1
|
Brito AM, Souto DO, Silva LC, Leite HR, Morais RLS. Social vulnerability among Brazilian children in early childhood: a scoping review. J Pediatr (Rio J) 2025; 101:7-20. [PMID: 39159916 PMCID: PMC11763581 DOI: 10.1016/j.jped.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 06/08/2024] [Accepted: 06/11/2024] [Indexed: 08/21/2024] Open
Abstract
OBJECTIVE To identify, map, and describe studies involving Brazilian children in early childhood in situations of social vulnerability. SOURCE OF DATA A scoping review including full articles published in Portuguese and English up to March 2023, with no temporal restrictions. Searches were conducted in the MEDLINE/PubMed, Scielo, EMBASE, Cochrane, Scopus, CINAHL, Web of Science, PEDro, and LILACS databases. Journal metrics, sample characteristics, study area, characterization of the situation of social vulnerability, and study outcomes were extracted. SUMMARY OF THE FINDINGS Seventy-six articles involving a total of 107.740 children in early childhood were included in this study. These studies presented relevant findings, including the temporal publication trend, the variability of social vulnerability indicators, the scarcity of intervention studies, and the fact that 100% of eligible studies were from the health area. Negative outcomes were associated with the condition of social vulnerability in almost the entire sample, reinforcing the need for government policies capable of protecting early childhood from the effects of social vulnerability. CONCLUSION This scoping review mapped important findings involving Brazilian children in social vulnerability. It also identified literature gaps such as the need for intervention and multisectoral studies among health, education, and social protection.
Collapse
Affiliation(s)
- Alcina M Brito
- Universidade Federal dos Vales do Jequitinhonha e Mucuri, Programa de Pós-Graduação em Saúde, Sociedade e Meio Ambiente, Diamantina, MG, Brazil
| | - Deisiane O Souto
- Universidade Federal de Minas Gerais (UFMG), Escola de Educação Física, Fisioterapia e Terapia Ocupacional (EEFFTO), Departamento de Fisioterapia, Programa de Pós-Graduação em Ciências da Reabilitação (PPGCR), Belo Horizonte, MG, Brazil.
| | - Luana C Silva
- Universidade Federal de Minas Gerais (UFMG), Escola de Educação Física, Fisioterapia e Terapia Ocupacional (EEFFTO), Departamento de Fisioterapia, Programa de Pós-Graduação em Ciências da Reabilitação (PPGCR), Belo Horizonte, MG, Brazil
| | - Hércules R Leite
- Universidade Federal de Minas Gerais (UFMG), Escola de Educação Física, Fisioterapia e Terapia Ocupacional (EEFFTO), Departamento de Fisioterapia, Programa de Pós-Graduação em Ciências da Reabilitação (PPGCR), Belo Horizonte, MG, Brazil
| | - Rosane L S Morais
- Universidade Federal dos Vales do Jequitinhonha e Mucuri, Programa de Pós-Graduação em Saúde, Sociedade e Meio Ambiente, Diamantina, MG, Brazil
| |
Collapse
|
2
|
Valdes AM, Louca P, Visconti A, Asnicar F, Bermingham K, Nogal A, Wong K, Michelotti GA, Wolf J, Segata N, Spector TD, Berry SE, Falchi M, Menni C. Vitamin A carotenoids, but not retinoids, mediate the impact of a healthy diet on gut microbial diversity. BMC Med 2024; 22:321. [PMID: 39113058 PMCID: PMC11304618 DOI: 10.1186/s12916-024-03543-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/28/2024] [Indexed: 08/11/2024] Open
Abstract
BACKGROUND Vitamin A is essential for physiological processes like vision and immunity. Vitamin A's effect on gut microbiome composition, which affects absorption and metabolism of other vitamins, is still unknown. Here we examined the relationship between gut metagenome composition and six vitamin A-related metabolites (two retinoid: -retinol, 4 oxoretinoic acid (oxoRA) and four carotenoid metabolites, including beta-cryptoxanthin and three carotene diols). METHODS We included 1053 individuals from the TwinsUK cohort with vitamin A-related metabolites measured in serum and faeces, diet history, and gut microbiome composition assessed by shotgun metagenome sequencing. Results were replicated in 327 women from the ZOE PREDICT-1 study. RESULTS Five vitamin A-related serum metabolites were positively correlated with microbiome alpha diversity (r = 0.15 to r = 0.20, p < 4 × 10-6). Carotenoid compounds were positively correlated with the short-chain fatty-acid-producing bacteria Faecalibacterium prausnitzii and Coprococcus eutactus. Retinol was not associated with any microbial species. We found that gut microbiome composition could predict circulating levels of carotenoids and oxoretinoic acid with AUCs ranging from 0.66 to 0.74 using random forest models, but not retinol (AUC = 0.52). The healthy eating index (HEI) was strongly associated with gut microbiome diversity and with all carotenoid compounds, but not retinoids. We investigated the mediating role of carotenoid compounds on the effect of a healthy diet (HEI) on gut microbiome diversity, finding that carotenoids significantly mediated between 18 and 25% of the effect of HEI on gut microbiome alpha diversity. CONCLUSIONS Our results show strong links between circulating carotene compounds and gut microbiome composition and potential links to a healthy diet pattern.
Collapse
Affiliation(s)
- Ana M Valdes
- Nottingham NIHR Biomedical Research Centre at the School of Medicine, University of Nottingham, Nottingham, NG5 1PB, UK.
- Inflammation, Recovery and Injury Sciences, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK.
- Department of Twin Research & Genetic Epidemiology, King's College London, London, SE1 7EH, UK.
| | - Panayiotis Louca
- Department of Twin Research & Genetic Epidemiology, King's College London, London, SE1 7EH, UK
- Human Nutrition and Exercise Research Centre, University of Newcastle, Newcastle Upon Tyne, NE2 4HH, UK
| | - Alessia Visconti
- Department of Twin Research & Genetic Epidemiology, King's College London, London, SE1 7EH, UK
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, 10124, Turin, Italy
| | - Francesco Asnicar
- Department CIBIO, University of Trento, Via Sommarive 9, 38123, Povo, Trento, Italy
| | - Kate Bermingham
- Department of Nutritional Sciences, King's College London, 150 Stamford Street, London, SE1 9NH, UK
- Zoe Limited, 164 Westminster Bridge Rd, London, SE1 7RW, UK
| | - Ana Nogal
- Department of Twin Research & Genetic Epidemiology, King's College London, London, SE1 7EH, UK
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Kari Wong
- Metabolon Inc, Research Triangle Park, Morrisville, NC, 27560, USA
| | | | - Jonathan Wolf
- Zoe Limited, 164 Westminster Bridge Rd, London, SE1 7RW, UK
| | - Nicola Segata
- Department CIBIO, University of Trento, Via Sommarive 9, 38123, Povo, Trento, Italy
| | - Tim D Spector
- Department of Twin Research & Genetic Epidemiology, King's College London, London, SE1 7EH, UK
- Zoe Limited, 164 Westminster Bridge Rd, London, SE1 7RW, UK
| | - Sarah E Berry
- Department of Nutritional Sciences, King's College London, 150 Stamford Street, London, SE1 9NH, UK
- Zoe Limited, 164 Westminster Bridge Rd, London, SE1 7RW, UK
| | - Mario Falchi
- Department of Twin Research & Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | - Cristina Menni
- Department of Twin Research & Genetic Epidemiology, King's College London, London, SE1 7EH, UK.
| |
Collapse
|
3
|
Schneider E, O'Riordan KJ, Clarke G, Cryan JF. Feeding gut microbes to nourish the brain: unravelling the diet-microbiota-gut-brain axis. Nat Metab 2024; 6:1454-1478. [PMID: 39174768 DOI: 10.1038/s42255-024-01108-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 07/15/2024] [Indexed: 08/24/2024]
Abstract
The prevalence of brain disorders, including stress-related neuropsychiatric disorders and conditions with cognitive dysfunction, is rising. Poor dietary habits contribute substantially to this accelerating trend. Conversely, healthy dietary intake supports mood and cognitive performance. Recently, the communication between the microorganisms within the gastrointestinal tract and the brain along the gut-brain axis has gained prominence as a potential tractable target to modulate brain health. The composition and function of the gut microbiota is robustly influenced by dietary factors to alter gut-brain signalling. To reflect this interconnection between diet, gut microbiota and brain functioning, we propose that a diet-microbiota-gut-brain axis exists that underpins health and well-being. In this Review, we provide a comprehensive overview of the interplay between diet and gut microbiota composition and function and the implications for cognition and emotional functioning. Important diet-induced effects on the gut microbiota for the development, prevention and maintenance of neuropsychiatric disorders are described. The diet-microbiota-gut-brain axis represents an uncharted frontier for brain health diagnostics and therapeutics across the lifespan.
Collapse
Affiliation(s)
| | | | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
4
|
Bjelakovic G, Nikolova D, Bjelakovic M, Pavlov CS, Sethi NJ, Korang SK, Gluud C. Effects of primary or secondary prevention with vitamin A supplementation on clinically important outcomes: a systematic review of randomised clinical trials with meta-analysis and trial sequential analysis. BMJ Open 2024; 14:e078053. [PMID: 38816049 PMCID: PMC11141198 DOI: 10.1136/bmjopen-2023-078053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 05/20/2024] [Indexed: 06/01/2024] Open
Abstract
OBJECTIVES This systematic review with meta-analyses of randomised trials evaluated the preventive effects of vitamin A supplements versus placebo or no intervention on clinically important outcomes, in people of any age. METHODS We searched different electronic databases and other resources for randomised clinical trials that had compared vitamin A supplements versus placebo or no intervention (last search 16 April 2024). We used Cochrane methodology. We used the random-effects model to calculate risk ratios (RRs), with 95% CIs. We analysed individually and cluster randomised trials separately. Our primary outcomes were mortality, adverse events and quality of life. We assessed risks of bias in the trials and used Grading of Recommendations, Assessment, Development, and Evaluations (GRADE) to assess the certainty of the evidence. RESULTS We included 120 randomised trials (1 671 672 participants); 105 trials allocated individuals and 15 allocated clusters. 92 trials included children (78 individually; 14 cluster randomised) and 28 adults (27 individually; 1 cluster randomised). 14/105 individually randomised trials (13%) and none of the cluster randomised trials were at overall low risk of bias. Vitamin A did not reduce mortality in individually randomised trials (RR 0.99, 95% CI 0.93 to 1.05; I²=32%; p=0.19; 105 trials; moderate certainty), and this effect was not affected by the risk of bias. In individually randomised trials, vitamin A had no effect on mortality in children (RR 0.96, 95% CI 0.88 to 1.04; I²=24%; p=0.28; 78 trials, 178 094 participants) nor in adults (RR 1.04, 95% CI 0.97 to 1.13; I²=24%; p=0.27; 27 trials, 61 880 participants). Vitamin A reduced mortality in the cluster randomised trials (0.84, 95% CI 0.76 to 0.93; I²=66%; p=0.0008; 15 trials, 14 in children and 1 in adults; 364 343 participants; very low certainty). No trial reported serious adverse events or quality of life. Vitamin A slightly increased bulging fontanelle of neonates and infants. We are uncertain whether vitamin A influences blindness under the conditions examined. CONCLUSIONS Based on moderate certainty of evidence, vitamin A had no effect on mortality in the individually randomised trials. Very low certainty evidence obtained from cluster randomised trials suggested a beneficial effect of vitamin A on mortality. If preventive vitamin A programmes are to be continued, supporting evidence should come from randomised trials allocating individuals and assessing patient-meaningful outcomes. PROSPERO REGISTRATION NUMBER CRD42018104347.
Collapse
Affiliation(s)
- Goran Bjelakovic
- Department of Internal Medicine, Medical Faculty, University of Nis, Nis, Serbia
- Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Clinic of Gastroenterohepatology, University Clinical Centre, Nis, Serbia
| | - Dimitrinka Nikolova
- Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Milica Bjelakovic
- Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Clinic of Gastroenterohepatology, University Clinical Centre, Nis, Serbia
| | - Chavdar S Pavlov
- Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Gastroenterology, Botkin Hospital, Moscow, Russian Federation
- Department of Therapy, I.M. Sechenov, First Moscow State Medical University, Moscow, Russian Federation
| | - Naqash J Sethi
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Steven Kwasi Korang
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Christian Gluud
- Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Regional Health Research, The Faculty of Heath Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
5
|
Khandelwal P, Langenberg L, Luebbering N, Lake KE, Butcher A, Bota K, Ramos KN, Taggart C, Choe H, Vasu S, Teusink-Cross A, Koo J, Wallace G, Romick-Rosendale L, Watanabe-Chailland M, Haslam DB, Lane A, Davies SM. A randomized phase 2 trial of oral vitamin A for graft-versus-host disease in children and young adults. Blood 2024; 143:1181-1192. [PMID: 38227933 DOI: 10.1182/blood.2023022865] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/02/2024] [Accepted: 01/02/2024] [Indexed: 01/18/2024] Open
Abstract
ABSTRACT Vitamin A plays a key role in the maintenance of gastrointestinal homeostasis and promotes a tolerogenic phenotype in tissue resident macrophages. We conducted a prospective randomized double-blinded placebo-controlled clinical trial in which 80 recipients of hematopoietic stem cell transplantation (HSCT) were randomized 1:1 to receive pretransplant high-dose vitamin A or placebo. A single oral dose of vitamin A of 4000 IU/kg, maximum 250 000 IU was given before conditioning. The primary end point was incidence of acute graft-versus-host disease (GVHD) at day +100. In an intent-to-treat analysis, incidence of acute GVHD was 12.5% in the vitamin A arm and 20% in the placebo arm (P = .5). Incidence of acute gastrointestinal (GI) GVHD was 2.5% in the vitamin A arm (P = .09) and 12.5% in the placebo arm at day +180. Incidence of chronic GVHD was 5% in the vitamin A arm and 15% in the placebo arm (P = .02) at 1 year. In an "as treated" analysis, cumulative incidence of acute GI GVHD at day +180 was 0% and 12.5% in recipients of vitamin A and placebo, respectively (P = .02), and cumulative incidence of chronic GVHD was 2.7% and 15% in recipients of vitamin A and placebo, respectively (P = .01). The only possibly attributable toxicity was asymptomatic grade 3 hyperbilirubinemia in 1 recipient of vitamin A at day +30, which self-resolved. Absolute CCR9+ CD8+ effector memory T cells, reflecting gut T-cell trafficking, were lower in the vitamin A arm at day +30 after HSCT (P = .01). Levels of serum amyloid A-1, a vitamin A transport protein with proinflammatory effects, were lower in the vitamin A arm. The vitamin A arm had lower interleukin-6 (IL-6), IL-8, and suppressor of tumorigenicity 2 levels and likely a more favorable gut microbiome and short chain fatty acids. Pre-HSCT oral vitamin A is inexpensive, has low toxicity, and reduces GVHD. This trial was registered at www.ClinicalTrials.gov as NCT03202849.
