1
|
Naftaly S, Pery T, Mhajne R, Ashkar A, Davidovich-Pinhas M, Zinger A. Harnessing the Potential of Human Breast Milk to Boost Intestinal Permeability for Nanoparticles and Macromolecules. J Control Release 2025; 379:768-785. [PMID: 39842727 DOI: 10.1016/j.jconrel.2025.01.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
The intricate interplay between human breast milk, nanoparticles, and macromolecules holds promise for innovative nutritional delivery strategies. Compared to bovine milk and infant formula, this study explores human breast milk's role in modulating intestinal permeability and its impact on nanoparticle and macromolecule transport. Comparative analysis with bovine milk and infant formula reveals significant elevations in permeability with human breast milk, accompanied by a decrease in transepithelial electrical resistance, suggesting enhanced paracellular transport. Mechanistically, human breast milk reduces Zonula occludens-1 levels, suggesting a regulatory role in intestinal barrier function. Through in vitro and ex vivo evaluations, we aim to understand better the mechanisms behind enhanced permeability and how human breast milk affects nanoparticle physicochemical properties, potentially modulating their behavior. Specifically, human breast milk improves the intestinal permeability of liposomes in a porcine intestinal model, with associated changes in the composition of milk proteins corona related to liposome charge. These findings underscore the unexploited potential of human breast milk in facilitating transport across the intestinal barrier, offering novel avenues for human nutritional delivery and therapeutic interventions.
Collapse
Affiliation(s)
- Si Naftaly
- Laboratory for Bioinspired Nano Engineering and Translational Therapeutics, Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Topaz Pery
- Laboratory for Bioinspired Nano Engineering and Translational Therapeutics, Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Rawan Mhajne
- Laboratory for Bioinspired Nano Engineering and Translational Therapeutics, Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Areen Ashkar
- Faculty of Biotechnology and Food Engineering, Technion, Israel
| | - Maya Davidovich-Pinhas
- Faculty of Biotechnology and Food Engineering, Technion, Israel; Russell-Berrie Nanotechnology Institute, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Assaf Zinger
- Laboratory for Bioinspired Nano Engineering and Translational Therapeutics, Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel; Russell-Berrie Nanotechnology Institute, Technion - Israel Institute of Technology, Haifa 3200003, Israel; Cardiovascular Sciences Department, Houston Methodist Academic Institute, Houston, TX 77030, United States; Neurosurgery Department, Houston Methodist Academic Institute, Houston, TX 77030, United States; Resnick Sustainability Center of Catalysis, Technion-Israel Institute of Technology, Haifa 3200003, Israel; Bruce and Ruth Rappaport Cancer Research Center, Technion-Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
2
|
Hildebrand F, Cukaci C, Schoeny H, Baumgartinger C, Stelzer B, Spedicato M, Frey T, Catani M, Schmetterer K, Frey R, Koellensperger G. Integrating the lactulose-mannitol test for intestinal permeability with untargeted metabolomics for drug monitoring through dual liquid chromatography-mass spectrometry. Anal Bioanal Chem 2025:10.1007/s00216-025-05790-7. [PMID: 40014070 DOI: 10.1007/s00216-025-05790-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/28/2025]
Abstract
In this study, we developed a customized high-resolution mass spectrometry metabolomics workflow integrating the dual sugar test employing lactulose and mannitol as test probes for intestinal permeability assessment with untargeted screening of small molecules. Urine samples were collected from patients with major depression and healthy controls as part of a clinical study at the psychiatric department. Using a dual injection/dual chromatography setup, the test probes were quantified by hydrophilic interaction liquid chromatography (HILIC) in a targeted assay, while drugs and their metabolites were profiled in an untargeted manner by reversed-phase separation. Rigorous method development and validation allowed for selective separation of sugar isomers and consequently accurate quantification of lactulose and mannitol in urine. Internal standardization with compound specific stable isotope-labeled standards enabled excellent analytical figures of merit such as high recoveries, precision (< 5%), and working range (5 orders of magnitude). Within one analytical run, intestinal permeability was assessed together with drugs and their metabolites, allowing to screen for confounding drugs and patient compliance to the therapeutic scheme.
Collapse
Affiliation(s)
- Felina Hildebrand
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090, Vienna, Austria
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Waehringer Str. 42, 1090, Vienna, Austria
| | - Cemre Cukaci
- Division of General Psychiatry, Department of Psychiatry and Psychotherapy, Medizinische Universität Wien/Medical University of Vienna, Vienna, Österreich
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medizinische Universität Wien/Medical University of Vienna, Vienna, Österreich
| | - Harald Schoeny
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090, Vienna, Austria
| | - Christoph Baumgartinger
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090, Vienna, Austria
| | - Bruno Stelzer
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090, Vienna, Austria
| | - Matteo Spedicato
- Department of Chemical, Pharmaceutical and Agricultural Sciences - DOCPAS, University of Ferrara, Via L.Borsari 46, 44121, Ferrara, Italy
| | - Tobias Frey
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Martina Catani
- Department of Chemical, Pharmaceutical and Agricultural Sciences - DOCPAS, University of Ferrara, Via L.Borsari 46, 44121, Ferrara, Italy
| | - Klaus Schmetterer
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Richard Frey
- Division of General Psychiatry, Department of Psychiatry and Psychotherapy, Medizinische Universität Wien/Medical University of Vienna, Vienna, Österreich
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medizinische Universität Wien/Medical University of Vienna, Vienna, Österreich
| | - Gunda Koellensperger
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090, Vienna, Austria.
- Vienna Metabolomics Center (VIME), University of Vienna, Althanstr. 14, 1090, Vienna, Austria.
| |
Collapse
|
3
|
Farella I, Fortunato M, Martinelli D, De Carlo C, Sparapano E, Stolfa S, Romanelli F, De Laurentiis V, Martinotti S, Capozzi L, Castellana S, Parisi A, Latorre G. Lactobacillus rhamnosus Sepsis in a Preterm Infant Following Probiotic Administration: Challenges in Diagnosis. Microorganisms 2025; 13:265. [PMID: 40005631 PMCID: PMC11857594 DOI: 10.3390/microorganisms13020265] [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: 01/10/2025] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Probiotic administration has become common practice in neonatal intensive care units (NICUs) to prevent necrotizing enterocolitis (NEC) and promote gut health in preterm infants. While probiotics are generally considered safe, rare cases of probiotic-related sepsis have been reported. We present a case of Lactobacillus rhamnosus sepsis in a preterm infant, highlighting the challenges involved in its diagnosis. The infant developed symptoms of sepsis on the 13th day of probiotic treatment. Laboratory analyses, including MALDI-TOF, BioFire BCID2 panel, and whole-genome sequencing (WGS), helped confirm the diagnosis and the presence of Lactobacillus rhamnosus. In this case, accurately identifying the Lactobacillus rhamnosus strain proved challenging, as initial analyses using the Vitek 2 system yielded incorrect identifications. This highlights the limitations of automated systems in distinguishing closely related species, reinforcing the need for advanced molecular techniques to achieve precise strain identification and confirm a probiotic-related infection. Given these diagnostic complexities, it is crucial for clinicians to maintain a high index of suspicion for probiotic-related infections in cases of unexplained sepsis, as this awareness can prompt further diagnostic investigations to ensure accurate pathogen identification. The infant responded to ampicillin therapy, showing clinical improvement within 10 days and was discharged in good health at 67 days of life. This case underscores the importance of advanced molecular diagnostic methods to confirm probiotic-related infections and highlights the need for caution in administering probiotics to vulnerable populations, such as preterm infants. Clinicians must maintain a high index of suspicion for probiotic-associated sepsis in unexplained cases of infection and tailor antibiotic therapy based on susceptibility profiles. These findings emphasize the need for rigorous monitoring, appropriate probiotic strain selection, and optimized safety protocols in NICUs to mitigate potential risks.
Collapse
Affiliation(s)
- Ilaria Farella
- Department of Medicine and Surgery, LUM University, Casamassima, 70010 Bari, Italy;
- Neonatology and Neonatal Intensive Care Unit, Ecclesiastical General Hospital F. Miulli, Acquaviva delle Fonti, 70021 Bari, Italy; (D.M.); (G.L.)
| | - Maria Fortunato
- Unit of Clinical Pathology and Microbiology, Miulli Regional General Hospital, Acquaviva delle Fonti, 70021 Bari, Italy;
| | - Domenico Martinelli
- Neonatology and Neonatal Intensive Care Unit, Ecclesiastical General Hospital F. Miulli, Acquaviva delle Fonti, 70021 Bari, Italy; (D.M.); (G.L.)
| | - Carmela De Carlo
- UOC Microbiology and Virology, Azienda Ospedaliera-Universitaria Policlinico of Bari, 70124 Bari, Italy; (C.D.C.); (E.S.); (S.S.); (F.R.); (V.D.L.)
| | - Eleonora Sparapano
- UOC Microbiology and Virology, Azienda Ospedaliera-Universitaria Policlinico of Bari, 70124 Bari, Italy; (C.D.C.); (E.S.); (S.S.); (F.R.); (V.D.L.)
| | - Stefania Stolfa
- UOC Microbiology and Virology, Azienda Ospedaliera-Universitaria Policlinico of Bari, 70124 Bari, Italy; (C.D.C.); (E.S.); (S.S.); (F.R.); (V.D.L.)
| | - Federica Romanelli
- UOC Microbiology and Virology, Azienda Ospedaliera-Universitaria Policlinico of Bari, 70124 Bari, Italy; (C.D.C.); (E.S.); (S.S.); (F.R.); (V.D.L.)
| | - Vittoriana De Laurentiis
- UOC Microbiology and Virology, Azienda Ospedaliera-Universitaria Policlinico of Bari, 70124 Bari, Italy; (C.D.C.); (E.S.); (S.S.); (F.R.); (V.D.L.)
| | - Stefano Martinotti
- Department of Medicine and Surgery, LUM University, Casamassima, 70010 Bari, Italy;
- Unit of Clinical Pathology and Microbiology, Miulli Regional General Hospital, Acquaviva delle Fonti, 70021 Bari, Italy;
| | - Loredana Capozzi
- Istituto Zooprofilattico Sperimentale della Puglia e della Basilicata, 71121 Foggia, Italy; (L.C.); (S.C.); (A.P.)
| | - Stefano Castellana
- Istituto Zooprofilattico Sperimentale della Puglia e della Basilicata, 71121 Foggia, Italy; (L.C.); (S.C.); (A.P.)
| | - Antonio Parisi
- Istituto Zooprofilattico Sperimentale della Puglia e della Basilicata, 71121 Foggia, Italy; (L.C.); (S.C.); (A.P.)
| | - Giuseppe Latorre
- Neonatology and Neonatal Intensive Care Unit, Ecclesiastical General Hospital F. Miulli, Acquaviva delle Fonti, 70021 Bari, Italy; (D.M.); (G.L.)
| |
Collapse
|
4
|
Colt S, Edielu A, Lewander D, Wu HW, Webb EL, Mawa PA, Nakyesige R, Ayebazibwe AGK, Friedman JF, Bustinduy AL. Associations of poor water, sanitation, and hygiene and parasite burden with markers of environmental enteric dysfunction in preschool-age children infected with Schistosoma mansoni in Uganda. Trop Med Int Health 2025; 30:14-21. [PMID: 39618064 PMCID: PMC11698645 DOI: 10.1111/tmi.14061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is an acquired subclinical condition of the small intestine with lasting health implications for nutritional status, linear growth and development among children. EED is characterised by structural and functional changes to the gut barrier. There are no standardised diagnostic criteria, however, a number of biomarkers have been evaluated to capture EED domains. While the causes of EED are not fully understood, risk factors include poor water, sanitation and hygiene conditions and exposure to enteric pathogens. Very few studies have evaluated the impact of schistosomiasis on EED despite repeated intestinal damage from parasite eggs passing across the gut barrier. METHODS In a cohort of 354 preschool-aged children aged 12-47 months with Schistosoma mansoni infection recruited from the Lake Albert region of Uganda, we assessed exposure to water, sanitation and hygiene conditions and measured markers from each EED domain: intestinal inflammation (faecal calprotectin), epithelial damage (serum intestinal fatty-acid binding-protein), increased permeability (urine lactulose to mannitol ratio and faecal alpha-1 antitrypsin) and microbial translocation (serum endotoxin core antibody). RESULTS In multivariable linear regression models, we found that children whose drinking water was sourced from Lake Albert had higher concentrations of intestinal fatty-acid binding-protein (β = 0.48, 95% CI 0.20-0.76, p < 0.001), and lack of toilet/latrine access was associated with higher concentrations of calprotectin (β = 0.48, 95% CI 0.18-0.78, p < 0.01). Higher schistosomiasis intensity (eggs per gram of stool) was associated with higher calprotectin (β = 0.10, 95% CI 0.02-0.17, p = 0.01), but not with other EED markers. CONCLUSIONS Few studies have investigated schistosomiasis-related morbidities in very young children infected with schistosomiasis. Our findings from Uganda show that poor water, sanitation and hygiene conditions and heavier schistosomiasis burden are associated with intestinal inflammation and damage, contributing to EED. Improved treatment coverage for preschool-aged children infected with schistosomiasis may reduce the burden from EED and associated long-term morbidities.
Collapse
Affiliation(s)
- Susannah Colt
- Center for International Health Research, Rhode Island Hospital, Providence, RI, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Andrew Edielu
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | - David Lewander
- Global Health Program, Boston Children’s Hospital, Boston, MA, USA
| | - Hannah W. Wu
- Center for International Health Research, Rhode Island Hospital, Providence, RI, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Emily L. Webb
- Medical Research Council International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Patrice A. Mawa
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Racheal Nakyesige
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - A. Gloria K. Ayebazibwe
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Jennifer F. Friedman
- Center for International Health Research, Rhode Island Hospital, Providence, RI, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Amaya L. Bustinduy
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
5
|
Roskes L, Chamzas A, Ma B, Medina AE, Gopalakrishnan M, Viscardi RM, Sundararajan S. Early human milk feeding: Relationship to intestinal barrier maturation and postnatal growth. Pediatr Res 2024:10.1038/s41390-024-03622-5. [PMID: 39397156 DOI: 10.1038/s41390-024-03622-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/15/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024]
Abstract
OBJECTIVES Early exposure to mother's own milk (MOM) promotes intestinal barrier maturation in preterm infants. We hypothesized (1) donor human milk (DHM) supplementation reduces intestinal permeability (IP) similar to exclusive MOM and (2) early HM exposure and low IP at 7-10 days postnatal age (PNA) are associated with improved growth outcomes. METHODS IP was measured by the standard sugar absorption test (SAT) in infants <33 weeks gestation between 7-10 days PNA. Nutritional and anthropometric data were recorded. Postnatal growth failure (PNGF) was defined as a decrease in weight z-score >1 from birth to discharge to home. RESULTS Of 158 preterm infants, the mean (SD) gestational age was 29.9(2.3) weeks and birthweight 1388(424) g. Diet prior to SAT was exclusive MOM [N = 55(35%)], DHM ± MOM [N = 52(33%)], or preterm formula±MOM [N = 51(32%)]. The mean Lactulose(La)/Rhamnose(Rh) ratio was lower in the exclusive MOM [0.06(0.07)] and DBM ± MOM [0.05(0.07)] groups compared to the preterm formula±MOM group [0.11(0.11)], p < 0.01). Cumulative intake >150 ml/kg MOM ± DHM, but not preterm formula within 7-10 days PNA was associated with early intestinal barrier maturation. Low IP was not associated with lower risk of PNGF at discharge. CONCLUSIONS Low IP is associated with cumulative intake of MOM alone or supplemented with DHM > 150 ml/kg within 7-10 days PNA. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov NCT01756040 ; web link to study on registry: https://clinicaltrials.gov/study/NCT01756040 . IMPACT Key message Early intestinal barrier maturation is associated with cumulative intake of exclusive MOM alone or supplemented with DHM > 150 ml/kg within 7-10 days after birth, but is not associated with lower risk of PNGF at time of discharge. What it adds to existing literature? This observational study is the first study to demonstrate that supplemental DHM promotes intestinal barrier maturation similar to MOM alone. What is the impact? The findings underscore the importance of early introduction of human milk feeds as MOM or MOM supplemented with DHM in sufficient volume to promote early intestinal barrier maturation.
