1
|
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
|
2
|
Li L, Sun W, Cai Y, Feng Z, Yu Y, Yang Z, Zhu X. The clinical characteristics and risk factors analysis within one week before the onset of necrotizing enterocolitis. Sci Rep 2024; 14:22380. [PMID: 39333703 PMCID: PMC11436905 DOI: 10.1038/s41598-024-73212-2] [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] [Received: 05/31/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
There are considerable researches on risk factors for necrotizing enterocolitis (NEC), focusing primarily on the entire course before onset. However, fewer studies address risk factors within the brief period before NEC occurrence. The current study aims to retrospectively analyze the clinical data of NEC patients while focusing on relevant risk factors in the preceding week of NEC onset. Infants born between January 2019 and December 2021 at Suzhou Municipal Hospital and Suzhou University Children's Hospital with a birth weight < 1500 g or a gestational age < 32 weeks were included. Around 54 NEC patients and 180 controls were recruited in the study. NEC patients satisfying the inclusion criteria formed the case group, while a 1:4 matching principle helped select the control group based on gestational age and birth weight. A statistically significant difference was observed between groups when red blood cell transfusions were compared the week before NEC onset (adjusted OR and 95% CI 2.16 (1.10, 4.24)). Broad-spectrum antibiotic usage before NEC occurrence was significantly lower in the NEC group than in the control group (adjusted OR and 95% CI 0.95 (0.91, 0.99)). A statistically significant difference was observed between groups while comparing patent ductus arteriosus (PDA) (adjusted OR and 95% CI 2.45 (1.23, 4.91)). The indication for packed red blood cell transfusion should be strictly controlled. Moreover, close monitoring of the patient's condition for NEC occurrence should be conducted within one-week post-transfusion. Accurately identifying infections and using broad-spectrum antibiotics can reduce the incidence of NEC.
Collapse
Affiliation(s)
- Lili Li
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
- Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Wenqiang Sun
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Yan Cai
- Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zongtai Feng
- Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yun Yu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Zuming Yang
- Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| | - Xueping Zhu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
3
|
Giugliano FP, Navis M, Ouahoud S, Garcia TM, Kreulen IA, Ferrantelli E, Meisner S, Vermeulen JL, van Roest M, Billaud JN, Koster J, Dawood Y, de Bakker BS, Picavet-Havik DI, Schimmel IM, van der Wel NN, Koelink PJ, Wildenberg ME, Derikx JP, de Jonge WJ, Renes IB, van Elburg RM, Muncan V. Pro-inflammatory T cells-derived cytokines enhance the maturation of the human fetal intestinal epithelial barrier. iScience 2024; 27:109909. [PMID: 38812539 PMCID: PMC11134877 DOI: 10.1016/j.isci.2024.109909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/31/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
Small intestine (SI) maturation during early life is pivotal in preventing the onset of gut diseases. In this study we interrogated the milestones of SI development by gene expression profiling and ingenuity pathway analyses. We identified a set of cytokines as main regulators of changes observed across different developmental stages. Upon cytokines stimulation, with IFNγ as the most contributing factor, human fetal organoids (HFOs) increase brush border gene expression and enzyme activity as well as trans-epithelial electrical resistance. Electron microscopy revealed developed brush border and loss of fetal cell characteristics in HFOs upon cytokine stimulation. We identified T cells as major source of IFNγ production in the fetal SI lamina propria. Co-culture of HFOs with T cells recapitulated the major effects of cytokine stimulation. Our findings underline pro-inflammatory cytokines derived from T cells as pivotal factors inducing functional SI maturation in vivo and capable of modulating the barrier maturation of HFOs in vitro.
Collapse
Affiliation(s)
- Francesca P. Giugliano
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Marit Navis
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Sarah Ouahoud
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Tânia Martins Garcia
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Irini A.M. Kreulen
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Evelina Ferrantelli
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Sander Meisner
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Jacqueline L.M. Vermeulen
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Manon van Roest
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-Noël Billaud
- QIAGEN Digital Insights, 1001 Marshall Street, Redwood City, CA, USA
- DNAnexus, 204 El Camino Real, Mountain View, CA, USA
| | - Jan Koster
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Yousif Dawood
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development research institute, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Bernadette S. de Bakker
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development research institute, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Daisy I. Picavet-Havik
- Department of Medical Biology, Electron Microscopy Center Amsterdam, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Irene M. Schimmel
- Department of Medical Biology, Electron Microscopy Center Amsterdam, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Nicole N. van der Wel
- Department of Medical Biology, Electron Microscopy Center Amsterdam, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Pim J. Koelink
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Manon E. Wildenberg
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Joep P.M. Derikx
- Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Wouter J. de Jonge
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Ingrid B. Renes
- Department of Pediatrics, Amsterdam University Medical Center (AUMC), Emma Children’s Hospital, Amsterdam, the Netherlands
- Danone Nutricia Research, Utrecht, the Netherlands
| | - Ruurd M. van Elburg
- Department of Pediatrics, Amsterdam University Medical Center (AUMC), Emma Children’s Hospital, Amsterdam, the Netherlands
| | - Vanesa Muncan
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
4
|
van Gorp C, de Lange IH, Hütten MC, López-Iglesias C, Massy KRI, Kessels L, Knoops K, Cuijpers I, Sthijns MMJPE, Troost FJ, van Gemert WG, Spiller OB, Birchenough GMH, Zimmermann LJI, Wolfs TGAM. Antenatal Ureaplasma Infection Causes Colonic Mucus Barrier Defects: Implications for Intestinal Pathologies. Int J Mol Sci 2024; 25:4000. [PMID: 38612809 PMCID: PMC11011967 DOI: 10.3390/ijms25074000] [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] [Received: 02/29/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
Chorioamnionitis is a risk factor for necrotizing enterocolitis (NEC). Ureaplasma parvum (UP) is clinically the most isolated microorganism in chorioamnionitis, but its pathogenicity remains debated. Chorioamnionitis is associated with ileal barrier changes, but colonic barrier alterations, including those of the mucus barrier, remain under-investigated, despite their importance in NEC pathophysiology. Therefore, in this study, the hypothesis that antenatal UP exposure disturbs colonic mucus barrier integrity, thereby potentially contributing to NEC pathogenesis, was investigated. In an established ovine chorioamnionitis model, lambs were intra-amniotically exposed to UP or saline for 7 d from 122 to 129 d gestational age. Thereafter, colonic mucus layer thickness and functional integrity, underlying mechanisms, including endoplasmic reticulum (ER) stress and redox status, and cellular morphology by transmission electron microscopy were studied. The clinical significance of the experimental findings was verified by examining colon samples from NEC patients and controls. UP-exposed lambs have a thicker but dysfunctional colonic mucus layer in which bacteria-sized beads reach the intestinal epithelium, indicating undesired bacterial contact with the epithelium. This is paralleled by disturbed goblet cell MUC2 folding, pro-apoptotic ER stress and signs of mitochondrial dysfunction in the colonic epithelium. Importantly, the colonic epithelium from human NEC patients showed comparable mitochondrial aberrations, indicating that NEC-associated intestinal barrier injury already occurs during chorioamnionitis. This study underlines the pathogenic potential of UP during pregnancy; it demonstrates that antenatal UP infection leads to severe colonic mucus barrier deficits, providing a mechanistic link between antenatal infections and postnatal NEC development.