Collapse
Affiliation(s)
- Pooja Khandelwal
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
| | - Lucille Langenberg
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
| | - Nathan Luebbering
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
| | - Kelly E Lake
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
| | - Abigail Butcher
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kylie Bota
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
| | - Kristie N Ramos
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
| | - Cynthia Taggart
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
| | - Hannah Choe
- Division of Hematology, The Ohio State Comprehensive Cancer Center, Columbus OH
| | - Sumithira Vasu
- Division of Hematology, The Ohio State Comprehensive Cancer Center, Columbus OH
| | - Ashley Teusink-Cross
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
- Division of Pharmacy, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Jane Koo
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
| | - Gregory Wallace
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
| | - Lindsey Romick-Rosendale
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Miki Watanabe-Chailland
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - David B Haslam
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
- Divison of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Adam Lane
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
| | - Stella M Davies
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH
| |
Collapse
|
6
|
Abstract
Retinoic acid (RA) is a metabolite of vitamin A and is essential for development and growth as well as cellular metabolism. Through genomic and nongenomic actions, RA regulates a variety of physiological functions. Dysregulation of RA signaling is associated with many diseases. Targeting RA signaling has been proven valuable to human health. All-trans retinoic acid (AtRA) and anthracycline-based chemotherapy are the standard treatment of acute promyelocytic leukemia (APL). Both human and animal studies have shown a significant relationship between RA signaling and the development and progression of nonalcoholic fatty liver disease (NAFLD). In this review article, we will first summarize vitamin A metabolism and then focus on the role of RA signaling in NAFLD. AtRA inhibits the development and progression of NAFLD via regulating lipid metabolism, inflammation, thermogenesis, etc.
Collapse
Affiliation(s)
- Fathima N Cassim Bawa
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA 44272
| | - Yanqiao Zhang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA 44272
| |
Collapse
|
7
|
Imdad A, Rogner J, Sherwani RN, Sidhu J, Regan A, Haykal MR, Tsistinas O, Smith A, Chan XHS, Mayo-Wilson E, Bhutta ZA. Zinc supplementation for preventing mortality, morbidity, and growth failure in children aged 6 months to 12 years. Cochrane Database Syst Rev 2023; 3:CD009384. [PMID: 36994923 PMCID: PMC10061962 DOI: 10.1002/14651858.cd009384.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
BACKGROUND Zinc deficiency is prevalent in low- and middle-income countries, and is considered a significant risk factor for morbidity, mortality, and linear growth failure. The effectiveness of preventive zinc supplementation in reducing prevalence of zinc deficiency needs to be assessed. OBJECTIVES To assess the effects of zinc supplementation for preventing mortality and morbidity, and for promoting growth, in children aged 6 months to 12 years. SEARCH METHODS A previous version of this review was published in 2014. In this update, we searched CENTRAL, MEDLINE, Embase, five other databases, and one trials register up to February 2022, together with reference checking and contact with study authors to identify additional studies. SELECTION CRITERIA Randomized controlled trials (RCTs) of preventive zinc supplementation in children aged 6 months to 12 years compared with no intervention, a placebo, or a waiting list control. We excluded hospitalized children and children with chronic diseases or conditions. We excluded food fortification or intake, sprinkles, and therapeutic interventions. DATA COLLECTION AND ANALYSIS Two review authors screened studies, extracted data, and assessed the risk of bias. We contacted study authors for missing information and used GRADE to assess the certainty of evidence. The primary outcomes of this review were all-cause mortality; and cause-specific mortality, due to all-cause diarrhea, lower respiratory tract infection (LRTI, including pneumonia), and malaria. We also collected information on a number of secondary outcomes, such as those related to diarrhea and LRTI morbidity, growth outcomes and serum levels of micronutrients, and adverse events. MAIN RESULTS We included 16 new studies in this review, resulting in a total of 96 RCTs with 219,584 eligible participants. The included studies were conducted in 34 countries; 87 of them in low- or middle-income countries. Most of the children included in this review were under five years of age. The intervention was delivered most commonly in the form of syrup as zinc sulfate, and the most common dose was between 10 mg and 15 mg daily. The median duration of follow-up was 26 weeks. We did not consider that the evidence for the key analyses of morbidity and mortality outcomes was affected by risk of bias. High-certainty evidence showed little to no difference in all-cause mortality with preventive zinc supplementation compared to no zinc (risk ratio (RR) 0.93, 95% confidence interval (CI) 0.84 to 1.03; 16 studies, 17 comparisons, 143,474 participants). Moderate-certainty evidence showed that preventive zinc supplementation compared to no zinc likely results in little to no difference in mortality due to all-cause diarrhea (RR 0.95, 95% CI 0.69 to 1.31; 4 studies, 132,321 participants); but probably reduces mortality due to LRTI (RR 0.86, 95% CI 0.64 to 1.15; 3 studies, 132,063 participants) and mortality due to malaria (RR 0.90, 95% CI 0.77 to 1.06; 2 studies, 42,818 participants); however, the confidence intervals around the summary estimates for these outcomes were wide, and we could not rule out a possibility of increased risk of mortality. Preventive zinc supplementation likely reduces the incidence of all-cause diarrhea (RR 0.91, 95% CI 0.90 to 0.93; 39 studies, 19,468 participants; moderate-certainty evidence) but results in little to no difference in morbidity due to LRTI (RR 1.01, 95% CI 0.95 to 1.08; 19 studies, 10,555 participants; high-certainty evidence) compared to no zinc. There was moderate-certainty evidence that preventive zinc supplementation likely leads to a slight increase in height (standardized mean difference (SMD) 0.12, 95% CI 0.09 to 0.14; 74 studies, 20,720 participants). Zinc supplementation was associated with an increase in the number of participants with at least one vomiting episode (RR 1.29, 95% CI 1.14 to 1.46; 5 studies, 35,192 participants; high-certainty evidence). We report a number of other outcomes, including the effect of zinc supplementation on weight and serum markers such as zinc, hemoglobin, iron, copper, etc. We also performed a number of subgroup analyses and there was a consistent finding for a number of outcomes that co-supplementation of zinc with iron decreased the beneficial effect of zinc. AUTHORS' CONCLUSIONS Even though we included 16 new studies in this update, the overall conclusions of the review remain unchanged. Zinc supplementation might help prevent episodes of diarrhea and improve growth slightly, particularly in children aged 6 months to 12 years of age. The benefits of preventive zinc supplementation may outweigh the harms in regions where the risk of zinc deficiency is relatively high.
Collapse
Affiliation(s)
- Aamer Imdad
- Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jaimie Rogner
- Departments of Medicine and Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Rida N Sherwani
- Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jasleen Sidhu
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Allison Regan
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Maya R Haykal
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Olivia Tsistinas
- Health Sciences Library, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Abigail Smith
- Health Sciences Library, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Xin Hui S Chan
- Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Evan Mayo-Wilson
- Department of Epidemiology, UNC Gillings School of Global Public HealthMcGavran-Greenberg Hall, Chapel Hill, NC, USA
| | - Zulfiqar A Bhutta
- Centre for Global Child Health, The Hospital for SickKids, Toronto, Canada
- Center of Excellence for Women and Child Health, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
8
|
Essential Oils and Terpenic Compounds as Potential Hits for Drugs against Amitochondriate Protists. Trop Med Infect Dis 2023; 8:tropicalmed8010037. [PMID: 36668944 PMCID: PMC9865018 DOI: 10.3390/tropicalmed8010037] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/26/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
The human anaerobic or microaerophilic protists Giardia duodenalis, Entamoeba histolytica, and Trichomonas vaginalis are classified as amitochondriate parasites, a group of unicellular organisms that lack canonical mitochondria organelles. These microorganisms suffered adaptations to survive in hostile microenvironments and together represent an increasing threat to public health in developing countries. Nevertheless, the current therapeutic drugs to manage the infections are scarce and often cause several side effects. Furthermore, refractory cases associated with the emergence of parasitic resistance are concerns that guide the search for new pharmacological targets and treatment alternatives. Herein, essential oils and terpenic compounds with activity against amitochondriate parasites with clinical relevance are summarized and insights into possible mechanisms of action are made. This review aims to contribute with future perspectives for research with these natural products as potential alternatives for the acquisition of new molecules for the treatment of amitochondriate protists.
Collapse
|
9
|
Immune Impairment Associated with Vitamin A Deficiency: Insights from Clinical Studies and Animal Model Research. Nutrients 2022; 14:nu14235038. [PMID: 36501067 PMCID: PMC9738822 DOI: 10.3390/nu14235038] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Vitamin A (VA) is critical for many biological processes, including embryonic development, hormone production and function, the maintenance and modulation of immunity, and the homeostasis of epithelium and mucosa. Specifically, VA affects cell integrity, cytokine production, innate immune cell activation, antigen presentation, and lymphocyte trafficking to mucosal surfaces. VA also has been reported to influence the gut microbiota composition and diversity. Consequently, VA deficiency (VAD) results in the imbalanced production of inflammatory and immunomodulatory cytokines, intestinal inflammation, weakened mucosal barrier functions, reduced reactive oxygen species (ROS) and disruption of the gut microbiome. Although VAD is primarily known to cause xerophthalmia, its role in the impairment of anti-infectious defense mechanisms is less defined. Infectious diseases lead to temporary anorexia and lower dietary intake; furthermore, they adversely affect VA status by interfering with VA absorption, utilization and excretion. Thus, there is a tri-directional relationship between VAD, immune response and infections, as VAD affects immune response and predisposes the host to infection, and infection decreases the intestinal absorption of the VA, thereby contributing to secondary VAD development. This has been demonstrated using nutritional and clinical studies, radiotracer studies and knockout animal models. An in-depth understanding of the relationship between VAD, immune response, gut microbiota and infections is critical for optimizing vaccine efficacy and the development of effective immunization programs for countries with high prevalence of VAD. Therefore, in this review, we have comprehensively summarized the existing knowledge regarding VAD impacts on immune responses to infections and post vaccination. We have detailed pathological conditions associated with clinical and subclinical VAD, gut microbiome adaptation to VAD and VAD effects on the immune responses to infection and vaccines.
Collapse
|
10
|
Imdad A, Mayo-Wilson E, Haykal MR, Regan A, Sidhu J, Smith A, Bhutta ZA. Vitamin A supplementation for preventing morbidity and mortality in children from six months to five years of age. Cochrane Database Syst Rev 2022; 3:CD008524. [PMID: 35294044 PMCID: PMC8925277 DOI: 10.1002/14651858.cd008524.pub4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Vitamin A deficiency (VAD) is a major public health problem in low- and middle-income countries, affecting 190 million children under five years of age and leading to many adverse health consequences, including death. Based on prior evidence and a previous version of this review, the World Health Organization has continued to recommend vitamin A supplementation (VAS) for children aged 6 to 59 months. The last version of this review was published in 2017, and this is an updated version of that review. OBJECTIVES To assess the effects of vitamin A supplementation (VAS) for preventing morbidity and mortality in children aged six months to five years. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, six other databases, and two trials registers up to March 2021. We also checked reference lists and contacted relevant organisations and researchers to identify additional studies. SELECTION CRITERIA Randomised controlled trials (RCTs) and cluster-RCTs evaluating the effect of synthetic VAS in children aged six months to five years living in the community. We excluded studies involving children in hospital and children with disease or infection. We also excluded studies evaluating the effects of food fortification, consumption of vitamin A rich foods, or beta-carotene supplementation. DATA COLLECTION AND ANALYSIS For this update, two review authors independently assessed studies for inclusion resolving discrepancies by discussion. We performed meta-analyses for outcomes, including all-cause and cause-specific mortality, disease, vision, and side effects. We used the GRADE approach to assess the quality of the evidence. MAIN RESULTS The updated search identified no new RCTs. We identified 47 studies, involving approximately 1,223,856 children. Studies were set in 19 countries: 30 (63%) in Asia, 16 of these in India; 8 (17%) in Africa; 7 (15%) in Latin America, and 2 (4%) in Australia. About one-third of the studies were in urban/periurban settings, and half were in rural settings; the remaining studies did not clearly report settings. Most studies included equal numbers of girls and boys and lasted about one year. The mean age of the children was about 33 months. The included studies were at variable overall risk of bias; however, evidence for the primary outcome was at low risk of bias. A meta-analysis for all-cause mortality included 19 trials (1,202,382 children). At longest follow-up, there was a 12% observed reduction in the risk of all-cause mortality for VAS compared with control using a fixed-effect model (risk ratio (RR) 0.88, 95% confidence interval (CI) 0.83 to 0.93; high-certainty evidence). Nine trials reported mortality due to diarrhoea and showed a 12% overall reduction for VAS (RR 0.88, 95% CI 0.79 to 0.98; 1,098,538 children; high-certainty evidence). There was no evidence of a difference for VAS on mortality due to measles (RR 0.88, 95% CI 0.69 to 1.11; 6 studies, 1,088,261 children; low-certainty evidence), respiratory disease (RR 0.98, 95% CI 0.86 to 1.12; 9 studies, 1,098,538 children; low-certainty evidence), and meningitis. VAS reduced the incidence of diarrhoea (RR 0.85, 95% CI 0.82 to 0.87; 15 studies, 77,946 children; low-certainty evidence), measles (RR 0.45, 95% CI 0.30 to 0.69; 2 studies, 1,982 children; low-certainty evidence), Bitot's spots (RR 0.42, 95% CI 0.33 to 0.53; 5 studies, 1,063,278 children; moderate-certainty evidence), night blindness (RR 0.32, 95% CI 0.21 to 0.50; 2 studies, 22,972 children; moderate-certainty evidence), and VAD (RR 0.71, 95% CI 0.65 to 0.78; 4 studies, 2262 children, moderate-certainty evidence). However, there was no evidence of a difference on incidence of respiratory disease (RR 0.99, 95% CI 0.92 to 1.06; 11 studies, 27,540 children; low-certainty evidence) or hospitalisations due to diarrhoea or pneumonia. There was an increased risk of vomiting within the first 48 hours of VAS (RR 1.97, 95% CI 1.44 to 2.69; 4 studies, 10,541 children; moderate-certainty evidence). AUTHORS' CONCLUSIONS This update identified no new eligible studies and the conclusions remain the same. VAS is associated with a clinically meaningful reduction in morbidity and mortality in children. Further placebo-controlled trials of VAS in children between six months and five years of age would not change the conclusions of this review, although studies that compare different doses and delivery mechanisms are needed. In populations with documented VAD, it would be unethical to conduct placebo-controlled trials.