Collapse
Affiliation(s)
- Lisa Roskes
- Department of Pediatrics, University of Maryland School of Medicine, 22 S, Greene St, Baltimore, Maryland, 21201, USA
| | - Athanasios Chamzas
- Center for Translational Medicine, University of Maryland School of Pharmacy, S Greene St, Baltimore, Maryland, 21201, USA
| | - Bing Ma
- Institute for Genome Sciences, Department of Microbiology and Immunology, University of Maryland School of Medicine, 22 S Greene St, Baltimore, Maryland, 21201, USA
| | - Alexandre E Medina
- Department of Pediatrics, University of Maryland School of Medicine, 22 S, Greene St, Baltimore, Maryland, 21201, USA
| | - Mathangi Gopalakrishnan
- Center for Translational Medicine, University of Maryland School of Pharmacy, S Greene St, Baltimore, Maryland, 21201, USA
| | - Rose M Viscardi
- Department of Pediatrics, University of Maryland School of Medicine, 22 S, Greene St, Baltimore, Maryland, 21201, USA
| | - Sripriya Sundararajan
- Department of Pediatrics, University of Maryland School of Medicine, 22 S, Greene St, Baltimore, Maryland, 21201, USA.
| |
Collapse
|
6
|
Koivusaari K, Niinistö S, Nevalainen J, Honkanen J, Ruohtula T, Koreasalo M, Ahonen S, Åkerlund M, Tapanainen H, Siljander H, Miettinen ME, Alatossava T, Ilonen J, Vaarala O, Knip M, Virtanen SM. Infant Feeding, Gut Permeability, and Gut Inflammation Markers. J Pediatr Gastroenterol Nutr 2023; 76:822-829. [PMID: 36913717 DOI: 10.1097/mpg.0000000000003756] [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] [Indexed: 03/15/2023]
Abstract
OBJECTIVES Increased gut permeability and gut inflammation have been linked to the development of type 1 diabetes. Little is known on whether and how intake of different foods is linked to these mechanisms in infancy. We investigated whether the amount of breast milk and intake of other foods are associated with gut inflammation marker concentrations and permeability. METHODS Seventy-three infants were followed from birth to 12 months of age. Their diet was assessed with structured questionnaires and 3-day weighed food records at the age of 3, 6, 9, and 12 months. Gut permeability was assessed with the lactulose/mannitol test and fecal calprotectin and human β-defensin-2 (HBD-2) concentrations were analyzed from stool samples at the age of 3, 6, 9, and 12 months. The associations between foods and gut inflammation marker concentrations and permeability were analyzed using generalized estimating equations. RESULTS Gut permeability and gut inflammation marker concentrations decreased during the first year of life. Intake of hydrolyzed infant formula ( P = 0.003) and intake of fruits and juices ( P = 0.001) were associated with lower intestinal permeability. Intake of fruits and juices ( P < 0.001), vegetables ( P < 0.001), and oats ( P = 0.003) were associated with lower concentrations of HBD-2. Higher intake of breast milk was associated with higher fecal calprotectin concentrations ( P < 0.001), while intake of fruits and juices ( P < 0.001), vegetables ( P < 0.001), and potatoes ( P = 0.007) were associated with lower calprotectin concentrations. CONCLUSIONS Higher intake of breast milk may contribute to higher calprotectin concentration, whereas several complementary foods may decrease gut permeability and concentrations of calprotectin and HBD-2 in infant gut.
Collapse
Affiliation(s)
- Katariina Koivusaari
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- the Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
| | - Sari Niinistö
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Jaakko Nevalainen
- the Faculty of Social Sciences, Unit of Health Sciences, Tampere University, Tampere, Finland
| | - Jarno Honkanen
- the Research Program for Translational Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Terhi Ruohtula
- the Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mirva Koreasalo
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Suvi Ahonen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Mari Åkerlund
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Heli Tapanainen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Heli Siljander
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Maija E Miettinen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Tapani Alatossava
- the Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
| | - Jorma Ilonen
- the Immunogenetics Laboratory, University of Turku, Turku, Finland
| | - Outi Vaarala
- the Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Knip
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Suvi M Virtanen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
7
|
Dilemmas in initiation of very preterm infant enteral feeds-when, what, how? J Perinatol 2023; 43:108-113. [PMID: 36447040 DOI: 10.1038/s41372-022-01564-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022]
Abstract
With limited clinical evidence available to guide common nutritional decisions, significant variation exists in approaches to enteral feeding for very preterm infants, specifically when feedings are initiated, what is fed, and the method used for feedings. Preclinical studies have highlighted the benefits associated with avoiding nil per os and providing early-stage mother's own milk or colostrum. However, these recommended approaches are often mutually exclusive due to the delays in lactation associated with very preterm delivery, resulting in uncertainty regarding which approach should be prioritized. Few studies have evaluated feeding frequency in preterm infants, with limited generalizability to extremely preterm infants. Therefore, even evidence-based approaches to very preterm infant feed initiation can differ. Future research is needed to identify optimal strategies for enteral nutrition in very preterm infants, but, until then, evidence-informed approaches may vary depending on each neonatal intensive care unit's assessment of risk and benefit.
Collapse
|
8
|
Intestinal epithelium in early life. Mucosal Immunol 2022; 15:1181-1187. [PMID: 36380094 DOI: 10.1038/s41385-022-00579-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022]
Abstract
Rapid development of the fetal and neonatal intestine is required to meet the growth requirements of early life and form a protective barrier against external insults encountered by the intestinal mucosa. The fetus receives nutrition via the placenta and is protected from harmful pathogens in utero, which leads to intestinal development in a relatively quiescent environment. Upon delivery, the intestinal mucosa is suddenly tasked with providing host defense and meeting nutritional demands. To serve these functions, an array of specialized epithelial cells develop from intestinal stem cells starting in utero and continuing postnatally. Intestinal disease results when these homeostatic processes are interrupted. For preterm neonates, the most common pathology resulting from epithelial barrier dysfunction is necrotizing enterocolitis (NEC). In this review, we discuss the normal development and function of the intestinal epithelium in early life as well as how disruption of these processes can lead to NEC.
Collapse
|
9
|
Andrews‐Trevino J, Webb P, Shrestha R, Pokharel A, Acharya S, Chandyo R, Davis D, Baral K, Wang J, Xue K, Ghosh S. Exposure to multiple mycotoxins, environmental enteric dysfunction and child growth: Results from the AflaCohort Study in Banke, Nepal. MATERNAL & CHILD NUTRITION 2022; 18:e13315. [PMID: 35020261 PMCID: PMC8932698 DOI: 10.1111/mcn.13315] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 12/07/2021] [Accepted: 12/14/2021] [Indexed: 11/30/2022]
Abstract
Evidence of the impact of exposure to multiple mycotoxins and environment enteric dysfunction (EED) on child growth is limited. Using data from a birth cohort study, the objectives of this study were to (a) quantify exposure to multiple mycotoxins (serum aflatoxin [AFB1] and ochratoxin A [OTA], urinary fumonisin [UFB1] and deoxynivalenol [DON]), as well EED (lactulose:mannitol [L:M] ratio); (b) examine the potential combined effects of multiple mycotoxin exposure and EED on growth. Multivariate regressions were used to identify associations between growth measurements (length, weight, anthropometric z‐scores, stunting and underweight) at 24–26 months of age and exposure to mycotoxins and EED at 18–22 months (n = 699). Prevalence of AFB1, DON, OTA and UFB1 exposure ranged from 85% to 100%; average L:M ratio was 0.29 ± 0.53. In individual mycotoxin models, AFB1 exposure was negatively associated with weight, WAZ, increased odds of stunting (odds ratio [OR]: 1.28, 95% confidence interval [CI]: 1.08, 1.52; p = 0.004) and underweight (OR: 1.18, 95% CI: 1.00, 1.38; p = 0.046). Irrespective of other mycotoxin exposure and presence of EED, AFB1 was negatively associated with length, weight, head circumference, LAZ and WAZ, and with increased odds of stunting and underweight, UFB1 was associated with increased odds of underweight, and DON was negatively associated with head circumference. EED was associated with the impaired length and weight. These findings suggest that certain mycotoxins and EED may have independent impacts on different facets of growth and that aflatoxin dominates such impacts. Thus, programs reducing exposure to mycotoxin and EED through multi‐sectoral nutrition‐sensitive interventions have the potential to improve child growth. Mycotoxin exposure was highly prevalent in children aged 18–22 months. Various mycotoxins and EED contribute independently to different manifestations of poor child growth AFB1 was negatively associated with length, weight, head circumference, LAZ, as well as WAZ, and associated with increased odds of stunting and underweight. UFB1 was also associated with increased odds of underweight. DON was negatively associated with head circumference.
Collapse
Affiliation(s)
- Johanna Andrews‐Trevino
- Division of Food and Nutrition Policy and Programs, Friedman School of Nutrition Science and Policy Tufts University Boston Massachusetts USA
| | - Patrick Webb
- Division of Food and Nutrition Policy and Programs, Friedman School of Nutrition Science and Policy Tufts University Boston Massachusetts USA
| | - Robin Shrestha
- Division of Food and Nutrition Policy and Programs, Friedman School of Nutrition Science and Policy Tufts University Boston Massachusetts USA
| | | | | | - Ram Chandyo
- Department of Community Medicine Kathmandu Medical College Kathmandu Nepal
| | - Dale Davis
- Helen Keller International‐Nepal Kathmandu Nepal
| | - Kedar Baral
- Department of Community Health Sciences Patan Academy of Health Sciences Lalitpur Nepal
| | - Jia‐Sheng Wang
- Department of Environmental Health Science University of Georgia Athens Georgia USA
| | - Kathy Xue
- Department of Environmental Health Science University of Georgia Athens Georgia USA
| | - Shibani Ghosh
- Division of Food and Nutrition Policy and Programs, Friedman School of Nutrition Science and Policy Tufts University Boston Massachusetts USA
| |
Collapse
|
10
|
How to Provide Breast Milk for the Preterm Infant and Avoid Symptomatic Cytomegalovirus Infection with Possible Long-Term Sequelae. Life (Basel) 2022; 12:life12040504. [PMID: 35454995 PMCID: PMC9031638 DOI: 10.3390/life12040504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 12/04/2022] Open
Abstract
Cytomegalovirus (CMV) is able to replicate in the breast milk of lactating mothers and thus the offspring might be affected by mild to severe symptoms of postnatal CMV disease in case of prematurity; not in term infants. Sepsis-like syndrome affects only very low birth infants; and few cases have been reported. The neurodevelopmental long-term outcome of those preterm infants revealed possible subtle deficiencies, but no major neurodevelopmental impairment. Neurodevelopmental sequelae are still in discussion and seem somewhat overestimated after careful evaluation of the published evidence. The main focus of postnatal CMV disease lies upon the extremely low birth weight of infants. Elimination of CMV is provided by short-term heating methods like the most widely used Holder pasteurization. Freezing and thawing methods leave a risk for CMV acquisition. The benefits of untreated breast milk have to be considered to outweigh the possible sequelae of postnatal CMV infection in the most vulnerable preterm infants.
Collapse
|
11
|
Athalye-Jape G, Esvaran M, Patole S, Simmer K, Nathan E, Doherty D, Keil A, Rao S, Chen L, Chandrasekaran L, Kok C, Schuster S, Conway P. Effect of single versus multistrain probiotic in extremely preterm infants: a randomised trial. BMJ Open Gastroenterol 2022; 9:bmjgast-2021-000811. [PMID: 35185013 PMCID: PMC8860036 DOI: 10.1136/bmjgast-2021-000811] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/12/2022] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE Evidence indicates that multistrain probiotics benefit preterm infants more than single-strain (SS) probiotics. We assessed the effects of SS versus triple-strain (TS) probiotic supplementation (PS) in extremely preterm (EP) infants. DESIGN EP infants (gestational age (GA) <28 weeks) were randomly allocated to TS or SS probiotic, assuring blinding. Reference (REF) group was EP infants in the placebo arm of our previous probiotic trial. PS was commenced with feeds and continued until 37 weeks' corrected GA. Primary outcome was time to full feed (TFF: 150 mL/kg/day). Secondary outcomes included short-chain fatty acids and faecal microbiota collected at T1 (first week) and T2 (after 3 weeks of PS) using 16S ribosomal RNA gene sequencing. RESULTS 173 EP (SS: 86, TS: 87) neonates with similar GA and birth weight (BW) were randomised. Median TFF was comparable (11 (IQR 8-16) vs 10 (IQR 8-16) days, p=0.92). Faecal propionate (SS, p<0.001, and TS, p=0.0009) and butyrate levels (TS, p=0.029) were significantly raised in T2 versus T1 samples. Secondary clinical outcomes were comparable. At T2, alpha diversity was comparable (p>0.05) between groups, whereas beta-diversity analysis revealed significant differences between PS and REF groups (both p=0.001). Actinobacteria were higher (both p<0.01), and Proteobacteria, Firmicutes and Bacteroidetes were lower in PS versus REF. Gammaproteobacteria, Clostridia and Negativicutes were lower in both PS versus REF. CONCLUSION TFF in EP infants was similar between SS and TS probiotics. Both probiotics were effective in reducing dysbiosis (higher bifidobacteria and lower Gammaproteobacteria). Long-term significance of increased propionate and butyrate needs further studies. TRIAL REGISTRATION NUMBER ACTRN 12615000940572.