Collapse
Affiliation(s)
- Charlotte van Gorp
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Ilse H. de Lange
- Department of Pediatrics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Matthias C. Hütten
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
- Neonatology, Department of Pediatrics, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Carmen López-Iglesias
- Microscopy CORE Lab, Maastricht Multimodal Molecular Imaging Institute (M4I), Maastricht University, 6211 LK Maastricht, The Netherlands; (C.L.-I.); (K.K.)
| | - Kimberly R. I. Massy
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Lilian Kessels
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Kèvin Knoops
- Microscopy CORE Lab, Maastricht Multimodal Molecular Imaging Institute (M4I), Maastricht University, 6211 LK Maastricht, The Netherlands; (C.L.-I.); (K.K.)
| | - Iris Cuijpers
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Mireille M. J. P. E. Sthijns
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Freddy J. Troost
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Wim G. van Gemert
- Department of Surgery, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Owen B. Spiller
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XW, UK;
| | - George M. H. Birchenough
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, 40530 Gothenburg, Sweden;
| | - Luc J. I. Zimmermann
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Tim G. A. M. Wolfs
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
- Department of Biomedical Engineering (BMT), School for Cardiovascular Diseases (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
5
|
Mortensen JS, Bohr SSR, Krog LS, Bøtker JP, Kapousidou V, Saaby L, Hatzakis NS, Mørck Nielsen H, Nguyen DN, Rønholt S. Neonatal intestinal mucus barrier changes in response to maturity, inflammation, and sodium decanoate supplementation. Sci Rep 2024; 14:7665. [PMID: 38561398 PMCID: PMC10985073 DOI: 10.1038/s41598-024-58356-5] [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] [Received: 12/20/2023] [Accepted: 03/28/2024] [Indexed: 04/04/2024] Open
Abstract
The integrity of the intestinal mucus barrier is crucial for human health, as it serves as the body's first line of defense against pathogens. However, postnatal development of the mucus barrier and interactions between maturity and its ability to adapt to external challenges in neonatal infants remain unclear. In this study, we unveil a distinct developmental trajectory of the mucus barrier in preterm piglets, leading to enhanced mucus microstructure and reduced mucus diffusivity compared to term piglets. Notably, we found that necrotizing enterocolitis (NEC) is associated with increased mucus diffusivity of our large pathogen model compound, establishing a direct link between the NEC condition and the mucus barrier. Furthermore, we observed that addition of sodium decanoate had varying effects on mucus diffusivity depending on maturity and health state of the piglets. These findings demonstrate that regulatory mechanisms governing the neonatal mucosal barrier are highly complex and are influenced by age, maturity, and health conditions. Therefore, our results highlight the need for specific therapeutic strategies tailored to each neonatal period to ensure optimal gut health.
Collapse
Affiliation(s)
- Janni Støvring Mortensen
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Søren S-R Bohr
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
- Department of Chemistry and Nanoscience Center, Faculty of Science, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
| | - Lasse Skjoldborg Krog
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Johan Peter Bøtker
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Vaya Kapousidou
- Department of Chemistry and Nanoscience Center, Faculty of Science, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
| | - Lasse Saaby
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
- Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| | - Nikos S Hatzakis
- Department of Chemistry and Nanoscience Center, Faculty of Science, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
- NovoNordisk Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Hanne Mørck Nielsen
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Dyrlægevej 68, 1870, Frederiksberg C, Denmark.
| | - Stine Rønholt
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark.
| |
Collapse
|
6
|
Zhang W, Zhang Q, Cao Z, Zheng L, Hu W. Physiologically Based Pharmacokinetic Modeling in Neonates: Current Status and Future Perspectives. Pharmaceutics 2023; 15:2765. [PMID: 38140105 PMCID: PMC10747965 DOI: 10.3390/pharmaceutics15122765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Rational drug use in special populations is a clinical problem that doctors and pharma-cists must consider seriously. Neonates are the most physiologically immature and vulnerable to drug dosing. There is a pronounced difference in the anatomical and physiological profiles be-tween neonates and older people, affecting the absorption, distribution, metabolism, and excretion of drugs in vivo, ultimately leading to changes in drug concentration. Thus, dose adjustments in neonates are necessary to achieve adequate therapeutic concentrations and avoid drug toxicity. Over the past few decades, modeling and simulation techniques, especially physiologically based pharmacokinetic (PBPK) modeling, have been increasingly used in pediatric drug development and clinical therapy. This rigorously designed and verified model can effectively compensate for the deficiencies of clinical trials in neonates, provide a valuable reference for clinical research design, and even replace some clinical trials to predict drug plasma concentrations in newborns. This review introduces previous findings regarding age-dependent physiological changes and pathological factors affecting neonatal pharmacokinetics, along with their research means. The application of PBPK modeling in neonatal pharmacokinetic studies of various medications is also reviewed. Based on this, we propose future perspectives on neonatal PBPK modeling and hope for its broader application.
Collapse
Affiliation(s)
| | | | | | - Liang Zheng
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| |
Collapse
|
7
|
Martins RDS, Kooi EMW, Poelstra K, Hulscher JBF. The role of intestinal alkaline phosphatase in the development of necrotizing enterocolitis. Early Hum Dev 2023; 183:105797. [PMID: 37300991 DOI: 10.1016/j.earlhumdev.2023.105797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating neonatal disease that affects neonates worldwide and often leads to high morbidity and mortality rates. Despite extensive research, the cause of NEC remains unclear, and current treatment options are limited. An important novel finding is the potential role of intestinal Alkaline Phosphatase (IAP) in both pathogenesis and treatment of NEC. IAP can play a vital role in detoxifying liposaccharides (LPS), a key mediator of many pathological processes, thereby reducing the inflammatory response associated with NEC. Furthermore, IAP can help prevent dysbiosis, improve intestinal perfusion, and promote autophagy. In this comprehensive review, we present evidence of the possible connection between IAP and the LPS/Toll-like receptor 4 (TLR4) pathway, impaired gut immunity, and dysbiosis in the preterm gut. Based on these findings, the administration of exogenous IAP might provide promising preventive and therapeutic avenues for the management of NEC.
