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 01/11/2023] Open
Abstract
Environmental enteric dysfunction (EED) is characterized by malabsorption and diarrhea that result in irreversible deficits in physical and intellectual growth. We sought to define the expression of transport and tight junction proteins by quantitative analysis of duodenal biopsies from patients with EED. Biopsies from Pakistani children with confirmed EED diagnoses were compared to those from age-matched North American healthy controls, patients with celiac disease, and patients with nonceliac disease with villous atrophy or intraepithelial lymphocytosis. Expression of brush border digestive and transport proteins and paracellular (tight junction) proteins was assessed by quantitative multiplex immunofluorescence microscopy. EED was characterized by partial villous atrophy and marked intraepithelial lymphocytosis. Epithelial proliferation and enteroendocrine, tuft, and Paneth cell numbers were unchanged, but there was significant goblet cell expansion in EED biopsies. Expression of proteins involved in nutrient and water absorption and that of the basolateral Cl- transport protein NKCC1 were also increased in EED. Finally, the barrier-forming tight junction protein claudin-4 (CLDN4) was significantly upregulated in EED, particularly within villous enterocytes. In contrast, expression of CFTR, CLDN2, CLDN15, JAM-A, occludin, ZO-1, and E-cadherin was unchanged. Upregulation of a barrier-forming tight junction protein and brush border and basolateral membrane proteins that support nutrient and water transport in EED is paradoxical, as their increased expression would be expected to be correlated with increased intestinal barrier function and enhanced absorption, respectively. These data suggest that EED activates adaptive intestinal epithelial responses to enhance nutrient absorption but that these changes are insufficient to restore health.
Collapse
Affiliation(s)
- Shabnam Abtahi
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anne Sailer
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Joseph T Roland
- Epithelial Biology Center, Vanderbilt University Medical Center; Nashville, Tennessee
| | - Xenia Haest
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sandra D Chanez-Paredes
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kumail Ahmad
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Kamran Sadiq
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Najeeha Talat Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - S Asad Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Department of Pathology, University of Chicago, Chicago, Illinois.
| |
Collapse
|
7
|
Lefebo BK, Kassa DH, Tarekegn BG. Factors associated with stunting: gut inflammation and child and maternal-related contributors among under-five children in Hawassa City, Sidama Region, Ethiopia. BMC Nutr 2023; 9:54. [PMID: 36945069 PMCID: PMC10031938 DOI: 10.1186/s40795-023-00701-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/03/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Under-nutrition remains a major global public health challenge, particularly among children under the age of five. Among the manifestations of under-nutrition, stunting accounts for the larger proportion, which is associated with multiple factors. In Ethiopia, however, the link between intestinal inflammation and childhood stunting was not well investigated. Therefore, the present study aimed to determine the association between gut inflammation and childhood stunting. METHOD A community-based cross-sectional study was conducted and a total of 82 children were included in the study. Anthropometric data were collected by measuring weight in underwear and without shoes with an electronic scale to the nearest 0.1 kg and their height in the Frankfort plane with a telescopic height instrument. Environmental risk factors for enteric bacterial exposure, access to improved sources of drinking water, and the presence of facilities for hygiene and sanitation conditions were assessed using a questionnaire. Gut inflammation was tested through fecal leukocyte count and each sample was stained with methylene blue. Stool samples were inoculated on MacConkey agar, Salmonella-Shigella agar, and Xylose Lysine Deoxycholate agar after enrichment with Selenite cystine broth and incubated at 37 °C for 18-24 h. Binary and multiple logistic regressions and Chi-square models were used to analyze the data. RESULT Data from the current study revealed that gut inflammation was (AOR: 5.28, 95% CI: 1.32-22.25) associated with stunting. On the other hand, children with reported diarrhea within the last week were 6 times more likely for the probability of being stunted (AOR: 6.21, 95% CI: 2.68-26.83). The findings of this study also demonstrated that children from a household with a family size of more than 5 members were three times more likely to be stunted than their counterparts (AOR: 3.21, 95% CI: 1.20 -10.13). Facts of the current study demonstrated that breastfeeding for 24 months and below was negatively associated (AOR: 0.3; 95% CI: -0.46-0.89) with gut inflammation. Detection of E.coli and Shigella species in the stool samples of children and Menaheria residents were positively associated with gut inflammation (AOR: 5.4, 95% CI: 1.32-22.25; AOR: 5, 95% CI: 1.47-24.21), respectively. CONCLUSION Therefore, there was a strong correlation between stunting and gastrointestinal inflammation. Moreover, stunting was associated with diarrhea, breastfeeding duration, residence, and family size. Similarly, intestinal inflammation was linked to residence, breastfeeding duration, and the prevalence of bacterial infections such as E. coli and Shigella species.
Collapse
Affiliation(s)
- Berhanu Kibemo Lefebo
- School of Nutrition, Food Science and Technology, College of Agriculture, University of Hawassa, Hawassa, Ethiopia
| | - Dejene Hailu Kassa
- School of Public Health, College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia
| | - Baye Gelaw Tarekegn
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
8
|
Citrulline and kynurenine to tryptophan ratio: potential EED (environmental enteric dysfunction) biomarkers in acute watery diarrhea among children in Bangladesh. Sci Rep 2023; 13:1416. [PMID: 36697429 PMCID: PMC9876903 DOI: 10.1038/s41598-023-28114-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023] Open
Abstract
Two emerging biomarkers of environmental enteric dysfunction (EED) include plasma citrulline (CIT), and the kynurenine (KYN): tryptophan (TRP)/ (KT) ratio. We sought to investigate the plasma concentration of CIT and KT ratio among the children having dehydrating diarrhea and examine associations between concentrations of CIT and KT ratio with concurrent factors. For this analysis, we used cross-sectional data from a total of 102, 6-36 months old male children who suffered from non-cholera acute watery diarrhea and had some dehydration admitted to an urban diarrheal hospital, in Bangladesh. CIT, TRP, and KYN concentrations were determined at enrollment from plasma samples using ELIZA. At enrollment, the mean plasma CIT concentration was 864.48 ± 388.55 µmol/L. The mean plasma kynurenine, tryptophan concentrations, and the KT ratio (× 1000) were 6.93 ± 3.08 µmol/L, 33.44 ± 16.39 µmol/L, and 12.12 ± 18.10, respectively. With increasing child age, KYN concentration decreased (coefficient: - 0.26; 95%CI: - 0.49, - 0.04; p = 0.021); with increasing lymphocyte count, CIT concentration decreased (coef.: - 0.01; 95% CI: - 0.02,0.001, p = 0.004); the wasted child had decreased KT ratio (coef.: - 0.6; 95% CI: - 1.18, - 0.02; p = 0.042) after adjusting for potential covariates. The CIT concentration was associated with blood neutrophils (coef.: 0.02; 95% CI: 0.01, 0.03; p < 0.001), lymphocytes (coef.: - 0.02; 95% CI: - 0.03, - 0.02; p < 0.001) and monocyte (coef.: 0.06; 95% CI: 0.01, 0.11; p = 0.021); KYN concentration was negatively associated with basophil (coef.: - 0.62; 95% CI: - 1.23, - 0.01; p = 0.048) after adjusting for age. In addition, total stool output (gm) increased (coef.: 793.84; 95% CI: 187.16, 1400.52; p = 0.011) and also increased duration of hospital stay (hour) (coef.: 22.89; 95% CI: 10.24, 35.54; p = 0.001) with increasing CIT concentration. The morphological changes associated with EED may increase the risk of enteric infection and diarrheal disease among children. Further research is critically needed to better understand the complex mechanisms by which EED biomarkers may impact susceptibility to dehydrating diarrhea in children.
Collapse
|
9
|
Past COVID-19: The Impact on IVF Outcomes Based on Follicular Fluid Lipid Profile. Int J Mol Sci 2022; 24:ijms24010010. [PMID: 36613453 PMCID: PMC9820085 DOI: 10.3390/ijms24010010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/12/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Follicular fluid is an important component of follicle growth and development. Negative effects of COVID-19 on follicular function are still open. The aim of this work was to study the features of the lipid profile of follicular fluid and evaluate the results of the in vitro fertilization (IVF) program in women after COVID-19 to identify biomarkers with prognostic potential. The study involved samples of follicular fluid collected from 237 women. Changes in the lipid composition of the follicular fluid of patients who underwent COVID-19 in mild and severe forms before entering the IVF program and women who did not have COVID-19 were studied by mass spectrometry. Several lipids were identified that significantly changed their level. On the basis of these findings, models were developed for predicting the threat of miscarriage in patients who had a severe course of COVID-19 and models for predicting the success of the IVF procedure, depending on the severity of COVID-19. Of practical interest is the possibility of using the developed predictive models in working with patients who have undergone COVID-19 before entering the IVF program. The results of the study suggest that the onset of pregnancy and its outcome after severe COVID-19 may be associated with changes in lipid metabolism in the follicular fluid.
Collapse
|
10
|
Kummerlowe C, Mwakamui S, Hughes TK, Mulugeta N, Mudenda V, Besa E, Zyambo K, Shay JES, Fleming I, Vukovic M, Doran BA, Aicher TP, Wadsworth MH, Bramante JT, Uchida AM, Fardoos R, Asowata OE, Herbert N, Yilmaz ÖH, Kløverpris HN, Garber JJ, Ordovas-Montanes J, Gartner Z, Wallach T, Shalek AK, Kelly P. Single-cell profiling of environmental enteropathy reveals signatures of epithelial remodeling and immune activation. Sci Transl Med 2022; 14:eabi8633. [PMID: 36044598 PMCID: PMC9594855 DOI: 10.1126/scitranslmed.abi8633] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Environmental enteropathy (EE) is a subclinical condition of the small intestine that is highly prevalent in low- and middle-income countries. It is thought to be a key contributing factor to childhood malnutrition, growth stunting, and diminished oral vaccine responses. Although EE has been shown to be the by-product of a recurrent enteric infection, its full pathophysiology remains unclear. Here, we mapped the cellular and molecular correlates of EE by performing high-throughput, single-cell RNA-sequencing on 33 small intestinal biopsies from 11 adults with EE in Lusaka, Zambia (eight HIV-negative and three HIV-positive), six adults without EE in Boston, United States, and two adults in Durban, South Africa, which we complemented with published data from three additional individuals from the same clinical site. We analyzed previously defined bulk-transcriptomic signatures of reduced villus height and decreased microbial translocation in EE and showed that these signatures may be driven by an increased abundance of surface mucosal cells-a gastric-like subset previously implicated in epithelial repair in the gastrointestinal tract. In addition, we determined cell subsets whose fractional abundances associate with EE severity, small intestinal region, and HIV infection. Furthermore, by comparing duodenal EE samples with those from three control cohorts, we identified dysregulated WNT and MAPK signaling in the EE epithelium and increased proinflammatory cytokine gene expression in a T cell subset highly expressing a transcriptional signature of tissue-resident memory cells in the EE cohort. Together, our work elucidates epithelial and immune correlates of EE and nominates cellular and molecular targets for intervention.