Collapse
Affiliation(s)
- Gayatri Athalye-Jape
- Neonatology directorate, King Edward Memorial Hospital for Women Perth, Subiaco, Western Australia, Australia
| | - Meera Esvaran
- Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Sanjay Patole
- Neonatal Clinical Care Unit, King Edward Memorial Hospital, Subiaco, Western Australia, Australia
| | - Karen Simmer
- Neonatal Clinical Care Unit, King Edward Memorial Hospital, Subiaco, Western Australia, Australia
| | - Elizabeth Nathan
- Biostatistics, Women and Infants Research Foundation Western Australia, Subiaco, Western Australia, Australia
| | - Dorota Doherty
- Biostatistics, Women and Infants Research Foundation Western Australia, Subiaco, Western Australia, Australia
| | - Anthony Keil
- Microbiology, PathWest Laboratory Medicine Western Australia, Nedlands, Western Australia, Australia
| | - Shripada Rao
- Neonatal Clinical Care Unit, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Liwei Chen
- Genomics and Bioinformatics, Nanyang Technological University, Singapore
| | | | - Chooi Kok
- Neonatal Clinical Care Unit, King Edward Memorial Hospital, Subiaco, Western Australia, Australia
| | - Stephan Schuster
- Genomics and Bioinformatics, Nanyang Technological University, Singapore
| | - Patricia Conway
- Genomics and Bioinformatics, Nanyang Technological University, Singapore
| |
Collapse
|
12
|
The effect of feeding patterns on serum zonulin levels in infants at 3-4 months of age. Eur J Pediatr 2021; 180:3273-3278. [PMID: 33973070 DOI: 10.1007/s00431-021-04102-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/09/2021] [Accepted: 05/02/2021] [Indexed: 12/22/2022]
Abstract
Zonulin so far is the only known endogenous modulator of intercellular tight junctions which regulate the intestinal permeability. Breastfeeding is considered to enhance the integrity of the gastrointestinal tract; however, limited data are available about the effect of feeding patterns on intestinal permeability. We aimed to investigate the potential association between the mode of feeding (breast versus formula milk) and the serum zonulin levels as a marker of intestinal permeability. One hundred fifty-seven full-term, healthy infants, born after an uncomplicated pregnancy, were enrolled within 72-96 h of life. Blood samples from 105 infants were obtained at 3 to 4 months of life. Serum zonulin levels were measured by ELISA. Out of 105 infants, 52.4% (55) were female, and 58.1% (61) were delivered by caesarian section at a mean gestational age of 38.9 (SD ± 1.0) weeks. At the time of blood sampling, median age was 3.4 (IQR 3.20-3.50) months, and mean weight was 6332 (SD ± 692) gr. Infants were divided in three groups according to the feeding patterns: exclusive breastfeeding (n = 42), mixed feeding (n = 41), and cow's milk formula (n = 22). The feeding pattern had no impact on infants' serum zonulin levels. Moreover, zonulin levels were not affected by infant's clinical and epidemiological characteristics such as body weight or family history of autoimmune disease.Conclusion: In our study, different feeding patterns were not associated with serum zonulin levels in healthy infants at 3-4 months of age. What is Known: • Serum zonulin is upregulated in conditions with increased intestinal permeability • Breast milk favors the physiological decline of the intestinal permeability after birth in the neonates What is New: • Serum zonulin levels were not affected by the feeding pattern (breast milk versus formula) in infants at 3-4 months of age • Clinical and epidemiological characteristics of infants had no impact on zonulin levels.
Collapse
|
13
|
Abramov VM, Kosarev IV, Priputnevich TV, Machulin AV, Abashina TN, Chikileva IO, Donetskova AD, Takada K, Melnikov VG, Vasilenko RN, Khlebnikov VS, Samoilenko VA, Nikonov IN, Sukhikh GT, Uversky VN, Karlyshev AV. S-layer protein 2 of vaginal Lactobacillus crispatus 2029 enhances growth, differentiation, VEGF production and barrier functions in intestinal epithelial cell line Caco-2. Int J Biol Macromol 2021; 189:410-419. [PMID: 34437917 DOI: 10.1016/j.ijbiomac.2021.08.150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022]
Abstract
We have previously demonstrated the ability of the human vaginal strain Lactobacillus crispatus 2029 (LC2029) for strong adhesion to cervicovaginal epithelial cells, expression of the surface layer protein 2 (Slp2), and antagonistic activity against urogenital pathogens. Slp2 forms regular two-dimensional structure around the LC2029 cells,which is secreted into the medium and inhibits intestinal pathogen-induced activation of caspase-9 and caspase-3 in the human intestinal Caco-2 cells. Here, we elucidated the effects of soluble Slp2 on adhesion of proteobacteria pathogens inducing necrotizing enterocolitis (NEC), such as Escherichia coli ATCC E 2348/69, E. coli ATCC 31705, Salmonella Enteritidis ATCC 13076, Campylobacter jejuni ATCC 29428, and Pseudomonas aeruginosa ATCC 27853 to Caco-2 cells, as well as on growth promotion, differentiation, vascular endothelial growth factor (VEGF) production, and intestinal barrier function of Caco-2 cell monolayers. Slp2 acts as anti-adhesion agent for NEC-inducing proteobacteria, promotes growth of immature Caco-2 cells and their differentiation, and enhances expression and functional activity of sucrase, lactase, and alkaline phosphatase. Slp2 stimulates VEGF production, decreases paracellular permeability, and increases transepithelial electrical resistance, strengthening barrier function of Caco-2 cell monolayers. These data support the important role of Slp2 in the early postnatal development of the human small intestine enterocytes.
Collapse
Affiliation(s)
- Vyacheslav M Abramov
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Igor V Kosarev
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Tatiana V Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Andrey V Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Tatiana N Abashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Irina O Chikileva
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Laboratory of Cell Immunity, Blokhin National Research, Center of Oncology Ministry of Health RF, Moscow 115478, Russia
| | | | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Vyacheslav G Melnikov
- Gabrichevsky Moscow Research Institute of Epidemiology and Microbiology, Federal Service for Supervision of Consumer Rights Protection and Human Welfare, Moscow 152212, Russia
| | - Raisa N Vasilenko
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia
| | | | - Vladimir A Samoilenko
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Ilya N Nikonov
- Federal State Education Institution of Higher Professional Education Moscow State Academy of Veterinary Medicine and Biotechnology named after K.I. Skryabin, Moscow 109472, Russia
| | - Gennady T Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Andrey V Karlyshev
- Department of Science, Engineering and Computing, Kingston University London, Kingston upon Thames KT1 2EE, UK
| |
Collapse
|
14
|
Boonma P, Shapiro JM, Hollister EB, Badu S, Wu Q, Weidler EM, Abraham BP, Devaraj S, Luna RA, Versalovic J, Heitkemper MM, Savidge TC, Shulman RJ. Probiotic VSL#3 Treatment Reduces Colonic Permeability and Abdominal Pain Symptoms in Patients With Irritable Bowel Syndrome. FRONTIERS IN PAIN RESEARCH 2021; 2:691689. [PMID: 35295488 PMCID: PMC8915646 DOI: 10.3389/fpain.2021.691689] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Little is known regarding the clinical impact of treatment and treatment duration of probiotic VSL#3 on gut and microbiome function in irritable bowel syndrome (IBS). As part of a safety trial, we assessed the effect of VSL#3 treatment duration on abdominal pain, stooling, gut permeability, microbiome composition and function. Methods: Adults with IBS were randomized into an open label trial to receive the probiotic VSL#3 for 4 or 8 weeks. Adverse events, abdominal pain, and stooling patterns were recorded daily. Gut permeability, fecal bile acid levels, and microbiome composition were profiled at baseline and after treatment. Results: Fifteen subjects completed the trial (4-week: n = 8; 8-week: n = 7). Number of pain episodes decreased in both groups (P = 0.049 and P = 0.034; 4- vs. 8-week, respectively). Probiotic organisms contained in VSL#3 were detected in feces by whole shotgun metagenomic sequencing analysis and relative abundances of Streptococcus thermophilus, Bifidobacterium animalis, Lactobacillus plantarum, and Lactobacillus casei subsp. paraccasei correlated significantly with improved abdominal pain symptoms and colonic permeability at study completion. Although abdominal pain correlated significantly with the detection of probiotic species at study completion, a composite view of gut microbiome structure showed no changes in community diversity or composition after VSL#3 treatment. Conclusions: Probiotic organisms identified in stool correlated significantly with improvement in colonic permeability and clinical symptoms, prompting future studies to investigate the mechanistic role of VSL#3 and colonic permeability in IBS pathophysiology in a larger randomized controlled trial. Clinical Trial Registration:www.clinicaltrials.gov, Identifier: NCT00971711.
Collapse
Affiliation(s)
- Prapaporn Boonma
- Department of Pathology, Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Jordan M. Shapiro
- Department of Medicine, Baylor College of Medicine,Houston, TX, United States
| | - Emily B. Hollister
- Department of Pathology, Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Shyam Badu
- Department of Pathology, Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Qinglong Wu
- Department of Pathology, Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Erica M. Weidler
- Center for Pediatric Abdominal Pain Research, Texas Children's Hospital, Houston, TX, United States
- Children's Nutrition Research Center, Houston, TX, United States
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Bincy P. Abraham
- Division of Gastroenterology, Houston Methodist Hospital, Houston, TX, United States
| | - Sridevi Devaraj
- Department of Pathology, Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Ruth Ann Luna
- Department of Pathology, Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - James Versalovic
- Department of Pathology, Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Margaret M. Heitkemper
- Department of Biobehavioral Nursing and Health Informatics, University of Washington, Seattle, WA, United States
| | - Tor C. Savidge
- Department of Pathology, Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Robert J. Shulman
- Center for Pediatric Abdominal Pain Research, Texas Children's Hospital, Houston, TX, United States
- Children's Nutrition Research Center, Houston, TX, United States
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Robert J. Shulman
| |
Collapse
|
15
|
Thoene M, Anderson-Berry A. Early Enteral Feeding in Preterm Infants: A Narrative Review of the Nutritional, Metabolic, and Developmental Benefits. Nutrients 2021; 13:nu13072289. [PMID: 34371799 PMCID: PMC8308411 DOI: 10.3390/nu13072289] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/25/2021] [Accepted: 06/03/2021] [Indexed: 01/01/2023] Open
Abstract
Enteral feeding is the preferred method of nutrient provision for preterm infants. Though parenteral nutrition remains an alternative to provide critical nutrition after preterm delivery, the literature suggests that enteral feeding still confers significant nutritional and non-nutritional benefits. Therefore, the purpose of this narrative review is to summarize health and clinical benefits of early enteral feeding within the first month of life in preterm infants. Likewise, this review also proposes methods to improve enteral delivery in clinical care, including a proposal for decision-making of initiation and advancement of enteral feeding. An extensive literature review assessed enteral studies in preterm infants with subsequent outcomes. The findings support the early initiation and advancement of enteral feeding impact preterm infant health by enhancing micronutrient delivery, promoting intestinal development and maturation, stimulating microbiome development, reducing inflammation, and enhancing brain growth and neurodevelopment. Clinicians must consider these short- and long-term implications when caring for preterm infants.
Collapse
|
16
|
Sadrudin Premji S, Chessell L, Stewart F. Continuous nasogastric milk feeding versus intermittent bolus milk feeding for preterm infants less than 1500 grams. Cochrane Database Syst Rev 2021; 6:CD001819. [PMID: 34165778 PMCID: PMC8223964 DOI: 10.1002/14651858.cd001819.pub3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Milk feedings can be given via nasogastric tube either intermittently, typically over 10 to 20 minutes every two or three hours, or continuously, using an infusion pump. Although the theoretical benefits and risks of each method have been proposed, their effects on clinically important outcomes remain uncertain. OBJECTIVES: To examine the evidence regarding the effectiveness of continuous versus intermittent bolus tube feeding of milk in preterm infants less than 1500 grams. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to run comprehensive searches in the Cochrane Central Register of Controlled Trials (CENTRAL 2020, Issue 7) in the Cochrane Library; Ovid MEDLINE and Epub Ahead of Print, In-Process & Other Non-Indexed Citations, Daily and Versions; and CINAHL (Cumulative Index to Nursing and Allied Health Literature) on 17 July 2020. We also searched clinical trials databases and the reference lists of retrieved articles for randomised controlled trials (RCTs) and quasi-RCTs. SELECTION CRITERIA We included RCTs and quasi-RCTs comparing continuous versus intermittent bolus nasogastric milk feeding in preterm infants less than 1500 grams. DATA COLLECTION AND ANALYSIS Two review authors independently assessed all trials for relevance and risk of bias. We used the standard methods of Cochrane Neonatal to extract data. We used the GRADE approach to assess the certainty of evidence. Primary outcomes were: age at full enteral feedings; feeding intolerance; days to regain birth weight; rate of gain in weight, length and head circumference; and risk of necrotising enterocolitis (NEC). MAIN RESULTS We included nine randomised trials (919 infants) in this updated Cochrane Review. One study is awaiting classification. Seven of the nine included trials reported data from infants with a maximum weight of between 1000 grams and 1400 grams. Two of the nine trials included infants weighing up to 1500 grams. Type(s) of milk feeds varied, including human milk (either mother's own milk or pasteurised donor human milk), preterm formula, or mixed feeding regimens. In some instances, preterm formula was initially diluted. Earlier studies also used water to initiate feedings. We judged six trials as unclear or high risk of bias for random sequence generation. We judged four trials as unclear for allocation concealment. We judged all trials as high risk of bias for blinding of care givers, and seven as unclear or high risk of bias for blinding of outcome assessors. We downgraded the certainty of evidence for imprecision, due to low numbers of participants in the trials, and/or wide 95% confidence intervals, and/or for risk of bias. Continuous compared to intermittent bolus (nasogastric and orogastric tube) milk feeding Babies receiving continuous feeding may reach full enteral feeding almost one day later than babies receiving intermittent feeding (mean difference (MD) 0.84 days, 95% confidence interval (CI) -0.13 to 1.81; 7 studies, 628 infants; low-certainty evidence). It is uncertain if there is any difference between continuous feeding and intermittent feeding in terms of number of days of feeding interruptions (MD -3.00 days, 95% CI -9.50 to 3.50; 1 study, 171 infants; very low-certainty evidence). It is uncertain if continuous feeding has any effect on days to regain birth weight (MD -0.38 days, 95% CI -1.16 to 0.41; 6 studies, 610 infants; low-certainty evidence). The certainty of evidence is low and the 95% confidence interval is consistent with possible benefit and possible harm. It is uncertain if continuous feeding has any effect on rate of gain in weight compared with intermittent feeding (standardised mean difference (SMD) 0.09, 95% CI -0.27 to 0.46; 5 studies, 433 infants; very low-certainty evidence). Continuous feeding may result in little to no difference in rate of gain in length compared with intermittent feeding (MD 0.02 cm/week, 95% CI -0.04 to 0.08; 5 studies, 433 infants; low-certainty evidence). Continuous feeding may result in little to no difference in rate of gain in head circumference compared with intermittent feeding (MD 0.01 cm/week, 95% CI -0.03 to 0.05; 5 studies, 433 infants; low-certainty evidence). It is uncertain if continuous feeding has any effect on the risk of NEC compared with intermittent feeding (RR 1.19, 95% CI 0.67 to 2.11; 4 studies, 372 infants; low-certainty evidence). The certainty of evidence is low and the 95% confidence interval is consistent with possible benefit and possible harm. AUTHORS' CONCLUSIONS Although babies receiving continuous feeding may reach full enteral feeding slightly later than babies receiving intermittent feeding, the evidence is of low certainty. However, the clinical risks and benefits of continuous and intermittent nasogastric tube milk feeding cannot be reliably discerned from current available randomised trials. Further research is needed to determine if either feeding method is more appropriate for the initiation of feeds. A rigorous methodology should be adopted, defining feeding protocols and feeding intolerance consistently for all infants. Infants should be stratified according to birth weight and gestation, and possibly according to illness.