Collapse
Affiliation(s)
- Raquel Dos Santos Martins
- Division of Pediatric Surgery, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Elisabeth M W Kooi
- Division of Neonatology, Department of Pediatrics, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Klaas Poelstra
- Department of Nanomedicine and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, the Netherlands
| | - Jan B F Hulscher
- Division of Pediatric Surgery, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
8
|
Developmental Pharmacokinetics of Antibiotics Used in Neonatal ICU: Focus on Preterm Infants. Biomedicines 2023; 11:biomedicines11030940. [PMID: 36979919 PMCID: PMC10046592 DOI: 10.3390/biomedicines11030940] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/22/2023] Open
Abstract
Neonatal Infections are among the most common reasons for admission to the intensive care unit. Neonatal sepsis (NS) significantly contributes to mortality rates. Empiric antibiotic therapy of NS recommended by current international guidelines includes benzylpenicillin, ampicillin/amoxicillin, and aminoglycosides (gentamicin). The rise of antibacterial resistance precipitates the growth of the use of antibiotics of the Watch (second, third, and fourth generations of cephalosporines, carbapenems, macrolides, glycopeptides, rifamycins, fluoroquinolones) and Reserve groups (fifth generation of cephalosporines, oxazolidinones, lipoglycopeptides, fosfomycin), which are associated with a less clinical experience and higher risks of toxic reactions. A proper dosing regimen is essential for effective and safe antibiotic therapy, but its choice in neonates is complicated with high variability in the maturation of organ systems affecting drug absorption, distribution, metabolism, and excretion. Changes in antibiotic pharmacokinetic parameters result in altered efficacy and safety. Population pharmacokinetics can help to prognosis outcomes of antibiotic therapy, but it should be considered that the neonatal population is heterogeneous, and this heterogeneity is mainly determined by gestational and postnatal age. Preterm neonates are common in clinical practice, and due to the different physiology compared to the full terms, constitute a specific neonatal subpopulation. The objective of this review is to summarize the evidence about the developmental changes (specific for preterm and full-term infants, separately) of pharmacokinetic parameters of antibiotics used in neonatal intensive care units.
Collapse
|
9
|
Arslanoglu S, Moro GE, Tonetto P, De Nisi G, Ambruzzi AM, Biasini A, Profeti C, Gagliardi L, Salvatori G, Bertino E. Recommendations for the establishment and operation of a donor human milk bank. Nutr Rev 2023; 81:1-28. [PMID: 36892193 PMCID: PMC9997086 DOI: 10.1093/nutrit/nuad012] [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: 03/10/2023] Open
Abstract
In Europe, an increasing number of human milk banks (HMBs) collect donor human milk to feed preterm infants when their mother's milk is not available or not enough. Moreover, donor milk is a bridge to breastfeeding, with positive clinical and psychological advantages for both mother and infant. Italy, with 41 HMBs actively operating in 2022, has the highest number of HMBs in Europe. The process of human milk donation is complex, so activity of HMBs must be regulated according to well-established rules. The present recommendations have been prepared as a tool to standardize the organization, management, and procedures of HMBs operating in Italy and to determine the minimal essential requirements to establish new HMBs. This article covers all the aspects of human milk donation and human milk banking, including general recommendations, donor recruitment and screening, expression, handling and storage of donor human milk, milk screening, and milk treatment (pasteurization). A pragmatic approach was taken to drafting the recommendations. Items for which there was consensus or robust published evidence on which to base recommendations were included. When there were differences that could not be resolved by reference to published research, a statement of explanation based on the expert opinion of the authors (all members of the Italian Association of Human Milk Banks) was included. Implementation of these recommendations can contribute to promotion of breastfeeding.
Collapse
Affiliation(s)
- Sertac Arslanoglu
- are with the Italian Association of Human Milk Banks (Associazione Italiana Banche del Latte Umano Donato), Milan, Italy
- is with the Division of Neonatology, Department of Pediatrics, İstanbul Medeniyet University, School of Medicine, İstanbul, Turkey
| | - Guido E Moro
- are with the Italian Association of Human Milk Banks (Associazione Italiana Banche del Latte Umano Donato), Milan, Italy
| | - Paola Tonetto
- are with the Italian Association of Human Milk Banks (Associazione Italiana Banche del Latte Umano Donato), Milan, Italy
- are with the Neonatal Care Unit of the University, City of Health and Science Hospital, Turin, Italy
| | - Giuseppe De Nisi
- are with the Italian Association of Human Milk Banks (Associazione Italiana Banche del Latte Umano Donato), Milan, Italy
| | - Amalia Maria Ambruzzi
- are with the Italian Association of Human Milk Banks (Associazione Italiana Banche del Latte Umano Donato), Milan, Italy
| | - Augusto Biasini
- are with the Italian Association of Human Milk Banks (Associazione Italiana Banche del Latte Umano Donato), Milan, Italy
| | - Claudio Profeti
- are with the Italian Association of Human Milk Banks (Associazione Italiana Banche del Latte Umano Donato), Milan, Italy
| | - Luigi Gagliardi
- is with the Woman and Child Health Department, Azienda USL Toscana Nord-Ovest, Lucca, Italy
| | - Guglielmo Salvatori
- are with the Italian Association of Human Milk Banks (Associazione Italiana Banche del Latte Umano Donato), Milan, Italy
- is with the Donor Human Milk Bank, Pediatric Hospital Bambino Gesù, Rome, Italy
| | - Enrico Bertino
- are with the Italian Association of Human Milk Banks (Associazione Italiana Banche del Latte Umano Donato), Milan, Italy
- are with the Neonatal Care Unit of the University, City of Health and Science Hospital, Turin, Italy
| |
Collapse
|
10
|
Sampath V, Martinez M, Caplan M, Underwood MA, Cuna A. Necrotizing enterocolitis in premature infants-A defect in the brakes? Evidence from clinical and animal studies. Mucosal Immunol 2023; 16:208-220. [PMID: 36804483 DOI: 10.1016/j.mucimm.2023.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023]
Abstract
A key aspect of postnatal intestinal adaptation is the establishment of symbiotic relationships with co-evolved gut microbiota. Necrotizing enterocolitis (NEC) is the most severe disease arising from failure in postnatal gut adaptation in premature infants. Although pathological activation of intestinal Toll-like receptors (TLRs) is believed to underpin NEC pathogenesis, the mechanisms are incompletely understood. We postulate that unregulated aberrant TLR activation in NEC arises from a failure in intestinal-specific mechanisms that tamponade TLR signaling (the brakes). In this review, we discussed the human and animal studies that elucidate the developmental mechanisms inhibiting TLR signaling in the postnatal intestine (establishing the brakes). We then evaluate evidence from preclinical models and human studies that point to a defect in the inhibition of TLR signaling underlying NEC. Finally, we provided a framework for the assessment of NEC risk by screening for signatures of TLR signaling and for NEC prevention by TLR-targeted therapy in premature infants.