Collapse
Affiliation(s)
- Conner Kummerlowe
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Simutanyi Mwakamui
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine; Lusaka, Zambia
| | - Travis K. Hughes
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Nolawit Mulugeta
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Victor Mudenda
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine; Lusaka, Zambia
| | - Ellen Besa
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine; Lusaka, Zambia
| | - Kanekwa Zyambo
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine; Lusaka, Zambia
| | - Jessica E. S. Shay
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital; Boston, MA, 02114, USA
| | - Ira Fleming
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Marko Vukovic
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Ben A. Doran
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital; Boston, MA 02115, USA
| | - Toby P. Aicher
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Marc H. Wadsworth
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | | | - Amiko M. Uchida
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital; Boston, MA 02115, USA
- Cancer Immunology and Virology, Dana Farber Cancer Institute; Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School; Boston MA, 02115, USA
| | - Rabiah Fardoos
- Africa Health Research Institute, Durban, 4001, South Africa
| | | | | | - Ömer H. Yilmaz
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Department of Pathology, MGH, Harvard Medical School, Boston, MA, 02115, USA
| | | | - John J. Garber
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital; Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School; Boston MA, 02115, USA
| | - Jose Ordovas-Montanes
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital; Boston, MA 02115, USA
- Program in Immunology, Harvard Medical School; Boston, MA, 02115, USA
- Harvard Stem Cell Institute; Cambridge, MA, 02138, USA
| | - Zev Gartner
- University of California San Francisco; San Francisco, CA, 94185 USA
| | - Thomas Wallach
- SUNY Downstate Health Sciences University; Department of Pediatrics, Brooklyn, NY, 11203, USA
| | - Alex K. Shalek
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Department of Pathology, MGH, Harvard Medical School, Boston, MA, 02115, USA
- Program in Immunology, Harvard Medical School; Boston, MA, 02115, USA
| | - Paul Kelly
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital; Boston, MA, 02114, USA
- Blizard Institute, Queen Mary University of London; London E1 2AT, United Kingdom
| |
Collapse
|
11
|
Bauer KC, Littlejohn PT, Ayala V, Creus-Cuadros A, Finlay BB. Nonalcoholic Fatty Liver Disease and the Gut-Liver Axis: Exploring an Undernutrition Perspective. Gastroenterology 2022; 162:1858-1875.e2. [PMID: 35248539 DOI: 10.1053/j.gastro.2022.01.058] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/31/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic condition affecting one quarter of the global population. Although primarily linked to obesity and metabolic syndrome, undernutrition and the altered (dysbiotic) gut microbiome influence NAFLD progression. Both undernutrition and NAFLD prevalence are predicted to considerably increase, but how the undernourished gut microbiome contributes to hepatic pathophysiology remains far less studied. Here, we present undernutrition conditions with fatty liver features, including kwashiorkor and micronutrient deficiency. We then review the gut microbiota-liver axis, highlighting key pathways linked to NAFLD progression within both overnutrition and undernutrition. To conclude, we identify challenges and collaborative possibilities of emerging multiomic research addressing the pathology and treatment of undernourished NAFLD.
Collapse
Affiliation(s)
- Kylynda C Bauer
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada; Thoracic and Gastrointestinal Malignancies Branch, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland
| | - Paula T Littlejohn
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Victoria Ayala
- Institut de Recerca Biomèdica de Lleida (IRB-Lleida), Lleida, Spain; Department of Experimental Medicine, Universitat de Lleida, Lleida, Spain
| | - Anna Creus-Cuadros
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada; Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
12
|
Gazi MA, Siddique MA, Alam MA, Hossaini F, Hasan MM, Fahim SM, Wahid BZ, Kabir MM, Das S, Mahfuz M, Ahmed T. Plasma Kynurenine to Tryptophan Ratio Is Not Associated with Undernutrition in Adults but Reduced after Nutrition Intervention: Results from a Community-Based Study in Bangladesh. Nutrients 2022; 14:nu14091708. [PMID: 35565678 PMCID: PMC9104876 DOI: 10.3390/nu14091708] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 12/10/2022] Open
Abstract
Infections and persistent immunological activation are linked to increased kynurenine (KYN) and the KYN-to-Tryptophan (TRP) or KT ratio and may be critical factors in undernutrition. We sought to determine the association between the KT ratio and adult malnutrition, as well as investigate if nutritional supplementation had any influence on the decrease of the KT ratio. A total of 525 undernourished adults aged 18–45 years were recruited and provided a nutrition intervention for 60 feeding days. TRP and KYN concentrations were determined from plasma samples using LC-MS/MS. At baseline, the median (interquartile range (IQR)) TRP, KYN and KT ratios were 24.1 (17.6, 34.3) µmol/L, 0.76 (0.53, 1.18) µmol/L and 30.9 (24.5, 41.7), respectively. Following intervention, the median (IQR) KYN and KT ratios were significantly reduced to 0.713 (0.46, 1.12) µmol/L and 27.5 (21.3, 35.8). The KT ratio was found to be inversely linked with adult BMI (coefficient: −0.09; 95% CI: −0.18, 0.004; p-value = 0.06) but not statistically significant. Additionally, Plasma CRP was correlated positively, while LRP1 was inversely correlated with the KT ratio. Our data suggest that in Bangladeshi adults, the KT ratio is not related to the pathophysiology of malnutrition but correlated with inflammatory and anti-inflammatory biomarkers, and the ratio can be reduced by a nutrition intervention.
Collapse
Affiliation(s)
- Md. Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Md. Abdullah Siddique
- Emerging Infections and Parasitology Laboratory, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.S.); (M.M.K.)
| | - Md. Ashraful Alam
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Farzana Hossaini
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Md. Mehedi Hasan
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Shah Mohammad Fahim
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Barbie Zaman Wahid
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Md. Mamun Kabir
- Emerging Infections and Parasitology Laboratory, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.S.); (M.M.K.)
| | - Subhasish Das
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
- Faculty of Medicine and Life Sciences, University of Tampere, 33100 Tampere, Finland
- Correspondence:
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (M.A.G.); (M.A.A.); (F.H.); (M.M.H.); (S.M.F.); (B.Z.W.); (S.D.); (T.A.)
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
13
|
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: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
|
14
|
Zambrana LE, Weber AM, Borresen EC, Zarei I, Perez J, Perez C, Rodríguez I, Becker-Dreps S, Yuan L, Vilchez S, Ryan EP. Daily Rice Bran Consumption for 6 Months Influences Serum Glucagon-Like Peptide 2 and Metabolite Profiles without Differences in Trace Elements and Heavy Metals in Weaning Nicaraguan Infants at 12 Months of Age. Curr Dev Nutr 2021; 5:nzab101. [PMID: 34514286 PMCID: PMC8421236 DOI: 10.1093/cdn/nzab101] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/09/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is associated with chronic gut inflammation affecting nutrient absorption and development of children, primarily in low- and middle-income countries. Several studies have shown that rice bran (RB) supplementation provides nutrients and modulates gut inflammation, which may reduce risk for undernutrition. OBJECTIVE The aim was to evaluate the effect of daily RB dietary supplementation for 6 mo on serum biomarkers in weaning infants and associated changes in serum and stool metabolites. METHODS A 6-mo randomized-controlled dietary intervention was conducted in a cohort of weaning 6-mo-old infants in León, Nicaragua. Anthropometric indices were obtained at 6, 8, and 12 mo. Serum and stool ionomics and metabolomics were completed at the end of the 6-mo intervention using inductively coupled plasma MS and ultra-high performance LC-tandem MS. The ɑ1-acid glycoprotein, C-reactive protein, and glucagon-like peptide 2 (GLP-2) serum EED biomarkers were measured by ELISA. RESULTS Twenty-four infants in the control group and 23 in the RB group successfully completed the 6-mo dietary intervention with 90% dietary compliance. RB participants had higher concentrations of GLP-2 as compared with control participants at 12 mo [median (IQR): 743.53 (380.54) pg/mL vs. 592.50 (223.59) pg/mL; P = 0.04]. Metabolite profiles showed significant fold differences of 39 serum metabolites and 44 stool metabolites from infants consuming RB compared with control, and with significant metabolic pathway enrichment scores of 4.7 for the tryptophan metabolic pathway, 5.7 for polyamine metabolism, and 5.7 for the fatty acid/acylcholine metabolic pathway in the RB group. No differences were detected in serum and stool trace elements or heavy metals following daily RB intake for 6 mo. CONCLUSIONS RB consumption influences a suite of metabolites associated with growth promotion and development, while also supporting nutrient absorption as measured by changes in serum GLP-2 in Nicaraguan infants. This clinical trial was registered at https://clinicaltrials.gov as NCT02615886.
Collapse
Affiliation(s)
- Luis E Zambrana
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Center of Infectious Diseases, Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León (UNAN-León), León, Nicaragua
| | - Annika M Weber
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Erica C Borresen
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Iman Zarei
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Johann Perez
- Center of Infectious Diseases, Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León (UNAN-León), León, Nicaragua
| | - Claudia Perez
- Center of Infectious Diseases, Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León (UNAN-León), León, Nicaragua
| | - Iker Rodríguez
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Biotic Products Development Center, National Polytechnic Institute, Morelos, Mexico
| | - Sylvia Becker-Dreps
- Departments of Family Medicine and Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Samuel Vilchez
- Center of Infectious Diseases, Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León (UNAN-León), León, Nicaragua
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
15
|
Parikh P, Semba R, Manary M, Swaminathan S, Udomkesmalee E, Bos R, Poh BK, Rojroongwasinkul N, Geurts J, Sekartini R, Nga TT. Animal source foods, rich in essential amino acids, are important for linear growth and development of young children in low- and middle-income countries. MATERNAL AND CHILD NUTRITION 2021; 18:e13264. [PMID: 34467645 PMCID: PMC8710096 DOI: 10.1111/mcn.13264] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/02/2021] [Accepted: 08/02/2021] [Indexed: 11/28/2022]
Abstract
Growth faltering under 5 years of age is unacceptably high worldwide, and even more children, while not stunted, fail to reach their growth potential. The time between conception and 2 years of age is critical for development. The period from 6 to 23 months, when complementary foods are introduced, coincides with a time when growth faltering and delayed neurocognitive developments are most common. Fortunately, this is also the period when diet exercises its greatest influence. Growing up in an adverse environment, with a deficient diet, as typically seen in low‐ and middle‐income countries (LMICs), hampers growth and development of children and prevents them from realising their full developmental and economic future potential. Sufficient nutrient availability and utilisation are paramount to a child's growth and development trajectory, especially in the period after breastfeeding. This review highlights the importance of essential amino acids (EAAs) in early life for linear growth and, likely, neurocognitive development. The paper further discusses signalling through mammalian target of rapamycin complex 1 (mTORC1) as one of the main amino acid (AA)‐sensing hubs and the master regulator of both growth and neurocognitive development. Children in LMICs, despite consuming sufficient total protein, do not meet their EAA requirements due to poor diet diversity and low‐quality dietary protein. AA deficiencies in early life can cause reductions in linear growth and cognition. Ensuring AA adequacy in diets, particularly through inclusion of nutrient‐dense animal source foods from 6 to 23 months, is strongly encouraged in LMICs in order to compensate for less than optimal growth during complementary feeding.