Collapse
Affiliation(s)
- Aamer Imdad
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Evan Mayo-Wilson
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Maya R Haykal
- College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Allison Regan
- College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jasleen Sidhu
- College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Abigail Smith
- Health Sciences Library, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Zulfiqar A Bhutta
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
11
|
Micronutrient Improvement of Epithelial Barrier Function in Various Disease States: A Case for Adjuvant Therapy. Int J Mol Sci 2022; 23:ijms23062995. [PMID: 35328419 PMCID: PMC8951934 DOI: 10.3390/ijms23062995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
The published literature makes a very strong case that a wide range of disease morbidity associates with and may in part be due to epithelial barrier leak. An equally large body of published literature substantiates that a diverse group of micronutrients can reduce barrier leak across a wide array of epithelial tissue types, stemming from both cell culture as well as animal and human tissue models. Conversely, micronutrient deficiencies can exacerbate both barrier leak and morbidity. Focusing on zinc, Vitamin A and Vitamin D, this review shows that at concentrations above RDA levels but well below toxicity limits, these micronutrients can induce cell- and tissue-specific molecular-level changes in tight junctional complexes (and by other mechanisms) that reduce barrier leak. An opportunity now exists in critical care—but also medical prophylactic and therapeutic care in general—to consider implementation of select micronutrients at elevated dosages as adjuvant therapeutics in a variety of disease management. This consideration is particularly pointed amidst the COVID-19 pandemic.
Collapse
|
12
|
Zhou J, Qin Y, Xiong X, Wang Z, Wang M, Wang Y, Wang QY, Yang HS, Yin Y. Effects of iron, vitamin A, and the interaction between the two nutrients on intestinal development and cell differentiation in piglets. J Anim Sci 2021; 99:6360813. [PMID: 34467981 DOI: 10.1093/jas/skab258] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022] Open
Abstract
This study aimed to investigate the effects of iron, vitamin A (VA) and their interaction on intestinal development and differentiation of cells in suckling piglets. Therefore, 32 Duroc × Landrace × Yorkshire 0-d-old newborn boars with similar body weights were randomly divided into four groups, with eight replicates in each group and one pig in each replicate. All the piglets were breastfed. In addition, the piglets were given normal saline (CON group) or ferrous sulfate (OAFe group) or VA (VA group) or ferrous sulfate and VA (OAFe + VA group) on the 2nd, 7th, 12th, and 17th day, respectively. The piglets were then slaughtered on the 21st day, and intestinal samples were collected. The results showed that: 1) iron (P < 0.001) significantly increased the length, weight, relative weight, and the length to weight ratio of the small intestine. On the other hand, VA had a significant effect on the weight to length ratio (P = 0.015) and relative weight (P < 0.001) of the small intestine; 2) with regard to intestinal morphology, supplementation with iron (P <0.05) had obvious effects on the villus height (VH), crypt depth (CD), villus width (VW), and surface area. Additionally, both VA and interaction of VA and iron increased the VH (P < 0.05) and surface area (P = 0.001). The results also showed that iron (P < 0.01) increased the number of crypt goblet cells, Ki67-positive cells, and endocrine cells. Moreover, both VA and the interaction between VA and iron increased the number of endocrine cells in the villi (P = 0.05); 3) With regard to the mRNA expression levels of stem cell differentiation marker genes, iron (P < 0.05) decreased the expression of trophinin 2 (Trop2), leucine-rich repeat containing G protein-coupled receptor 5 positive (Lgr5+), male-specific lethal 1(Msl1), BMI 1 proto-oncogene, polycomb ring finger (Bmi1), and achaete-scute family bHLH transcription factor 2 (Ascl2). On the other hand, VA increased the expression of Ascl2 (P = 0.001) although the interaction of VA and iron (P < 0.05) had an effect on the expression of secreted phosphoprotein 1 (Spp1) and Bmi1. In addition, VA decreased the gene or mRNA expression of aconitase 1 (Aco1; P < 0.001), transferrin receptor (TFRC; P = 0.001), and solute carrier family 11 member 2 (DMT1; P = 0.003) in the Iron Reactive Element/Iron Regulatory Protein (IRE/IRP) signaling pathway although iron and the interaction of VA and iron had no effect on the genes' expression. The results therefore showed that VA, iron, and their interaction can promote intestinal development and epithelial cell differentiation in piglets.
Collapse
Affiliation(s)
- Jing Zhou
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Yan Qin
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Xia Xiong
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Zhaobin Wang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Min Wang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Yancan Wang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Qiye Y Wang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Huansheng S Yang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Yulong Yin
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China.,Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| |
Collapse
|
13
|
High-fat diet increases the severity of Giardia infection in association with low-grade inflammation and gut microbiota dysbiosis. Sci Rep 2021; 11:18842. [PMID: 34552170 PMCID: PMC8458452 DOI: 10.1038/s41598-021-98262-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/01/2021] [Indexed: 12/26/2022] Open
Abstract
Exogenous factors that may influence the pathophysiology of Giardia infection remain incompletely understood. We have investigated the role of dietary fat in the pathogenesis of Giardia infection. Male 3 to 4-week-old C57BL/6 mice were fed either a low fat (LF) or a high fat (HF) diet for 12 days and challenged with G. duodenalis. In infected animals, the trophozoite burden was higher in HF + Giardia mice compared to the LF + Giardia group at day 7 post infection. Fatty acids exerted direct pro-growth effects on Giardia trophozoites. Analysis of disease parameters showed that HF + Giardia mice exhibited more mucosal infiltration by inflammatory cells, decreased villus/crypt ratios, goblet cell hyperplasia, mucus disruption, increased gut motility, and elevated fecal water content compared with LF + Giardia. HF diet-dependent exacerbation of Giardia-induced goblet cell hyperplasia was associated with elevated Atoh1 and Muc2 gene expression. Gut microbiota analysis revealed that the HF diet alone induces a taxonomic shift. HF + Giardia mice exhibited microbiota dysbiosis characterized by an increase of Firmicutes and a decrease of Bacteroidetes and significant changes in α- and β-diversity metrics. Taken together, the findings suggest that a HF diet exacerbates the outcome of Giardia infection. The data demonstrate that elevated dietary fat represents an important exogenous factor promoting the pathophysiology of giardiasis.
Collapse
|
14
|
Fekete E, Allain T, Siddiq A, Sosnowski O, Buret AG. Giardia spp. and the Gut Microbiota: Dangerous Liaisons. Front Microbiol 2021; 11:618106. [PMID: 33510729 PMCID: PMC7835142 DOI: 10.3389/fmicb.2020.618106] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Alteration of the intestinal microbiome by enteropathogens is commonly associated with gastrointestinal diseases and disorders and has far-reaching consequences for overall health. Significant advances have been made in understanding the role of microbial dysbiosis during intestinal infections, including infection with the protozoan parasite Giardia duodenalis, one of the most prevalent gut protozoa. Altered species composition and diversity, functional changes in the commensal microbiota, and changes to intestinal bacterial biofilm structure have all been demonstrated during the course of Giardia infection and have been implicated in Giardia pathogenesis. Conversely, the gut microbiota has been found to regulate parasite colonization and establishment and plays a critical role in immune modulation during mono and polymicrobial infections. These disruptions to the commensal microbiome may contribute to a number of acute, chronic, and post-infectious clinical manifestations of giardiasis and may account for variations in disease presentation within and between infected populations. This review discusses recent advances in characterizing Giardia-induced bacterial dysbiosis in the gut and the roles of dysbiosis in Giardia pathogenesis.
Collapse
Affiliation(s)
- Elena Fekete
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Thibault Allain
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Affan Siddiq
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Olivia Sosnowski
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Andre G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
15
|
de Freitas REM, Medeiros PHQS, Rodrigues FADP, Clementino MADF, Fernandes C, da Silva AVA, Prata MDMG, Cavalcante PA, Lima AÂM, Havt A. Retinoids delay cell cycle progression and promote differentiation of intestinal epithelial cells exposed to nutrient deprivation. Nutrition 2020; 85:111087. [PMID: 33545543 DOI: 10.1016/j.nut.2020.111087] [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: 07/13/2020] [Revised: 10/10/2020] [Accepted: 11/12/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Vitamin A is commonly recommended as a treatment for diarrhea and undernutrition; however, little is known about the underlying cellular mechanisms. The aim of this study was to investigate the modulation of cell cycle by vitamin A derivatives (retinyl palmitate or retinol) in undernourished intestinal epithelial crypts (IEC-6). METHODS IEC-6 cells were exposed to nutrient deprivation (no serum and no glutamine) and supplemented with retinyl palmitate or retinol at a range of 2 to 20 μM. Proliferation, apoptosis/necrosis, cell cycle process, and gene transcription were assessed. RESULTS Nutrient deprivation for 6, 12, 24, or 48 h decreased cell proliferation, and retinyl palmitate further decreased it after 24 and 48 h. Apoptosis rates were reduced by undernourishment and further reduced by retinyl palmitate after 48 h; whereas necrosis rates were unaltered. Undernourishment induced overall cell quiescence, increased percentage of cells in G0/G1 phase and decreased percentage of cells in S phase after 12 h and in G2/M phases at 6, 12, and 24 h after treatment. Both retinoids also showed cell quiescence induction with less cells in G2/M phases after 48 h, whereas only retinol showed significant modulation of G0/G1 and S phases. Both retinoids also increased markers of cell differentiation Fabp and Iap gene transcriptions in about fivefold rates after 42 h. Furthermore, specific gene transcriptions related to MAP kinase signaling pathway regulation of cell differentiation and cell cycle regulation were triggered by retinoids in undernourished IEC-6, with higher levels of expression for Atf2 and C-jun genes. CONCLUSIONS These findings indicated that both vitamin A derivatives induce further survival mechanisms in undernourished intestinal epithelial crypt cells. These mechanisms include increased cell quiescence, decreased apoptosis, increased cell differentiation, and transcription of genes related to MAP kinase signaling pathway.
Collapse
Affiliation(s)
- Rosa Elayne Marques de Freitas
- Institute of Biomedicine and Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | | | | | | | - Camila Fernandes
- Institute of Biomedicine and Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Antonio Vinicios Alves da Silva
- Institute of Biomedicine and Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Mara de Moura Gondim Prata
- Institute of Biomedicine and Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | | | | | - Alexandre Havt
- Institute of Biomedicine and Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
| |
Collapse
|
16
|
Khoshbin K, Camilleri M. Effects of dietary components on intestinal permeability in health and disease. Am J Physiol Gastrointest Liver Physiol 2020; 319:G589-G608. [PMID: 32902315 PMCID: PMC8087346 DOI: 10.1152/ajpgi.00245.2020] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Altered intestinal permeability plays a role in many pathological conditions. Intestinal permeability is a component of the intestinal barrier. This barrier is a dynamic interface between the body and the food and pathogens that enter the gastrointestinal tract. Therefore, dietary components can directly affect this interface, and many metabolites produced by the host enzymes or the gut microbiota can act as signaling molecules or exert direct effects on this barrier. Our aim was to examine the effects of diet components on the intestinal barrier in health and disease states. Herein, we conducted an in-depth PubMed search based on specific key words (diet, permeability, barrier, health, disease, and disorder), as well as cross references from those articles. The normal intestinal barrier consists of multiple components in the lumen, epithelial cell layer and the lamina propria. Diverse methods are available to measure intestinal permeability. We focus predominantly on human in vivo studies, and the literature is reviewed to identify dietary factors that decrease (e.g., emulsifiers, surfactants, and alcohol) or increase (e.g., fiber, short-chain fatty acids, glutamine, and vitamin D) barrier integrity. Effects of these dietary items in disease states, such as metabolic syndrome, liver disease, or colitis are documented as examples of barrier dysfunction in the multifactorial diseases. Effects of diet on intestinal barrier function are associated with precise mechanisms in some instances; further research of those mechanisms has potential to clarify the role of dietary interventions in treating diverse pathologic states.