Collapse
Affiliation(s)
| | | | - Fiona Stewart
- Cochrane Children and Families Network, c/o Cochrane Pregnancy and Childbirth, Department of Women's and Children's Health, The University of Liverpool, Liverpool, UK
| |
Collapse
|
17
|
Bifidobacterium breve BBG-001 and intestinal barrier function in preterm babies: Exploratory Studies from the PiPS Trial. Pediatr Res 2021; 89:1818-1824. [PMID: 32947603 DOI: 10.1038/s41390-020-01135-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/09/2020] [Accepted: 08/11/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND Uncertainty remains about the role of probiotics to prevent necrotising enterocolitis (NEC) some of which arises from the variety of probiotic interventions used in different trials, many with no prior evidence of potential efficacy. Mechanistic studies of intestinal barrier function embedded in a large probiotic trial could provide evidence about which properties of probiotics might be important for NEC prevention thus facilitating identification of strains with therapeutic potential. METHODS Intestinal permeability, stool microbiota, SCFAs and mucosal inflammation were assessed from the second postnatal week in babies enrolled to a randomised controlled trial of B. breve BBG-001 (the PiPS trial). Results were compared by allocation and by stool colonisation with the probiotic. RESULTS Ninety-four preterm babies were recruited across six nested studies. B. breve BBG-001 content was higher by allocation and colonisation; Enterobacteriaceae and acetic acid levels were higher by colonisation. No measure of intestinal barrier function showed differences. The PiPS trial found no evidence of efficacy to reduce NEC. CONCLUSIONS That the negative results of the PiPS trial were associated with failure of this probiotic to modify intestinal barrier function supports the possibility that the tests described here have the potential to identify strains to progress to large clinical trials. IMPACT Uncertainty about the therapeutic role of probiotics to prevent necrotising enterocolitis is in part due to the wide range of bacterial strains with no previous evidence of efficacy used in clinical trials. We hypothesised that mechanistic studies embedded in a probiotic trial would provide evidence about which properties of probiotics might be important for NEC prevention. The finding that the probiotic strain tested, Bifidobacterium breve BBG-001, showed neither effects on intestinal barrier function nor clinical efficacy supports the possibility that these tests have the potential to identify strains to progress to large clinical trials.
Collapse
|
18
|
Kaczmarczyk M, Löber U, Adamek K, Węgrzyn D, Skonieczna-Żydecka K, Malinowski D, Łoniewski I, Markó L, Ulas T, Forslund SK, Łoniewska B. The gut microbiota is associated with the small intestinal paracellular permeability and the development of the immune system in healthy children during the first two years of life. J Transl Med 2021; 19:177. [PMID: 33910577 PMCID: PMC8082808 DOI: 10.1186/s12967-021-02839-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/16/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The intestinal barrier plays an important role in the defense against infections, and nutritional, endocrine, and immune functions. The gut microbiota playing an important role in development of the gastrointestinal tract can impact intestinal permeability and immunity during early life, but data concerning this problem are scarce. METHODS We analyzed the microbiota in fecal samples (101 samples in total) collected longitudinally over 24 months from 21 newborns to investigate whether the markers of small intestinal paracellular permeability (zonulin) and immune system development (calprotectin) are linked to the gut microbiota. The results were validated using data from an independent cohort that included the calprotectin and gut microbiota in children during the first year of life. RESULTS Zonulin levels tended to increase for up to 6 months after childbirth and stabilize thereafter remaining at a high level while calprotectin concentration was high after childbirth and began to decline from 6 months of life. The gut microbiota composition and the related metabolic potentials changed during the first 2 years of life and were correlated with zonulin and calprotectin levels. Faecal calprotectin correlated inversely with alpha diversity (Shannon index, r = - 0.30, FDR P (Q) = 0.039). It also correlated with seven taxa; i.a. negatively with Ruminococcaceae (r = - 0.34, Q = 0.046), and Clostridiales (r = - 0.34, Q = 0.048) and positively with Staphylococcus (r = 0.38, Q = 0.023) and Staphylococcaceae (r = 0.35, Q = 0.04), whereas zonulin correlated with 19 taxa; i.a. with Bacillales (r = - 0.52, Q = 0.0004), Clostridiales (r = 0.48, Q = 0.001) and the Ruminococcus (torques group) (r = 0.40, Q = 0.026). When time intervals were considered only changes in abundance of the Ruminococcus (torques group) were associated with changes in calprotectin (β = 2.94, SE = 0.8, Q = 0.015). The dynamics of stool calprotectin was negatively associated with changes in two MetaCyc pathways: pyruvate fermentation to butanoate (β = - 4.54, SE = 1.08, Q = 0.028) and Clostridium acetobutylicum fermentation (β = - 4.48, SE = 1.16, Q = 0.026). CONCLUSIONS The small intestinal paracellular permeability, immune system-related markers and gut microbiota change dynamically during the first 2 years of life. The Ruminococcus (torques group) seems to be especially involved in controlling paracellular permeability. Staphylococcus, Staphylococcaceae, Ruminococcaceae, and Clostridiales, may be potential biomarkers of the immune system. Despite observed correlations their clear causation and health consequences were not proven. Mechanistic studies are required.
Collapse
Affiliation(s)
- Mariusz Kaczmarczyk
- Department of Clinical Biochemistry, Pomeranian Medical University in Szczecin, 70-111, Szczecin, Poland
| | - Ulrike Löber
- Experimental and Clinical Research Center, A Cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 14195, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Karolina Adamek
- Department of Neonatal Diseases, Pomeranian Medical University in Szczecin, 70-111, Szczecin, Poland
| | - Dagmara Węgrzyn
- Department of Neonatal Diseases, Pomeranian Medical University in Szczecin, 70-111, Szczecin, Poland
| | | | - Damian Malinowski
- Department of Pharmacology, Pomeranian Medical University in Szczecin, 70-111, Szczecin, Poland
| | - Igor Łoniewski
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, 71-460, Szczecin, Poland.
- Department of Human Nutrition and Metabolomics, Broniewskiego 24, 71-460, Szczecin, Poland.
| | - Lajos Markó
- Experimental and Clinical Research Center, A Cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 14195, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
| | - Thomas Ulas
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn, 53127, Bonn, Germany
| | - Sofia K Forslund
- Experimental and Clinical Research Center, A Cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 14195, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117, Heidelberg, Germany
| | - Beata Łoniewska
- Department of Neonatal Diseases, Pomeranian Medical University in Szczecin, 70-111, Szczecin, Poland
| |
Collapse
|
19
|
Singh A, Ghosh S, Ward H, Manary MJ, Rogers BL, Rosenberg IH. Biomarkers of environmental enteric dysfunction are differently associated with recovery and growth among children with moderate acute malnutrition in Sierra Leone. Am J Clin Nutr 2021; 113:1556-1564. [PMID: 33668048 PMCID: PMC8243924 DOI: 10.1093/ajcn/nqaa434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) may influence growth during and recovery from moderate acute malnutrition (MAM), however, biomarkers to assess these relations have yet to be identified. OBJECTIVES The objectives of this study were to: 1) develop a score for EED based on host fecal mRNA transcripts, 2) compare biomarkers of EED with each other, and 3) examine associations between the EED biomarkers and recovery from MAM and growth outcomes. METHODS In a cohort of 520 Sierra Leonean MAM children, biomarkers of EED included the lactulose: mannitol (L: M) test, 15 host fecal mRNA transcripts, and host fecal proteins [α-1-antitrypsin (AAT), myeloperoxidase (MPO), neopterin (NEO)]. Anthropometry data were also collected and z scores were computed for length-for-age (LAZ) and weight-for-length (WLZ). Recovery from MAM was defined as midupper arm circumference ≥12.5 cm. Factor analysis was used to identify EED scores using the mRNA transcripts, and mixed effects regression was conducted to test for associations. RESULTS The 15 host fecal mRNA transcripts were clustered into 3 scores: gut inflammation (GI) score, gut structure (GS) score, and gut defense (GD) score. We found agreement between certain inflammation markers (GI score and MPO), and permeability markers (GS score and AAT; AAT and the L: M excretion ratio). Antimicrobial gut defense (GD score) was inversely associated with percent lactulose excreted, a measure of intestinal permeability. LAZ (β: -0.08; 95% CI: -0.14, -0.02) and WLZ (β: -0.03; 95% CI: -0.06, -0.01) were negatively associated with GI score. A high GD score (β: 0.36; 95% CI: 0.08, 0.64) and low AAT (β: -1.35; 95% CI: -2.35, -0.36) were associated with recovery from MAM. CONCLUSIONS Scores derived from host fecal mRNA transcript variably correlated with the L: M test and host fecal proteins. Markers of intestinal inflammation, permeability, and defense were associated with growth outcomes and recovery from MAM.
Collapse
Affiliation(s)
| | - Shibani Ghosh
- Friedman School of Nutrition Science and Policy, Tufts
University, Boston, MA, USA
| | - Honorine Ward
- Tufts Medical Center and Tufts University School of
Medicine, Boston, MA, USA
| | - Mark J Manary
- Department of Pediatrics, Washington University,
St Louis, MO, USA
| | - Beatrice L Rogers
- Friedman School of Nutrition Science and Policy, Tufts
University, Boston, MA, USA
| | - Irwin H Rosenberg
- Friedman School of Nutrition Science and Policy, Tufts
University, Boston, MA, USA
| |
Collapse
|
20
|
Statelova M, Holm R, Fotaki N, Reppas C, Vertzoni M. Successful Extrapolation of Paracetamol Exposure from Adults to Infants After Oral Administration of a Pediatric Aqueous Suspension Is Highly Dependent on the Study Dosing Conditions. AAPS JOURNAL 2020; 22:126. [DOI: 10.1208/s12248-020-00504-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/18/2020] [Indexed: 01/10/2023]
|
21
|
Jayasinghe TN, Vatanen T, Chiavaroli V, Jayan S, McKenzie EJ, Adriaenssens E, Derraik JGB, Ekblad C, Schierding W, Battin MR, Thorstensen EB, Cameron-Smith D, Forbes-Blom E, Hofman PL, Roy NC, Tannock GW, Vickers MH, Cutfield WS, O'Sullivan JM. Differences in Compositions of Gut Bacterial Populations and Bacteriophages in 5-11 Year-Olds Born Preterm Compared to Full Term. Front Cell Infect Microbiol 2020; 10:276. [PMID: 32612960 PMCID: PMC7309444 DOI: 10.3389/fcimb.2020.00276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/11/2020] [Indexed: 12/17/2022] Open
Abstract
Preterm infants are exposed to major perinatal, post-natal, and early infancy events that could impact on the gut microbiome. These events include infection, steroid and antibiotic exposure, parenteral nutrition, necrotizing enterocolitis, and stress. Studies have shown that there are differences in the gut microbiome during the early months of life in preterm infants. We hypothesized that differences in the gut microbial composition and metabolites in children born very preterm persist into mid-childhood. Participants were healthy prepubertal children aged 5-11 years who were born very preterm (≤32 weeks of gestation; n = 51) or at term (37-41 weeks; n = 50). We recorded the gestational age, birth weight, mode of feeding, mode of birth, age, sex, and the current height and weight of our cohort. We performed a multi'omics [i.e., 16S rRNA amplicon and shotgun metagenomic sequencing, SPME-GCMS (solid-phase microextraction followed by gas chromatography-mass spectrometry)] analysis to investigate the structure and function of the fecal microbiome (as a proxy of the gut microbiota) in our cross-sectional cohort. Children born very preterm were younger (7.8 vs. 8.3 years; p = 0.034), shorter [height-standard deviation score (SDS) 0.31 vs. 0.92; p = 0.0006) and leaner [BMI (body mass index) SDS -0.20 vs. 0.29; p < 0.0001] than the term group. Children born very preterm had higher fecal calprotectin levels, decreased fecal phage richness, lower plasma arginine, lower fecal branched-chain amino acids and higher fecal volatile (i.e., 3-methyl-butanoic acid, butyrolactone, butanoic acid and pentanoic acid) profiles. The bacterial microbiomes did not differ between preterm and term groups. We speculate that the observed very preterm-specific changes were established in early infancy and may impact on the capacity of the very preterm children to respond to environmental changes.
Collapse
Affiliation(s)
| | - Tommi Vatanen
- Liggins Institute, University of Auckland, Auckland, New Zealand
- The Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | | | - Sachin Jayan
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | | | - José G. B. Derraik
- Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start—National Science Challenge, University of Auckland, Auckland, New Zealand
| | - Cameron Ekblad
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | - Paul L. Hofman
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Nicole C. Roy
- AgResearch, Palmerston North, New Zealand
- The Riddet Institute, Massey University, Palmerston North, New Zealand
- The High-Value Nutrition Challenge, Auckland, New Zealand
| | - Gerald W. Tannock
- The Riddet Institute, Massey University, Palmerston North, New Zealand
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Mark H. Vickers
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Wayne S. Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand
- Quadram Institute Bioscience, Norwich, United Kingdom
| | | |
Collapse
|
22
|
Łoniewska B, Adamek K, Węgrzyn D, Kaczmarczyk M, Skonieczna-Żydecka K, Clark J, Adler G, Tousty J, Uzar I, Tousty P, Łoniewski I. Analysis of Faecal Zonulin and Calprotectin Concentrations in Healthy Children During the First Two Years of Life. An Observational Prospective Cohort Study. J Clin Med 2020; 9:jcm9030777. [PMID: 32178435 PMCID: PMC7141325 DOI: 10.3390/jcm9030777] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
Factors affecting the intestinal-barrier permeability of newborns, such as body mass index (BMI), nutrition and antibiotics, are assumed to affect intestinal-barrier permeability in the first two years of life. This study assessed 100 healthy, full-term newborns to 24 months old. Faecal zonulin/calprotectin concentrations were measured at 1, 6, 12, 24 months as gut-permeability markers. Zonulin concentrations increased between 1 and 12 months (medians: 114.41, 223.7 ng/mL; respectively), whereas calprotectin concentrations decreased between one and six months (medians: 149. 29, 109.28 µg/mL); both then stabilized (24 months: 256.9 ng/mL zonulin; 59.5 µg/mL calprotectin). In individual children, high levels at one month gave high levels at older ages (correlations: calprotectin: between 1 and 6 or 12 months: correlation coefficient (R) = 0.33, statistical significance (p) = 0.0095; R = 0.28, p = 0.032; zonulin: between 1 and 24 months: R = 0.32; p = 0.022, respectively). Parameters which gave marker increases: antibiotics during pregnancy (calprotectin; six months: by 80%, p = 0.038; 12 months: by 48%, p = 0.028); vaginal birth (calprotectin: 6 months: by 140%, p = 0.005); and > 5.7 pregnancy-BMI increase (zonulin: 12 months: by 74%, p = 0.049). Conclusions: “Closure of the intestines” is spread over time and begins between the sixth and twelfth month of life. Antibiotic therapy, BMI increase > 5.7 during pregnancy and vaginal birth are associated with increased intestinal permeability during the first two years of life. Stool zonulin and calprotectin concentrations were much higher compared with previous measurements at older ages; clinical interpretation and validation are needed (no health associations found).