Collapse
Affiliation(s)
| | - Richard Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark Manary
- Department of Paediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sumathi Swaminathan
- St John's Research Institute, St John's National Academy of Health Sciences, Bangalore, Karnataka, India
| | | | - Rolf Bos
- FrieslandCampina, Amersfoort, The Netherlands
| | - Bee Koon Poh
- Nutritional Sciences Programme & Centre for Community Health, Faculty of Health Sciences, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | - Jan Geurts
- FrieslandCampina, Amersfoort, The Netherlands
| | - Rini Sekartini
- Faculty of Medicine, Department of Pediatrics, University of Indonesia and Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Tran Thuy Nga
- Department of Occupational and School Nutrition, National Institute of Nutrition (NIN), Hanoi, Vietnam
| |
Collapse
|
16
|
Derakhshandeh-Rishehri PhD Student SM, Shenavar MSc R, Farmani Bs A, Hemmati PhD A, Faghih PhD S. The effects of nutritional support baskets on growth parameters of under five years old children with malnutrition and low socio-economic status. J Trop Pediatr 2021; 67:6139356. [PMID: 33594420 DOI: 10.1093/tropej/fmab007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AND OBJECTIVES Although malnutrition is globally widespread among children, there is no consensus on the most effective intervention for improving a child's growth. The present study is designed to investigate the effects of nutritional support basketson growth indices of malnourished children. METHODS This is a field trial conducted on 3667 malnourished children (0-60 months) for 9months, between 2017 and 2018, in Fars province of Iran. Weight-for-age, height-for-age, weight-for-height and body mass index-for age indices were assessed before and after the intervention with nutritional support baskets, which contains 600 kcal/day. RESULTS The baseline prevalence of moderate/severeunderweight, stunting and wasting were 31.5%, 33.3% and 28.9%, respectively. After the intervention, the prevalence non-significantly reduced to 25.5%, 31.7% and 20.35%, respectively (p > 0.05). The intervention is associated with a non-significant reduction in the prevalence of underweight and wasting in 0-23 months children, and a non-significant reduction in the prevalence of underweight, stunting and wasting in 24-60 months children. Furthermore, the intervention is associated with a non-significant reduction in the prevalence of underweight, stunting and wasting in girls, and a non-significant reduction in the prevalence of underweight and wasting in boys. CONCLUSIONS Nutrition support was effective in improving malnutrition indices of children; however, the results were non-significant. Further studies with longer period and control group areneeded to support the effectiveness of nutrition support in children.
Collapse
Affiliation(s)
- Seyedeh-Masomeh Derakhshandeh-Rishehri PhD Student
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Community Nutrition, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Shenavar MSc
- Department of Community Nutrition, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azam Farmani Bs
- Department of Community Nutrition, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Shiva Faghih PhD
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Community Nutrition, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
17
|
Selimoglu MA, Kansu A, Aydogdu S, Sarioglu AA, Erdogan S, Dalgic B, Yuce A, Cullu Cokugras F. Nutritional Support in Malnourished Children With Compromised Gastrointestinal Function: Utility of Peptide-Based Enteral Therapy. Front Pediatr 2021; 9:610275. [PMID: 34164352 PMCID: PMC8215107 DOI: 10.3389/fped.2021.610275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 05/07/2021] [Indexed: 12/20/2022] Open
Abstract
This review focuses on nutritional support in malnourished children with compromised gastrointestinal function addressing the interplay between malnutrition and gastrointestinal dysfunction, and the specific role of peptide-based enteral therapy in pediatric malnutrition. Malnutrition is associated with impaired gut functions such as increased intestinal permeability, malabsorption, and diarrhea, while pre-existing functional gastrointestinal disorders may also lead to malnutrition. Presence of compromised gastrointestinal function in malnourished children is critical given that alterations such as malabsorption and increased intestinal permeability directly interfere with efficacy of nutritional support and recovery from malnutrition. Appropriate nutritional intervention is the key step in the management of malnutrition, while alterations in gastrointestinal functions in malnourished children are likely even in those with mild degree malnutrition. Therefore, nutritional therapy in children with compromised gastrointestinal function is considered to involve gut-protective interventions that address the overlapping and interacting effects of diarrhea, enteropathy and malnutrition to improve child survival and developmental potential in the long-term. Peptide-based enteral formulas seem to have clinical applications in malnourished children with compromised gastrointestinal function, given their association with improved gastrointestinal tolerance and absorption, better nitrogen retention/ balance, reduced diarrhea and bacterial translocation, enhanced fat absorption, and maintained/restored gut integrity as compared with free amino acid or whole-protein formulas.
Collapse
Affiliation(s)
- Mukadder Ayse Selimoglu
- Department of Pediatric Gastroenterology, Atasehir and Bahcelievler Memorial Hospitals, Istanbul, Turkey
| | - Aydan Kansu
- Department of Pediatric Gastroenterology, Ankara University School of Medicine, Ankara, Turkey
| | - Sema Aydogdu
- Department of Pediatric Gastroenterology, Ege University Faculty of Medicine, Izmir, Turkey
| | | | | | - Buket Dalgic
- Department of Pediatric Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Aysel Yuce
- Department of Pediatric Gastroenterology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Fugen Cullu Cokugras
- Department of Pediatric Gastroenterology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
18
|
Parpia TC, Elwood SE, Scharf RJ, McDermid JM, Wanjuhi AW, Rogawski McQuade ET, Gratz J, Svensen E, Swann JR, Donowitz JR, Jatosh S, Katengu S, Mdoe P, Kivuyo S, Houpt ER, DeBoer MD, Mduma E, Platts-Mills JA. Baseline Characteristics of Study Participants in the Early Life Interventions for Childhood Growth and Development in Tanzania (ELICIT) Trial. Am J Trop Med Hyg 2020; 103:1397-1404. [PMID: 32783799 PMCID: PMC7543831 DOI: 10.4269/ajtmh.19-0918] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Recurrent enteric infections and micronutrient deficiencies, including deficiencies in the tryptophan–kynurenine–niacin pathway, have been associated with environmental enteric dysfunction, potentially contributing to poor child growth and development. We are conducting a randomized, placebo-controlled, 2 × 2 factorial interventional trial in a rural population in Haydom, Tanzania, to determine the effect of 1) antimicrobials (azithromycin and nitazoxanide) and/or 2) nicotinamide, a niacin vitamer, on attained length at 18 months. Mother/infant dyads were enrolled within 14 days of the infant’s birth from September 2017 to September 2018, with the follow-up to be completed in February 2020. Here, we describe the baseline characteristics of the study cohort, risk factors for low enrollment weight, and neonatal adverse events (AEs). Risk factors for a low enrollment weight included being a firstborn child (−0.54 difference in weight-for-age z-score [WAZ] versus other children, 95% CI: −0.71, −0.37), lower socioeconomic status (−0.28, 95% CI: −0.43, −0.12 difference in WAZ), and birth during the preharvest season (November to March) (−0.22, 95% CI: −0.33, −0.11 difference in WAZ). The most common neonatal serious AEs were respiratory tract infections and neonatal sepsis (2.2 and 1.4 events per 100 child-months, respectively). The study cohort represents a high-risk population for whom interventions to improve child growth and development are urgently needed. Further analyses are needed to understand the persistent impacts of seasonal malnutrition and the interactions between seasonality, socioeconomic status, and the study interventions.
Collapse
Affiliation(s)
- Tarina C Parpia
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia
| | - Sarah E Elwood
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia
| | - Rebecca J Scharf
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Joann M McDermid
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia
| | - Anne W Wanjuhi
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | | | - Jean Gratz
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia
| | | | - Jonathan R Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jeffrey R Donowitz
- Division of Infectious Disease, Children's Hospital of Richmond at Virginia Commonwealth University, Richmond, Virginia
| | - Samwel Jatosh
- Haydom Global Health Research Centre, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Siphael Katengu
- Haydom Global Health Research Centre, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Paschal Mdoe
- Haydom Global Health Research Centre, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Sokoine Kivuyo
- National Institute for Medical Research, Muhimbili Medical Research Centre, Dar es Salaam, Tanzania
| | - Eric R Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia
| | - Mark D DeBoer
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Estomih Mduma
- Haydom Global Health Research Centre, Haydom Lutheran Hospital, Haydom, Tanzania
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
19
|
Gazi MA, Das S, Siddique MA, Alam MA, Fahim SM, Hasan MM, Hossaini F, Kabir MM, Noor Z, Haque R, Mahfuz M, Ahmed T. Plasma Kynurenine to Tryptophan Ratio Is Negatively Associated with Linear Growth of Children Living in a Slum of Bangladesh: Results from a Community-Based Intervention Study. Am J Trop Med Hyg 2020; 104:766-773. [PMID: 33236707 PMCID: PMC7866305 DOI: 10.4269/ajtmh.20-0049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 09/26/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic exposure to infectious agents results in environmental enteric dysfunction-a significant contributor to childhood stunting. Low plasma tryptophan (TRP), increased kynurenine (KYN), and KYN-TRP (KT) ratio are associated with infections and chronic immune activation. We postulated that both these conditions are interlinked, and therefore aimed to identify the association between KT ratio and the linear growth of Bangladeshi children. A total of 480 stunted and at risk of being stunted children aged 12-18 months were enrolled and provided nutrition intervention for 90 days. Plasma samples were assessed using liquid chromatography tandem mass spectrometry to measure TRP and KYN concentrations. Multivariable linear regression with generalized estimating equations was applied to analyze association between the KT ratio and linear growth. Tryptophan, KYN, and KT ratio were significantly higher in stunted children than in children at risk of being stunted both at baseline and at the end of nutrition intervention. Following intervention, the median (interquartile range [IQR]) KYN concentration was significantly reduced from 4.6 (3.6, 5.4) µmol/L to 3.9 (0.3, 7.6) µmol/L, and median (IQR) KT ratio decreased from 104 (80.9, 131) to 92.8 (6.6, 247) in stunted children. We also found KT ratio to be negatively associated (coefficient = -0.7; 95% CI = -1.13, -0.26; P-value = 0.002) with linear growth. In addition, KYN and KT ratio were positively correlated with fecal neopterin and plasma C-reactive protein, whereas TRP was negatively correlated with both of these biomarkers and alpha-1-acid glycoprotein. Our findings imply that KT ratio is associated in the pathophysiology of stunting as well as with biomarkers of inflammation in Bangladeshi children.
Collapse
Affiliation(s)
- Md. Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Subhasish Das
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Abdullah Siddique
- Emerging Infection and Parasitology Laboratory, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Ashraful Alam
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Shah Mohammad Fahim
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Mehedi Hasan
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Farzana Hossaini
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Mamun Kabir
- Emerging Infection and Parasitology Laboratory, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Zannatun Noor
- Emerging Infection and Parasitology Laboratory, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Rashidul Haque
- Emerging Infection and Parasitology Laboratory, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- James P. Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| |
Collapse
|
20
|
Roediger R, Stein H, Callaghan‐Gillespie M, Blackman JK, Kohlmann K, Maleta K, Manary M. Protein quality in ready-to-use supplementary foods for moderate wasting. MATERNAL & CHILD NUTRITION 2020; 16:e13019. [PMID: 32426949 PMCID: PMC7507576 DOI: 10.1111/mcn.13019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 11/26/2022]
Abstract
There are no guidelines for the optimal protein quality of ready-to-supplementary food (RUSF) for moderate acute malnutrition (MAM). This randomized, controlled, double-blinded, clinical effectiveness trial evaluated two RUSFs in the treatment of MAM. Both foods contained greater than 7% dairy protein, but the protein-optimized RUSF had a calculated digestible indispensable amino acid score (DIAAS) of 95%, whereas the control RUSF had a calculated DIAAS of 63%. There were 1,737 rural Malawian children 6-59 months of age treated with 75 kcal/kg/day of either control or protein quality-optimized RUSF for up to 12 weeks. There was no difference in the proportion of children who recovered from MAM between the group that received protein-optimized RUSF (759/860, 88%) and the group that received control RUSF (766/877, 87%, difference 1%, 95% CI, -2.1 to 4.1, p = 0.61). There were no differences in time to recovery or average weight gain; nor were adverse effects reported. Both RUSFs showed indistinguishable clinical outcomes, with recovery rates higher than typically seen in treatment for MAM. The DIAAS of these two RUSFs was measured using a pig model. Unexpectedly, the protein quality of the optimized RUSF was inferior to the control RUSF: DIAAS = 82% for the protein quality optimized RUSF and 96% for control RUSF. The controlled conditions of this trial suggest that in supplementary food products for MAM, protein quality is not an independent predictor of clinical effectiveness.