Collapse
Affiliation(s)
- Katayoun Khoshbin
- Clinical Enteric Neuroscience Translational and Epidemiological Research, Mayo Clinic, Rochester, Minnesota
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
17
|
Snyder LM, Arora J, Kennett MJ, Weaver V, Cantorna MT. Retinoid Signaling in Intestinal Epithelial Cells Is Essential for Early Survival From Gastrointestinal Infection. Front Immunol 2020; 11:559635. [PMID: 33117344 PMCID: PMC7578243 DOI: 10.3389/fimmu.2020.559635] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022] Open
Abstract
Vitamin A deficiency (A–) increases morbidity and mortality to gastrointestinal (GI) infection. Blocking retinoid signaling (dominant negative retinoic acid receptor, dnRAR) in intestinal epithelial cells (IEC, IECdnRAR) had no effect on vitamin A absorption, the expression of tight junction proteins or the integrity of the barrier. Immune cells in the gut were present in normal frequencies in the IECdnRAR mice, with the exception of the T cell receptor (TCR)αβ+/CD8αα cells, which were significantly lower than in wildtype littermates. Challenging the IECdnRAR mice with dextran sodium sulfate to induce colitis or Citrobacter rodentium infection resulted in similar disease to wildtype littermates. Feeding mice vitamin A deficient diets reduced vitamin A status and the A– IECdnRAR mice developed more severe colitis and C. rodentium infection. In particular, retinoid signaling in the IEC was crucial for the A– host to survive early infection following C. rodentium. Treating A– mice with retinoic acid (RA) beginning on the day of infection protects most mice from early lethality. However, RA treatment of the A– IECdnRAR mice was ineffective for preventing lethality following C. rodentium infection. Retionid signaling in IEC is critical, especially when there are reduced levels of dietary vitamin A. IEC are direct targets of vitamin A for mounting early defense against infection.
Collapse
Affiliation(s)
- Lindsay M Snyder
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States.,Center for Evolutionary & Theoretical Immunology, University of New Mexico, Albuquerque, NM, United States
| | - Juhi Arora
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Mary J Kennett
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Veronika Weaver
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Margherita T Cantorna
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
18
|
Van Wayenbergh E, Struyf N, Rezaei MN, Sagalowicz L, Bel-Rhlid R, Moccand C, Courtin CM. Cereal bran protects vitamin A from degradation during simmering and storage. Food Chem 2020; 331:127292. [PMID: 32559599 DOI: 10.1016/j.foodchem.2020.127292] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 05/07/2020] [Accepted: 06/07/2020] [Indexed: 11/30/2022]
Abstract
Food supplementation with vitamin A is an efficient strategy to combat vitamin A deficiency. The stability of vitamin A during cooking and storage is, however, low. We here show that cereal bran protects retinyl palmitate (RP) during simmering and storage. Native wheat bran stabilized RP the most during simmering. About 75% RP was recovered after 120 min of cooking, while all RP was lost after 80 min in the absence of bran. Heat-treated rice bran protected RP the best during forced storage, with a 35% recovery after 8 weeks. RP was degraded entirely in the absence of bran in less than one week. Results suggested that the physical entrapment of oil within the large wheat bran particles protects RP from the action of water and pro-oxidants during simmering. During storage, the high amount and diversity of lipid components present in rice bran are presumably responsible for its protective effect.
Collapse
Affiliation(s)
- Eline Van Wayenbergh
- Laboratory of Food Chemistry and Biochemistry & Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Kasteelpark Arenberg 20, B-3001 Heverlee, Belgium
| | - Nore Struyf
- Laboratory of Food Chemistry and Biochemistry & Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Kasteelpark Arenberg 20, B-3001 Heverlee, Belgium
| | - Mohammad N Rezaei
- Laboratory of Food Chemistry and Biochemistry & Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Kasteelpark Arenberg 20, B-3001 Heverlee, Belgium
| | | | - Rachid Bel-Rhlid
- Nestlé Research, Vers-Chez-Les-Blanc, 1000 Lausanne 26, Switzerland
| | - Cyril Moccand
- Nestlé Research, Vers-Chez-Les-Blanc, 1000 Lausanne 26, Switzerland
| | - Christophe M Courtin
- Laboratory of Food Chemistry and Biochemistry & Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Kasteelpark Arenberg 20, B-3001 Heverlee, Belgium.
| |
Collapse
|
19
|
Wang Z, Li J, Wang Y, Wang L, Yin Y, Yin L, Yang H, Yin Y. Dietary vitamin A affects growth performance, intestinal development, and functions in weaned piglets by affecting intestinal stem cells. J Anim Sci 2020; 98:skaa020. [PMID: 31955210 PMCID: PMC7023621 DOI: 10.1093/jas/skaa020] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
Vitamin A (VA) is an important nutrient for weaning piglets. It plays a significant role in the normal formation, development, and maintenance of epithelial cells. Previous studies have shown that VA supplements could improve the host's intestinal barrier function. Therefore, we hypothesized that VA supplements can affect intestinal function in weaned piglets by regulating intestinal stem cells. Thirty-two 21-d-old weaned [(Yorkshire × Landrace) × Duroc] piglets with an average weight of 8.34 ± 0.13 kg were randomly divided into 4 treatment groups, with 1) 2 mg/kg (control), 2) 4 mg/kg, 3) 8 mg/kg, and 4) 16 mg/kg doses of VA, respectively. The experiment lasted for 14 d. Weaned piglets were given ad libitum access to food and water during the test. The ADG (linear, P = 0.020) and G:F (linear, P = 0.005) of the piglets were found to increase significantly from days 8 to 14. The Lgr5+ gene expression (P = 0.012) in the jejunum mucosa of the 16 mg/kg VA group was increased. The jejunum villus height (P = 0.027) and villi surface area (P = 0.035) were significantly increased in the 4 mg/kg VA treatment group. The crypt depth increased significantly in the 4 and 8 mg/kg VA treatment groups (quadratic, P = 0.043), and the ratios of villus height to crypt depth significantly increased in the 16 mg/kg VA group (quadratic, P = 0.015). The maltase (P = 0.032), sucrose (P = 0.041), and alkaline phosphatase activity (linear, P = 0.024) were significantly increased when further supplemented with 4 mg/kg VA. Slc2a2 mRNA abundance was significantly increased in the 2 mg/kg VA group (linear, P = 0.024). Moreover, the budding rates, buddings number per organoid, and Chromogranin A and Muc2 expression of piglet intestinal organoids were significantly reduced (P < 0.05) by VA and its metabolites (retinoic acid). Compared with the control group, the expression of Spp1 and Trop2 increased. These results indicated that VA may increase the stemness of intestinal stem cell in vitro. This study suggested that VA could affect growth performance and intestinal function by regulating intestinal stem cells in the jejunum of weaned piglets.
Collapse
Affiliation(s)
- Zhaobin Wang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Changsha, Hunan, People’s Republic of China
| | - Jia Li
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Changsha, Hunan, People’s Republic of China
| | - Yu Wang
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, Hubei, People’s Republic of China
| | - Lei Wang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Changsha, Hunan, People’s Republic of China
| | - Yuebang Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Changsha, Hunan, People’s Republic of China
| | - Lanmei Yin
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, People’s Republic of China
| | - Huansheng Yang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Changsha, Hunan, People’s Republic of China
| | - Yulong Yin
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
20
|
Allain T, Buret AG. Pathogenesis and post-infectious complications in giardiasis. ADVANCES IN PARASITOLOGY 2019; 107:173-199. [PMID: 32122529 DOI: 10.1016/bs.apar.2019.12.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Giardia is an important cause of diarrhoea, and results in post-infectious and extra-intestinal complications. This chapter presents a state-of-the art of our understanding of how this parasite may cause such abnormalities, which appear to develop at least in part in Assemblage-dependent manner. Findings from prospective longitudinal cohort studies indicate that Giardia is one of the four most prevalent enteropathogens in early life, and represents a risk factor for stunting at 2 years of age. This may occur independently of diarrheal disease, in strong support of the pathophysiological significance of the intestinal abnormalities induced by this parasite. These include epithelial malabsorption and maldigestion, increased transit, mucus depletion, and disruptions of the commensal microbiota. Giardia increases epithelial permeability and facilitates the invasion of gut bacteria. Loss of intestinal barrier function is at the core of the acute and post-infectious complications associated with this infection. Recent findings demonstrate that the majority of the pathophysiological responses triggered by this parasite can be recapitulated by the effects of its membrane-bound and secreted cysteine proteases.
Collapse
Affiliation(s)
- Thibault Allain
- University of Calgary, Host-Parasite Interactions Program, Inflammation Research Network, Department of Biological Sciences, Calgary, Canada
| | - André G Buret
- University of Calgary, Host-Parasite Interactions Program, Inflammation Research Network, Department of Biological Sciences, Calgary, Canada.
| |
Collapse
|
21
|
Cantorna MT, Snyder L, Arora J. Vitamin A and vitamin D regulate the microbial complexity, barrier function, and the mucosal immune responses to ensure intestinal homeostasis. Crit Rev Biochem Mol Biol 2019; 54:184-192. [PMID: 31084433 DOI: 10.1080/10409238.2019.1611734] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diet is an important regulator of the gastrointestinal microbiota. Vitamin A and vitamin D deficiencies result in less diverse, dysbiotic microbial communities and increased susceptibility to infection or injury of the gastrointestinal tract. The vitamin A and vitamin D receptors are nuclear receptors expressed by the host, but not the microbiota. Vitamin A- and vitamin D-mediated regulation of the intestinal epithelium and mucosal immune cells underlies the effects of these nutrients on the microbiota. Vitamin A and vitamin D regulate the expression of tight junction proteins on intestinal epithelial cells that are critical for barrier function in the gut. Other shared functions of vitamin A and vitamin D include the support of innate lymphoid cells that produce IL-22, suppression of IFN-γ and IL-17 by T cells, and induction of regulatory T cells in the mucosal tissues. There are some unique functions of vitamin A and D; for example, vitamin A induces gut homing receptors on T cells, while vitamin D suppresses gut homing receptors on T cells. Together, vitamin A- and vitamin D-mediated regulation of the intestinal epithelium and mucosal immune system shape the microbial communities in the gut to maintain homeostasis.
Collapse
Affiliation(s)
- Margherita T Cantorna
- a Department of Veterinary and Biomedical Science , The Pennsylvania State University , University Park , PA , USA.,b Center for Molecular Immunology and Infectious Disease , The Pennsylvania State University , University Park , PA , USA
| | - Lindsay Snyder
- a Department of Veterinary and Biomedical Science , The Pennsylvania State University , University Park , PA , USA.,b Center for Molecular Immunology and Infectious Disease , The Pennsylvania State University , University Park , PA , USA
| | - Juhi Arora
- a Department of Veterinary and Biomedical Science , The Pennsylvania State University , University Park , PA , USA.,b Center for Molecular Immunology and Infectious Disease , The Pennsylvania State University , University Park , PA , USA
| |
Collapse
|
22
|
Strobbe S, De Lepeleire J, Van Der Straeten D. From in planta Function to Vitamin-Rich Food Crops: The ACE of Biofortification. FRONTIERS IN PLANT SCIENCE 2018; 9:1862. [PMID: 30619424 PMCID: PMC6305313 DOI: 10.3389/fpls.2018.01862] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/03/2018] [Indexed: 05/11/2023]
Abstract
Humans are highly dependent on plants to reach their dietary requirements, as plant products contribute both to energy and essential nutrients. For many decades, plant breeders have been able to gradually increase yields of several staple crops, thereby alleviating nutritional needs with varying degrees of success. However, many staple crops such as rice, wheat and corn, although delivering sufficient calories, fail to satisfy micronutrient demands, causing the so called 'hidden hunger.' Biofortification, the process of augmenting nutritional quality of food through the use of agricultural methodologies, is a pivotal asset in the fight against micronutrient malnutrition, mainly due to vitamin and mineral deficiencies. Several technical advances have led to recent breakthroughs. Nutritional genomics has come to fruition based on marker-assisted breeding enabling rapid identification of micronutrient related quantitative trait loci (QTL) in the germplasm of interest. As a complement to these breeding techniques, metabolic engineering approaches, relying on a continuously growing fundamental knowledge of plant metabolism, are able to overcome some of the inevitable pitfalls of breeding. Alteration of micronutrient levels does also require fundamental knowledge about their role and influence on plant growth and development. This review focuses on our knowledge about provitamin A (beta-carotene), vitamin C (ascorbate) and the vitamin E group (tocochromanols). We begin by providing an overview of the functions of these vitamins in planta, followed by highlighting some of the achievements in the nutritional enhancement of food crops via conventional breeding and genetic modification, concluding with an evaluation of the need for such biofortification interventions. The review further elaborates on the vast potential of creating nutritionally enhanced crops through multi-pathway engineering and the synergistic potential of conventional breeding in combination with genetic engineering, including the impact of novel genome editing technologies.
Collapse
|
23
|
de Medeiros PHQS, Pinto DV, de Almeida JZ, Rêgo JMC, Rodrigues FAP, Lima AÂM, Bolick DT, Guerrant RL, Oriá RB. Modulation of Intestinal Immune and Barrier Functions by Vitamin A: Implications for Current Understanding of Malnutrition and Enteric Infections in Children. Nutrients 2018; 10:nu10091128. [PMID: 30134532 PMCID: PMC6164597 DOI: 10.3390/nu10091128] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/11/2018] [Accepted: 08/17/2018] [Indexed: 12/24/2022] Open
Abstract
The micronutrient vitamin A refers to a group of compounds with pleiotropic effects on human health. These molecules can modulate biological functions, including development, vision, and regulation of the intestinal barrier. The consequences of vitamin A deficiency and supplementation in children from developing countries have been explored for several years. These children live in an environment that is highly contaminated by enteropathogens, which can, in turn, influence vitamin A status. Vitamin A has been described to modulate gene expression, differentiation and function of diverse immune cells; however, the underlying mechanisms are not fully elucidated. This review aims to summarize the most updated advances on elucidating the vitamin A effects targeting intestinal immune and barrier functions, which may help in further understanding the burdens of malnutrition and enteric infections in children. Specifically, by covering both clinical and in vivo/in vitro data, we describe the effects of vitamin A related to gut immune tolerance/homeostasis, intestinal barrier integrity, and responses to enteropathogens in the context of the environmental enteric dysfunction. Some of the gaps in the literature that require further research are also highlighted.