Collapse
Affiliation(s)
- Beata Łoniewska
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
- Correspondence: ; Tel.: +48-(91)-466-1375
| | - Karolina Adamek
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
| | - Dagmara Węgrzyn
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
| | - Mariusz Kaczmarczyk
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin 70-111, Poland; (M.K.); (J.C.)
| | - Karolina Skonieczna-Żydecka
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, Szczecin 71-460, Poland; (K.S.-Ż.); (I.Ł.)
| | - Jeremy Clark
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin 70-111, Poland; (M.K.); (J.C.)
| | - Grażyna Adler
- Department of Studies in Anthropogenetics and Biogerontology, Pomeranian Medical University, Szczecin 71-210, Poland;
| | - Joanna Tousty
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
| | - Izabela Uzar
- Department of General Pharmacology and Pharmacoeconomics, Pomeranian Medical University, Szczecin 71-230, Poland;
| | - Piotr Tousty
- Department of Obstetrics and Gynecology, Pomeranian Medical University, Szczecin 70-111, Poland;
| | - Igor Łoniewski
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, Szczecin 71-460, Poland; (K.S.-Ż.); (I.Ł.)
| |
Collapse
|
23
|
Burrin D, Sangild PT, Stoll B, Thymann T, Buddington R, Marini J, Olutoye O, Shulman RJ. Translational Advances in Pediatric Nutrition and Gastroenterology: New Insights from Pig Models. Annu Rev Anim Biosci 2020; 8:321-354. [PMID: 32069436 DOI: 10.1146/annurev-animal-020518-115142] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pigs are increasingly important animals for modeling human pediatric nutrition and gastroenterology and complementing mechanistic studies in rodents. The comparative advantages in size and physiology of the neonatal pig have led to new translational and clinically relevant models of important diseases of the gastrointestinal tract and liver in premature infants. Studies in pigs have established the essential roles of prematurity, microbial colonization, and enteral nutrition in the pathogenesis of necrotizing enterocolitis. Studies in neonatal pigs have demonstrated the intestinal trophic effects of akey gut hormone, glucagon-like peptide 2 (GLP-2), and its role in the intestinal adaptation process and efficacy in the treatment of short bowel syndrome. Further, pigs have been instrumental in elucidating the physiology of parenteral nutrition-associated liver disease and the means by which phytosterols, fibroblast growth factor 19, and a new generation of lipid emulsions may modify disease. The premature pig will continue to be a valuable model in the development of optimal infant diets (donor human milk, colostrum), specific milk bioactives (arginine, growth factors), gut microbiota modifiers (pre-, pro-, and antibiotics), pharmaceutical drugs (GLP-2 analogs, FXR agonists), and novel diagnostic tools (near-infrared spectroscopy) to prevent and treat these pediatric diseases.
Collapse
Affiliation(s)
- Douglas Burrin
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| | - Per Torp Sangild
- Comparative Pediatrics and Nutrition, University of Copenhagen, DK-1870 Frederiksberg C., Copenhagen, Denmark
| | - Barbara Stoll
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, University of Copenhagen, DK-1870 Frederiksberg C., Copenhagen, Denmark
| | - Randal Buddington
- College of Nursing, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Juan Marini
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
- Department of Pediatrics, Section of Critical Care Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Oluyinka Olutoye
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Robert J Shulman
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| |
Collapse
|
24
|
Relationships of Microbiome Markers With Extraintestinal, Psychological Distress and Gastrointestinal Symptoms, and Quality of Life in Women With Irritable Bowel Syndrome. J Clin Gastroenterol 2020; 54:175-183. [PMID: 30148765 PMCID: PMC6387862 DOI: 10.1097/mcg.0000000000001107] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Altered microbial diversity has been associated with gastrointestinal (GI) symptoms in persons with irritable bowel syndrome (IBS). Less is known about the relationship of microbiome with extraintestinal pain and psychological distress symptoms and quality of life (QOL) in persons with IBS. We aimed to evaluate the relationship of fecal microbiota to GI symptoms, stool consistency, psychological distress, extraintestinal pain, and QOL in participants meeting Rome III criteria for IBS. METHODS Seventy-six women completed a 28-day diary that included GI, stool consistency, psychological distress, and extraintestinal pain ratings. Participants completed the IBS-Specific Quality of Life questionnaire. Stool samples were collected and analyzed by 16S rRNA gene sequencing. Principal component analysis was performed and the first 2 components (PC1, PC2) were used to test relationships among bacterial families and clinical measures. RESULTS Participants were categorized as IBS constipation (n=22), IBS diarrhea (n=39), IBS mixed (n=13), and IBS unsubtyped (n=2). There was a significant group effect for the Firmicutes to Bacteroidetes ratio and PC1. Lower microbial diversity and richness were associated with increased urgency and extraintestinal pain, worse QOL, and looser stools. Lower extraintestinal pain was associated with increased Rikenellaceae, Christensenellaceae, Dehalobabacteriaceae, Oscillospiraceae, Mogibacteriaceae, Ruminococcaceae, Sutterellaceae, Desulfovibrionaceae, and Erysipelotrichaceae abundances. QOL was positively associated with many of these same bacterial families. Higher Firmicutes to Bacteroidetes ratio was positively associated with loose stools. There were no statistically significant relationships between daily psychological distress or abdominal pain and bacterial families. CONCLUSIONS Stool microbial diversity and composition are linked to daily extraintestinal symptoms, stool consistency, and QOL in women with IBS.
Collapse
|
25
|
Arnaud AP, Rome V, Richard M, Formal M, David-Le Gall S, Boudry G. Post-natal co-development of the microbiota and gut barrier function follows different paths in the small and large intestine in piglets. FASEB J 2019; 34:1430-1446. [PMID: 31914707 DOI: 10.1096/fj.201902514r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/04/2019] [Accepted: 11/13/2019] [Indexed: 12/17/2022]
Abstract
Gut microbiota and intestinal barrier co-develop after birth, establishing a homeostatic state whereby mucosal cells cohabit with commensal bacteria. We hypothesized that this post-natal co-development follows different timings depending on the intestinal site considered. Jejunal, ileal, and colonic luminal contents and mucosa were sampled in suckling piglets at post-natal day (PND) 0, 2, 7, 14, and 28. Jejunal, ileal, and colonic luminal microbiota (evaluated by 16S DNA sequencing followed by beta-diversity analysis) clustered at PND2 but colonic microbiota diverge afterwards (P < .05). Mucosal permeability, evaluated in Ussing chambers, increased with age in the jejunum and ileum (P < .05) but not the colon. Expression of pattern recognition receptor (PRR) exhibited different patterns (gradual or sharp increase, decrease, or no change with age, P < .05) depending on PRR and intestinal site considered. Principal component analysis of mucosa data revealed clear clustering of colonic samples, irrespective of the age and clustering of jejunal and ileal samples, with gradual changes with age. Correlation analysis highlighted three families correlating with mucosal parameters: Enterobacteriaceae in the jejunum, Peptostreptococcaceae in the ileum, and Micrococcaceae in the colon. In conclusion, small and large intestine display close microbiota composition early in life but distinct mucosal phenotype and follow very different post-natal development.
Collapse
Affiliation(s)
- Alexis Pierre Arnaud
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France.,Service de chirurgie pédiatrique, CHU rennes, University of Rennes 1, Rennes, France
| | - Véronique Rome
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| | - Marion Richard
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| | - Michèle Formal
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| | | | - Gaëlle Boudry
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| |
Collapse
|
26
|
Lauer JM, Duggan CP, Ausman LM, Griffiths JK, Webb P, Bashaasha B, Agaba E, Turyashemererwa FM, Ghosh S. Unsafe Drinking Water Is Associated with Environmental Enteric Dysfunction and Poor Growth Outcomes in Young Children in Rural Southwestern Uganda. Am J Trop Med Hyg 2019; 99:1606-1612. [PMID: 30350765 PMCID: PMC6283503 DOI: 10.4269/ajtmh.18-0143] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Environmental enteric dysfunction (EED), a subclinical disorder of the small intestine, and poor growth are associated with living in poor water, sanitation, and hygiene (WASH) conditions, but specific risk factors remain unclear. Nested within a birth cohort study, this study investigates relationships among water quality, EED, and growth in 385 children living in southwestern Uganda. Water quality was assessed using a portable water quality test when children were 6 months, and safe water was defined as lacking Escherichia coli contamination. Environmental enteric dysfunction was assessed using the lactulose:mannitol (L:M) test at 12-16 months. Anthropometry and covariate data were extracted from the cohort study, and associations were assessed using linear and logistic regression models. Less than half of the households (43.8%) had safe water, and safe versus unsafe water did not correlate with improved versus unimproved water source. In adjusted linear regression models, children from households with safe water had significantly lower log-transformed (ln) L:M ratios (β: -0.22, 95% confidence interval (CI): -0.44, -0.00) and significantly higher length-for-age (β: 0.29, 95% CI: 0.00, 0.58) and weight-for-age (β: 0.20, 95% CI: 0.05, 0.34) Z-scores at 12-16 months. Furthermore, in adjusted linear regression models, ln L:M ratios at 12-16 months significantly decreased with increasing length-for-age Z-scores at birth, 6 months, and 9 months (β: -0.05, 95% CI: -0.10, -0.004; β: -0.06, 95% CI: -0.11, -0.006; and β: -0.05, 95% CI: -0.09, -0.005, respectively). Overall, our data suggest that programs seeking to improve nutrition should address poor WASH conditions simultaneously, particularly related to household drinking water quality.
Collapse
Affiliation(s)
- Jacqueline M Lauer
- United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts.,Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts.,Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| | - Christopher P Duggan
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts.,United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts
| | - Lynne M Ausman
- United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts.,Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| | - Jeffrey K Griffiths
- Tufts University Cummings School of Veterinary Medicine, Tufts University School of Engineering, Medford, Massachusetts.,Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts
| | - Patrick Webb
- United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts.,Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| | - Bernard Bashaasha
- Department of Agribusiness and Natural Resource Economics, Makerere University, Kampala, Uganda
| | - Edgar Agaba
- United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts
| | | | - Shibani Ghosh
- United States Agency for International Development (USAID) Feed the Future Innovation Lab for Nutrition, Tufts University, Boston, Massachusetts.,Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| |
Collapse
|
27
|
Neal-Kluever A, Fisher J, Grylack L, Kakiuchi-Kiyota S, Halpern W. Physiology of the Neonatal Gastrointestinal System Relevant to the Disposition of Orally Administered Medications. Drug Metab Dispos 2019; 47:296-313. [PMID: 30567878 DOI: 10.1124/dmd.118.084418] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/14/2018] [Indexed: 02/13/2025] Open
Abstract
A thorough knowledge of the newborn (age, birth to 1 month postpartum) infant's gastrointestinal tract (GIT) is critical to the evaluation of the absorption, distribution, metabolism, and excretion (ADME) of orally administered drugs in this population. Developmental changes in the GIT during the newborn period are important for nutrient uptake as well as the disposition of orally administered medications. Some aspects of gastrointestinal function do not mature until driven by increased dietary complexity and nutritional demands later in the postnatal period. The functionalities present at birth, and subsequent maturation, can also impact the ADME parameters of orally administered compounds. This review will examine some specific contributors to the ADME processes in human neonates, as well as what is currently understood about the drivers for their maturation. Key species differences will be highlighted, with a focus on laboratory animals used in juvenile toxicity studies. Because of the gaps and inconsistencies in our knowledge, we will also highlight areas where additional study is warranted to better inform the appropriate use of medicines specifically intended for neonates.
Collapse
Affiliation(s)
- April Neal-Kluever
- US Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, Maryland (A.N.-K.); US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas (J.F.); Independent Consultant, Vienna, Virginia (L.G.); and Genentech Inc., South San Francisco, California (S.K.-K., W.H.)
| | - Jeffrey Fisher
- US Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, Maryland (A.N.-K.); US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas (J.F.); Independent Consultant, Vienna, Virginia (L.G.); and Genentech Inc., South San Francisco, California (S.K.-K., W.H.)
| | - Lawrence Grylack
- US Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, Maryland (A.N.-K.); US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas (J.F.); Independent Consultant, Vienna, Virginia (L.G.); and Genentech Inc., South San Francisco, California (S.K.-K., W.H.)
| | - Satoko Kakiuchi-Kiyota
- US Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, Maryland (A.N.-K.); US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas (J.F.); Independent Consultant, Vienna, Virginia (L.G.); and Genentech Inc., South San Francisco, California (S.K.-K., W.H.)
| | - Wendy Halpern
- US Food and Drug Administration, Center for Food Safety and Applied Nutrition, College Park, Maryland (A.N.-K.); US Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas (J.F.); Independent Consultant, Vienna, Virginia (L.G.); and Genentech Inc., South San Francisco, California (S.K.-K., W.H.)
| |
Collapse
|
28
|
Calarge CA, Devaraj S, Shulman RJ. Gut permeability and depressive symptom severity in unmedicated adolescents. J Affect Disord 2019; 246:586-594. [PMID: 30605877 DOI: 10.1016/j.jad.2018.12.077] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/13/2018] [Accepted: 12/24/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVE This study examined gut permeability in unmedicated adolescents with and without major depressive disorder. METHOD Medically healthy, non-medicated, 12-17 year-old females in a major depressive episode (MDE) or healthy controls, without any psychiatric condition, were enrolled. They completed the Children's Depression Rating Scale-Revised (CDRS-R) and underwent a clinical interview. Preejection period (PEP) and respiratory sinus arrhythmia (RSA) data were collected to measure autonomic nervous system activity. Following an overnight fast, participants ingested lactulose and mannitol and collected urine for 4 hours while still fasting, to examine gut permeability. Plasma cytokines (interleukin 1β, interleukin 6, and tumor necrosis factor α) were measured. Correlational analyses were used to examine the associations between relevant variables. RESULTS 41 female participants (age: 14.8 ± 1.6 years, n = 25 with MDE) were enrolled. PEP, but not RSA, was inversely associated with neurovegetative symptom severity on the CDRS-R (r = -0.31, p < 0.06). In the 30 participants with gut permeability data, the lactulose to mannitol ratio (LMR) was significantly positively associated with depression severity, particularly neurovegetative symptom severity (r = 0.37, p < 0.05). Notably, the association between neurovegetative symptom severity and PEP was substantially reduced after adjusting for LMR. Additionally, depression severity was significantly associated with circulating cytokines. CONCLUSIONS This is the first study to examine gut permeability in unmedicated adolescents, offering preliminary support for a mechanistic pathway linking sympathetic nervous system activation to increased gut permeability and activation of the innate immune system, likely contributing to the emergence of neurovegetative symptoms of depression.
Collapse
Affiliation(s)
- Chadi A Calarge
- Menninger Department of Psychiatry and Behavioral Sciences and Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, 1102 Bates Ave, Suite 790 (C-0790.03) Houston, TX 77030, USA.
| | - Sridevi Devaraj
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Robert J Shulman
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
29
|
Lauer JM, Duggan CP, Ausman LM, Griffiths JK, Webb P, Agaba E, Nshakira N, Tran HQ, Gewirtz AT, Ghosh S. Biomarkers of maternal environmental enteric dysfunction are associated with shorter gestation and reduced length in newborn infants in Uganda. Am J Clin Nutr 2018; 108:889-896. [PMID: 30247538 PMCID: PMC6186209 DOI: 10.1093/ajcn/nqy176] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 07/09/2018] [Indexed: 12/21/2022] Open
Abstract
Background Adverse birth outcomes, including preterm birth and stunting at birth, have long-term health implications. The relation between adverse birth outcomes and chronic, asymptomatic gastrointestinal inflammation (environmental enteric dysfunction-EED) is poorly understood. Objective We aimed to examine the relation between maternal EED and adverse birth outcomes in a sample of pregnant Ugandan women and their newborn infants. Design We conducted a prospective cohort study in Mukono, Uganda. A total of 258 pregnant women were enrolled at their first prenatal visit (∼18 weeks of gestation). EED was measured by urinary lactulose:mannitol (L:M) ratio and serum concentrations of antibodies to the bacterial components flagellin and LPS. Covariates were obtained from survey data collected at 2 time points. Associations were assessed through the use of unadjusted and adjusted simple linear regression models. Results Complete birth outcome data were recorded for 220 infants within 48 h of delivery. Mean ± SD gestational age was 39.7 ± 2.1 wk, and 7% were born preterm. Mean ± SD length and length-for-age z score (LAZ) at birth were 48.1 ± 3.2 cm and -0.44 ± 1.07, respectively. L:M ratio was not associated with any birth outcome. In adjusted models, higher concentrations of natural log-transformed anti-flagellin immunoglobin G (IgG) and anti-LPS IgG were significantly associated with shorter length of gestation (β: -0.89 wk; 95% CI: -1.77, -0.01 wk, and β: -1.01 wk; 95% CI: -1.87, -0.17 wk, respectively) and with reduced length (β: -0.80 cm; 95% CI: -1.55, -0.05 cm, and β: -0.79 cm; 95% CI: -1.54, -0.04 cm, respectively) and LAZ at birth (β -0.44 z score; 95% CI: -0.83, -0.05, and β: -0.40 z score; 95% CI: -0.79, -0.01, respectively). Conclusion Maternal anti-flagellin and anti-LPS IgG concentrations in pregnancy, but not L:M ratio, were associated with shorter gestation and reduced infant length at birth. Further research on the relation between maternal EED and birth outcomes is warranted.