Collapse
Affiliation(s)
- Rebecca Roediger
- Division of Gastroenterology, Department of MedicineWashington UniversitySt. LouisMissouriUSA
| | | | | | | | - Kristin Kohlmann
- Department of PediatricsWashington UniversitySt. LouisMissouriUSA
| | - Kenneth Maleta
- Department of Community Health, College of MedicineUniversity of MalawiBlantyreMalawi
| | - Mark Manary
- Department of PediatricsWashington UniversitySt. LouisMissouriUSA
- Department of Community Health, College of MedicineUniversity of MalawiBlantyreMalawi
| |
Collapse
|
21
|
CHEN L, ZHANG J, LI N, ZHANG L, XU XF. Suckling calves (Bos taurus) with pica exhibit blood metabolome alterations. THE INDIAN JOURNAL OF ANIMAL SCIENCES 2020. [DOI: 10.56093/ijans.v90i5.104613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The aim of this study was to evaluate the changes of blood metabolomics in calves with pica, which causes serious harm to livestock. Two groups, each comprising 12 calves of approximately 3 weeks old and of similar weight were selected as subjects; Group A calves were control animals in good condition and Group B calves (pica animals) had rough hair, emaciation, flaccid forestomach, diarrhoea, and stunted development. Blood samples were collected from the tail root vein. Masslynx 4.1 software (Waters Company) was used to pre-process data, which were then analysed by principal component analysis, partial least squares discriminant analysis, and orthogonal partial least squares discriminant analysis. Twenty potential biomarkers were closely related to the occurrence of pica, viz. GDP-glucose, UDP-glucose, proline, creatine, arginine, glutamine, citrulline, urea, alanine, methionine, serine, glycerate, cysteine, spermine, spermidine, carnitine, xanthurenic acid, kynurenine, and thyroxine. Metabolic pathway analysis showed that, in calves, pica resulted in decreased antioxidant capacity; disruption of the mutual transformation between pentose and glucuronic acid; abnormal metabolism of cysteine, methionine, serine, arginine, and proline; impairment of lipid metabolism; reduced immunity; increased intestinal permeability; and elevated central nervous excitability. Calves with pica exhibit disruption of various metabolic pathways.
Collapse
|
22
|
Bauer KC, Huus KE, Brown EM, Bozorgmehr T, Petersen C, Cirstea MS, Woodward SE, McCoy J, Hun J, Pamplona R, Ayala V, Finlay BB. Dietary Intervention Reverses Fatty Liver and Altered Gut Microbiota during Early-Life Undernutrition. mSystems 2020; 5:e00499-20. [PMID: 32900869 PMCID: PMC7483509 DOI: 10.1128/msystems.00499-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/01/2020] [Indexed: 01/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), largely studied as a condition of overnutrition, also presents in undernourished populations. Like NAFLD, undernutrition disrupts systemic metabolism and has been linked to gut microbiota dysbiosis. Indeed, chronic exposures to fecal microbes contribute to undernutrition pathology in regions with poor sanitation. Despite a growing prevalence of fatty liver disease, the influence of undernutrition and the gut microbiota remain largely unexplored. Here, we utilize an established murine model (C57BL/6J mice placed on a malnourished diet that received iterative Escherichia coli/Bacteroidales gavage [MBG mice]) that combines a protein/fat-deficient diet and iterative exposure to specific, fecal microbes. Fecal-oral contamination exacerbates triglyceride accumulation in undernourished mice. MBG livers exhibit diffuse lipidosis accompanied by striking shifts in fatty acid, glycerophospholipid, and retinol metabolism. Multiomic analyses revealed metabolomic pathways linked to the undernourished gut microbiome and hepatic steatosis, including phenylacetate metabolism. Intriguingly, fatty liver features were observed only in the early-life, but not adult, MBG model despite similar liver metabolomic profiles. Importantly, we demonstrate that dietary intervention largely mitigates aberrant metabolomic and microbiome features in MBG mice. These findings indicate a crucial window in early-life development that, when disrupted by nutritional deficiency, may significantly influence liver function. Our work provides a multifaceted study of how diet and gut microbes inform fatty liver progression and reversal during undernutrition.IMPORTANCE Nonalcoholic fatty liver disease (NAFLD) remains a global epidemic, but it is often studied in the context of obesity and aging. Nutritional deficits, however, also trigger hepatic steatosis, influencing health trajectories in undernourished pediatric populations. Here, we report that exposure to specific gut microbes impacts fatty liver pathology in mice fed a protein/fat-deficient diet. We utilize a multiomics approach to (i) characterize NAFLD in the context of early undernutrition and (ii) examine the impact of diet and gut microbes in the pathology and reversal of hepatic steatosis. We provide compelling evidence that an early-life, critical development window facilitates undernutrition-induced fatty liver pathology. Moreover, we demonstrate that sustained dietary intervention largely reverses fatty liver features and microbiome shifts observed during early-life malnutrition.
Collapse
Affiliation(s)
- K C Bauer
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - K E Huus
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - E M Brown
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - T Bozorgmehr
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - C Petersen
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - M S Cirstea
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - S E Woodward
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - J McCoy
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - J Hun
- The Metabolomics Innovation Centre, University of Victoria, British Columbia, Canada
| | - R Pamplona
- Institut de Recerca Biomèdica de Lleida (IRB-Lleida), Lleida, Spain
- Department of Metabolomic Physiology, Universitat de Lleida, Lleida, Spain
| | - V Ayala
- Institut de Recerca Biomèdica de Lleida (IRB-Lleida), Lleida, Spain
- Department of Metabolomic Physiology, Universitat de Lleida, Lleida, Spain
| | - B B Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
- Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, Canada
| |
Collapse
|
23
|
Zhang X, Wang T, Song J, Deng J, Sun Z. Study on follicular fluid metabolomics components at different ages based on lipid metabolism. Reprod Biol Endocrinol 2020; 18:42. [PMID: 32398082 PMCID: PMC7216654 DOI: 10.1186/s12958-020-00599-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/24/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Follicular fluid is an important external environment for the growth and development of oocytes. A thorough identification of specific components in follicular fluid can better the existing understand of intracellular signal transduction and reveal potential biomarkers of oocyte health in women undergoing assisted reproductive therapy. To study on follicular fluid metabolomics components at different ages based on lipid metabolism, we have adopted a new method of SWATH to MRM(the sequential window acquisition of all theoretical fragment-ion spectra to multiple reaction monitor)metabolomics to provide extensive coverage and excellent quantitative data. This was done to investigate the differences in follicular fluid of patients undergoing in vitro fertilization (IVF) and embryo transfer in different age groups and to further explore the relationship between follicular fluid, age and reproductive function. METHOD A combination of Ultra-high-performance liquid chromatography and high resolution mass spectrometry techniques were used to analyze the follicular fluid of 230 patients enrolled for the IVF cycle. The patients were of different ages grouped into two groups:the younger and older patients.The obtained multidimensional chromatographic data were processed by principal component analysis (PCA) and partial least squares discriminant analysis (PLS-DA). The charge ratios and mass numbers enabled for the identification of different fragments in the samples. Matching information obtained through database search and the fragment information obtained by fragment ion scan structurally identified substances in the samples. This was used to determine the differential compounds. RESULTS The quality of oocytes decline with age,and the lipid composition in follicular fluid also changes,The lipid metabolism that changes with age may be related to the quality of oocytes.The main differences were in lipid metabolites. Some were up-regulated: Arachidonate, LysoPC(16:1), LysoPC(20:4) and LysoPC(20:3) while others were down-regulated: LysoPC(18:3) and LysoPC(18:1). CONCLUSIONS Metabolomic analysis of follicular fluid revealed that with the increase in age, several differential metabolites are at play. Among these metabolites, lipid metabolism undergoes significant changes that affect the development of oocytes thus causing reduced fertility in older women. These differential metabolites related to follicular development may provide possible detection and treatment targets for promoting oocyte health, and provide scientific basis for understanding the environment of oocyte development.
Collapse
Affiliation(s)
- Xingxing Zhang
- Maternity and Child Health Care of ZaoZhuang, ZaoZhuang, 277100, Shandong, China
| | - Tianqi Wang
- Traditional Chinese Medicine History and Literature, Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jingyan Song
- Department of Gynecology and Obstetrics of Traditional Chinese Medicine, The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jifeng Deng
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou, 510640, China
| | - Zhengao Sun
- Reproductive and Genetic Center of Integrated Traditional and Western Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China.
| |
Collapse
|
24
|
Devi S, Varkey A, Dharmar M, Holt RR, Allen LH, Sheshshayee MS, Preston T, Keen CL, Kurpad AV. Amino Acid Digestibility of Extruded Chickpea and Yellow Pea Protein is High and Comparable in Moderately Stunted South Indian Children with Use of a Dual Stable Isotope Tracer Method. J Nutr 2020; 150:1178-1185. [PMID: 32006007 PMCID: PMC7198288 DOI: 10.1093/jn/nxaa004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/19/2019] [Accepted: 01/07/2020] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Legumes are an excellent plant source of the limiting indispensable amino acid (IAA) lysine in vegetarian, cereal-based diets. However, their digestibility is poor largely because of their antiprotease content. Extrusion can enhance digestibility by inactivating trypsin inhibitors and thus potentially improve the protein quality of legumes. OBJECTIVE We measured the digestibility of extruded chickpea and yellow pea protein with use of a dual stable isotope method in moderately stunted South Indian primary school children. METHODS Twenty-eight moderately stunted children (height-for-age z scores <-2.0 SD and >-3.0 SD) aged 6-11 y from low to middle socioeconomic status were randomly assigned to receive a test protein (extruded intrinsically [2H]-labeled chickpea or yellow pea) along with a standard of U-[13C]-spirulina protein to measure amino acid (AA) digestibility with use of a dual stable isotope method. Individual AA digestibility in the test protein was calculated by the ratios of AA enrichments in the test protein to the standard protein in the food and their appearance in blood plasma collected at 6 and 6.5 h during the experiment, representing a plateau state. RESULTS The mean AA digestibility of extruded chickpea and yellow pea protein in moderately stunted children (HAZ; -2.86 to -1.2) was high and similar in both extruded test proteins (89.0% and 88.0%, respectively, P = 0.83). However, lysine and proline digestibilities were higher in extruded chickpea than yellow pea (79.2% compared with 76.5% and 75.0% compared with 72.0%, respectively, P < 0.02). CONCLUSION Extruded chickpea and yellow pea protein had good IAA digestibility in moderately stunted children, which was 20% higher than an earlier report of their digestibility when pressure-cooked, measured by the same method in adults. Higher digestibility of lysine and proline highlights better retention of these AA in chickpea during extrusion-based processing. Extrusion might be useful for developing high-quality protein foods from legumes. This trial was registered at www.ctri.nic.in as CTRI/2018/03/012439.