Collapse
Affiliation(s)
- Pedro Henrique Q S de Medeiros
- Laboratory of Infectious Diseases, Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Daniel V Pinto
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
| | - Juliana Zani de Almeida
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
| | - Juliana M C Rêgo
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
- Department of Nutrition, Christus University Center, Fortaleza 60190-060 CE, Brazil.
| | - Francisco A P Rodrigues
- Laboratory of Infectious Diseases, Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
| | - Aldo Ângelo M Lima
- Laboratory of Infectious Diseases, Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
| | - David T Bolick
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Richard L Guerrant
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Reinaldo B Oriá
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza 60430-270 CE, Brazil.
| |
Collapse
|
24
|
Abdelhamid L, Luo XM. Retinoic Acid, Leaky Gut, and Autoimmune Diseases. Nutrients 2018; 10:E1016. [PMID: 30081517 PMCID: PMC6115935 DOI: 10.3390/nu10081016] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/25/2018] [Accepted: 07/25/2018] [Indexed: 12/19/2022] Open
Abstract
A leaky gut has been observed in a number of autoimmune diseases including type 1 diabetes, multiple sclerosis, inflammatory bowel disease, and systemic lupus erythematosus. Previous studies from our laboratory have shown that lupus mice also bear a leaky gut and that the intestinal barrier function can be enhanced by gut colonization of probiotics such as Lactobacillus spp. Retinoic acid (RA) can increase the relative abundance of Lactobacillus spp. in the gut. Interestingly, RA has also been shown to strengthen the barrier function of epithelial cells in vitro and in the absence of probiotic bacteria. These reports bring up an interesting question of whether RA exerts protective effects on the intestinal barrier directly or through regulating the microbiota colonization. In this review, we will discuss the roles of RA in immunomodulation, recent literature on the involvement of a leaky gut in different autoimmune diseases, and how RA shapes the outcomes of these diseases.
Collapse
Affiliation(s)
- Leila Abdelhamid
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Xin M Luo
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
25
|
Xiao L, Cui T, Liu S, Chen B, Wang Y, Yang T, Li T, Chen J. Vitamin A supplementation improves the intestinal mucosal barrier and facilitates the expression of tight junction proteins in rats with diarrhea. Nutrition 2018; 57:97-108. [PMID: 30153586 DOI: 10.1016/j.nut.2018.06.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 06/11/2018] [Accepted: 06/19/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The aim of this study is to investigate the specific effects of vitamin A (VA) on diarrhea in rats and its potential targets to protect the intestinal mucosa. METHODS Specific pathogen-free Sprague Dawley rats were fed a VA deficient (VAD) or VA normal (VAN) diet for 4 wk. Then, half of the VAN rats were treated with a VAN diet and the other half with a lactose VAN diet. VAD rats were randomly assigned to one of four groups and fed a VAD diet, lactose VAD diet, VAN diet with VA supplementation (VAS) via daily intragastric administration, or a lactose VAN diet with daily VAS. Rat weight and degree of diarrhea were evaluated daily. After 15 d, the serum retinol level was measured by high-performance liquid chromatography, and the serum diamine oxidase (DAO) and zonulin concentrations were analyzed by enzyme-linked immunosorbent assays. The small intestine mucosal pathology was observed by hematoxylin and eosin staining. Western blotting was performed to detect the protein expression levels of occludin and claudin-1 in the intestinal mucosa, and the zonula-occludens 1 expression was assessed using immunohistochemistry. RESULTS VAD limited weight gain in rats and increased the degree of diarrhea. The serum retinol levels and the level of tight junction (TJ) proteins claudin-1 and occludin and grip strength were affected by the interaction between lactose-induced diarrhea and the VA diet. Diarrhea, independent of VAD, significantly decreased rat weight, increased serum DAO levels, damaged small intestine villi, and impaired zonula-occludens 1 protein expression. VAD significantly increased the concentration of zonulin independently of diarrhea, but VAS increased the serum retinol level, reduced the severity of diarrhea, increased the expression levels of the TJ proteins, facilitated the restoration of the small intestine villi that were damaged by the diarrhea, and decreased the concentrations of serum DAO and zonulin. CONCLUSIONS VAD may aggravate the degree of diarrhea and intestinal mucosal damage during the duration of diarrhea, and VAS helps relieve diarrhea and improves intestinal damage likely by regulating the expression of TJ proteins. Therefore, VA plays a pivotal role in the protection of the intestinal mucosa during instances of diarrhea.
Collapse
Affiliation(s)
- Lu Xiao
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Ting Cui
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Shu Liu
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Baolin Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Yuting Wang
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China; Department of Digestive, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ting Yang
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Tingyu Li
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Jie Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China.
| |
Collapse
|
26
|
Rerksuppaphol S, Rerksuppaphol L. Mid-Upper-Arm Circumference and Arm-to-Height Ratio to Identify Obesity in School-Age Children. Clin Med Res 2017; 15:53-58. [PMID: 29018004 PMCID: PMC5849437 DOI: 10.3121/cmr.2017.1365] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 07/24/2017] [Accepted: 10/02/2017] [Indexed: 11/18/2022]
Abstract
BACKGROUND In resource-poor settings, anthropometric parameters are evaluated as potential alternatives to the body mass index (BMI) for detecting overweight and obesity in children. To this end, the mid-upper-arm circumference (MUAC) and the mid-upper-arm circumference-to-height ratio (AHtR) were evaluated as proxies to BMI in Thai school-age children. STUDY DESIGN An observational, cross-sectional study was performed on school-aged children. PARTICIPANTS Children in grades 1 through 6 at all public elementary schools in the Ongkharak district, Nakhon Nayok, Thailand during May and June 2013 were included. This is a rural district with low per capita income. METHODS Weight, height, and MUAC were measured in school-age children and analyzed to identify optimal cut-off values for MUAC and AHtR for detection of overweight and obesity in comparison to BMI. Receiver operating characteristic (ROC) curve analysis determined the validity of MUAC and AHtR use. RESULTS Data from 3,618 children, aged 6.0-12.99 years, were analyzed. MUAC correlated with age and height (P < 0.001), but especially with body weight (r = 0.888 to 0.914) and BMI (r = 0.859 to 0.908) in both genders, while AHtR correlated with body weight and BMI (P < 0.001), but not with age. Cut-off values of MUAC for obesity diagnosis ranged from 18.9 to 25.5 cm for boys and from 19.8 to 25.4 cm for girls. Accuracy was excellent for both boys (AUC = 0.952-0.991) and girls (AUC = 0.917-0.990). Cut-off of MUAC for overweight diagnosis ranged from 17.2 to 22.4 cm for boys (AUC = 0.883-0.965) and from 18.0 to 23.2 cm for girls (AUC = 0.905-0.931). AHtR cut-off values for obesity and overweight diagnosis at 0.16 and 0.145, respectively, were determined with excellent diagnostic accuracy (AUC ranged from 0.920 to 0.975). CONCLUSION MUAC and AHtR were reliable tools to detect overweight and obesity in Thai school-age children. Cut-off points for MUAC were age and gender specific, while AHtR at 0.16 and 0.145 were the optimal values for both genders, independent of age. These anthropometric measurements showed excellent accuracy in predicting overweight and obesity with high specificity and sensitivity.
Collapse
Affiliation(s)
- Sanguansak Rerksuppaphol
- Department of Pediatrics, Faculty of Medicine, Srinakharinwirot University, Nakorn Nayok, Thailand, E-mail: .
| | - Lakkana Rerksuppaphol
- Department of Preventive Medicine, Faculty of Medicine, Srinakharinwirot University, Nakorn Nayok, Thailand, E-mail:
| |
Collapse
|
27
|
Fink MY, Singer SM. The Intersection of Immune Responses, Microbiota, and Pathogenesis in Giardiasis. Trends Parasitol 2017; 33:901-913. [PMID: 28830665 DOI: 10.1016/j.pt.2017.08.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/18/2017] [Accepted: 08/01/2017] [Indexed: 02/07/2023]
Abstract
Giardia lamblia is one of the most common infectious protozoans in the world. Giardia rarely causes severe life-threatening diarrhea, and may even have a slight protective effect in this regard, but it is a major contributor to malnutrition and growth faltering in children in the developing world. Giardia infection also appears to be a significant risk factor for postinfectious irritable bowel and chronic fatigue syndromes. In this review we highlight recent work focused on the impact of giardiasis and the mechanisms that contribute to the various outcomes of this infection, including changes in the composition of the microbiota, activation of immune responses, and immunopathology.
Collapse
Affiliation(s)
- Marc Y Fink
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Steven M Singer
- Department of Biology, Georgetown University, Washington, DC, USA.
| |
Collapse
|
28
|
Wiseman EM, Bar-El Dadon S, Reifen R. The vicious cycle of vitamin a deficiency: A review. Crit Rev Food Sci Nutr 2017; 57:3703-3714. [DOI: 10.1080/10408398.2016.1160362] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Elina Manusevich Wiseman
- The Center of Nutrigenomics, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Shimrit Bar-El Dadon
- The Center of Nutrigenomics, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Ram Reifen
- The Center of Nutrigenomics, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
29
|
Watson S, Gong YY, Routledge M. Interventions targeting child undernutrition in developing countries may be undermined by dietary exposure to aflatoxin. Crit Rev Food Sci Nutr 2017; 57:1963-1975. [PMID: 26176888 DOI: 10.1080/10408398.2015.1040869] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Child undernutrition, a form of malnutrition, is a major public health burden in developing countries. Supplementation interventions targeting the major micronutrient deficiencies have only reduced the burden of child undernutrition to a certain extent, indicating that there are other underlying determinants that need to be addressed. Aflatoxin exposure, which is also highly prevalent in developing countries, may be considered an aggravating factor for child undernutrition. Increasing evidence suggests that aflatoxin exposure can occur in any stage of life, including in utero through a trans-placental pathway and in early childhood (through contaminated weaning food and family food). Early life exposure to aflatoxin is associated with adverse effects on low birth weight, stunting, immune suppression, and the liver function damage. The mechanisms underlying impaired growth and aflatoxin exposure are still unclear but intestinal function damage, reduced immune function, and alteration in the insulin-like growth factor axis caused by the liver damage are the suggested hypotheses. Given the fact that both aflatoxin and child undernutrition are common in sub-Saharan Africa, effective interventions aimed at reducing undernutrition cannot be satisfactorily achieved until the interactive relationship between aflatoxin and child undernutrition is clearly understood, and an aflatoxin mitigation strategy takes effect in those vulnerable mothers and children.
Collapse
Affiliation(s)
- Sinead Watson
- a Institute for Global Food Safety, Queen's University Belfast , Belfast , United Kingdom
| | - Yun Yun Gong
- a Institute for Global Food Safety, Queen's University Belfast , Belfast , United Kingdom
| | | |
Collapse
|
30
|
Imdad A, Mayo‐Wilson E, Herzer K, Bhutta ZA, Cochrane Developmental, Psychosocial and Learning Problems Group. Vitamin A supplementation for preventing morbidity and mortality in children from six months to five years of age. Cochrane Database Syst Rev 2017; 3:CD008524. [PMID: 28282701 PMCID: PMC6464706 DOI: 10.1002/14651858.cd008524.pub3] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Vitamin A deficiency (VAD) is a major public health problem in low- and middle-income countries, affecting 190 million children under five years of age and leading to many adverse health consequences, including death. Based on prior evidence and a previous version of this review, the World Health Organization has continued to recommend vitamin A supplementation for children aged 6 to 59 months. There are new data available from recently published randomised trials since the previous publication of this review in 2010, and this update incorporates this information and reviews the evidence. OBJECTIVES To assess the effects of vitamin A supplementation (VAS) for preventing morbidity and mortality in children aged six months to five years. SEARCH METHODS In March 2016 we searched CENTRAL, Ovid MEDLINE, Embase, six other databases, and two trials registers. We also checked reference lists and contacted relevant organisations and researchers to identify additional studies. SELECTION CRITERIA Randomised controlled trials (RCTs) and cluster-RCTs evaluating the effect of synthetic VAS in children aged six months to five years living in the community. We excluded studies involving children in hospital and children with disease or infection. We also excluded studies evaluating the effects of food fortification, consumption of vitamin A rich foods, or beta-carotene supplementation. DATA COLLECTION AND ANALYSIS For this update, two reviewers independently assessed studies for inclusion and abstracted data, resolving discrepancies by discussion. We performed meta-analyses for outcomes, including all-cause and cause-specific mortality, disease, vision, and side effects. We used the GRADE approach to assess the quality of the evidence. MAIN RESULTS We identified 47 studies (4 of which are new to this review), involving approximately 1,223,856 children. Studies took place in 19 countries: 30 (63%) in Asia, 16 of these in India; 8 (17%) in Africa; 7 (15%) in Latin America, and 2 (4%) in Australia. About one-third of the studies were in urban/periurban settings, and half were in rural settings; the remaining studies did not clearly report settings. Most of the studies included equal numbers of girls and boys and lasted about a year. The included studies were at variable overall risk of bias; however, evidence for the primary outcome was at low risk of bias. A meta-analysis for all-cause mortality included 19 trials (1,202,382 children). At longest follow-up, there was a 12% observed reduction in the risk of all-cause mortality for vitamin A compared with control using a fixed-effect model (risk ratio (RR) 0.88, 95% confidence interval (CI) 0.83 to 0.93; high-quality evidence). This result was sensitive to choice of model, and a random-effects meta-analysis showed a different summary estimate (24% reduction: RR 0.76, 95% CI 0.66 to 0.88); however, the confidence intervals overlapped with that of the fixed-effect model. Nine trials reported mortality due to diarrhoea and showed a 12% overall reduction for VAS (RR 0.88, 95% CI 0.79 to 0.98; 1,098,538 participants; high-quality evidence). There was no significant effect for VAS on mortality due to measles, respiratory disease, and meningitis. VAS reduced incidence of diarrhoea (RR 0.85, 95% CI 0.82 to 0.87; 15 studies; 77,946 participants; low-quality evidence) and measles (RR 0.50, 95% CI 0.37 to 0.67; 6 studies; 19,566 participants; moderate-quality evidence). However, there was no significant effect on incidence of respiratory disease or hospitalisations due to diarrhoea or pneumonia. There was an increased risk of vomiting within the first 48 hours of VAS (RR 1.97, 95% CI 1.44 to 2.69; 4 studies; 10,541 participants; moderate-quality evidence). AUTHORS' CONCLUSIONS Vitamin A supplementation is associated with a clinically meaningful reduction in morbidity and mortality in children. Therefore, we suggest maintaining the policy of universal supplementation for children under five years of age in populations at risk of VAD. Further placebo-controlled trials of VAS in children between six months and five years of age would not change the conclusions of this review, although studies that compare different doses and delivery mechanisms are needed. In populations with documented vitamin A deficiency, it would be unethical to conduct placebo-controlled trials.