Collapse
Affiliation(s)
- Jacqueline M Lauer
- Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy and,USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA,Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA,Address correspondence to JML (e-mail: )
| | - Christopher P Duggan
- USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA,Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Lynne M Ausman
- Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy and,USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA
| | - Jeffrey K Griffiths
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA,Tufts University Cummings School of Veterinary Medicine and Tufts University School of Engineering, Medford, MA
| | - Patrick Webb
- Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy and,USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA
| | - Edgar Agaba
- USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA
| | | | - Hao Q Tran
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA
| | - Andrew T Gewirtz
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA
| | - Shibani Ghosh
- Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy and,USAID Feed the Future Innovation Lab for Nutrition at Tufts University, Boston, MA
| |
Collapse
|
30
|
Telang S. Lactoferrin: A Critical Player in Neonatal Host Defense. Nutrients 2018; 10:nu10091228. [PMID: 30181493 PMCID: PMC6165050 DOI: 10.3390/nu10091228] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/30/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
Newborn infants are at a high risk for infection due to an under-developed immune system, and human milk has been shown to exhibit substantial anti-infective properties that serve to bolster neonatal defenses against multiple infections. Lactoferrin is the dominant whey protein in human milk and has been demonstrated to perform a wide array of antimicrobial and immunomodulatory functions and play a critical role in protecting the newborn infant from infection. This review summarizes data describing the structure and important functions performed by lactoferrin in protecting the neonate from infection and contributing to the maturation of the newborn innate and adaptive immune systems. We also briefly discuss clinical trials examining the utility of lactoferrin supplementation in the prevention of sepsis and necrotizing enterocolitis in newborn infants. The data reviewed provide rationale for the continuation of studies to examine the effects of lactoferrin administration on the prevention of sepsis in the neonate.
Collapse
Affiliation(s)
- Sucheta Telang
- Division of Neonatology, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA.
- Division of Hematology/Oncology, Department of Medicine, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
31
|
Heitkemper MM, Cain KC, Shulman RJ, Burr RL, Ko C, Hollister EB, Callen N, Zia J, Han CJ, Jarrett ME. Stool and urine trefoil factor 3 levels: associations with symptoms, intestinal permeability, and microbial diversity in irritable bowel syndrome. Benef Microbes 2018; 9:345-355. [PMID: 29633639 DOI: 10.3920/bm2017.0059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previously we showed that urine trefoil factor 3 (TFF3) levels were higher in females with irritable bowel syndrome (IBS) compared to non-IBS females. To assess if TFF3 is associated with symptoms and/or reflect alterations in gastrointestinal permeability and gut microbiota in an IBS population, we correlated stool and urine TFF3 levels with IBS symptoms, intestinal permeability, stool microbial diversity and relative abundance of predominant bacterial families and genera. We also tested the relationship of stool TFF3 to urine TFF3, and compared results based on hormone contraception use. Samples were obtained from 93 females meeting Rome III IBS criteria and completing 4-week symptom diaries. TFF3 levels were measured by ELISA. Permeability was assessed with the urine lactulose/mannitol (L/M) ratio. Stool microbiota was assessed using 16S rRNA. Stool TFF3, but not urine TFF3, was associated positively with diarrhoea and loose stool consistency. Higher stool TFF3 was also associated with lower L/M ratio and microbial diversity. Of the 20 most abundant bacterial families Mogibacteriaceae and Christensenellaceae were inversely related to stool TFF3, with only Christensenellaceae remaining significant after multiple comparison adjustment. There were no significant relationships between stool or urine TFF3 levels and other symptoms, nor between stool and urine levels. In premenopausal females, urine TFF3 levels were higher in those reporting hormone contraception. Collectively these results suggest that higher stool TFF3 levels are associated with IBS symptoms (loose/diarrhoeal stools), lower gut permeability, and altered stool bacteria composition (decreased diversity and decreased Christensenellaceae), which further suggests that TFF3 may be an important marker of host-bacteria interaction.
Collapse
Affiliation(s)
- M M Heitkemper
- 1 Department of Biobehavioral Nursing and Health Informatics, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195-7266, USA
| | - K C Cain
- 2 Department of Biostatistics and Office of Nursing Research, University of Washington, 1959 NE Pacific Street, Seattle, WA, USA
| | - R J Shulman
- 3 Children's Nutrition Research Center, Baylor College of Medicine, Texas Children's Hospital, 6621 Fannin Street, Houston, TX 77030, USA
| | - R L Burr
- 1 Department of Biobehavioral Nursing and Health Informatics, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195-7266, USA
| | - C Ko
- 4 University of Washington Medical Center, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - E B Hollister
- 5 Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Microbiome Center, Texas Children's Hospital, 6621 Fannin Street, Houston, TX 77030, USA
| | - N Callen
- 6 Department of Family and Child Nursing, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - J Zia
- 4 University of Washington Medical Center, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - C J Han
- 1 Department of Biobehavioral Nursing and Health Informatics, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195-7266, USA
| | - M E Jarrett
- 1 Department of Biobehavioral Nursing and Health Informatics, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195-7266, USA
| |
Collapse
|
32
|
Abstract
Diarrhoea is a common problem in the neonatal and suckling foal. In certain circumstances supplemental nutrition is necessary depending on the age of foal, severity of diarrhoea and presence of other systemic manifestations. Nutritional supplementation can be provided either enterally or parenterally. Enteral nutrition is superior to parenteral nutrition because it is the most natural and physiologically sound means to provide nutritional support. Parenteral nutrition may be warranted if the foal is unable to receive or tolerate enteral nutrition. Dextrose alone or with amino acids and lipids can provide appropriate nutrition when enteral feeding is not tolerated. As soon as the foal stabilises enteral feeding can be reintroduced.
Collapse
Affiliation(s)
- B Barr
- Rood and Riddle Equine Hospital Lexington Kentucky USA
| |
Collapse
|
33
|
Agapova SE, Stephenson KB, Divala O, Kaimila Y, Maleta KM, Thakwalakwa C, Ordiz MI, Trehan I, Manary MJ. Additional Common Bean in the Diet of Malawian Children Does Not Affect Linear Growth, but Reduces Intestinal Permeability. J Nutr 2018; 148:267-274. [PMID: 29490090 DOI: 10.1093/jn/nxx013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/18/2017] [Indexed: 01/26/2023] Open
Abstract
Background Chronic malnutrition, as manifested by linear growth faltering, is pervasive among rural African children. Improvements in complementary feeding may decrease the burden of environmental enteric dysfunction (EED) and thus improve growth in children during the critical first 1000 d of development. Objective We tested the hypothesis that systematically including common bean or cowpea into complementary feeding would reduce EED and growth faltering among children in rural Malawi. Methods This was a double-blind clinical trial in which children 12-23 mo of age were randomly assigned to receive complementary feeding with 1 of 3 foods: roasted cowpea or common bean flour, or an isoenergetic amount of corn-soy blend as a control food for 48 wk. Children aged 12-23 mo received 155 kcal/d and thereafter until 35 mo received 200 kcal/d. The primary outcomes were change in length-for-age z score (LAZ) and improvements in a biomarker of EED, the percentage of lactulose (%L) excreted as part of the lactulose:mannitol dual-sugar absorption test. Anthropometric measurements and urinary %L excretion were compared between the 2 intervention groups and the control group separately with the use of linear mixed model analyses for repeated measures. Results A total of 331 children completed the clinical trial. Compliance with the study interventions was excellent, with >90% of the intervention flour consumed as intended. No significant effects on LAZ, change in LAZ, or weight-for-length z score were observed due to either intervention legume, compared to the control. %L was reduced with common bean consumption (effect estimate was -0.07 percentage points of lactulose, P = 0.0007). The lactulose:mannitol test was not affected by the legume intervention. Conclusion The addition of common bean to complementary feeding of rural Malawian children during the second year of life led to an improvement in a biomarker of gut health, although this did not directly translate into improved linear growth. This trial was registered at clinicaltrials.gov as NCT02472301.
Collapse
Affiliation(s)
| | | | - Oscar Divala
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Yankho Kaimila
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Kenneth M Maleta
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Chrissie Thakwalakwa
- School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - M Isabel Ordiz
- Department of Pediatrics, Washington University, St Louis, MO
| | - Indi Trehan
- Department of Pediatrics, Washington University, St Louis, MO.,Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Mark J Manary
- Department of Pediatrics, Washington University, St Louis, MO.,School of Public Health and Family Medicine and Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi.,Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
34
|
Abstract
Human donor milk is the feeding of choice for preterm infants, when own mother's milk is not available. Holder pasteurization is necessary to secure the safety of donor milk, although it can affect milk quality by reduction of nutritional and bioactive components. Recently, research has focused on the potential role of breast milk glucocorticoids for infant development. At this moment, it is unknown whether pasteurization affects milk glucocorticoid levels. Therefore, we assessed whether Holder pasteurization, the most frequently used method nowadays, reduces breast milk cortisol and cortisone levels, using breast milk samples from 30 women who delivered at term. We found tight correlations between pre- and postpasteurization levels of cortisol (R = 0.99) and cortisone (R = 0.98), and good agreement in Passing and Bablok regression analysis. In conclusion, cortisol and cortisone in human term breast milk are not significantly affected by Holder pasteurization.
Collapse
|
35
|
Stephenson KB, Agapova SE, Divala O, Kaimila Y, Maleta KM, Thakwalakwa C, Ordiz MI, Trehan I, Manary MJ. Complementary feeding with cowpea reduces growth faltering in rural Malawian infants: a blind, randomized controlled clinical trial. Am J Clin Nutr 2017; 106:1500-1507. [PMID: 29092882 PMCID: PMC6482976 DOI: 10.3945/ajcn.117.160986] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/04/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Growth faltering is common in rural African children and is attributed to inadequate dietary intake and environmental enteric dysfunction (EED).Objective: We tested the hypothesis that complementary feeding with cowpea or common bean flour would reduce growth faltering and EED in 6-mo-old rural Malawians compared with the control group receiving a corn-soy blend.Design: A prospective, double-blind, randomized controlled clinical trial was conducted in which children received daily feeding for 6 mo (200 kcal/d when 6-9 mo old and 300 kcal/d when 10-12 mo old). The primary outcomes were change in length-for-age z score (LAZ) and improvements in EED, as measured by percentage of lactulose excretion (%L). %L <0.2% was considered normal. Anthropometric measurements and %L through urine were compared between each legume group and the control group with Student's t test.Results: Of the 355 infants enrolled, 291 infants completed the trial, and 288 were breastfed throughout the duration of the study. Cowpea and common bean added 4.6-5.2 g protein/d and 4-5 g indigestible carbohydrate/d to the diet. LAZ and weight-for-height z score were reduced in all 3 groups from 6 to 12 mo of age. The changes in LAZ [mean (95% CI)] for the cowpea, common bean, and control groups from 6 to 9 mo were -0.14 (-0.24, -0.04), -0.27 (-0.38, -0.16), and -0.27 (-0.35, -0.19), respectively. LAZ was reduced less in infants receiving cowpea than in those receiving control food from 6 to 9 mo (P = 0.048). The absolute value of %L did not differ between the dietary groups at 9 mo of age (mean ± SD: 0.30 ± 0.43, 0.23 ± 0.21, and 0.26 ± 0.31 for cowpea, common bean, and control, respectively), nor did the change in %L from 6 to 9 mo.Conclusion: Addition of cowpea to complementary feeding in Malawian infants resulted in less linear growth faltering. This trial was registered at clinicaltrials.gov as NCT02472262.
Collapse
Affiliation(s)
| | | | - Oscar Divala
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - Yankho Kaimila
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - Kenneth M Maleta
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - Chrissie Thakwalakwa
- School of Public Health and Family Medicine, University of Malawi, Blantyre, Malawi
| | - M Isabel Ordiz
- Department of Pediatrics, Washington University, St. Louis, MO
| | - Indi Trehan
- Department of Pediatrics, Washington University, St. Louis, MO,Department of Paediatrics, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Mark J Manary
- Department of Pediatrics, Washington University, St. Louis, MO; .,School of Public Health and Family Medicine and.,Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
36
|
Lee GO, McCormick BJJ, Seidman JC, Kosek MN, Haque R, Olortegui MP, Lima AAM, Bhutta ZA, Kang G, Samie A, Amour C, Mason CJ, Ahmed T, Yori PP, Oliveira DB, Alam D, Babji S, Bessong P, Mduma E, Shrestha SK, Ambikapathi R, Lang DR, Gottlieb M, Guerrant RL, Caulfield LE. Infant Nutritional Status, Feeding Practices, Enteropathogen Exposure, Socioeconomic Status, and Illness Are Associated with Gut Barrier Function As Assessed by the Lactulose Mannitol Test in the MAL-ED Birth Cohort. Am J Trop Med Hyg 2017; 97:281-290. [PMID: 28719336 PMCID: PMC5508897 DOI: 10.4269/ajtmh.16-0830] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 01/31/2017] [Indexed: 12/20/2022] Open
Abstract
The lactulose mannitol (LM) dual sugar permeability test is the most commonly used test of environmental enteropathy in developing countries. However, there is a large but conflicting literature on its association with enteric infection and host nutritional status. We conducted a longitudinal cohort using a single field protocol and comparable laboratory procedures to examine intestinal permeability in multiple, geographically diverse pediatric populations. Using a previously published systematic review to guide the selection of factors potentially associated with LM test results, we examined the relationships between these factors and mucosal breach, represented by percent lactulose excretion; absorptive area, represented by percent mannitol excretion; and gut barrier function, represented by the L/M ratio. A total of 6,602 LM tests were conducted in 1,980 children at 3, 6, 9, and 15 months old; percent lactulose excretion, percent mannitol excretion, and the L/M ratio were expressed as age- and sex-specific normalized values using the Brazil cohort as the reference population. Among the factors considered, recent severe diarrhea, lower socioeconomic status, and recent asymptomatic enteropathogen infections were associated with decreased percent mannitol excretion and higher L/M ratios. Poorer concurrent weight-for-age, infection, and recent breastfeeding were associated with increased percent lactulose excretion and increased L/M ratios. Our results support previously reported associations between the L/M ratio and factors related to child nutritional status and enteropathogen exposure. These results were remarkably consistent across sites and support the hypothesis that the frequency of these exposures in communities living in poverty leads to alterations in gut barrier function.