Collapse
Affiliation(s)
- Sarita Devi
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - Aneesia Varkey
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - Madan Dharmar
- Betty Irene Moore School of Nursing and the Department of Pediatrics, University of California, Davis School of Medicine, Sacramento, CA, USA
| | - Roberta R Holt
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Lindsay H Allen
- United States Department of Agriculture, Agricultural Research Service Western Human Nutrition Research Center, Davis, CA, USA
| | - M S Sheshshayee
- Department of Crop Physiology, University of Agricultural Sciences, Bangalore, India
| | - Thomas Preston
- Scottish Universities Environmental Research Centre, East Kilbride, UK
| | - Carl L Keen
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Anura V Kurpad
- Department of Physiology, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| |
Collapse
|
25
|
Mahmud I, Kabir M, Haque R, Garrett TJ. Decoding the Metabolome and Lipidome of Child Malnutrition by Mass Spectrometric Techniques: Present Status and Future Perspectives. Anal Chem 2019; 91:14784-14791. [PMID: 31682425 DOI: 10.1021/acs.analchem.9b03338] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Child malnutrition (CM) is a global public health problem. It contributes to poor health in one in four children under five years worldwide and causes serious health problems in children, including stunted, wasted, and overweight growth. These serious public health issues lead to a higher chance of living in poverty in adulthood. Malnutrition is related with reduced economic productivity and increases the serious national and international burden. Currently, there is no meaningful therapeutic intervention of CM, and the use of different therapeutic foods has shown poor outcomes among supplemented malnourished children. The role of metabolites and lipids has been extensively recognized as early determinants of child health, but their contribution in CM and its pathobiology are poorly understood. This perspective provides a most recent update on these aspects. After briefly introducing the disciplines of metabolomics and lipidomics, we describe a mass spectrometry-based metabolic workflow for analysis of both metabolites and lipids and summarize several recent applications of metabolomics and lipidomics in CM. Finally, we discuss the future directions of the field toward the development of meaningful interventions for CM through metabolomics and lipidomics advances.
Collapse
Affiliation(s)
- Iqbal Mahmud
- Department of Pathology, Immunology, and Laboratory Medicine , University of Florida, College of Medicine , Gainesville , Florida 32608 , United States.,Southeast Center for Integrated Metabolomics (SECIM), Clinical and Translational Science Institute , University of Florida , Gainesville , Florida 32608 , United States
| | - Mamun Kabir
- Emerging Infections and Parasitology Laboratory, Infectious Disease Division , International Centre for Diarrheal Disease Research , Dhaka 1213 , Bangladesh
| | - Rashidul Haque
- Emerging Infections and Parasitology Laboratory, Infectious Disease Division , International Centre for Diarrheal Disease Research , Dhaka 1213 , Bangladesh
| | - Timothy J Garrett
- Department of Pathology, Immunology, and Laboratory Medicine , University of Florida, College of Medicine , Gainesville , Florida 32608 , United States.,Southeast Center for Integrated Metabolomics (SECIM), Clinical and Translational Science Institute , University of Florida , Gainesville , Florida 32608 , United States
| |
Collapse
|
26
|
Sun H, Zhang AH, Zhang HL, Zhou XH, Wang XQ, Liu L, Wang XJ. Ultra-performance liquid chromatography/mass spectrometry technology and high-throughput metabolomics for deciphering the preventive mechanism of mirabilite on colorectal cancer via the modulation of complex metabolic networks. RSC Adv 2019; 9:35356-35363. [PMID: 35528071 PMCID: PMC9074663 DOI: 10.1039/c9ra07687e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 10/17/2019] [Indexed: 01/22/2023] Open
Abstract
Colorectal cancer (CRC) is a highly virulent and malignant disease and always accompanied by metabolic disorders. Currently, there are no effective therapeutic drugs for the treatment of CRC. High-throughput metabolomics approaches have been used to unveil the metabolic pathways related to several diseases. In this study, ultra-performance liquid chromatography/mass spectrometry-based high-throughput metabolomics was used for deciphering the potential preventive mechanism of mirabilite on CRC via the modulation of the associated metabolic disorders; a total of 28 differential biomarkers, including indole acetaldehyde, 5-hydroxyindoleacetic acid, hypoxanthine, retinal, retinal ester, linoleic acid, stearic acid, 6-deoxocastasterone, 2-hydroxybutyric acid and LysoPC, were identified in the APCmin/+ mice. These differential biomarkers are involved in the tryptophan metabolism, glycerophospholipid metabolism and biosynthesis of unsaturated fatty acids. Note that these biomarkers and their disturbed metabolic pathways were also regulated by mirabilite. It has been found that the prevention of CRC by mirabilite is mainly associated with tryptophan metabolism; this study shows that high-throughput metabolomics can reveal the perturbed metabolic disorders targeted in the action mechanism of drug treatment.
Collapse
Affiliation(s)
- Hui Sun
- National Chinmedomics Research Center, Sino-America Chinmedomics Technology Collaboration Center, National TCM Key Laboratory of Serum Pharmacochemistry, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin China +86-451-82110818 +86-451-82110818
| | - Ai-Hua Zhang
- National Chinmedomics Research Center, Sino-America Chinmedomics Technology Collaboration Center, National TCM Key Laboratory of Serum Pharmacochemistry, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin China +86-451-82110818 +86-451-82110818
| | - Hong-Lian Zhang
- National Chinmedomics Research Center, Sino-America Chinmedomics Technology Collaboration Center, National TCM Key Laboratory of Serum Pharmacochemistry, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin China +86-451-82110818 +86-451-82110818
| | - Xiao-Hang Zhou
- National Chinmedomics Research Center, Sino-America Chinmedomics Technology Collaboration Center, National TCM Key Laboratory of Serum Pharmacochemistry, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin China +86-451-82110818 +86-451-82110818
| | - Xiang-Qian Wang
- National Chinmedomics Research Center, Sino-America Chinmedomics Technology Collaboration Center, National TCM Key Laboratory of Serum Pharmacochemistry, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin China +86-451-82110818 +86-451-82110818
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology Avenida Wai Long, Taipa Macau China
| | - Xi-Jun Wang
- National Chinmedomics Research Center, Sino-America Chinmedomics Technology Collaboration Center, National TCM Key Laboratory of Serum Pharmacochemistry, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin China +86-451-82110818 +86-451-82110818
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology Avenida Wai Long, Taipa Macau China
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant Nanning Guangxi China
| |
Collapse
|
27
|
Correlates of Gut Function in Children Hospitalized for Severe Acute Malnutrition, a Cross-sectional Study in Uganda. J Pediatr Gastroenterol Nutr 2019; 69:292-298. [PMID: 31169661 DOI: 10.1097/mpg.0000000000002381] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVE Children with severe acute malnutrition (SAM) may have impaired intestinal function, which can result in malabsorption, diarrhoea, and poor growth. This study evaluated the gut function of children with SAM using fecal and blood biomarkers and assessed their correlates. METHODS A cross-sectional study, nested in a randomized trial (www.isrctn.com, ISRCTN 16454889), was conducted at Mulago hospital, Uganda among subgroups of 400 children with complicated SAM and 30 community controls. Gut function was evaluated by 5 biomarkers: plasma citrulline, fecal myeloperoxidase and fecal neopterin, bacterially derived 16S rRNA gene and internal transcribed Spacer region (ITS) specific for Candida spp. in blood. RESULTS Compared with controls, children with SAM had lower median plasma citrulline (5.14 vs 27.4 μmol/L, P < 0.001), higher median fecal myeloperoxidase (18083 vs 7482 ng/mL, P = 0.001), and fecal neopterin (541 vs 210 nmol/L, P < 0.001). A higher blood concentration of 16S rRNA gene copy numbers was observed among children with SAM (95 vs 28 copies/μl, P = 0.05), whereas there was no difference in the blood concentration of Candida-specific ITS fragment.Among those with SAM, plasma citrulline was lower in children with edema, diarrhoea, dermatosis, and plasma C-reactive protein (CRP) >10 mg/L. Fecal neopterin was positively correlated with symptoms of fever and cough whereas it was negatively correlated with mid-upper arm circumference (MUAC), weight-for-height z score (WHZ), edema, and dermatosis. CONCLUSIONS Children with complicated SAM seem to have impaired gut function characterized by reduced enterocyte mass, intestinal inflammation, and increased bacterial translocation.
Collapse
|
28
|
Bourke CD, Jones KDJ, Prendergast AJ. Current Understanding of Innate Immune Cell Dysfunction in Childhood Undernutrition. Front Immunol 2019; 10:1728. [PMID: 31417545 PMCID: PMC6681674 DOI: 10.3389/fimmu.2019.01728] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022] Open
Abstract
Undernutrition affects millions of children in low- and middle-income countries (LMIC) and underlies almost half of all deaths among children under 5 years old. The growth deficits that characterize childhood undernutrition (stunting and wasting) result from simultaneous underlying defects in multiple physiological processes, and current treatment regimens do not completely normalize these pathways. Most deaths among undernourished children are due to infections, indicating that their anti-pathogen immune responses are impaired. Defects in the body's first-line-of-defense against pathogens, the innate immune system, is a plausible yet understudied pathway that could contribute to this increased infection risk. In this review, we discuss the evidence for innate immune cell dysfunction from cohort studies of childhood undernutrition in LMIC, highlighting knowledge gaps in almost all innate immune cell types. We supplement these gaps with insights from relevant experimental models and make recommendations for how human and animal studies could be improved. A better understanding of innate immune function could inform future tractable immune-targeted interventions for childhood undernutrition to reduce mortality and improve long-term health, growth and development.