Collapse
Affiliation(s)
- Aamer Imdad
- Vanderbilt University School of MedicineDepartment of Pediatrics, D. Brent Polk Division of Gastroenterology, Hepatology and NutritionNashvilleTNUSA37212
| | - Evan Mayo‐Wilson
- Johns Hopkins University Bloomberg School of Public HealthDepartment of Epidemiology615 North Wolfe StreetBaltimoreMarylandUSA21205
| | - Kurt Herzer
- Johns Hopkins School of MedicineTower 711600 North Wolfe St.BaltimoreMDUSA21287
| | - Zulfiqar A Bhutta
- Hospital for Sick ChildrenCentre for Global Child HealthTorontoONCanadaM5G A04
| | | |
Collapse
|
31
|
Lower levels of vitamin A are associated with increased gastrointestinal graft-versus-host disease in children. Blood 2017; 129:2801-2807. [PMID: 28279965 DOI: 10.1182/blood-2017-02-765826] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/03/2017] [Indexed: 12/15/2022] Open
Abstract
Vitamin A promotes development of mucosal tolerance and enhances differentiation of regulatory T cells. Vitamin A deficiency impairs epithelial integrity, increasing intestinal permeability. We hypothesized that higher vitamin A levels would reduce the risk of graft-versus-host disease (GVHD) through reduced gastrointestinal (GI) permeability, reduced mucosal injury, and reduced lymphocyte homing to the gut. We tested this hypothesis in a cohort study of 114 consecutive patients undergoing allogeneic stem cell transplant. Free vitamin A levels were measured in plasma at day 30 posttransplant. GI GVHD was increased in patients with vitamin A levels below the median (38% vs 12.4% at 100 days, P = .0008), as was treatment-related mortality (17.7% vs 7.4% at 1 year, P = .03). Bloodstream infections were increased in patients with vitamin A levels below the median (24% vs 8% at 1 year, P = .03), supporting our hypothesis of increased intestinal permeability. The GI mucosal intestinal fatty acid-binding protein was decreased after transplant, confirming mucosal injury, but was not correlated with vitamin A levels, indicating that vitamin A did not protect against mucosal injury. Expression of the gut homing receptor CCR9 on T-effector memory cells 30 days after transplant was increased in children with vitamin A levels below the median (r = -0.34, P = .03). Taken together, these data support our hypothesis that low levels of vitamin A actively promote GI GVHD and are not simply a marker of poor nutritional status or a sicker patient. Vitamin A supplementation might improve transplant outcomes.
Collapse
|
32
|
Faubion WA, Camilleri M, Murray JA, Kelly P, Amadi B, Kosek MN, Enders F, Larson J, Grover M, Boe G, Dyer R, Singh R. Improving the detection of environmental enteric dysfunction: a lactulose, rhamnose assay of intestinal permeability in children aged under 5 years exposed to poor sanitation and hygiene. BMJ Glob Health 2016; 1:e000066. [PMID: 28588929 PMCID: PMC5321325 DOI: 10.1136/bmjgh-2016-000066] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 12/01/2022] Open
Abstract
Background Environmental enteric dysfunction (EED) is an asymptomatic intestinal disorder affecting populations living in conditions of poor sanitation and hygiene. The study tested intestinal barrier function in infants with EED. Methods We prospectively studied an advanced high-performance liquid chromatography mass spectrometry assay of urine collected after oral intake of the monosaccharide, L-rhamnose and the disaccharide, lactulose, in 112 children from three continents. Findings Compared to the US cohort (n=27), the cohorts of children from Peru (n=19) and Zambia (n=85) were older with evidence of growth impairment. The median (range) of age (months) was 8.0 (2.0 to 13.0), 27.0 (15.0 to 29.0) and 21.0 (12.0 to 36.0), respectively. The median (range) of height for age Z score was −0.1 (−1.8 to 2.4), −1.8 (−3.3 to −0.2) and −2.3 (−8.5 to 1.2), respectively. Among children with valid sugar data (n=22 USA, n=19 Peru, n=73 Zambia), there were no significant differences in the median rhamnose urine concentrations between the three groups. The median (range) lactulose concentration (µg/mL) was 6.78 (0.29 to 31.90), 47.60 (4.23 to 379.00) and 75.40 (0.67 to 873.00) in the US, Peruvian and Zambian cohorts, respectively (p<0.001). The lactulose/rhamnose ratio (LRR) was higher in cohorts from Peru (0.75, 0.15, 5.02) and Zambia (2.26, 0.08, 14.48) compared to the US (0.14, 0.06, 1.00) cohort (p<0.001). In a multivariate effect modification model, higher weight-for-age z scores were associated with lower post-dose lactulose when rhamnose excretion was constant (p=0.003). Conclusions This non-invasive two saccharide permeability protocol measures changes in intestinal permeability in children with EED and permits the identification of individuals for interventional trials.
Collapse
Affiliation(s)
- W A Faubion
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - M Camilleri
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - J A Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - P Kelly
- Queen Mary, University of London, University of Zambia School of Medicine, London, UK
| | - B Amadi
- Queen Mary, University of London, University of Zambia School of Medicine, London, UK
| | - M N Kosek
- Department of International Health, John's Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - F Enders
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - J Larson
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - G Boe
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - R Dyer
- Immunochemical Core Laboratory, Mayo Clinic, Rochester, Minnesota, USA
| | - R Singh
- Immunochemical Core Laboratory, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
33
|
Abstract
BACKGROUND Environmental enteric dysfunction (EED) refers to an incompletely defined syndrome of inflammation, reduced absorptive capacity, and reduced barrier function in the small intestine. It is widespread among children and adults in low- and middle-income countries. Understanding of EED and its possible consequences for health is currently limited. OBJECTIVE A narrative review of the current understanding of EED: epidemiology, pathogenesis, therapies, and relevance to child health. METHODS Searches for key papers and ongoing trials were conducted using PUBMED 1966-June 2014; ClinicalTrials.gov; the WHO Clinical Trials Registry; the Cochrane Library; hand searches of the references of retrieved literature; discussions with experts; and personal experience from the field. RESULTS EED is established during infancy and is associated with poor sanitation, certain gut infections, and micronutrient deficiencies. Helicobacter pylori infection, small intestinal bacterial overgrowth (SIBO), abnormal gut microbiota, undernutrition, and toxins may all play a role. EED is usually asymptomatic, but it is important due to its association with stunting. Diagnosis is frequently by the dual sugar absorption test, although other biomarkers are emerging. EED may partly explain the reduced efficacy of oral vaccines in low- and middle-income countries and the increased risk of serious infection seen in children with undernutrition. CONCLUSIONS Despite its potentially significant impacts, it is currently unclear exactly what causes EED and how it can be treated or prevented. Ongoing trials involve nutritional supplements, water and sanitation interventions, and immunomodulators. Further research is needed to better understand this condition, which is of likely crucial importance for child health and development in low- and middle-income settings.
Collapse
|
34
|
Patel S, Vajdy M. Induction of cellular and molecular immunomodulatory pathways by vitamin A and flavonoids. Expert Opin Biol Ther 2015; 15:1411-28. [PMID: 26185959 DOI: 10.1517/14712598.2015.1066331] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION A detailed study of reports on the immunomodulatory properties of vitamin A and select flavonoids may pave the way for using these natural compounds or compounds with similar structures in novel drug and vaccine designs against infectious and autoimmune diseases and cancers. AREAS COVERED Intracellular transduction pathways, cellular differentiation and functional immunomodulatory responses have been reviewed. The reported studies encompass in vitro, in vivo preclinical and clinical studies that address the role of vitamin A and select flavonoids in induction of innate and adaptive B- and T-cell responses, including TH1, TH2 and regulatory T cells (Treg). EXPERT OPINION While the immunomodulatory role of vitamin A, and related compounds, is well-established in many preclinical studies, its role in humans has begun to gain wider acceptance. In contrast, the role of flavonoids is mostly controversial in clinical trials, due to the diversity of the various classes of these compounds, and possibly due to the purity and the selected doses of the compounds. However, current preclinical and clinical studies warrant further detailed studies of these promising immunomodulatory compounds.
Collapse
Affiliation(s)
- Sapna Patel
- a EpitoGenesis, Inc. , 1392 Storrs Rd Unit 4213, ATL Building, Rm 101, Storrs, CT 06269, USA
| | - Michael Vajdy
- a EpitoGenesis, Inc. , 1392 Storrs Rd Unit 4213, ATL Building, Rm 101, Storrs, CT 06269, USA
| |
Collapse
|
35
|
Sá ACC, Gómez MM, Lima IFN, Quetz JS, Havt A, Oriá RB, Lima AA, Leite JPG. Group a rotavirus and norovirus genotypes circulating in the northeastern Brazil in the post-monovalent vaccination era. J Med Virol 2015; 87:1480-90. [DOI: 10.1002/jmv.24144] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Ana Caroline C. Sá
- Laboratory of Comparative and Environmental Virology; Oswaldo Cruz Institute- Fiocruz; Rio de Janeiro Brazil
| | - Mariela M. Gómez
- Laboratory of Comparative and Environmental Virology; Oswaldo Cruz Institute- Fiocruz; Rio de Janeiro Brazil
| | - Ila Fernanda N. Lima
- Institute of Biomedicine for Brazilian Semi-Arid & Clinical Research Unit; Federal University of Ceara; Fortaleza Brazil
| | - Josiane S. Quetz
- Institute of Biomedicine for Brazilian Semi-Arid & Clinical Research Unit; Federal University of Ceara; Fortaleza Brazil
| | - Alexandre Havt
- Institute of Biomedicine for Brazilian Semi-Arid & Clinical Research Unit; Federal University of Ceara; Fortaleza Brazil
| | - Reinaldo B. Oriá
- Institute of Biomedicine for Brazilian Semi-Arid & Clinical Research Unit; Federal University of Ceara; Fortaleza Brazil
| | - Aldo A. Lima
- Institute of Biomedicine for Brazilian Semi-Arid & Clinical Research Unit; Federal University of Ceara; Fortaleza Brazil
| | - José Paulo G. Leite
- Laboratory of Comparative and Environmental Virology; Oswaldo Cruz Institute- Fiocruz; Rio de Janeiro Brazil
| |
Collapse
|
36
|
Raiten DJ, Sakr Ashour FA, Ross AC, Meydani SN, Dawson HD, Stephensen CB, Brabin BJ, Suchdev PS, van Ommen B. Inflammation and Nutritional Science for Programs/Policies and Interpretation of Research Evidence (INSPIRE). J Nutr 2015; 145:1039S-1108S. [PMID: 25833893 PMCID: PMC4448820 DOI: 10.3945/jn.114.194571] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/08/2014] [Accepted: 10/29/2014] [Indexed: 02/06/2023] Open
Abstract
An increasing recognition has emerged of the complexities of the global health agenda—specifically, the collision of infections and noncommunicable diseases and the dual burden of over- and undernutrition. Of particular practical concern are both 1) the need for a better understanding of the bidirectional relations between nutritional status and the development and function of the immune and inflammatory response and 2) the specific impact of the inflammatory response on the selection, use, and interpretation of nutrient biomarkers. The goal of the Inflammation and Nutritional Science for Programs/Policies and Interpretation of Research Evidence (INSPIRE) is to provide guidance for those users represented by the global food and nutrition enterprise. These include researchers (bench and clinical), clinicians providing care/treatment, those developing and evaluating programs/interventions at scale, and those responsible for generating evidence-based policy. The INSPIRE process included convening 5 thematic working groups (WGs) charged with developing summary reports around the following issues: 1) basic overview of the interactions between nutrition, immune function, and the inflammatory response; 2) examination of the evidence regarding the impact of nutrition on immune function and inflammation; 3) evaluation of the impact of inflammation and clinical conditions (acute and chronic) on nutrition; 4) examination of existing and potential new approaches to account for the impact of inflammation on biomarker interpretation and use; and 5) the presentation of new approaches to the study of these relations. Each WG was tasked with synthesizing a summary of the evidence for each of these topics and delineating the remaining gaps in our knowledge. This review consists of a summary of the INSPIRE workshop and the WG deliberations.