Collapse
Affiliation(s)
- Venkatesh Sampath
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, Missouri, USA; School of Medicine, University of Missouri Kansas City, Kansas City, Missouri, USA.
| | - Maribel Martinez
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, Missouri, USA; School of Medicine, University of Missouri Kansas City, Kansas City, Missouri, USA
| | - Michael Caplan
- Department of Pediatrics, North Shore University Health System, Evanston, Illinois, USA
| | - Mark A Underwood
- Department of Pediatrics, University of California Davis, Sacramento, California, USA
| | - Alain Cuna
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, Missouri, USA; School of Medicine, University of Missouri Kansas City, Kansas City, Missouri, USA
| |
Collapse
|
11
|
Cuna A, Morowitz MJ, Sampath V. Early antibiotics and risk for necrotizing enterocolitis in premature infants: A narrative review. Front Pediatr 2023; 11:1112812. [PMID: 36865691 PMCID: PMC9971631 DOI: 10.3389/fped.2023.1112812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/24/2023] [Indexed: 02/16/2023] Open
Abstract
While prompt initiation of antibiotics at birth due to concerns for early onset sepsis is common, it often leads to many preterm infants being exposed to treatment despite negative blood cultures. Such exposure to early antibiotics can impact the developing gut microbiome putting infants at increased risk of several diseases. Necrotizing enterocolitis (NEC), a devastating inflammatory bowel disease that affects preterm infants, is among the most widely studied neonatal disease that has been linked to early antibiotics. While some studies have demonstrated an increased risk of NEC, other studies have demonstrated seemingly contrary findings of decreased NEC with early antibiotics. Studies using animal models have also yielded differing findings of benefit vs. harm of early antibiotic exposure on subsequent NEC susceptibility. We thus sought to conduct this narrative review to help clarify the relationship between early antibiotics exposure and future risk of NEC in preterm infants. Our objectives are to: (1) summarize findings from human and animal studies that investigated the relationship between early antibiotics and NEC, (2) highlight important limitations of these studies, (3) explore potential mechanisms that can explain why early antibiotics may increase or decrease NEC risk, and (4) identify future directions for research.
Collapse
Affiliation(s)
- Alain Cuna
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MOUnited States
- School of Medicine, University of Missouri-Kansas City, Kansas City, MOUnited States
| | - Michael J. Morowitz
- Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PAUnited States
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Venkatesh Sampath
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MOUnited States
- School of Medicine, University of Missouri-Kansas City, Kansas City, MOUnited States
| |
Collapse
|
12
|
Shen R, Embleton N, Lyng Forman J, Gale C, Griesen G, Sangild PT, Uthaya S, Berrington J. Early antibiotic use and incidence of necrotising enterocolitis in very preterm infants: a protocol for a UK based observational study using routinely recorded data. BMJ Open 2022; 12:e065934. [PMID: 36379645 PMCID: PMC9667987 DOI: 10.1136/bmjopen-2022-065934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Necrotising enterocolitis (NEC) remains a major contributor to preterm mortality and morbidity. Prolonged duration of antibiotic therapy after delivery is associated with later NEC development but recent evidence suggests that absence of antibiotic treatment after delivery may also increase NEC risk. We will explore this controversy using a large pre-existing dataset of preterm infants in the UK. METHODS AND ANALYSIS This is a retrospective cohort study using data from UK National Neonatal Research Database (NNRD) for infants born 1 January 2012 to 31 December 2020. Eligible infants will be <32 weeks gestation, alive on day 3. Primary outcome is development of severe NEC, compared in infants receiving early antibiotics (days 1-2 after birth) and those not. Subgroup analysis on duration of early antibiotic exposure will also occur. Secondary outcomes are: late onset sepsis, total antibiotic use, predischarge mortality, retinopathy of prematurity, intraventricular haemorrhage, bronchopulmonary dysplasia, focal intestinal perforation and any abdominal surgery. To address competing risks, incidence of death before day 7, 14 and 28 will be analysed. We will perform logistic regression and propensity score matched analyses. Statistical analyses will be guided by NEC risk factors, exposures and outcome presented in a causal diagram. These covariates include but are not limited to gestational age, birth weight, small for gestational age, sex, ethnicity, delivery mode, delivery without labour, Apgar score, early feeding and probiotic use. Sensitivity analyses of alternate NEC definitions, specific antibiotics and time of initiation will occur. ETHICS AND DISSEMINATION We will use deidentified data from NNRD, which holds permissions for the original data, from which parents can opt out and seek study-specific research ethics approval. The results will help to determine optimal use of early antibiotics for very preterm infants. IMPLICATIONS This data will help optimise early antibiotic use in preterm infants. TRIAL REGISTRATION NUMBER ISRCTN55101779.
Collapse
Affiliation(s)
- Rene Shen
- Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhgaen, Denmark
| | - Nicholas Embleton
- Neonatology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
- Population Health Sciences, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | | | - Chris Gale
- Neonatal Medicine, School of Public Health, Imperial College London, London, UK
| | - Gorm Griesen
- Neonatology, Odense University Hospital, Odense, Denmark
| | - Per Torp Sangild
- Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhgaen, Denmark
- Department of Neonatology, Rigshospitalet, Copenhagen, Denmark
| | - Sabita Uthaya
- Faculty of Medicine, Imperial College London, London, UK
| | - Janet Berrington
- Neonatology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
- Clinical and Translational Research Institute, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
| |
Collapse
|
13
|
Niewiem M, Grzybowska-Chlebowczyk U. Assessment of Selected Intestinal Permeability Markers in Children with Food Allergy Depending on the Type and Severity of Clinical Symptoms. Nutrients 2022; 14:nu14204385. [PMID: 36297068 PMCID: PMC9608842 DOI: 10.3390/nu14204385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 12/01/2022] Open
Abstract
Background: Food allergy (FA) has a broad range of symptoms, and clinical manifestations may concern several reactions from one system or organ. Aim: The aim of the study was to assess intestinal permeability (IP) based on the analysis of serum zonulin and bacterial lipopolysaccharides (LPS) levels in children with FA, taking into account the pathomechanism of immune reaction, clinical symptoms of FA and their severity. Material and methods: The study comprised 103 patients aged 7–60 months (median 34); 49 children with IgE-mediated allergy and 25 children with non-IgE-mediated allergy; the reference group comprised 29 children with functional gastrointestinal disorders. IP markers were determined using ELISA. Results: There was no correlation between the severity of clinical symptoms and the level of IP markers in children with FA. Zonulin and LPS levels were significantly higher in children with FA and gastrointestinal symptoms. Zonulin levels in the subgroup of children with non-IgE-mediated FA and gastrointestinal symptoms were significantly higher than in the subgroup of children with IgE-mediated FA and these symptoms. The level of LPS was significantly higher in the subgroup with IgE-mediated FA and atopic dermatitis. Conclusions: Zonulin and LPS levels were significantly higher in children with FA compared to children from the reference group. Zonulin levels were significantly higher in children with non-IgE-mediated FA than in children with IgE-mediated FA.
Collapse
|
14
|
Dimitroglou M, Iliodromiti Z, Christou E, Volaki P, Petropoulou C, Sokou R, Boutsikou T, Iacovidou N. Human Breast Milk: The Key Role in the Maturation of Immune, Gastrointestinal and Central Nervous Systems: A Narrative Review. Diagnostics (Basel) 2022; 12:diagnostics12092208. [PMID: 36140609 PMCID: PMC9498242 DOI: 10.3390/diagnostics12092208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 12/22/2022] Open
Abstract
Premature birth is a major cause of mortality and morbidity in the pediatric population. Because their immune, gastrointestinal and nervous systems are not fully developed, preterm infants (<37 weeks of gestation) and especially very preterm infants (VPIs, <32 weeks of gestation) are more prone to infectious diseases, tissue damage and future neurodevelopmental impairment. The aim of this narrative review is to report the immaturity of VPI systems and examine the role of Human Breast Milk (HBM) in their development and protection against infectious diseases, inflammation and tissue damage. For this purpose, we searched and synthesized the data from the existing literature published in the English language. Studies revealed the significance of HBM and indicate HBM as the best dietary choice for VPIs.