Collapse
Affiliation(s)
- Gwenyth O. Lee
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan
| | | | - Jessica C. Seidman
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland
| | - Margaret N. Kosek
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | | | - Aldo A. M. Lima
- Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Zulfiqar A. Bhutta
- Center of Excellence in Women and Child Health, the Aga Khan University, Karachi, Pakistan
| | - Gagandeep Kang
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Amidou Samie
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | | | - Carl J. Mason
- Walter Reed/Armed Forces Research Institute of Medical Sciences, Kathmandu, Nepal
| | | | - Pablo Peñataro Yori
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Didar Alam
- Center of Excellence in Women and Child Health, the Aga Khan University, Karachi, Pakistan
| | - Sudhir Babji
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Pascal Bessong
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | | | - Sanjaya K. Shrestha
- Walter Reed/Armed Forces Research Institute of Medical Sciences, Kathmandu, Nepal
| | - Ramya Ambikapathi
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Dennis R. Lang
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland
- Foundation for the NIH, Bethesda, Maryland
| | | | - Richard L. Guerrant
- Division of Infectious Diseases, University of Virginia, Charlottesville, Virginia
| | - Laura E. Caulfield
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | |
Collapse
|
37
|
Saleem B, Okogbule-Wonodi AC, Fasano A, Magder LS, Ravel J, Kapoor S, Viscardi RM. Intestinal Barrier Maturation in Very Low Birthweight Infants: Relationship to Feeding and Antibiotic Exposure. J Pediatr 2017; 183:31-36.e1. [PMID: 28159311 PMCID: PMC5367935 DOI: 10.1016/j.jpeds.2017.01.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 12/07/2016] [Accepted: 01/04/2017] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To test the hypothesis that feeding and antibiotic exposures affect intestinal barrier maturation in preterm infants, we serially measured intestinal permeability (IP) biomarkers in infants <33 weeks gestation (gestational age [GA]) during the first 2 weeks of life. STUDY DESIGN Eligible infants <33 weeks GA were enrolled within 4 days of birth in a prospective study of IP biomarkers (NCT01756040). Study participants received the nonmetabolized sugars lactulose/rhamnose enterally on study days 1, 8, and 15 and lactulose/rhamnose were measured in urine by high-performance liquid chromatography. Serum zonulin and fecal alpha-1-anti-trypsin, 2 other IP markers, were measured by semiquantitative Western blot and ELISA, respectively. RESULTS In a cohort of 43 subjects, the lactulose/rhamnose ratio was increased on day 1 and decreased over 2 weeks, but remained higher in infants born at ≤28 weeks of gestation compared with IP in infants born at >28 weeks of gestation. Exclusive breastmilk feeding was associated with more rapid maturation in intestinal barrier function. A cluster analysis of 35 subjects who had urine samples from all time points revealed 3 IP patterns (cluster 1, normal maturation: n = 20 [57%]); cluster 2, decreased IP during the first week and subsequent substantial increase: n = 5 [14%]); and cluster 3, delayed maturation: n = 10 [29%]). There were trends toward more prolonged antibiotic exposure (P = .092) and delayed initiation of feeding ≥4 days (P = .064) in infants with abnormal IP patterns. CONCLUSIONS Intestinal barrier maturation in preterm infants is GA and postnatal age dependent, and is influenced by feeding with a maturational effect of breastmilk feeding and possibly by antibiotic exposures. TRIAL REGISTRATION ClinicalTrials.gov: NCT01756040.
Collapse
Affiliation(s)
- Bushra Saleem
- Departments of Pediatrics, University of Maryland School of Medicine, Baltimore, MD
| | | | - Alessio Fasano
- MassGeneral Hospital for Children, Center for Celiac Research and Treatment, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA
| | - Laurence S. Magder
- Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD
| | - Jacques Ravel
- Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD,Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - Shiv Kapoor
- Departments of Pediatrics, University of Maryland School of Medicine, Baltimore, MD
| | - Rose M. Viscardi
- Departments of Pediatrics, University of Maryland School of Medicine, Baltimore, MD,Corresponding author (No reprints): Rose M. Viscardi, M.D., University of Maryland School of Medicine, 110 S. Paca Street, 8th Floor, Baltimore, MD 21201, Telephone: (410) 706-1913; Fax: (410) 706-0404;
| |
Collapse
|
38
|
Abstract
Human milk is the preferred diet for preterm infants as it protects against a multitude of NICU challenges, specifically necrotizing enterocolitis. Infants who receive greater than 50% of mother's own milk (MOM) in the 2 weeks after birth have a significantly decreased risk of NEC. An additional factor in the recent declining rates of NEC is the increased utilization of donor human milk (DHM). This creates a bridge until MOM is readily available, thus decreasing the exposure to cow milk protein. Preterm infants are susceptible to NEC due to the immaturity of their gastrointestinal and immune systems. An exclusive human milk diet compensates for these immature systems in many ways such as lowering gastric pH, enhancing intestinal motility, decreasing epithelial permeability, and altering the composition of bacterial flora. Ideally, preterm infants should be fed human milk and avoid bovine protein. A diet consisting of human milk-based human milk fortifier is one way to provide the additional nutritional supplements necessary for adequate growth while receiving the protective benefits of a human milk diet.
Collapse
Affiliation(s)
- Diana Maffei
- Division of Neonatal-Perinatal Medicine, Cohen Children's Medical Center of New York, 269-01 76th Ave, New Hyde Park, NY 11040; Hofstra Northwell School of Medicine, New Hyde Park, NY
| | - Richard J Schanler
- Division of Neonatal-Perinatal Medicine, Cohen Children's Medical Center of New York, 269-01 76th Ave, New Hyde Park, NY 11040; Hofstra Northwell School of Medicine, New Hyde Park, NY.
| |
Collapse
|
39
|
Stoll B, Price PT, Reeds PJ, Chang X, Henry JF, van Goudoever JB, Holst JJ, Burrin DG. Feeding an Elemental Dietvsa Milk-Based Formula Does Not Decrease Intestinal Mucosal Growth in Infant Pigs. JPEN J Parenter Enteral Nutr 2017; 30:32-9. [PMID: 16387897 DOI: 10.1177/014860710603000132] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND We previously showed that the level of enteral nutrient intake determines the rate of intestinal growth in piglets. Our objective was to determine whether providing enteral nutrition in the form of elemental nutrients (glucose, amino acids, lipid [ED]) rather than cow's milk formula (lactose, protein, lipid [FORM]) reduces small intestinal growth and lactase activity. METHODS Three-week-old piglets were fed either ED (n = 7) intragastrically or FORM (n = 6) orally for 6 days. RESULTS Intestinal protein and DNA masses, villus height, and crypt depth were not different in ED and FORM pigs. Crypt cell proliferation, measured by in vivo bromodeoxyuridine labeling, was significantly (p < .05) higher (+37%) in ED than in FORM pigs. Rates of mucosal protein synthesis (%/d), measured by in vivo 2H-leucine incorporation, were higher (p < .05) in ED than FORM (147 vs 89) pigs. Circulating concentrations (pmol/L) of the intestinotrophic peptide, glucagon-like peptide-2 (GLP-2), were also higher (p < .05) in ED than in FORM (148 vs 87) pigs. The mean lactase-specific activity (micromol/min/g) in proximal and distal segments was higher (p < .05) in FORM than in ED (124 vs 58) pigs. CONCLUSIONS We conclude that intestinal mucosal growth and villus morphology are similar in pigs fed ED and FORM, despite higher cell proliferation and protein synthesis rates and lower lactase activity with ED. This implies that elemental diets may be as trophic as polymeric formulas to simultaneously provide nutrition and a stimulus for intestinal growth during bowel rest.
Collapse
Affiliation(s)
- Barbara Stoll
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang AZ, Shulman RJ, Crocker AH, Thakwalakwa C, Maleta KM, Devaraj S, Manary MJ, Trehan I. A Combined Intervention of Zinc, Multiple Micronutrients, and Albendazole Does Not Ameliorate Environmental Enteric Dysfunction or Stunting in Rural Malawian Children in a Double-Blind Randomized Controlled Trial. J Nutr 2017; 147:97-103. [PMID: 27807040 DOI: 10.3945/jn.116.237735] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/09/2016] [Accepted: 10/11/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) and linear growth stunting affect many rural agrarian children in the developing world and contribute to the persistently high rates of stunting that are observed worldwide. Effective interventions to consistently ameliorate EED are lacking. OBJECTIVE We tested whether a bundle of safe and affordable interventions would decrease EED and stunting over 12-24 wk in a cohort of rural Malawian children 12-35 mo old. METHODS This was a randomized, double-blind, placebo-controlled clinical trial in which the intervention group received a single dose of albendazole and 14 d of zinc at enrollment and after 20 wk. The intervention group also received a daily multiple micronutrient powder throughout the 24 wk of study. The primary outcomes were improvements in EED, as measured by the urinary lactulose-to-mannitol ratio (L:M ratio) from dual-sugar absorption testing, and linear growth. Urinary L:M ratios and anthropometric measurements were evaluated after 12 and 24 wk of intervention and compared with a placebo group that did not receive any of these interventions. RESULTS A total of 254 children were enrolled at a mean age of 24 mo; 55% were female. Their mean weight-for-age z score was -1.5, and their mean length-for-age z score was -0.9. After 12 and 24 wk of study, increases in the L:M ratio did not differ between the intervention group (0.071 and 0.088 units, respectively) and the placebo group (0.073 and 0.080 units, respectively) (P = 0.87 and 0.19, respectively). Relative changes in length and weight also did not differ significantly between groups at any time point. CONCLUSION The combined usage of albendazole, zinc, and a daily multiple micronutrient powder did not decrease EED or stunting in this population of agrarian children 12-35 mo old in rural Malawi. Alternative interventions to improve these diseases should be investigated. This trial was registered at clinicaltrials.gov as NCT02253095.
Collapse
Affiliation(s)
- Alfred Z Wang
- University of Texas Southwestern Medical School, Dallas, TX.,Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | - Robert J Shulman
- USDA/Agricultural Research Service Children's Nutrition Research Center, Houston, TX; Departments of.,Pediatrics and
| | - Audrey H Crocker
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | | | | | | | - Mark J Manary
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO; .,USDA/Agricultural Research Service Children's Nutrition Research Center, Houston, TX; Departments of.,School of Public Health and Family Medicine and
| | - Indi Trehan
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO; .,Department of Paediatrics and Child Health, University of Malawi, Blantyre, Malawi
| |
Collapse
|
41
|
McCormick BJJ, Lee GO, Seidman JC, Haque R, Mondal D, Quetz J, Lima AAM, Babji S, Kang G, Shrestha SK, Mason CJ, Qureshi S, Bhutta ZA, Olortegui MP, Yori PP, Samie A, Bessong P, Amour C, Mduma E, Patil CL, Guerrant RL, Lang DR, Gottlieb M, Caulfield LE, Kosek MN. Dynamics and Trends in Fecal Biomarkers of Gut Function in Children from 1-24 Months in the MAL-ED Study. Am J Trop Med Hyg 2016; 96:465-472. [PMID: 27994110 PMCID: PMC5303054 DOI: 10.4269/ajtmh.16-0496] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/06/2016] [Indexed: 12/28/2022] Open
Abstract
Growth and development shortfalls that are disproportionately prevalent in children living in poor environmental conditions are postulated to result, at least in part, from abnormal gut function. Using data from The Etiology, Risk Factors, and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health and Development (MAL-ED) longitudinal cohort study, we examine biomarkers of gut inflammation and permeability in relation to environmental exposures and feeding practices. Trends in the concentrations of three biomarkers, myeloperoxidase (MPO), neopterin (NEO), and α-1-antitrypsin (AAT), are described from fecal samples collected during the first 2 years of each child's life. A total of 22,846 stool samples were processed during the longitudinal sampling of 2,076 children 0–24 months of age. Linear mixed models were constructed to examine the relationship between biomarker concentrations and recent food intake, symptoms of illness, concurrent enteropathogen infection, and socioeconomic status. Average concentrations of MPO, NEO, and AAT were considerably higher than published references for healthy adults. The concentration of each biomarker tended to decrease over the first 2 years of life and was highly variable between samples from each individual child. Both MPO and AAT were significantly elevated by recent breast milk intake. All three biomarkers were associated with pathogen presence, although the strength and direction varied by pathogen. The interpretation of biomarker concentrations is subject to the context of their collection. Herein, we identify that common factors (age, breast milk, and enteric infection) influence the concentration of these biomarkers. Within the context of low- and middle-income communities, we observe concentrations that indicate gut abnormalities, but more appropriate reference standards are needed.
Collapse
Affiliation(s)
| | - Gwenyth O Lee
- Tulane University, New Orleans, Louisiana.,Fogarty International Center/National Institutes of Health, Bethesda, Maryland
| | - Jessica C Seidman
- Fogarty International Center/National Institutes of Health, Bethesda, Maryland
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Dinesh Mondal
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | | | | | | | | | - Sanjaya K Shrestha
- Walter Reed/Armed Forces Research Institute of Medical Science (AFRIMS) Research Unit, Kathmandu, Nepal
| | - Carl J Mason
- Walter Reed/Armed Forces Research Institute of Medical Science (AFRIMS) Research Unit, Kathmandu, Nepal
| | | | | | - Maribel Paredes Olortegui
- Asociacion Benéfica Proyectos en Informatica, Salud, Medicina, y Agricultura (A. B. PRISMA), Iquitos, Peru
| | - Pablo Peñataro Yori
- Asociacion Benéfica Proyectos en Informatica, Salud, Medicina, y Agricultura (A. B. PRISMA), Iquitos, Peru
| | | | | | | | | | | | | | - Dennis R Lang
- Foundation for the National Institutes of Health, Bethesda, Maryland.,Fogarty International Center/National Institutes of Health, Bethesda, Maryland
| | - Michael Gottlieb
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | - Laura E Caulfield
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Margaret N Kosek
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
42
|
Schuck-Phan A, Phan T, Dawson PA, Dial EJ, Bell C, Liu Y, Rhoads JM, Lichtenberger LM. Formula Feeding Predisposes Gut to NSAID-Induced Small Intestinal Injury. ACTA ACUST UNITED AC 2016; 6. [PMID: 31565540 DOI: 10.4172/2161-1459.1000222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objectives Breast feeding protects infants from many diseases, including necrotizing enterocolitis, peptic ulceration and infectious diarrhea. Conversely, maternal separation stress and Non-Steroidal Anti-Inflammatory Drugs (NSAID's) can induce intestinal injury and bleeding. This study aimed to evaluate in suckling rats if maternal separation/formula feeding leads to increased intestinal sensitivity to indomethacin (indo)-induced intestinal injury and to look at potential mechanisms involved. Methods Nine-day-old rats were dam-fed or separated/trained to formula-feed for 6 days prior to indo administration (5 mg/kg/day) or saline (control) for 3 days. Intestinal bleeding and injury were assessed by measuring luminal and Fecal Hemoglobin (Hob) and jejunal histology. Maturation of the intestine was assessed by measuring luminal bile acids, jejunal sucrase, serum corticosterone, and mRNA expression of ileal Apical Sodium-Dependent Bile Acid Transporter (ASBT). Results At 17 days, formula-fed indo-treated pups had a 2-fold increase in luminal Hb compared to formula-fed control pups and had evidence of morphological injury to the small intestinal mucosa as observed at the light microscopic level, whereas indo had no effect on dam-fed littermates. In addition, formula-fed rats had significant increases in luminal bile acid, sucrase specific activity, serum corticosterone, and expression of ASBT mRNA compared to dam-fed rats. Conclusion Maternal separation stress may cause early intestinal maturational changes induced by corticosteroid release, including increased epithelial exposure to bile acids. These maturational changes may have a sensitizing rather than protective effect against indo-induced injury in the new-born.