Collapse
Affiliation(s)
- Daniel J Raiten
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD;
| | - Fayrouz A Sakr Ashour
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD
| | - A Catharine Ross
- Departments of Nutritional Sciences and Veterinary and Biomedical Science and Center for Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, PA
| | - Simin N Meydani
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Harry D Dawson
- USDA-Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, MD
| | - Charles B Stephensen
- Agricultural Research Service, Western Human Nutrition Research Center, USDA, Davis, CA
| | - Bernard J Brabin
- Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, United Kingdom; Global Child Health Group, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Parminder S Suchdev
- Department of Pediatrics and Global Health, Emory University, Atlanta, GA; and
| | | |
Collapse
|
37
|
Talachian E, Bidari A, Noorbakhsh S, Tabatabaei A, Salari F. Serum levels of vitamins A and D, and zinc in children with acute diarrhea: A cross-sectional study. Med J Islam Repub Iran 2015; 29:207. [PMID: 26157725 PMCID: PMC4476211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/11/2015] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Diarrhea is a leading cause of mortality and morbidity during the early life period especially in developing countries. Micronutrients deficiencies have been proposed either as a risk factor or a consequence of diarrhea. Association studies highlight the relation of vitamins and minerals' deficiencies with acute diarrhea. In this regard we aimed to evaluate the status of vitamins A and D, and zinc serum levels in children with acute diarrhea. METHODS In this cross sectional study performed in a referral teaching hospital, we measured and compared baseline vitamin A, 25-hydroxy vitamin D (25(OH)D), and zinc serum levels in 25 children admitted with acute diarrhea and 25 other children who were admitted for undergoing elective surgeries. RESULTS 25-(OH)D levels were significantly lower in the diarrhea group (p=0.03). We were unable to demonstrate a significant difference in the levels of vitamin A and zinc between the case and control groups (p= 0.14 and p=0.07, respectively). CONCLUSION We observed lower serum 25(OH)D levels in children with acute diarrhea. Whether this finding indicates a premorbid risk factor or simply a consequence of diarrhea needs further studies. Regardless of the cause and effect relationship, supplementation with vitamin D in acute diarrhea remains as a plausible consideration.
Collapse
Affiliation(s)
- Elham Talachian
- MD, Associate Professor, Pediatric Gastroenterology division Ali-Asghar Children's Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Ali Bidari
- MD, Associate Professor, Emergency Department, Hazrat-e-Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Samileh Noorbakhsh
- MD, Associate Professor, Pediatric Infectious Disease Research Center, Hazrat-e-Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Azardokht Tabatabaei
- Pediatric Infectious Disease Research Center, Hazrat-e-Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
38
|
Denno DM, VanBuskirk K, Nelson ZC, Musser CA, Hay Burgess DC, Tarr PI. Use of the lactulose to mannitol ratio to evaluate childhood environmental enteric dysfunction: a systematic review. Clin Infect Dis 2015; 59 Suppl 4:S213-9. [PMID: 25305289 DOI: 10.1093/cid/ciu541] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Childhood gut dysfunction (enteropathy) is common in resource-poor environments. Stunting is its presumed major consequence. Identification of biomarkers of gut dysfunction could identify the presence of, and, ideally, assess interventions for, enteropathy. Classically, enteropathy has been identified histopathologically. However, less invasive assays may be more sensitive for detecting earlier perturbations reflecting specific functional derangements. The most commonly used test has been the urinary lactulose to mannitol ratio (L:M), which primarily assesses gut leakiness, and which also measures absorption. We systematically reviewed the L:M literature published from 2000 to 2010 pertinent to children in developing country settings, and identified 25 relevant publications representing heterogeneous studies. We conclude that the L:M test has many attributes, including reflecting 2 physiologic processes (absorption and permeability) and likely correlation with growth failure consequent to child gut dysfunction. However, improved test technical performance, data reporting, and correlation with host phenotypes are needed to maximize the utility of this test.
Collapse
Affiliation(s)
- Donna M Denno
- Departments of Pediatrics and Global Health, University of Washington, Seattle
| | | | | | | | | | - Phillip I Tarr
- Departments of Pediatrics and Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
39
|
Intestinal permeability--a new target for disease prevention and therapy. BMC Gastroenterol 2014; 14:189. [PMID: 25407511 PMCID: PMC4253991 DOI: 10.1186/s12876-014-0189-7] [Citation(s) in RCA: 1199] [Impact Index Per Article: 109.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 10/17/2014] [Indexed: 02/06/2023] Open
Abstract
Data are accumulating that emphasize the important role of the intestinal barrier and intestinal permeability for health and disease. However, these terms are poorly defined, their assessment is a matter of debate, and their clinical significance is not clearly established. In the present review, current knowledge on mucosal barrier and its role in disease prevention and therapy is summarized. First, the relevant terms 'intestinal barrier' and 'intestinal permeability' are defined. Secondly, the key element of the intestinal barrier affecting permeability are described. This barrier represents a huge mucosal surface, where billions of bacteria face the largest immune system of our body. On the one hand, an intact intestinal barrier protects the human organism against invasion of microorganisms and toxins, on the other hand, this barrier must be open to absorb essential fluids and nutrients. Such opposing goals are achieved by a complex anatomical and functional structure the intestinal barrier consists of, the functional status of which is described by 'intestinal permeability'. Third, the regulation of intestinal permeability by diet and bacteria is depicted. In particular, potential barrier disruptors such as hypoperfusion of the gut, infections and toxins, but also selected over-dosed nutrients, drugs, and other lifestyle factors have to be considered. In the fourth part, the means to assess intestinal permeability are presented and critically discussed. The means vary enormously and probably assess different functional components of the barrier. The barrier assessments are further hindered by the natural variability of this functional entity depending on species and genes as well as on diet and other environmental factors. In the final part, we discuss selected diseases associated with increased intestinal permeability such as critically illness, inflammatory bowel diseases, celiac disease, food allergy, irritable bowel syndrome, and--more recently recognized--obesity and metabolic diseases. All these diseases are characterized by inflammation that might be triggered by the translocation of luminal components into the host. In summary, intestinal permeability, which is a feature of intestinal barrier function, is increasingly recognized as being of relevance for health and disease, and therefore, this topic warrants more attention.
Collapse
|
40
|
Chikhungu LC, Madise NJ. Seasonal variation of child under nutrition in Malawi: is seasonal food availability an important factor? Findings from a national level cross-sectional study. BMC Public Health 2014; 14:1146. [PMID: 25373873 PMCID: PMC4246561 DOI: 10.1186/1471-2458-14-1146] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 10/20/2014] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Child under nutrition is an underlying factor in millions of under-five child deaths and poor cognitive development worldwide. Whilst many studies have examined the levels and factors associated with child under nutrition in different settings, very little has been written on the variation of child under nutrition across seasons. This study explored seasonal food availability and child morbidity as influences of child nutritional status in Malawi. METHODS The study used the 2004 Malawi Integrated Household Survey data. Graphical analysis of the variation of child under nutrition, child morbidity and food availability across the 12 months of the year was undertaken to display seasonal patterns over the year. Multivariate analysis was used to explore the importance of season after controlling for well-established factors that are known to influence a child's nutritional status. RESULTS A surprising finding is that children were less likely to be stunted and less likely to be underweight in the lean cropping season (September to February) compared to the post-harvest season (March to August). The odds ratio for stunting were 0.80 (0.72, 0.90) and the odds ratio for underweight were 0.77 (0.66, 0.90). The season when child under nutrition levels were high coincided with the period of high child morbidity in line with previous studies. Children that were ill in the two weeks prior to survey were more likely to be underweight compared to children that were not ill 1.18 (1.01, 1.38). CONCLUSION In Malawi child nutritional status varies across seasons and follows a seasonal pattern of childhood illness but not that of household food availability.
Collapse
Affiliation(s)
- Lana Clara Chikhungu
- Division of Social Statistics and Demography, Centre for Global Health, Population, Poverty and Policy, Faculty of Social and Human Sciences, University of Southampton, Building 58, Social Sciences Academic Unit, Highfield, Southampton SO17 1BJ, UK.
| | | |
Collapse
|
41
|
Al-Mekhlafi HM, Anuar TS, Al-Zabedi EM, Al-Maktari MT, Mahdy MAK, Ahmed A, Sallam AA, Abdullah WA, Moktar N, Surin J. Does vitamin A supplementation protect schoolchildren from acquiring soil-transmitted helminthiasis? A randomized controlled trial. Parasit Vectors 2014; 7:367. [PMID: 25127885 PMCID: PMC4141119 DOI: 10.1186/1756-3305-7-367] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 07/29/2014] [Indexed: 11/27/2022] Open
Abstract
Background Despite the intensive global efforts to control intestinal parasitic infections, the prevalence of soil-transmitted helminth (STH) infections is still very high in many developing countries particularly among children in rural areas. Methods A randomized, double-blind, placebo-controlled trial was conducted on 250 Aboriginal schoolchildren in Malaysia to investigate the effects of a single high-dose of vitamin A supplementation (200 000 IU) on STH reinfection. The effect of the supplement was assessed at 3 and 6 months after receiving interventions; after a complete 3-day deworming course of 400 mg/daily of albendazole tablets. Results Almost all children (98.6%) were infected with at least one STH species. The overall prevalence of ascariasis, trichuriasis and hookworm infection was 67.8%, 95.5% and 13.4%, respectively. Reinfection rates of Ascaris, Trichuris and hookworm were high; at 6 months, assessment reached 80% of the prevalence reported before treatment. There were no significant differences in the reinfection rates and intensities of STH between vitamin A supplemented-children and those who received placebo at 3 and 6 months (p > 0.05). Conclusions Vitamin A supplementation showed no protective effect against STH reinfection and this could be due to the high endemicity of STH in this community. Long-term interventions to reduce poverty will help significantly in reducing this continuing problem and there is no doubt that reducing intestinal parasitic infection would have a positive impact on the health, nutrition and education of these children. Trial registration This trial was registered at clinicaltrials.gov as NCT00936091.
Collapse
Affiliation(s)
- Hesham M Al-Mekhlafi
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Queiroz DD, Paiva ADA, Gama JSDFA, Lima ZN, Pedraza DF. Índices antropométricos e retinolemia em crianças menores de cinco anos do e Estado da Paraíba. REV NUTR 2013. [DOI: 10.1590/s1415-52732013000500007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJETIVO: Avaliar a relação entre os índices antropométricos e os níveis de retinol sérico em crianças menores de cinco anos do Estado da Paraíba. MÉTODOS: Estudo de corte transversal, de base populacional, envolvendo 1 205 crianças. O estado nutricional de vitamina A foi avaliado pelas concentrações séricas de retinol. O estado nutricional antropométrico foi avaliado utilizando-se os índices peso/altura e altura/idade. RESULTADOS: Crianças na faixa etária de 6 a 12 meses com deficit de altura apresentaram concentrações de retinol sérico significantemente inferiores quando comparadas às crianças sem deficit de altura (p=0,02). CONCLUSÃO: A associação observada entre deficit de altura e menores níveis de retinol sérico em crianças de menor faixa etária mostra a maior vulnerabilidade desse grupo para sofrer deficits vitamínicos e de crescimento.
Collapse
|
43
|
Lander RL, Lander AG, Houghton L, Williams SM, Costa-Ribeiro H, Barreto DL, Mattos AP, Gibson RS. Factors influencing growth and intestinal parasitic infections in preschoolers attending philanthropic daycare centers in Salvador, Northeast Region of Brazil. CAD SAUDE PUBLICA 2013; 28:2177-88. [PMID: 23147959 DOI: 10.1590/s0102-311x2012001100017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Accepted: 07/02/2012] [Indexed: 11/22/2022] Open
Abstract
Poor growth and intestinal parasitic infections are widespread in disadvantaged urban children. This cross-sectional study assessed factors influencing poor growth and intestinal parasites in 376 children aged three to six years in daycare centers in Salvador, in the Northeast Region of Brazil. Data was obtained from seven daycare centers on child weight, height, socio-economic status, health and intestinal parasites in stool samples. Prevalence of moderate underweight (< -1SD > -2SD), wasting and stunting was 12%, 16% and 6% respectively. Socioeconomic status, birth order, and maternal weight were predictors of poor anthropometric status. Almost 30% of children were infected with more than one intestinal parasite. Helminths (17.8%), notably Trichuris trichiura (12%) and Ascaris lumbricoides (10.5%), and protozoan Giardia duodenalis (13%) were the most common types of parasites detected. One percent of children had hookworm and Cryptosporidium sp. and 25% had non-pathogenic protozoan cysts. Boys from families with very low socio-economic status had lower linear growth and presented a greater risk of helminth infection. Deworming is considered an alternative for reducing the prevalence of intestinal parasitic infections in this age group.
Collapse
Affiliation(s)
- Rebecca L Lander
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Multiple micronutrient deficiencies are still common worldwide and often occur at an early age, negatively affecting both physical and cognitive development. Even though specific effects of a variety of different micronutrients have been studied extensively, it is understood that a combination of multiple micronutrients may be more beneficial in most instances, as deficiencies usually do not occur in isolation. The aim of this review was to summarize the most recent evidence of the effects of micronutrient interventions on growth, mental, and physical performance as well as morbidity in children. RECENT FINDINGS For growth, cognitive or motor function, and morbidity, it appears that providing sufficient amounts of micronutrients especially to the most vulnerable and deficient groups of children can make a difference, but also that the intervention has to be planned carefully. However, findings are still variable, more particularly with reference to morbidity. Two reports of increased diagnoses of infection with micronutrient supplementation are also of concern. SUMMARY There are still difficulties in coming to a universal conclusion about benefit in all populations, because reports vary in study design, population, and so on. Although micronutrients have a role in depleted groups, it is also important to not only consider the group of children targeted, but also the regime of administration.
Collapse
Affiliation(s)
- Anura V Kurpad
- Division of Nutrition, St John's Research Institute, St John's National Academy of Health Sciences, Bangalore, India.
| | | | | |
Collapse
|
45
|
Lima AAM, Kvalsund MP, Souza PPED, Figueiredo ÍL, Soares AM, Mota RMS, Lima NL, Pinkerton RC, Patrick PP, Guerrant RL, Oriá RB. Zinc, vitamin A, and glutamine supplementation in Brazilian shantytown children at risk for diarrhea results in sex-specific improvements in verbal learning. Clinics (Sao Paulo) 2013; 68:351-8. [PMID: 23644855 PMCID: PMC3611743 DOI: 10.6061/clinics/2013(03)oa11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 11/19/2012] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE To identify the impact of supplemental zinc, vitamin A, and glutamine, alone or in combination, on long-term cognitive outcomes among Brazilian shantytown children with low median height-for-age z-scores. METHODS A randomized, double-blind, placebo-controlled trial was conducted in children aged three months to nine years old from the urban shanty compound community of Fortaleza, Brazil. Demographic and anthropometric information was assessed. The random treatment groups available for cognitive testing (total of 167 children) were: (1) placebo, n = 25; (2) glutamine, n = 23; (3) zinc, n = 18; (4) vitamin A, n = 19; (5) glutamine+zinc, n = 20; (6) glutamine+vitamin A, n = 21; (7) zinc+vitamin A, n = 23; and (8) glutamine+zinc+vitamin A, n = 18. Neuropsychological tests were administered for the cognitive domains of non-verbal intelligence and abstraction, psychomotor speed, verbal memory and recall ability, and semantic and phonetic verbal fluency. Statistical analyses were performed using SPSS, version 16.0. ClinicalTrials.gov: NCT00133406. RESULTS Girls receiving a combination of glutamine, zinc, and vitamin A had higher mean age-adjusted verbal learning scores than girls receiving only placebo (9.5 versus 6.4, p = 0.007) and girls receiving zinc+vitamin A (9.5 versus 6.5, p = 0.006). Similar group differences were not found between male study children. CONCLUSIONS The findings suggest that combination therapy offers a sex-specific advantage on tests of verbal learning, similar to that seen among female patients following traumatic brain injury.