Collapse
|
15
|
Liu Z, Ashorn U, Chingwanda C, Maleta K, Hallamaa L, Matchado A, Kortekangas E, Dewey KG, Ashorn P, Fan Y. Provision of small‐quantity lipid‐based nutrient supplements does not improve intestinal health among rural Malawian children. MATERNAL & CHILD NUTRITION 2022; 18:e13331. [PMID: 35128820 PMCID: PMC9218311 DOI: 10.1111/mcn.13331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Zhifei Liu
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | - Ulla Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | | | - Kenneth Maleta
- Department of Public Health University of Malawi Zomba Malawi
| | - Lotta Hallamaa
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | - Andrew Matchado
- Department of Public Health University of Malawi Zomba Malawi
| | - Emma Kortekangas
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | - Kathryn G Dewey
- Department of Nutrition University of California Davis CA USA
| | - Per Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
- Department of Paediatrics Tampere University Hospital Tampere Finland
| | - Yue‐Mei Fan
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
| |
Collapse
|
16
|
Intestinal epithelium in early life. Mucosal Immunol 2022; 15:1181-1187. [PMID: 36380094 DOI: 10.1038/s41385-022-00579-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [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
|
17
|
Niewiem M, Grzybowska-Chlebowczyk U. Intestinal Barrier Permeability in Allergic Diseases. Nutrients 2022; 14:nu14091893. [PMID: 35565858 PMCID: PMC9101724 DOI: 10.3390/nu14091893] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023] Open
Abstract
The role of intestinal permeability (IP) markers among children and adults with food allergies is not fully understood, and the identification of biological indicators/markers that predict growth retardation in children with allergic diseases and atopy has not been well explained. Studies have shown that patients with atopic diseases respond abnormally to food allergens. Accordingly, differences in the types of immune complexes formed in response to antigen challenges are significant, which seems to underlie the systemic signs of the food allergy. Increased intestinal permeability over the course of a food allergy allows allergens to penetrate through the intestinal barrier and stimulate the submucosal immune system. Additionally, the release of cytokines and inflammatory mediators enhances the degradation of the epithelial barrier and leads to an improper cycle, resulting in increased intestinal permeability. Several studies have also demonstrated increased permeability of the epithelial cells in those afflicted with atopic eczema and bronchial asthma. Ongoing research is aimed at finding various indicators to assess IP in patients with atopic diseases.
Collapse
|
18
|
Chaaban H, Patel MM, Burge K, Eckert JV, Lupu C, Keshari RS, Silasi R, Regmi G, Trammell M, Dyer D, McElroy SJ, Lupu F. Early Antibiotic Exposure Alters Intestinal Development and Increases Susceptibility to Necrotizing Enterocolitis: A Mechanistic Study. Microorganisms 2022; 10:519. [PMID: 35336095 PMCID: PMC8951210 DOI: 10.3390/microorganisms10030519] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 12/24/2022] Open
Abstract
Increasing evidence suggests that prolonged antibiotic therapy in preterm infants is associated with increased mortality and morbidities, such as necrotizing enterocolitis (NEC), a devastating gastrointestinal pathology characterized by intestinal inflammation and necrosis. While a clinical correlation exists between antibiotic use and the development of NEC, the potential causality of antibiotics in NEC development has not yet been demonstrated. Here, we tested the effects of systemic standard-of-care antibiotic therapy for ten days on intestinal development in neonatal mice. Systemic antibiotic treatment impaired the intestinal development by reducing intestinal cell proliferation, villi height, crypt depth, and goblet and Paneth cell numbers. Oral bacterial challenge in pups who received antibiotics resulted in NEC-like intestinal injury in more than half the pups, likely due to a reduction in mucous-producing cells affecting microbial-epithelial interactions. These data support a novel mechanism that could explain why preterm infants exposed to prolonged antibiotics after birth have a higher incidence of NEC and other gastrointestinal disorders.
Collapse
Affiliation(s)
- Hala Chaaban
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.V.E.)
| | - Maulin M. Patel
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (M.M.P.); (C.L.); (R.S.K.); (R.S.); (G.R.); (F.L.)
| | - Kathryn Burge
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.V.E.)
| | - Jeffrey V. Eckert
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.V.E.)
| | - Cristina Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (M.M.P.); (C.L.); (R.S.K.); (R.S.); (G.R.); (F.L.)
| | - Ravi S. Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (M.M.P.); (C.L.); (R.S.K.); (R.S.); (G.R.); (F.L.)
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (M.M.P.); (C.L.); (R.S.K.); (R.S.); (G.R.); (F.L.)
| | - Girija Regmi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (M.M.P.); (C.L.); (R.S.K.); (R.S.); (G.R.); (F.L.)
| | - MaJoi Trammell
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA; (M.T.); (D.D.)
| | - David Dyer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA; (M.T.); (D.D.)
| | - Steven J. McElroy
- Department of Pediatrics, UC Davis Health, Sacramento, CA 95817, USA;
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (M.M.P.); (C.L.); (R.S.K.); (R.S.); (G.R.); (F.L.)
| |
Collapse
|
19
|
Lemme-Dumit JM, Song Y, Lwin HW, Hernandez-Chavez C, Sundararajan S, Viscardi RM, Ravel J, Pasetti MF, Ma B. Altered Gut Microbiome and Fecal Immune Phenotype in Early Preterm Infants With Leaky Gut. Front Immunol 2022; 13:815046. [PMID: 35280991 PMCID: PMC8905226 DOI: 10.3389/fimmu.2022.815046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Intestinal barrier immaturity, or "leaky gut", is the proximate cause of susceptibility to necrotizing enterocolitis in preterm neonates. Exacerbated intestinal immune responses, gut microbiota dysbiosis, and heightened barrier injury are considered primary triggers of aberrant intestinal maturation in early life. Inordinate host immunity contributes to this process, but the precise elements remain largely uncharacterized, leaving a significant knowledge gap in the biological underpinnings of gut maturation. In this study, we investigated the fecal cytokine profile and gut microbiota in a cohort of 40 early preterm infants <33-weeks-gestation to identify immune markers of intestinal barrier maturation. Three distinct microbiota types were demonstrated to be differentially associated with intestinal permeability (IP), maternal breast milk feeding, and immunological profiles. The Staphylococcus epidermidis- and Enterobacteriaceae-predominant microbiota types were associated with an elevated IP, reduced breast milk feeding, and less defined fecal cytokine profile. On the other hand, a lower IP was associated with increased levels of fecal IL-1α/β and a microbiota type that included a wide array of anaerobes with expanded fermentative capacity. Our study demonstrated the critical role of both immunological and microbiological factors in the early development of intestinal barrier that collectively shape the intestinal microenvironment influencing gut homeostasis and postnatal intestinal maturation in early preterm newborns.