Collapse
Affiliation(s)
- Claire D Bourke
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, London, United Kingdom.,Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Kelsey D J Jones
- Kennedy Institute for Rheumatology, University of Oxford, Oxford, United Kingdom.,Department of Paediatric Gastroenterology & Nutrition, University of Oxford NHS Foundation Trust, Oxford, United Kingdom
| | - Andrew J Prendergast
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, London, United Kingdom.,Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| |
Collapse
|
29
|
Keski-Rahkonen P, Kolehmainen M, Lappi J, Micard V, Jokkala J, Rosa-Sibakov N, Pihlajamäki J, Kirjavainen PV, Mykkänen H, Poutanen K, Gunter MJ, Scalbert A, Hanhineva K. Decreased plasma serotonin and other metabolite changes in healthy adults after consumption of wholegrain rye: an untargeted metabolomics study. Am J Clin Nutr 2019; 109:1630-1639. [PMID: 31136658 DOI: 10.1093/ajcn/nqy394] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/31/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Wholegrain consumption has been associated with beneficial health effects including reduction of diabetes and cancer risk; however, the underlying mechanisms are not fully understood. OBJECTIVE The aim of this study was to characterize the effects of wholegrain rye intake on circulating metabolites in a human intervention study using untargeted metabolomics. METHODS The intervention consisted of 2 successive 4-wk periods in a randomized crossover design, where 15 adults consumed wholegrain rye bread (WGR) or white wheat bread enriched with fermented rye bran (WW+RB), following a 4-wk rye-free period with white wheat bread (WW). Fasting plasma samples were collected at the end of each period and analyzed using liquid chromatography-mass spectrometry. Metabolic profiles were compared to identify compounds discriminating WGR from the WW+RB and WW periods. Because peripheral serotonin is produced mainly in the gut, a hypothesis of its altered biosynthesis as a response to increased cereal fiber intake was tested by measuring intestinal serotonin of mice fed for 9 wk on a high-fat diet supplemented with different sources of fiber (rye bran flour, ground wheat aleurone, or powdered cellulose). RESULTS Five endogenous metabolites and 15 rye phytochemicals associated with WGR intake were identified. Plasma concentrations of serotonin, taurine, and glycerophosphocholine were significantly lower after the WGR than WW period (Q < 0.05). Concentrations of 2 phosphatidylethanolamine plasmalogens, PE(18:2/P-18:0) and PE(18:2/P-16:0), were lower after the WGR period than the WW+RB period (Q < 0.05). The concentration of serotonin was significantly lower in the colonic tissue of mice that consumed rye bran or wheat aleurone compared with cellulose (P < 0.001). CONCLUSIONS Wholegrain rye intake decreases plasma serotonin in healthy adults when compared with refined wheat. Intake of rye bran and wheat aleurone decreases colonic serotonin in mice. These results suggest that peripheral serotonin could be a potential link between wholegrain consumption and its associated health effects.Data used in the study were derived from a trial registered at www.clinicaltrials.gov as NCT03550365.
Collapse
Affiliation(s)
| | - Marjukka Kolehmainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Jenni Lappi
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Valerie Micard
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Montpellier SupAgro-INRA-University of Montpellier-CIRAD, Montpellier, France
| | - Jenna Jokkala
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Natalia Rosa-Sibakov
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Montpellier SupAgro-INRA-University of Montpellier-CIRAD, Montpellier, France
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Clinical Nutrition and Obesity Center, Kuopio University Hospital, Kuopio, Finland
| | - Pirkka V Kirjavainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Environmental Health Unit, The National Institute for Health and Welfare, Kuopio, Finland
| | - Hannu Mykkänen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Kaisa Poutanen
- VTT Technical Research Centre of Finland, Espoo, Finland
| | - Marc J Gunter
- International Agency for Research on Cancer, Lyon, France
| | | | - Kati Hanhineva
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
30
|
Moreau GB, Ramakrishnan G, Cook HL, Fox TE, Nayak U, Ma JZ, Colgate ER, Kirkpatrick BD, Haque R, Petri WA. Childhood growth and neurocognition are associated with distinct sets of metabolites. EBioMedicine 2019; 44:597-606. [PMID: 31133540 PMCID: PMC6604877 DOI: 10.1016/j.ebiom.2019.05.043] [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: 04/08/2019] [Revised: 05/10/2019] [Accepted: 05/18/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Undernutrition is a serious global problem that contributes to increased child morbidity and mortality, impaired neurocognitive development, and decreased educational and economic attainment. Current interventions are only marginally effective, and identification of associated metabolic pathways can offer new strategies for intervention. METHODS Plasma samples were collected at 9 and 36 months from a subset of the PROVIDE child cohort (n = 130). Targeted metabolomics was performed on bile acids, acylcarnitines, amino acids, phosphatidylcholines, and sphingomyelins. Metabolic associations with linear growth and neurocognitive outcomes at four years were evaluated using correlation and penalized-linear regression analysis as well as conditional random forest modeling. FINDINGS Different metabolites were associated with growth and neurocognitive outcomes. Improved growth outcomes were associated with higher concentrations of hydroxy-sphingomyelin and essential amino acids and lower levels of acylcarnitines and bile acid conjugation. Neurocognitive scores were largely associated with phosphatidylcholine species and early metabolic indicators of inflammation. All metabolites identified explain ~45% of growth and neurocognitive variation. INTERPRETATION Growth outcomes were predominantly associated with metabolites measured early in life (9 months), many of which were biomarkers of insufficient diet, environmental enteric dysfunction, and microbiome disruption. Hydroxy-sphingomyelin was a significant predictor of improved growth. Neurocognitive outcome was predominantly associated with 36 month phosphatidylcholines and inflammatory metabolites, which may serve as important biomarkers of optimal neurodevelopment. The distinct sets of metabolites associated with growth and neurocognition suggest that intervention may require targeted approaches towards distinct metabolic pathways. FUND: Bill & Melinda Gates Foundation (OP1173478); National Institutes of Health (AI043596, CA044579).
Collapse
Affiliation(s)
- G Brett Moreau
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Girija Ramakrishnan
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Heather L Cook
- Department of Statistics, University of Virginia, Charlottesville, VA, USA
| | - Todd E Fox
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Uma Nayak
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jennie Z Ma
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - E Ross Colgate
- Vaccine Testing Center, Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Beth D Kirkpatrick
- Vaccine Testing Center, Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Rashidul Haque
- International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - William A Petri
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
31
|
Semba RD, Zhang P, Adelnia F, Sun K, Gonzalez‐Freire M, Salem N, Brennan N, Spencer RG, Fishbein K, Khadeer M, Shardell M, Moaddel R, Ferrucci L. Low plasma lysophosphatidylcholines are associated with impaired mitochondrial oxidative capacity in adults in the Baltimore Longitudinal Study of Aging. Aging Cell 2019; 18:e12915. [PMID: 30719830 PMCID: PMC6413748 DOI: 10.1111/acel.12915] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 12/04/2018] [Accepted: 12/30/2018] [Indexed: 01/21/2023] Open
Abstract
The decrease in skeletal muscle mitochondrial oxidative capacity with age adversely affects muscle strength and physical performance. Factors that are associated with this decrease have not been well characterized. Low plasma lysophosphatidylcholines (LPC), a major class of systemic bioactive lipids, are predictive of aging phenotypes such as cognitive impairment and decline of gait speed in older adults. Therefore, we tested the hypothesis that low plasma LPC are associated with impaired skeletal muscle mitochondrial oxidative capacity. Skeletal muscle mitochondrial oxidative capacity was measured using in vivo phosphorus magnetic resonance spectroscopy (31P‐MRS) in 385 participants (256 women, 129 men), aged 24–97 years (mean 72.5) in the Baltimore Longitudinal Study of Aging. Postexercise recovery rate of phosphocreatine (PCr), kPCr, was used as a biomarker of mitochondrial oxidative capacity. Plasma LPC were measured using liquid chromatography–tandem mass spectrometry. Adults in the highest quartile of kPCr had higher plasma LPC 16:0 (p = 0.04), 16:1 (p = 0.004), 17:0 (p = 0.01), 18:1 (p = 0.0002), 18:2 (p = 0.002), and 20:3 (p = 0.0007), but not 18:0 (p = 0.07), 20:4 (p = 0.09) compared with those in the lower three quartiles in multivariable linear regression models adjusting for age, sex, and height. Multiple machine‐learning algorithms showed an area under the receiver operating characteristic curve of 0.638 (95% confidence interval, 0.554, 0.723) comparing six LPC in adults in the lower three quartiles of kPCr with the highest quartile. Low plasma LPC are associated with impaired mitochondrial oxidative capacity in adults.
Collapse
Affiliation(s)
- Richard D. Semba
- Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore Maryland
| | - Pingbo Zhang
- Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore Maryland
| | - Fatemeh Adelnia
- National Institute on Aging, National Institutes of Health Baltimore Maryland
| | - Kai Sun
- Wilmer Eye Institute Johns Hopkins University School of Medicine Baltimore Maryland
| | | | | | - Nicholas Brennan
- National Institute on Aging, National Institutes of Health Baltimore Maryland
| | - Richard G. Spencer
- National Institute on Aging, National Institutes of Health Baltimore Maryland
| | - Kenneth Fishbein
- National Institute on Aging, National Institutes of Health Baltimore Maryland
| | - Mohammed Khadeer
- National Institute on Aging, National Institutes of Health Baltimore Maryland
| | - Michelle Shardell
- National Institute on Aging, National Institutes of Health Baltimore Maryland
| | - Ruin Moaddel
- National Institute on Aging, National Institutes of Health Baltimore Maryland
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health Baltimore Maryland
| |
Collapse
|
32
|
Supplementation With Lactoferrin and Lysozyme Ameliorates Environmental Enteric Dysfunction: A Double-Blind, Randomized, Placebo-Controlled Trial. Am J Gastroenterol 2019; 114:671-678. [PMID: 30829679 DOI: 10.14309/ajg.0000000000000170] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Environmental enteric dysfunction (EED) predisposes children throughout the developing world to high rates of systemic exposure to enteric pathogens and stunting. Effective interventions that treat or prevent EED may help children achieve their full physical and cognitive potential. The objective of this study is to test whether 2 components of breast milk would improve a biomarker of EED and linear growth during the second year of life. METHODS A prospective, randomized, double-blind, placebo-controlled clinical trial among children aged 12-23 months was conducted in rural Malawi. The experimental group received a daily supplement of 1.5 g of lactoferrin and 0.2 g of lysozyme for 16 weeks. The primary outcome was an improvement in EED, as measured by the change in the percentage of ingested lactulose excreted into the urine (Δ%L). RESULTS Among 214 children who completed the study, there was a significant difference in Δ%L between the control and experimental groups over 8 weeks (an increase of 0.23% vs 0.14%, respectively; P = 0.04). However, this relative improvement was not as strongly sustained over the full 16 weeks of the study (an increase of 0.16% vs 0.11%, respectively; P = 0.17). No difference in linear growth over this short period was observed. The experimental intervention group had significantly lower rates of hospitalization and the development of acute malnutrition during the course of the study (2.5% vs 10.3%, relative risk 0.25; P < 0.02). DISCUSSION Supplementation with lactoferrin and lysozyme in a population of agrarian children during the second year of life has a beneficial effect on gut health. This intervention also protected against hospitalization and the development of acute malnutrition, a finding with a significant clinical and public health importance. This finding should be pursued in larger studies with longer follow-up and optimized dosing.
Collapse
|
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
|
Kashyap S, Shivakumar N, Varkey A, Duraisamy R, Thomas T, Preston T, Devi S, Kurpad AV. Ileal digestibility of intrinsically labeled hen's egg and meat protein determined with the dual stable isotope tracer method in Indian adults. Am J Clin Nutr 2018; 108:980-987. [PMID: 30272112 PMCID: PMC6250983 DOI: 10.1093/ajcn/nqy178] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/04/2018] [Indexed: 01/18/2023] Open
Abstract
Background Protein quality assessment through the Digestible Indispensable Amino Acid Score requires accurate measurements of true ileal protein and amino acid digestibility, for which a dual isotope technique was recently developed. However, the ileal digestibility of indispensable amino acids (IAA) in humans from high-quality proteins is not well known. Objective The aim of this study was to intrinsically label hen's egg and meat protein by the use of uniformly 2H-labeled amino acids, and to measure their true ileal indispensable amino acid (IAA) digestibility via the dual isotope method in humans. Design 2H-labeled lyophilized boiled egg white protein, whole boiled egg, and cooked meat were obtained from layer hens (BV-300) administered a uniformly 2H-labeled amino acid mix orally for 35 d with their daily feed. The ileal IAA digestibility of these proteins was determined with reference to digestibility of previously characterized [U-13C]spirulina in a dual tracer method in healthy Indian subjects whose intestinal health was measured by the plasma kynurenine-to-tryptophan (KT) ratio. Results All subjects had normal KT ratios. The mean ± SD true ileal IAA digestibility of 2H-labeled egg white protein, whole boiled egg, and cooked meat was 86.3% ± 4.6%, 89.4% ± 4.5%, and 92.0% ± 2.8%, respectively. Leucine digestibility correlated with the KT ratio (r = -0.772; P = 0.009). Conclusions Uniformly 2H-labeled hen's egg and meat protein can be used to measure ileal IAA digestibility by the dual isotope tracer approach in humans. The mean IAA digestibility values for these high-quality proteins in the healthy Indians studied were similar to values obtained in earlier human and animal experiments. Leucine digestibility in these meal matrices correlated with the KT ratio, but this aspect needs further evaluation. This trial was registered at the Clinical Trials Registry of India (http://ctri.nic.in) as CTRI/2018/03/012265.