Collapse
Affiliation(s)
- A Schuck-Phan
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Texas Health Science Center, Houston, TX, USA
| | - T Phan
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
| | - P A Dawson
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - E J Dial
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
| | - C Bell
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Texas Health Science Center, Houston, TX, USA
| | - Y Liu
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Texas Health Science Center, Houston, TX, USA
| | - J M Rhoads
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Texas Health Science Center, Houston, TX, USA
| | - L M Lichtenberger
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
43
|
Environmental Enteric Dysfunction Is Associated With Poor Linear Growth and Can Be Identified by Host Fecal mRNAs. J Pediatr Gastroenterol Nutr 2016; 63:453-459. [PMID: 27347722 PMCID: PMC5084633 DOI: 10.1097/mpg.0000000000001315] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Environmental enteric dysfunction (EED) can be assessed by the lactulose:mannitol (L:M) test. Our objective was to determine if selected host fecal transcripts were correlated with EED, and whether transcripts and clinical characteristics could be used to predict EED in rural African children. METHODS Demographic and sanitation characteristics, along with L:M testing and host fecal transcript analyses from 798 asymptomatic Malawian children aged 12 to 61 months were compared with linear growth over the subsequent 3 months. Fecal host mRNA analysis included quantification of expression of 18 transcripts associated with L:M. Permeability was categorized as normal (L:M ≤ 0.15), moderate (0.15<L:M<0.45) and severe (L:M ≥ 0.45), and random forest predictive models were created. RESULTS L:M was inversely correlated with linear growth over the subsequent 3 months (r = -0.32, P < 0.001) and severe EED was associated with stunting (P < 0.0001). Age younger than 24 months, weight-for-height z score <0, domesticated animals in the child's sleep environment, lack of a pit latrine combined with a potentially contaminated water source, and a recent history of diarrhea were associated with severe EED. A random forest model using CD53, HLA-DRA, MUC12, and TNF was 84% sensitive for severe EED and 83% sensitive for no EED. CONCLUSIONS Selected host fecal transcripts can be used in a random forest model as a noninvasive biomarker for categories of EED in rural African children.
Collapse
|
44
|
|
45
|
Abstract
To discuss the potential clinical benefits of lactoferrin in preterm and term infants, as well as in young children and to review information on the burden of neonatal sepsis. Current evidence on the mechanisms that explain the role of human milk in the neonatal and infant anti-infective responses will be briefly reviewed and preclinical research data on the potential mechanisms of action by which lactoferrin may impact infant gut health, gut immune development and functions, including the lactoferrin effects on the neonatal microbiome, will be examined. Finally, updated translational research on lactoferrin will be presented and discussed and the current evidence from prospective randomized controlled trials in neonates, infants, and toddlers will be analyzed. These randomized controlled trials demonstrate that lactoferrin has a clinically significant impact on feeding, the microbiome, and clinical outcomes in neonates and infants.
Collapse
Affiliation(s)
- Paolo Manzoni
- Neonatology and Neonatal Intensive Care Unit, S. Anna Hospital, AO Città della Salute e della Scienza, Torino, Italy; Charity and Scientific Foundation "Crescere insieme al S.Anna-ONLUS", Torino, Italy.
| |
Collapse
|
46
|
Zhang GQ, Hu HJ, Liu CY, Shakya S, Li ZY. Probiotics for Preventing Late-Onset Sepsis in Preterm Neonates: A PRISMA-Compliant Systematic Review and Meta-Analysis of Randomized Controlled Trials. Medicine (Baltimore) 2016; 95:e2581. [PMID: 26937897 PMCID: PMC4778994 DOI: 10.1097/md.0000000000002581] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The effect of probiotics on late-onset sepsis (LOS) in preterm neonates remains controversial. The authors systematically reviewed the literature to investigate whether enteral probiotic supplementation reduced the risk of LOS in preterm neonates in neonatal intensive care units.PubMed, Embase, and Cochrane Central Register of Controlled Trials were systematically searched for randomized controlled trials (RCTs) regarding the effect of probiotics in preterm neonates. The primary outcome was culture-proven bacterial and/or fungal sepsis. The Mantel-Haenszel method with random-effects model was used to calculate pooled relative risks (RRs) and 95% confidence intervals (CIs).Twenty-seven trials were included in our review, and 25 trials involving 6104 preterm neonates were statistically analyzed. Pooled analysis indicated that enteral probiotic supplementation significantly reduced the risk of any sepsis (25 RCTs; RR 0.83, 95% CI 0.73-0.94; I = 26%), bacterial sepsis (11 RCTs; RR 0.82, 95% CI 0.71-0.95; I = 0%), and fungal sepsis (6 RCTs; RR 0.57, 95% CI 0.41-0.78; I = 0%). This beneficial effect remains in very low birth weight infants (<1500 g) (19 RCTs; RR 0.86, 95% CI 0.75-0.97; I = 18%), but not in extremely low birth weight infants (<1000 g) (3 RCTs; RR 0.73, 95% CI 0.45-1.19; I = 53%). All the included trials reported no systemic infection caused by the supplemental probiotic organisms.Current evidence indicates that probiotic supplementation is safe, and effective in reducing the risk of LOS in preterm neonates in neonatal intensive care units. Further studies are needed to address the optimal probiotic organism, dosing, timing, and duration. High-quality and adequately powered RCTs regarding the efficacy and safety of the use of probiotics in extremely low birth weight infants are still warranted.
Collapse
Affiliation(s)
- Guo-Qiang Zhang
- From the Department of Gastroenterology (G-QZ, H-JH, SS, Z-YL) and Department of Nephrology (C-YL), Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Children's Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
47
|
Somani AA, Thelen K, Zheng S, Trame MN, Coboeken K, Meyer M, Schnizler K, Ince I, Willmann S, Schmidt S. Evaluation of changes in oral drug absorption in preterm and term neonates for Biopharmaceutics Classification System (BCS) class I and II compounds. Br J Clin Pharmacol 2015; 81:137-47. [PMID: 26302359 DOI: 10.1111/bcp.12752] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/11/2015] [Accepted: 08/17/2015] [Indexed: 12/24/2022] Open
Abstract
AIMS Evidence suggests that the rate of oral drug absorption changes during early childhood. Yet, respective clinical implications are currently unclear, particularly for preterm neonates. The objective of this study was to evaluate changes in oral drug absorption after birth for different Biopharmaceutics Classification System (BCS) class I and II compounds to better understand respective implications for paediatric pharmacotherapy. METHODS Two paradigm compounds were selected for BCS class I (paracetamol (acetaminophen) and theophylline) and II (indomethacin and ibuprofen), respectively, based on the availability of clinical literature data following intravenous and oral dosing. A comparative population pharmacokinetic analysis was performed in a step-wise manner in NONMEM® 7.2 to characterize and predict changes in oral drug absorption after birth for paracetamol, theophylline and indomethacin. RESULTS A one compartment model with an age-dependent maturation function for oral drug absorption was found appropriate to characterize the pharmacokinetics of paracetamol. Our findings indicate that the rate at which a drug is absorbed from the GI tract reaches adult levels within about 1 week after birth. The maturation function for paracetamol was found applicable to theophylline and indomethacin once solubility limitations were overcome via drug formulation. The influence of excipients on solubility and, hence, oral bioavailability was confirmed for ibuprofen, a second BCS class II compound. CONCLUSIONS The findings of our study suggest that the processes underlying changes in oral drug absorption after birth are drug-independent and that the maturation function identified for paracetamol may be generally applicable to other BCS class I and II compounds for characterizing drug absorption in preterm as well as term neonates.
Collapse
Affiliation(s)
- Amit A Somani
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Kirstin Thelen
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - Songmao Zheng
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Mirjam N Trame
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Katrin Coboeken
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - Michaela Meyer
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - Katrin Schnizler
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - Ibrahim Ince
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - Stefan Willmann
- Computational Systems Biology, Bayer Technology Services GmbH, Leverkusen, Germany
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| |
Collapse
|
48
|
Clinical characteristics associated with postoperative intestinal epithelial barrier dysfunction in children with congenital heart disease. Pediatr Crit Care Med 2015; 16:37-44. [PMID: 25162512 PMCID: PMC4286428 DOI: 10.1097/pcc.0000000000000256] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Children with congenital heart disease have loss of intestinal epithelial barrier function, which increases their risk for postoperative sepsis and organ dysfunction. We do not understand how postoperative cardiopulmonary support or the inflammatory response to cardiopulmonary bypass might alter intestinal epithelial barrier function. We examined variation in a panel of plasma biomarkers to reflect intestinal epithelial barrier function (cellular and paracellular) after cardiopulmonary bypass and in response to routine ICU care. DESIGN Prospective cohort. SETTING University medical center cardiac ICU. PATIENTS Twenty children aged between newborn and 18 years undergoing repair or palliation of congenital heart disease with cardiopulmonary bypass. INTERVENTIONS We measured baseline and repeated plasma intestinal fatty acid-binding protein, citrulline, claudin 3, and dual sugar permeability testing to reflect intestinal epithelial integrity, epithelial function, paracellular integrity, and paracellular function, respectively. We measured baseline and repeated plasma proinflammatory (interleukin-6, tumor necrosis factor-α, and interferon-γ) and anti-inflammatory (interleukin-4 and interleukin-10) cytokines, known to modulate intestinal epithelial barrier function in murine models of cardiopulmonary bypass. MEASUREMENTS AND MAIN RESULTS All patients had abnormal baseline intestinal fatty acid-binding protein concentrations (mean, 3,815.5 pg/mL; normal, 41-336 pg/mL). Cytokine response to cardiopulmonary bypass was associated with early, but not late, changes in plasma concentrations of intestinal fatty acid-binding protein 2 and citrulline. Variation in biomarker concentrations over time was associated with aspects of ICU care indicating greater severity of illness: claudin 3, intestinal fatty acid-binding protein 2, and dual sugar permeability test ratio were associated with symptoms of feeding intolerance (p < 0.05), whereas intestinal fatty acid-binding protein was positively associated with vasoactive-inotrope score (p = 0.04). Citrulline was associated with larger arteriovenous oxygen saturation difference (p = 0.04) and had a complex relationship with vasoactive-inotrope score. CONCLUSIONS Children undergoing cardiopulmonary bypass for repair or palliation of congenital heart disease are at risk for intestinal injury and often present with evidence for loss of intestinal epithelial integrity preoperatively. Greater severity of illness requiring increased cardiopulmonary support rather than the inflammatory response to cardiopulmonary bypass seems to mediate late postoperative intestinal epithelial barrier function.
Collapse
|
49
|
Smith HE, Ryan KN, Stephenson KB, Westcott C, Thakwalakwa C, Maleta K, Cheng JY, Brenna JT, Shulman RJ, Trehan I, Manary MJ. Multiple micronutrient supplementation transiently ameliorates environmental enteropathy in Malawian children aged 12-35 months in a randomized controlled clinical trial. J Nutr 2014; 144:2059-65. [PMID: 25411039 DOI: 10.3945/jn.114.201673] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Environmental enteropathy (EE) is subclinical, diffuse villous atrophy characterized by T cell infiltration of the small intestinal mucosa associated with nutrient malabsorption and stunting. EE is assessed by the lactulose:mannitol (L:M) test, whereby nonmetabolized sugars are ingested and quantified in the urine. Multiple micronutrient (MN) deficiency morphologically mimics EE, and ω-3 (n-3) polyunsaturated fatty acids reduce mucosal inflammation in Crohn disease. OBJECTIVE We tested the hypothesis that supplementary MNs, with or without fish oil (FO), would improve L:M in rural Malawian children aged 1-3 y compared with a control (C) group receiving a placebo. METHODS The MNs and FO provided the Recommended Dietary Intake for 26 vitamins, minerals, eicosapentaenoic acid, and docosahexaenoic acid. This was a 3-arm, randomized, double-blind, placebo-controlled clinical trial, with the primary outcomes being the change in L:M (ΔL:M) after 12 and 24 wk of supplementation. Comparisons were made for ΔL:M after 12 and 24 wk within each group by using a Wilcoxon matched pairs signed rank test, because the data are not normally distributed. RESULTS A total of 230 children had specimens adequate for analysis; all had an abnormal baseline L:M, defined as >0.10. After 12 wk, children who received MNs + FO had a ΔL:M [mean (95% CI)] of -0.10 (-0.04, -0.15; P = 0.001), and children receiving only MNs had ΔL:M of -0.12 (-0.03, -0.21; P = 0.002). After 24 wk, children who received MNs + FO had a ΔL:M of -0.09 (-0.03, -0.15; P = 0.001); children receiving only MNs had a ΔL:M of -0.11 (-0.02, -0.20; P = 0.001), and the C group had ΔL:M of -0.07 (0.02, -0.16); P = 0.002). Linear growth was similar in all groups, ∼4.3 cm over 24 wk. CONCLUSION Although the effect was modest, these data suggest MNs can transiently ameliorate EE in rural African children. The trial was registered at clinicaltrials.gov as NCT01593033.
Collapse
Affiliation(s)
- Hannah E Smith
- Department of Pediatrics, Washington University, St. Louis, MO
| | - Kelsey N Ryan
- Department of Pediatrics, Washington University, St. Louis, MO
| | | | - Claire Westcott
- Department of Pediatrics, Washington University, St. Louis, MO
| | | | - Ken Maleta
- College of Medicine, University of Malawi, Blantyre, Malawi
| | | | - J Thomas Brenna
- Division of Nutritional Sciences, Cornell University, Ithaca, NY; and
| | - Robert J Shulman
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Indi Trehan
- Department of Pediatrics, Washington University, St. Louis, MO; College of Medicine, University of Malawi, Blantyre, Malawi
| | - Mark J Manary
- Department of Pediatrics, Washington University, St. Louis, MO; College of Medicine, University of Malawi, Blantyre, Malawi; Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
50
|
Reducing necrotizing enterocolitis in very low birth weight infants using quality-improvement methods. J Perinatol 2014; 34:850-7. [PMID: 25010221 PMCID: PMC4216600 DOI: 10.1038/jp.2014.123] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 05/12/2014] [Accepted: 05/14/2014] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Owing to a rise in necrotizing enterocolitis (NEC, stage ⩾ 2) among very low birth weight (VLBW, birth weight <1500 g) infants from 4% in 2005 to 2006 to 10% in 2007 to 2008, we developed and implemented quality improvement (QI) initiatives. The objective was to evaluate the impact of QI initiatives on NEC incidence in VLBW infants. STUDY DESIGN In September 2009, we developed an NEC QI multidisciplinary team that conducted literature reviews and reviewed practices from other institutions to develop a feeding protocol, which was implemented in December 2009. The team tracked intervention compliance and occurrence of NEC stage ⩾ 2. In May 2010, we reviewed our nasogastric tube practice and relevant literature to develop a second intervention that reduced nasogastric tube indwelling time. The infants were divided into three groups: baseline (January 2008 to Novovember 2009, n219), QI phase 1 (December 2009 to May 2010, n62) and QI phase 2 (June 2010 to November 2011, n170). RESULT The NEC incidence did not decrease after implementation of the feeding protocol in QI phase 1 (19.4%) but did decline significantly after changing nasogastric tube management in QI phase 2 (2.9%). Multivariable logistic regression analysis demonstrated a significant relationship between QI phase and the incidence of NEC. CONCLUSION QI initiatives were effective in decreasing NEC incidence in our high human milk-feeding NICU. Nasogastric tube bacterial contamination may have contributed to our peak in NEC incidence.
Collapse
|