Collapse
Affiliation(s)
- Jose M Lemme-Dumit
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Hnin Wai Lwin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Claudia Hernandez-Chavez
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sripriya Sundararajan
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rose M Viscardi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcela F Pasetti
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Bing Ma
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
20
|
Siljander H, Jason E, Ruohtula T, Selvenius J, Koivusaari K, Salonen M, Ahonen S, Honkanen J, Ilonen J, Vaarala O, Virtanen SM, Lähdeaho ML, Knip M. Effect of Early Feeding on Intestinal Permeability and Inflammation Markers in Infants with Genetic Susceptibility to Type 1 Diabetes: A Randomized Clinical Trial. J Pediatr 2021; 238:305-311.e3. [PMID: 34293372 DOI: 10.1016/j.jpeds.2021.07.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/16/2021] [Accepted: 07/15/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To assess whether weaning to an extensively hydrolyzed formula (EHF) decreases gut permeability and/or markers of intestinal inflammation in infants with HLA-conferred diabetes susceptibility, when compared with conventional formula. STUDY DESIGN By analyzing 1468 expecting biological parent pairs for HLA-conferred susceptibility for type 1 diabetes, 465 couples (32 %) potentially eligible for the study were identified. After further parental consent, 332 babies to be born were randomized at 35th gestational week. HLA genotyping was performed at birth in 309 infants. Out of 87 eligible children, 73 infants participated in the intervention study: 33 in the EHF group and 40 in the control group. Clinical visits took place at 3, 6, 9, and 12 months of age. The infants were provided either EHF or conventional formula whenever breastfeeding was not available or additional feeding was required over the first 9 months of life. The main outcome was the lactulose to mannitol ratio (L/M ratio) at 9 months. The secondary outcomes were L/M ratio at 3, 6, and 12 months of age, and fecal calprotectin and human beta-defensin 2 (HBD-2) levels at each visit. RESULTS Compared with controls, the median L/M ratio was lower in the EHF group at 9 months (.006 vs .028; P = .005). Otherwise, the levels of intestinal permeability, fecal calprotectin, and HBD-2 were comparable between the two groups, although slight differences in the age-related dynamics of these markers were observed. CONCLUSIONS It is possible to decrease intestinal permeability in infancy through weaning to an extensively hydrolyzed formula. This may reduce the early exposure to dietary antigens. TRIAL REGISTRATION Clinicaltrials.gov: NCT01735123.
Collapse
Affiliation(s)
- Heli Siljander
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eeva Jason
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Center for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Terhi Ruohtula
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Clinicum, University of Helsinki, Helsinki, Finland
| | - Jenni Selvenius
- Center for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Katariina Koivusaari
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
| | - Marja Salonen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Suvi Ahonen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Tampere University Hospital, Research, Development and Innovation Center, Tampere, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Jarno Honkanen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Clinicum, University of Helsinki, Helsinki, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, University of Turku and Turku University of Hospital, Turku, Finland
| | - Outi Vaarala
- Clinicum, University of Helsinki, Helsinki, Finland
| | - Suvi M Virtanen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Tampere University Hospital, Research, Development and Innovation Center, Tampere, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Marja-Leena Lähdeaho
- Center for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland; Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| | - Mikael Knip
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Center for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland; Department of Pediatrics, Tampere University Hospital, Tampere, Finland.
| |
Collapse
|
21
|
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: 1.7] [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
|
22
|
Chiale F, Maggiora E, Aceti A, Liotto N, Coscia A, Peila C, Baldassarre ME, Bertino E, Cresi F. Complementary Feeding: Recommendations for the Introduction of Allergenic Foods and Gluten in the Preterm Infant. Nutrients 2021; 13:2477. [PMID: 34371985 PMCID: PMC8308791 DOI: 10.3390/nu13072477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/30/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The aim of this systematic review is to analyze the available literature on the introduction of allergenic foods and gluten among preterm infants. METHODS A systematic review of published studies concerning the introduction of gluten and allergenic foods in preterm infants was performed on PubMed and on the Cochrane Library. RESULTS Of the 174 PubMed results, 15 papers were considered suitable for the review. A total of 83 records were identified through the Cochrane Library search; eight papers were included in the review. Additional papers were identified from the reference lists of included studies. A secondary search was conducted on the same databases to find recommendations and advice regarding healthy full-term infants that could be translated to preterm infants. Therefore, 59 additional papers were included in the review. CONCLUSIONS Current guidelines for the introduction of solid food cannot be directly transposed to preterm infants. Further research is needed to provide evidence-based guidelines regarding weaning in preterm infants. To date, we can suggest that in preterm infants allergenic foods and gluten may be introduced when complementary feeding is started, any time after 4 months of corrected age, avoiding delayed introduction and irrespective of infants' relative risk of developing allergy. Avoiding large amounts of gluten during the first few weeks after gluten introduction and during infancy is advised, despite limited evidence to support this recommendation.
Collapse
Affiliation(s)
- Federica Chiale
- Neonatal Pathology and Neonatal Intensive Care Unit, Sant’Anna Hospital, City of Health and Science University Hospital of Turin, University of Turin, 10126 Turin, Italy; (F.C.); (A.C.); (C.P.); (E.B.); (F.C.)
| | - Elena Maggiora
- Neonatal Pathology and Neonatal Intensive Care Unit, Sant’Anna Hospital, City of Health and Science University Hospital of Turin, University of Turin, 10126 Turin, Italy; (F.C.); (A.C.); (C.P.); (E.B.); (F.C.)
| | - Arianna Aceti
- Neonatal Intensive Care Unit, AOU Bologna, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Nadia Liotto
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, 20122 Milan, Italy;
| | - Alessandra Coscia
- Neonatal Pathology and Neonatal Intensive Care Unit, Sant’Anna Hospital, City of Health and Science University Hospital of Turin, University of Turin, 10126 Turin, Italy; (F.C.); (A.C.); (C.P.); (E.B.); (F.C.)
| | - Chiara Peila
- Neonatal Pathology and Neonatal Intensive Care Unit, Sant’Anna Hospital, City of Health and Science University Hospital of Turin, University of Turin, 10126 Turin, Italy; (F.C.); (A.C.); (C.P.); (E.B.); (F.C.)
| | | | - Enrico Bertino
- Neonatal Pathology and Neonatal Intensive Care Unit, Sant’Anna Hospital, City of Health and Science University Hospital of Turin, University of Turin, 10126 Turin, Italy; (F.C.); (A.C.); (C.P.); (E.B.); (F.C.)
| | - Francesco Cresi
- Neonatal Pathology and Neonatal Intensive Care Unit, Sant’Anna Hospital, City of Health and Science University Hospital of Turin, University of Turin, 10126 Turin, Italy; (F.C.); (A.C.); (C.P.); (E.B.); (F.C.)