Collapse
Affiliation(s)
- Sindhu Kashyap
- Division of Nutrition, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| | - Nirupama Shivakumar
- Departments of Physiology, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| | - Aneesia Varkey
- Division of Nutrition, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| | - Rajendran Duraisamy
- Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Adugodi, Bangalore, India
| | - Tinku Thomas
- Departments of Biostatistics, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| | - Thomas Preston
- Scottish Universities Environmental Research Centre, East Kilbride, Scotland, United Kingdom
| | - Sarita Devi
- Division of Nutrition, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| | - Anura V Kurpad
- Division of Nutrition, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India,Departments of Physiology, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India,Address correspondence to AVK (e-mail: )
| |
Collapse
|
35
|
Agapova SE, Stephenson KB, Divala O, Kaimila Y, Maleta KM, Thakwalakwa C, Ordiz MI, Trehan I, Manary MJ. Additional Common Bean in the Diet of Malawian Children Does Not Affect Linear Growth, but Reduces Intestinal Permeability. J Nutr 2018; 148:267-274. [PMID: 29490090 DOI: 10.1093/jn/nxx013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/18/2017] [Indexed: 01/26/2023] Open
Abstract
Background Chronic malnutrition, as manifested by linear growth faltering, is pervasive among rural African children. Improvements in complementary feeding may decrease the burden of environmental enteric dysfunction (EED) and thus improve growth in children during the critical first 1000 d of development. Objective We tested the hypothesis that systematically including common bean or cowpea into complementary feeding would reduce EED and growth faltering among children in rural Malawi. Methods This was a double-blind clinical trial in which children 12-23 mo of age were randomly assigned to receive complementary feeding with 1 of 3 foods: roasted cowpea or common bean flour, or an isoenergetic amount of corn-soy blend as a control food for 48 wk. Children aged 12-23 mo received 155 kcal/d and thereafter until 35 mo received 200 kcal/d. The primary outcomes were change in length-for-age z score (LAZ) and improvements in a biomarker of EED, the percentage of lactulose (%L) excreted as part of the lactulose:mannitol dual-sugar absorption test. Anthropometric measurements and urinary %L excretion were compared between the 2 intervention groups and the control group separately with the use of linear mixed model analyses for repeated measures. Results A total of 331 children completed the clinical trial. Compliance with the study interventions was excellent, with >90% of the intervention flour consumed as intended. No significant effects on LAZ, change in LAZ, or weight-for-length z score were observed due to either intervention legume, compared to the control. %L was reduced with common bean consumption (effect estimate was -0.07 percentage points of lactulose, P = 0.0007). The lactulose:mannitol test was not affected by the legume intervention. Conclusion The addition of common bean to complementary feeding of rural Malawian children during the second year of life led to an improvement in a biomarker of gut health, although this did not directly translate into improved linear growth. This trial was registered at clinicaltrials.gov as NCT02472301.
Collapse
Affiliation(s)
| | | | - Oscar Divala
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Yankho Kaimila
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Kenneth M Maleta
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Chrissie Thakwalakwa
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - M Isabel Ordiz
- Department of Pediatrics, Washington University, St Louis, MO
| | - Indi Trehan
- Department of Pediatrics, Washington University, St Louis, MO.,Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Mark J Manary
- Department of Pediatrics, Washington University, St Louis, MO.,School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi.,Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
36
|
Harper KM, Mutasa M, Prendergast AJ, Humphrey J, Manges AR. Environmental enteric dysfunction pathways and child stunting: A systematic review. PLoS Negl Trop Dis 2018; 12:e0006205. [PMID: 29351288 PMCID: PMC5792022 DOI: 10.1371/journal.pntd.0006205] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/31/2018] [Accepted: 01/03/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is commonly defined as an acquired subclinical disorder of the small intestine, characterized by villous atrophy and crypt hyperplasia. EED has been proposed to underlie stunted growth among children in developing countries. A collection of biomarkers, organized into distinct domains, has been used to measure different aspects of EED. Here, we examine whether these hypothesized relationships, among EED domains and between each domain and stunting, are supported by data from recent studies. METHODOLOGY A systematic literature search was conducted using PubMed, MEDLINE, EMBASE, Web of Science, and CINAHL between January 1, 2010 and April 20, 2017. Information on study objective, design, population, location, biomarkers, and results were recorded, as well as qualitative and quantitative definitions of EED. Biomarkers were organized into five EED domains, and the number of studies that support or do not support relationships among domains and between each domain with stunting were summarized. RESULTS There was little evidence to support the pathway from intestinal permeability to microbial translocation and from microbial translocation to stunting, but stronger support existed for the link between intestinal inflammation and systemic inflammation and for intestinal inflammation and stunting. There was conflicting evidence for the pathways from intestinal damage to intestinal permeability and intestinal damage to stunting. CONCLUSIONS These results suggest that certain EED biomarkers may require reconsideration, particularly those most difficult to measure, such as microbial translocation and intestinal permeability. We discuss several issues with currently used biomarkers and recommend further analysis of pathogen-induced changes to the intestinal microbiota as a pathway leading to stunting.
Collapse
Affiliation(s)
- Kaitlyn M. Harper
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Maxine Mutasa
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Andrew J. Prendergast
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jean Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Amee R. Manges
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
- British Columbia Centre for Disease Control, Vancouver, Canada
| |
Collapse
|
37
|
Cheng WD, Wold KJ, Benzoni NS, Thakwalakwa C, Maleta KM, Manary MJ, Trehan I. Lactoferrin and lysozyme to reduce environmental enteric dysfunction and stunting in Malawian children: study protocol for a randomized controlled trial. Trials 2017; 18:523. [PMID: 29110675 PMCID: PMC5674751 DOI: 10.1186/s13063-017-2278-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/25/2017] [Indexed: 12/11/2022] Open
Abstract
Background Chronic childhood malnutrition, as manifested by stunted linear growth, remains a persistent barrier to optimal child growth and societal development. Environmental enteric dysfunction (EED) is a significant underlying factor in the causal pathway to stunting, delayed cognitive development, and ultimately morbidity and mortality. Effective therapies against EED and stunting are lacking and further clinical trials are warranted to effectively identify and operationalize interventions. Methods/design A prospective randomized placebo-controlled parallel-group randomized controlled trial will be conducted to determine if a daily supplement of lactoferrin and lysozyme, two important proteins found in breast milk, can decrease the burden of EED and stunting in rural Malawian children aged 12–23 months old. The intervention and control groups will have a sample size of 86 subjects each. All field and laboratory researchers will be blinded to the assigned intervention group, as will the subjects and their caregivers. The percentage of ingested lactulose excreted in the urine (Δ%L) after 4 h will be used as the biomarker for EED and linear growth as the measure of chronic malnutrition (stunting). The primary outcomes of interest will be change in Δ%L from baseline to 8 weeks and to 16 weeks. Intention-to-treat analyses will be used. Discussion A rigorous clinical trial design will be used to assess the biologically plausible use of lactoferrin and lysozyme as dietary supplements for children at high risk for EED. If proven effective, these safe proteins may serve to markedly reduce the burden of childhood malnutrition and improve survival. Trial Registration Clinicaltrials.gov, NCT02925026. Registered on 4 October 2016. Electronic supplementary material The online version of this article (doi:10.1186/s13063-017-2278-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- William D Cheng
- Department of Pediatrics, Washington University in St. Louis, One Children's Place, Campus Box 8116, Saint Louis, MO, 63110, USA
| | - Karl J Wold
- Department of Pediatrics, Washington University in St. Louis, One Children's Place, Campus Box 8116, Saint Louis, MO, 63110, USA
| | - Nicole S Benzoni
- Department of Pediatrics, Washington University in St. Louis, One Children's Place, Campus Box 8116, Saint Louis, MO, 63110, USA
| | - Chrissie Thakwalakwa
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - Kenneth M Maleta
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - Mark J Manary
- Department of Pediatrics, Washington University in St. Louis, One Children's Place, Campus Box 8116, Saint Louis, MO, 63110, USA.,School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi.,Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Indi Trehan
- Department of Pediatrics, Washington University in St. Louis, One Children's Place, Campus Box 8116, Saint Louis, MO, 63110, USA. .,Department of Paediatrics and Child Health, University of Malawi, Blantyre, Malawi. .,Lao Friends Hospital for Children, Luang Prabang, Lao PDR.
| |
Collapse
|
38
|
Lustgarten MS, Fielding RA. Metabolites related to renal function, immune activation, and carbamylation are associated with muscle composition in older adults. Exp Gerontol 2017; 100:1-10. [PMID: 29030163 DOI: 10.1016/j.exger.2017.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/28/2017] [Accepted: 10/01/2017] [Indexed: 12/25/2022]
Abstract
Reduced skeletal muscle density in older adults is associated with insulin resistance, decreased physical function, and an increased all-cause mortality risk. To elucidate mechanisms that may underlie the maintenance of skeletal muscle density, we conducted a secondary analysis of previously published muscle composition and serum metabolomic data in 73 older adults (average age, 78y). Multivariable-adjusted linear regression was used to examine associations between 321 metabolites with muscle composition, defined as the ratio between normal density (NDM) with low density (LDM) thigh muscle cross sectional area (NDM/LDM). Sixty metabolites were significantly (p≤0.05 and q<0.30) associated with NDM/LDM. Decreased renal function and the immune response have been previously linked with reduced muscle density, but the mechanisms underlying these connections are less clear. Metabolites that were significantly associated with muscle composition were then tested for their association with circulating markers of renal function (blood urea nitrogen, creatinine, uric acid), and with the immune response (neutrophils/lymphocytes) and activation (kynurenine/tryptophan). 43 significant NDM/LDM metabolites (including urea) were co-associated with at least 1 marker of renal function; 23 of these metabolites have been previously identified as uremic solutes. The neutrophil/lymphocyte ratio was significantly associated with NDM/LDM (β±SE: -0.3±0.1, p=0.01, q=0.04). 35 significant NDM/LDM metabolites were co-associated with immune activation. Carbamylation (defined as homocitrulline/lysine) was identified as a pathway that may link renal function and immune activation with muscle composition, as 29 significant NDM/LDM metabolites were co-associated with homocitrulline/lysine, with at least 2 markers of renal function, and with kynurenine/tryptophan. When considering that elevated urea and uremic metabolites have been linked with an increased systemic microbial burden, that antimicrobial defense can be reduced in the presence of carbamylation, and that adipocytes can promote host defense, we propose the novel hypothesis that the age-related increase in adipogenesis within muscle may be a compensatory antimicrobial response to protect against an elevated microbial burden.