Collapse
Affiliation(s)
- Aldo A M Lima
- Department of Physiology and Pharmacology, Clinical Research Unit and Institute of Biomedicine/Center for Global Health, School of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
DeBoer MD, Lima AAM, Oría RB, Scharf RJ, Moore SR, Luna MA, Guerrant RL. Early childhood growth failure and the developmental origins of adult disease: do enteric infections and malnutrition increase risk for the metabolic syndrome? Nutr Rev 2012; 70:642-53. [PMID: 23110643 DOI: 10.1111/j.1753-4887.2012.00543.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hypotheses regarding the developmental origins of health and disease postulate that developing fetuses - and potentially young children - undergo adaptive epigenetic changes that have longstanding effects on metabolism and other processes. Ongoing research explores whether these adaptations occur during early life following early childhood malnutrition. In the developing world, there remains a high degree of nutritional stunting, defined as linear growth failure caused by inadequate caloric intake, which may be exacerbated by inflammation from ongoing infections. In areas with poor sanitation, children experience vicious cycles of enteric infections and malnutrition, resulting in poor nutrient absorption as a result of changes in the intestinal mucosa, now termed "environmental enteropathy." Emerging evidence links early childhood diarrhea and/or growth failure with an increased occurrence of risk factors for cardiovascular disease in later life, including dyslipidemia, hypertension, and glucose intolerance. The mechanisms for these associations remain poorly understood and may relate to epigenetic responses to poor nutrition, increased inflammation, or both. Given the increased incidence of cardiovascular disease in developing areas of the world, associations between childhood malnutrition, early-life infections, and the increased occurrence of risk factors for cardiovascular disease underscore further reasons to improve nutrition and infection-related outcomes for young children worldwide.
Collapse
Affiliation(s)
- Mark D DeBoer
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Virginia, Charlottesville, VA, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Mitter SS, Oriá RB, Kvalsund MP, Pamplona P, Joventino ES, Mota RMS, Gonçalves DC, Patrick PD, Guerrant RL, Lima AAM. Apolipoprotein E4 influences growth and cognitive responses to micronutrient supplementation in shantytown children from northeast Brazil. Clinics (Sao Paulo) 2012; 67:11-8. [PMID: 22249475 PMCID: PMC3248595 DOI: 10.6061/clinics/2012(01)03] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 09/12/2011] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Apolipoprotein E4 may benefit children during early periods of life when the body is challenged by infection and nutritional decline. We examined whether apolipoprotein E4 affects intestinal barrier function, improving short-term growth and long-term cognitive outcomes in Brazilian shantytown children. METHODS A total of 213 Brazilian shantytown children with below-median height-for-age z-scores (HAZ) received 200,000 IU of retinol (every four months), zinc (40 mg twice weekly), or both for one year, with half of each group receiving glutamine supplementation for 10 days. Height-for-age z-scores, weight-for-age z-scores, weight-for-height z-scores, and lactulose:mannitol ratios were assessed during the initial four months of treatment. An average of four years (range 1.4-6.6) later, the children underwent cognitive testing to evaluate non-verbal intelligence, coding, verbal fluency, verbal learning, and delayed verbal learning. Apolipoprotein E4 carriage was determined by PCR analysis for 144 children. RESULTS Thirty-seven children were apolipoprotein E4(+), with an allele frequency of 13.9%. Significant associations were found for vitamin A and glutamine with intestinal barrier function. Apolipoprotein E4(+) children receiving glutamine presented significant positive Pearson correlations between the change in height-for-age z-scores over four months and delayed verbal learning, along with correlated changes over the same period in weight-for-age z-scores and weight-for-height z-scores associated with non-verbal intelligence quotients. There was a significant correlation between vitamin A supplementation of apolipoprotein E4(+) children and improved delta lactulose/mannitol. Apolipoprotein E4(-) children, regardless of intervention, exhibited negative Pearson correlations between the change in lactulose-to-mannitol ratio over four months and verbal learning and non-verbal intelligence. CONCLUSIONS During development, apolipoprotein E4 may function concomitantly with gut-tropic nutrients to benefit immediate nutritional status, which can translate into better long-term cognitive outcomes.
Collapse
Affiliation(s)
- Sumeet S Mitter
- Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ueno PM, Oriá RB, Maier EA, Guedes M, de Azevedo OG, Wu D, Willson T, Hogan SP, Lima AAM, Guerrant RL, Polk DB, Denson LA, Moore SR. Alanyl-glutamine promotes intestinal epithelial cell homeostasis in vitro and in a murine model of weanling undernutrition. Am J Physiol Gastrointest Liver Physiol 2011; 301:G612-22. [PMID: 21799183 PMCID: PMC3191556 DOI: 10.1152/ajpgi.00531.2010] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Alanyl-glutamine (Ala-Gln) has recently been shown to enhance catch-up growth and gut integrity in undernourished children from Northeast Brazil. We hypothesized that the intestinal epithelial effects of Ala-Gln in malnourished weanling mice and mouse small intestinal epithelial (MSIE) cells would include modulation of barrier function, proliferation, and apoptosis. Dams of 10-day-old suckling C57BL/6 pups were randomized to a standard diet or an isocaloric Northeast Brazil "regional basic diet," moderately deficient in protein, fat, and minerals. Upon weaning to their dam's diet on day of life 21, pups were randomized to Ala-Gln solution or water. At 6 wk of age, mice were killed, and jejunal tissue was collected for morphology, immunohistochemistry, and Ussing chamber analysis of transmucosal resistance and permeability. Proliferation of MSIE cells in the presence or absence of Ala-Gln was measured by MTS and bromodeoxyuridine assays. MSIE apoptosis was assessed by annexin and 7-amino-actinomycin D staining. Pups of regional basic diet-fed dams exhibited failure to thrive. Jejunal specimens from undernourished weanlings showed decreased villous height and crypt depth, decreased transmucosal resistance, increased permeability to FITC-dextran, increased claudin-3 expression, and decreased epithelial proliferation and increased epithelial apoptosis (as measured by bromodeoxyuridine and cleaved caspase-3 staining, respectively). Undernourished weanlings supplemented with Ala-Gln showed improvements in weight velocity, villous height, crypt depth, transmucosal resistance, and epithelial proliferation/apoptosis compared with unsupplemented controls. Similarly, Ala-Gln increased proliferation and reduced apoptosis in MSIE cells. In summary, Ala-Gln promotes intestinal epithelial homeostasis in a mouse model of malnutrition-associated enteropathy, mimicking key features of the human disease.
Collapse
Affiliation(s)
- Priscilla M. Ueno
- 1Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | | | - Elizabeth A. Maier
- 1Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | - Marjorie Guedes
- 3Institute of Biomedicine, Federal University of Ceará, Ceará, Brazil;
| | | | - David Wu
- 4Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | - Tara Willson
- 1Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | - Simon P. Hogan
- 4Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | - Aldo A. M. Lima
- 3Institute of Biomedicine, Federal University of Ceará, Ceará, Brazil;
| | - Richard L. Guerrant
- 5Center for Global Health, University of Virginia, Charlottesville, Virginia; and
| | - D. Brent Polk
- 6Department of Pediatrics, University of Southern California, Los Angeles, California
| | - Lee A. Denson
- 1Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | - Sean R. Moore
- 1Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| |
Collapse
|
49
|
Mayo-Wilson E, Imdad A, Herzer K, Yakoob MY, Bhutta ZA. Vitamin A supplements for preventing mortality, illness, and blindness in children aged under 5: systematic review and meta-analysis. BMJ 2011; 343:d5094. [PMID: 21868478 PMCID: PMC3162042 DOI: 10.1136/bmj.d5094] [Citation(s) in RCA: 190] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/07/2011] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To determine if vitamin A supplementation is associated with reductions in mortality and morbidity in children aged 6 months to 5 years. DESIGN Systematic review and meta-analysis. Two reviewers independently assessed studies for inclusion. Data were double extracted; discrepancies were resolved by discussion. Meta-analyses were performed for mortality, illness, vision, and side effects. DATA SOURCES Cochrane Central Register of Controlled Trials (CENTRAL) in the Cochrane Library, Medline, Embase, Global Health, Latin American and Caribbean Health Sciences, metaRegister of Controlled Trials, and African Index Medicus. Databases were searched to April 2010 without restriction by language or publication status. ELIGIBILITY CRITERIA FOR SELECTING STUDIES Randomised trials of synthetic oral vitamin A supplements in children aged 6 months to 5 years. Studies of children with current illness (such as diarrhoea, measles, and HIV), studies of children in hospital, and studies of food fortification or β carotene were excluded. RESULTS 43 trials with about 215,633 children were included. Seventeen trials including 194,483 participants reported a 24% reduction in all cause mortality (rate ratio=0.76, 95% confidence interval 0.69 to 0.83). Seven trials reported a 28% reduction in mortality associated with diarrhoea (0.72, 0.57 to 0.91). Vitamin A supplementation was associated with a reduced incidence of diarrhoea (0.85, 0.82 to 0.87) and measles (0.50, 0.37 to 0.67) and a reduced prevalence of vision problems, including night blindness (0.32, 0.21 to 0.50) and xerophthalmia (0.31, 0.22 to 0.45). Three trials reported an increased risk of vomiting within the first 48 hours of supplementation (2.75, 1.81 to 4.19). CONCLUSIONS Vitamin A supplementation is associated with large reductions in mortality, morbidity, and vision problems in a range of settings, and these results cannot be explained by bias. Further placebo controlled trials of vitamin A supplementation in children between 6 and 59 months of age are not required. However, there is a need for further studies comparing different doses and delivery mechanisms (for example, fortification). Until other sources are available, vitamin A supplements should be given to all children at risk of deficiency, particularly in low and middle income countries.
Collapse
Affiliation(s)
- Evan Mayo-Wilson
- Centre for Evidence-Based Intervention, Department of Social Policy and Intervention, University of Oxford, UK
| | | | | | | | | |
Collapse
|
50
|
Imdad A, Herzer K, Mayo-Wilson E, Yakoob MY, Bhutta ZA. Vitamin A supplementation for preventing morbidity and mortality in children from 6 months to 5 years of age. Cochrane Database Syst Rev 2010:CD008524. [PMID: 21154399 DOI: 10.1002/14651858.cd008524.pub2] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Vitamin A deficiency (VAD) is a major public health problem in low and middle income countries affecting 190 million children under 5. VAD can lead to many adverse health consequences, including death. OBJECTIVES To evaluate the effect of vitamin A supplementation (VAS) for preventing morbidity and mortality in children aged 6 months to 5 years. SEARCH STRATEGY We searched the Cochrane Central Register of Controlled Trials (CENTRAL 2010 Issue 2), MEDLINE (1950 to April Week 2 2010), EMBASE (1980 to 2010 Week 16), Global Health (1973 to March 2010), Latin American and Caribbean Health Sciences (LILACS), metaRegister of Controlled Trials and African Index Medicus (27 April 2010). SELECTION CRITERIA Randomised controlled trials (RCTs) and cluster RCTs evaluating the effect of synthetic VAS in children aged 6 months to 5 years living in the community. We excluded studies concerned with children in hospital and children with disease or infection. We excluded studies evaluating the effects of food fortification, consumption of vitamin A rich foods or beta-carotene supplementation. DATA COLLECTION AND ANALYSIS Two review authors independently assessed studies for inclusion. Data were double abstracted and discrepancies were resolved by discussion. Meta-analyses were performed for outcomes including all-cause and cause-specific mortality, disease, vision, and side-effects. MAIN RESULTS 43 trials involving 215,633 children were included. A meta-analysis for all-cause mortality included 17 trials comprising 194,795 children with 3536 deaths in both groups. At follow-up, there was a 24% observed reduction in the risk of all-cause mortality for Vitamin A compared with Control (Relative risk (RR) = 0.76 [95% confidence interval (CI) 0.69, 0.83]). Seven trials reported diarrhoea mortality and a 28% overall reduction for VAS (RR = 0.72 [0.57, 0.91]). There was no significant effect of VAS on cause specific mortality of measles, respiratory disease and meningitis. VAS reduced incidence of diarrhoea (RR = 0.85 [0.82, 0.87]) and measles morbidity (RR = 0.50 [0.37, 0.67]); however, there was no significant effect on incidence of respiratory disease or hospitalisations due to diarrhoea or pneumonia. There was an increased risk of vomiting within the first 48 hours of VAS (RR = 2.75 [1.81, 4.19]). AUTHORS' CONCLUSIONS VAS is effective in reducing all-cause mortality by about 24% compared to no treatment. In our opinion, given the evidence that VAS causes considerable reduction in child mortality, further placebo-controlled trials of VAS in children between 6 months and 5 years of age are not required. There is a need for further studies comparing different doses and delivery mechanisms (for example, fortification).
Collapse
Affiliation(s)
- Aamer Imdad
- Division of Women and Child Health, Aga Khan University Hospital, Stadium Road, PO Box 3500, Karachi, Pakistan, 74800
| | | | | | | | | |
Collapse
|