| |
Collapse
|
23
|
Henderickx JGE, Zwittink RD, Renes IB, van Lingen RA, van Zoeren-Grobben D, Jebbink LJG, Boeren S, van Elburg RM, Knol J, Belzer C. Maturation of the preterm gastrointestinal tract can be defined by host and microbial markers for digestion and barrier defense. Sci Rep 2021; 11:12808. [PMID: 34140588 PMCID: PMC8211855 DOI: 10.1038/s41598-021-92222-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/01/2021] [Indexed: 12/19/2022] Open
Abstract
Functionality of the gastrointestinal tract is essential for growth and development of newborns. Preterm infants have an immature gastrointestinal tract, which is a major challenge in neonatal care. This study aims to improve the understanding of gastrointestinal functionality and maturation during the early life of preterm infants by means of gastrointestinal enzyme activity assays and metaproteomics. In this single-center, observational study, preterm infants born between 24 and 33 weeks (n = 40) and term infants born between 37 and 42 weeks (n = 3), who were admitted to Isala (Zwolle, the Netherlands), were studied. Enzyme activity analyses identified active proteases in gastric aspirates of preterm infants. Metaproteomics revealed human milk, digestive and immunological proteins in gastric aspirates of preterm infants and feces of preterm and term infants. The fecal proteome of preterm infants was deprived of gastrointestinal barrier-related proteins during the first six postnatal weeks compared to term infants. In preterm infants, bacterial oxidative stress proteins were increased compared to term infants and higher birth weight correlated to higher relative abundance of bifidobacterial proteins in postnatal week 3 to 6. Our findings indicate that gastrointestinal and beneficial microbial proteins involved in gastrointestinal maturity are associated with gestational and postnatal age.
Collapse
Affiliation(s)
- Jannie G E Henderickx
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Romy D Zwittink
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ingrid B Renes
- Danone Nutricia Research, Utrecht, the Netherlands
- Emma Children's Hospital, Amsterdam UMC, Location AMC Amsterdam, Amsterdam, The Netherlands
| | - Richard A van Lingen
- Department of Neonatology, Isala Women and Children's Hospital, Zwolle, The Netherlands
| | | | | | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University and Research, Wageningen, The Netherlands
| | - Ruurd M van Elburg
- Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan Knol
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
- Danone Nutricia Research, Utrecht, the Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| |
Collapse
|
24
|
Chaaban H, Burge K, Eckert J, Trammell M, Dyer D, Keshari RS, Silasi R, Regmi G, Lupu C, Good M, McElroy SJ, Lupu F. Acceleration of Small Intestine Development and Remodeling of the Microbiome Following Hyaluronan 35 kDa Treatment in Neonatal Mice. Nutrients 2021; 13:2030. [PMID: 34204790 PMCID: PMC8231646 DOI: 10.3390/nu13062030] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
The beneficial effects of human milk suppressing the development of intestinal pathologies such as necrotizing enterocolitis in preterm infants are widely known. Human milk (HM) is rich in a multitude of bioactive factors that play major roles in promoting postnatal maturation, differentiation, and the development of the microbiome. Previous studies showed that HM is rich in hyaluronan (HA) especially in colostrum and early milk. This study aims to determine the role of HA 35 KDa, a HM HA mimic, on intestinal proliferation, differentiation, and the development of the intestinal microbiome. We show that oral HA 35 KDa supplementation for 7 days in mouse pups leads to increased villus length and crypt depth, and increased goblet and Paneth cells, compared to controls. We also show that HA 35 KDa leads to an increased predominance of Clostridiales Ruminococcaceae, Lactobacillales Lactobacillaceae, and Clostridiales Lachnospiraceae. In seeking the mechanisms involved in the changes, bulk RNA seq was performed on samples from the terminal ileum and identified upregulation in several genes essential for cellular growth, proliferation, and survival. Taken together, this study shows that HA 35 KDa supplemented to mouse pups promotes intestinal epithelial cell proliferation, as well as the development of Paneth cells and goblet cell subsets. HA 35 KDa also impacted the intestinal microbiota; the implications of these responses need to be determined.
Collapse
Affiliation(s)
- Hala Chaaban
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.)
| | - Kathryn Burge
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.)
| | - Jeffrey Eckert
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.)
| | - MaJoi Trammell
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (M.T.); (D.D.)
| | - David Dyer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (M.T.); (D.D.)
| | - Ravi S. Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.S.K.); (R.S.); (G.R.); (C.L.); (F.L.)
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.S.K.); (R.S.); (G.R.); (C.L.); (F.L.)
| | - Girija Regmi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.S.K.); (R.S.); (G.R.); (C.L.); (F.L.)
| | - Cristina Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.S.K.); (R.S.); (G.R.); (C.L.); (F.L.)
| | - Misty Good
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Steven J. McElroy
- Department of Microbiology and Immunology, Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA;
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.S.K.); (R.S.); (G.R.); (C.L.); (F.L.)
| |
Collapse
|
25
|
Nunez N, Réot L, Menu E. Neonatal Immune System Ontogeny: The Role of Maternal Microbiota and Associated Factors. How Might the Non-Human Primate Model Enlighten the Path? Vaccines (Basel) 2021; 9:584. [PMID: 34206053 PMCID: PMC8230289 DOI: 10.3390/vaccines9060584] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/23/2022] Open
Abstract
Interactions between the immune system and the microbiome play a crucial role on the human health. These interactions start in the prenatal period and are critical for the maturation of the immune system in newborns and infants. Several factors influence the composition of the infant's microbiota and subsequently the development of the immune system. They include maternal infection, antibiotic treatment, environmental exposure, mode of delivery, breastfeeding, and food introduction. In this review, we focus on the ontogeny of the immune system and its association to microbial colonization from conception to food diversification. In this context, we give an overview of the mother-fetus interactions during pregnancy, the impact of the time of birth and the mode of delivery, the neonate gastrointestinal colonization and the role of breastfeeding, weaning, and food diversification. We further review the impact of the vaccination on the infant's microbiota and the reciprocal case. Finally, we discuss several potential therapeutic interventions that might help to improve the newborn and infant's health and their responses to vaccination. Throughout the review, we underline the main scientific questions that are left to be answered and how the non-human primate model could help enlighten the path.
Collapse
Affiliation(s)
- Natalia Nunez
- CEA, Université Paris-Sud, Inserm, U1184 “Immunology of Viral Infections and Autoimmune Diseases” (IMVA-HB), IDMIT Department, IBFJ, 92265 Fontenay-aux-Roses, France; (N.N.); (L.R.)
| | - Louis Réot
- CEA, Université Paris-Sud, Inserm, U1184 “Immunology of Viral Infections and Autoimmune Diseases” (IMVA-HB), IDMIT Department, IBFJ, 92265 Fontenay-aux-Roses, France; (N.N.); (L.R.)
| | - Elisabeth Menu
- CEA, Université Paris-Sud, Inserm, U1184 “Immunology of Viral Infections and Autoimmune Diseases” (IMVA-HB), IDMIT Department, IBFJ, 92265 Fontenay-aux-Roses, France; (N.N.); (L.R.)