Collapse
Affiliation(s)
- Michael S Lustgarten
- Nutrition, Exercise Physiology, and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center, Tufts University, Boston, MA, USA.
| | - Roger A Fielding
- Nutrition, Exercise Physiology, and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center, Tufts University, Boston, MA, USA
| |
Collapse
|
39
|
Semba RD, Trehan I, Li X, Moaddel R, Ordiz MI, Maleta KM, Kraemer K, Shardell M, Ferrucci L, Manary M. Environmental Enteric Dysfunction is Associated with Carnitine Deficiency and Altered Fatty Acid Oxidation. EBioMedicine 2017; 17:57-66. [PMID: 28122695 PMCID: PMC5360565 DOI: 10.1016/j.ebiom.2017.01.026] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/03/2017] [Accepted: 01/17/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED), a condition characterized by small intestine inflammation and abnormal gut permeability, is widespread in children in developing countries and a major cause of growth failure. The pathophysiology of EED remains poorly understood. METHODS We measured serum metabolites using liquid chromatography-tandem mass spectrometry in 400 children, aged 12-59months, from rural Malawi. Gut permeability was assessed by the dual-sugar absorption test. FINDINGS 80.7% of children had EED. Of 677 serum metabolites measured, 21 were negatively associated and 56 were positively associated with gut permeability, using a false discovery rate approach (q<0.05, p<0.0095). Increased gut permeability was associated with elevated acylcarnitines, deoxycarnitine, fatty acid β-oxidation intermediates, fatty acid ω-oxidation products, odd-chain fatty acids, trimethylamine-N-oxide, cystathionine, and homocitrulline, and with lower citrulline, ornithine, polyphenol metabolites, hippurate, tryptophan, and indolelactate. INTERPRETATION EED is a syndrome characterized by secondary carnitine deficiency, abnormal fatty acid oxidation, alterations in polyphenol and amino acid metabolites, and metabolic dysregulation of sulfur amino acids, tryptophan, and the urea cycle. Future studies are needed to corroborate the presence of secondary carnitine deficiency among children with EED and to understand how these metabolic derangements may negatively affect the growth and development of young children.
Collapse
Affiliation(s)
- Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Indi Trehan
- Department of Pediatrics, Washington University at St. Louis, St. Louis, MO, USA
| | - Ximin Li
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ruin Moaddel
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - M Isabel Ordiz
- Department of Pediatrics, Washington University at St. Louis, St. Louis, MO, USA
| | | | - Klaus Kraemer
- Sight and Life, Basel, Switzerland; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Michelle Shardell
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Mark Manary
- Department of Pediatrics, Washington University at St. Louis, St. Louis, MO, USA
| |
Collapse
|
40
|
Wang AZ, Shulman RJ, Crocker AH, Thakwalakwa C, Maleta KM, Devaraj S, Manary MJ, Trehan I. A Combined Intervention of Zinc, Multiple Micronutrients, and Albendazole Does Not Ameliorate Environmental Enteric Dysfunction or Stunting in Rural Malawian Children in a Double-Blind Randomized Controlled Trial. J Nutr 2017; 147:97-103. [PMID: 27807040 DOI: 10.3945/jn.116.237735] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/09/2016] [Accepted: 10/11/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) and linear growth stunting affect many rural agrarian children in the developing world and contribute to the persistently high rates of stunting that are observed worldwide. Effective interventions to consistently ameliorate EED are lacking. OBJECTIVE We tested whether a bundle of safe and affordable interventions would decrease EED and stunting over 12-24 wk in a cohort of rural Malawian children 12-35 mo old. METHODS This was a randomized, double-blind, placebo-controlled clinical trial in which the intervention group received a single dose of albendazole and 14 d of zinc at enrollment and after 20 wk. The intervention group also received a daily multiple micronutrient powder throughout the 24 wk of study. The primary outcomes were improvements in EED, as measured by the urinary lactulose-to-mannitol ratio (L:M ratio) from dual-sugar absorption testing, and linear growth. Urinary L:M ratios and anthropometric measurements were evaluated after 12 and 24 wk of intervention and compared with a placebo group that did not receive any of these interventions. RESULTS A total of 254 children were enrolled at a mean age of 24 mo; 55% were female. Their mean weight-for-age z score was -1.5, and their mean length-for-age z score was -0.9. After 12 and 24 wk of study, increases in the L:M ratio did not differ between the intervention group (0.071 and 0.088 units, respectively) and the placebo group (0.073 and 0.080 units, respectively) (P = 0.87 and 0.19, respectively). Relative changes in length and weight also did not differ significantly between groups at any time point. CONCLUSION The combined usage of albendazole, zinc, and a daily multiple micronutrient powder did not decrease EED or stunting in this population of agrarian children 12-35 mo old in rural Malawi. Alternative interventions to improve these diseases should be investigated. This trial was registered at clinicaltrials.gov as NCT02253095.
Collapse
Affiliation(s)
- Alfred Z Wang
- University of Texas Southwestern Medical School, Dallas, TX.,Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | - Robert J Shulman
- USDA/Agricultural Research Service Children's Nutrition Research Center, Houston, TX; Departments of.,Pediatrics and
| | - Audrey H Crocker
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | | | | | | | - Mark J Manary
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO; .,USDA/Agricultural Research Service Children's Nutrition Research Center, Houston, TX; Departments of.,School of Public Health and Family Medicine and
| | - Indi Trehan
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO; .,Department of Paediatrics and Child Health, University of Malawi, Blantyre, Malawi
| |
Collapse
|
41
|
Di Giovanni V, Bourdon C, Wang DX, Seshadri S, Senga E, Versloot CJ, Voskuijl W, Semba RD, Trehan I, Moaddel R, Ordiz MI, Zhang L, Parkinson J, Manary MJ, Bandsma RH. Metabolomic Changes in Serum of Children with Different Clinical Diagnoses of Malnutrition. J Nutr 2016; 146:2436-2444. [PMID: 27807038 PMCID: PMC5118769 DOI: 10.3945/jn.116.239145] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/28/2016] [Accepted: 10/04/2016] [Indexed: 12/04/2022] Open
Abstract
Background: Mortality in children with severe acute malnutrition (SAM) remains high despite standardized rehabilitation protocols. Two forms of SAM are classically distinguished: kwashiorkor and marasmus. Children with kwashiorkor have nutritional edema and metabolic disturbances, including hypoalbuminemia and hepatic steatosis, whereas marasmus is characterized by severe wasting. The metabolic changes underlying these phenotypes have been poorly characterized, and whether homeostasis is achieved during hospital stay is unclear. Objectives: We aimed to characterize metabolic differences between children with marasmus and kwashiorkor at hospital admission and after clinical stabilization and to compare them with stunted and nonstunted community controls. Methods: We studied children aged 9–59 mo from Malawi who were hospitalized with SAM (n = 40; 21 with kwashiorkor and 19 with marasmus) or living in the community (n = 157; 78 stunted and 79 nonstunted). Serum from patients with SAM was obtained at hospital admission and 3 d after nutritional stabilization and from community controls. With the use of targeted metabolomics, 141 metabolites, including amino acids, biogenic amines, acylcarnitines, sphingomyelins, and phosphatidylcholines, were measured. Results: At admission, most metabolites (128 of 141; 91%) were lower in children with kwashiorkor than in those with marasmus, with significant differences in several amino acids and biogenic amines, including those of the kynurenine-tryptophan pathway. Several phosphatidylcholines and some acylcarnitines also differed. Patients with SAM had profiles that were profoundly different from those of stunted and nonstunted controls, even after clinical stabilization. Amino acids and biogenic amines generally improved with nutritional rehabilitation, but most sphingomyelins and phosphatidylcholines did not. Conclusions: Children with kwashiorkor were metabolically distinct from those with marasmus, and were more prone to severe metabolic disruptions. Children with SAM showed metabolic profiles that were profoundly different from stunted and nonstunted controls, even after clinical stabilization. Therefore, metabolic recovery in children with SAM likely extends beyond discharge, which may explain the poor long-term outcomes in these children. This trial was registered at isrctn.org as ISRCTN13916953.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wieger Voskuijl
- Department of Pediatrics and Child Health, and.,Global Child Health Group, Emma Children's Hospital, Academic Medical Centre, University of Amsterdam, Netherlands
| | - Richard D Semba
- Wilmer Eye Institute, John Hopkins University School of Medicine, Baltimore, MD
| | - Indi Trehan
- Department of Pediatrics and Child Health, and.,National Institute of Aging, NIH, Baltimore, MD
| | | | - M Isabel Ordiz
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO; and
| | - Ling Zhang
- Department of Physiology and Experimental Medicine
| | - John Parkinson
- Program in Molecular Structure and Function.,Department of Biochemistry and Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Mark J Manary
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi.,Department of Pediatrics, Washington University in St. Louis, St. Louis, MO; and
| | - Robert Hj Bandsma
- Department of Physiology and Experimental Medicine, .,Division of Gastroenterology, Hepatology and Nutrition, and.,Centre for Global Child Health, Hospital for Sick Children, Toronto, Canada.,Department of Biomedical Sciences
| |
Collapse
|
42
|
Semba RD, Trehan I, Gonzalez-Freire M, Kraemer K, Moaddel R, Ordiz MI, Ferrucci L, Manary MJ. Perspective: The Potential Role of Essential Amino Acids and the Mechanistic Target of Rapamycin Complex 1 (mTORC1) Pathway in the Pathogenesis of Child Stunting. Adv Nutr 2016; 7:853-65. [PMID: 27633102 PMCID: PMC5015042 DOI: 10.3945/an.116.013276] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Stunting is the best summary measure of chronic malnutrition in children. Approximately one-quarter of children under age 5 worldwide are stunted. Lipid-based or micronutrient supplementation has little to no impact in reducing stunting, which suggests that other critical dietary nutrients are missing. A dietary pattern of poor-quality protein is associated with stunting. Stunted children have significantly lower circulating essential amino acids than do nonstunted children. Inadequate dietary intakes of essential amino acids could adversely affect growth, because amino acids are required for synthesis of proteins. The master growth regulation pathway, the mechanistic target of rapamycin complex 1 (mTORC1) pathway, is exquisitely sensitive to amino acid availability. mTORC1 integrates cues such as nutrients, growth factors, oxygen, and energy to regulate growth of bone, skeletal muscle, nervous system, gastrointestinal tract, hematopoietic cells, immune effector cells, organ size, and whole-body energy balance. mTORC1 represses protein and lipid synthesis and cell and organismal growth when amino acids are deficient. Over the past 4 decades, the main paradigm for child nutrition in developing countries has been micronutrient malnutrition, with relatively less attention paid to protein. In this Perspective, we present the view that essential amino acids and the mTORC1 pathway play a key role in child growth. The current assumption that total dietary protein intake is adequate for growth among most children in developing countries needs re-evaluation.
Collapse
Affiliation(s)
- Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD;
| | - Indi Trehan
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | | | - Klaus Kraemer
- Sight and Life, Basel, Switzerland; and Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | | | - M Isabel Ordiz
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | | | - Mark J Manary
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|