- MISTIC Group, Department of Virology, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
26
|
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: 2.7] [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
|
27
|
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: 10.3] [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
|
28
|
Human Milk Exosomal MicroRNA: Associations with Maternal Overweight/Obesity and Infant Body Composition at 1 Month of Life. Nutrients 2021; 13:nu13041091. [PMID: 33801634 PMCID: PMC8066780 DOI: 10.3390/nu13041091] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 12/11/2022] Open
Abstract
Among all the body fluids, breast milk is one of the richest sources of microRNAs (miRNAs). MiRNAs packaged within the milk exosomes are bioavailable to breastfeeding infants. The role of miRNAs in determining infant growth and the impact of maternal overweight/obesity on human milk (HM) miRNAs is poorly understood. The objectives of this study were to examine the impact of maternal overweight/obesity on select miRNAs (miR-148a, miR-30b, miR-29a, miR-29b, miR-let-7a and miR-32) involved in adipogenesis and glucose metabolism and to examine the relationship of these miRNAs with measures of infant body composition in the first 6 months of life. Milk samples were collected from a cohort of 60 mothers (30 normal-weight [NW] and 30 overweight [OW]/obese [OB]) at 1-month and a subset of 48 of these at 3 months of lactation. Relative abundance of miRNA was determined using real-time PCR. The associations between the miRNAs of interest and infant weight and body composition at one, three, and six months were examined after adjusting for infant gestational age, birth weight, and sex. The abundance of miR-148a and miR-30b was lower by 30% and 42%, respectively, in the OW/OB group than in the NW group at 1 month. miR-148a was negatively associated with infant weight, fat mass, and fat free mass, while miR-30b was positively associated with infant weight, percent body fat, and fat mass at 1 month. Maternal obesity is negatively associated with the content of select miRNAs in human milk. An association of specific miRNAs with infant body composition was observed during the first month of life, suggesting a potential role in the infant's adaptation to enteral nutrition.
Collapse
|
29
|
Zhao X, Zhou J, Liang W, Sheng Q, Lu L, Chen T, Chen J, Tan K, Lv Z. Probiotics mixture reinforces barrier function to ameliorate necrotizing enterocolitis by regulating PXR-JNK pathway. Cell Biosci 2021; 11:20. [PMID: 33482929 PMCID: PMC7824920 DOI: 10.1186/s13578-021-00530-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023] Open
Abstract
Background Intestinal dysbiosis is believed to be one of the factors inducing neonatal necrotizing enterocolitis (NEC). Probiotics have been employed to treat NEC in a number of animal experiments and clinical trials, and some significant benefits of utilizing probiotics for the prevention or alleviation of NEC have been confirmed. However, the mechanism underlying the efficacy of probiotics in treating NEC has not been elucidated. Results Impairment of the intestinal barrier, which was characterized by the decreased expression of tight junction components, was observed in the pathogenesis of NEC. The probiotic mixture alleviated this intestinal damage by enhancing the function of the barrier. Meanwhile, the probiotics remodeled the composition of the intestinal microbiota in NEC mice. Furthermore, increased expression of the pregnane X receptor (PXR) was observed after treatment with the probiotic mixture, and PXR overexpression in Caco-2 cells protected the barrier from lipopolysaccharide (LPS) damage. Further research showed that PXR could inhibit the phosphorylation of c-Jun N-terminal kinase (JNK) and could increase the expression of tight junction components. Conclusions Our study confirmed that probiotics could ameliorate intestinal lesions by enhancing the function of the mucosal barrier. Specifically, probiotics may target PXR, which may subsequently enhance the expression of tight junction components by inhibiting the phosphorylation of JNK and enhance the function of the barrier.
Collapse
Affiliation(s)
- Xiuhao Zhao
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Road, Putuo, Shanghai, China
| | - Jin Zhou
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Road, Putuo, Shanghai, China
| | - Wenhua Liang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Sheng
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Road, Putuo, Shanghai, China
| | - Li Lu
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Road, Putuo, Shanghai, China
| | - Tong Chen
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Road, Putuo, Shanghai, China
| | - Jianglong Chen
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Road, Putuo, Shanghai, China
| | - Kezhe Tan
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Road, Putuo, Shanghai, China
| | - Zhibao Lv
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Road, Putuo, Shanghai, China.
| |
Collapse
|
30
|
Li Y, Shen RL, Ayede AI, Berrington J, Bloomfield FH, Busari OO, Cormack BE, Embleton ND, van Goudoever JB, Greisen G, He Z, Huang Y, Li X, Lin HC, Mei J, Meier PP, Nie C, Patel AL, Sangild PT, Skeath T, Simmer K, Uhlenfeldt S, de Waard M, Ye S, Ye X, Zhang C, Zhu Y, Zhou P. Early Use of Antibiotics Is Associated with a Lower Incidence of Necrotizing Enterocolitis in Preterm, Very Low Birth Weight Infants: The NEOMUNE-NeoNutriNet Cohort Study. J Pediatr 2020; 227:128-134.e2. [PMID: 32553865 PMCID: PMC7686259 DOI: 10.1016/j.jpeds.2020.06.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To determine whether commencement of antibiotics within 3 postnatal days in preterm, very low birth weight (VLBW; ≤1500 g) infants is associated with the development of necrotizing enterocolitis (NEC). STUDY DESIGN Preplanned statistical analyses were done to study the association between early antibiotic treatment and later NEC development, using the NEOMUNE-NeoNutriNet cohort of VLBW infants from 13 neonatal intensive care units (NICUs) in 5 continents (n = 2831). NEC incidence was compared between infants who received early antibiotics and those who did not, with statistical adjustments for NICU, gestational age, birth weight, sex, delivery mode, antenatal steroid use, Apgar score, and type and initiation of enteral nutrition. RESULTS The incidence of NEC was 9.0% in the group of infants who did not receive early antibiotics (n = 269), compared with 3.9% in those who did receive early antibiotics (n = 2562). The incidence remained lower in the early antibiotic group after stepwise statistical adjustments for NICU (OR, 0.57; 95% CI, 0.35-0.94, P < .05) and other potential confounders (OR, 0.25; 95% CI, 0.12-0.47; P < .0001). CONCLUSIONS In this large international cohort of preterm VLBW infants, a small proportion of infants did not receive antibiotics just after birth, and these infants had a higher incidence of NEC. It is important to better understand the role of such variables as time, type, and duration of antibiotic treatment on NEC incidence, immune development, gut colonization, and antibiotic resistance in the NICU.
Collapse
Affiliation(s)
- Yanqi Li
- Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark
| | | | - Adejumoke I. Ayede
- Department of Pediatrics-Neonatology Unit, College of Medicine, University of Ibadan and University College Hospital, Ibadan, Nigeria
| | - Janet Berrington
- Department of Neonatology, Newcastle Hospitals NHS Foundation Trust, Newcastle, UK
| | - Frank H. Bloomfield
- Liggins Institute, University of Auckland and Newborn Service, National Women’s Health, Auckland, New Zealand
| | - Olubunmi O. Busari
- Department of Pediatrics-Neonatology Unit, College of Medicine, University of Ibadan and University College Hospital, Ibadan, Nigeria
| | - Barbara E. Cormack
- Paediatric Dietitians, Starship Child Health or Liggins Institute, University of Auckland
| | - Nicholas D. Embleton
- Department of Neonatology, Newcastle Hospitals NHS Foundation Trust, Newcastle, UK
| | - Johannes B. van Goudoever
- Department of Pediatrics, Amsterdam UMC, Vrije Universiteit, Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Gorm Greisen
- Department of Neonatology, Rigshospitalet, Copenhagen, Denmark.
| | - Zhongqian He
- Department of Neonatology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Yan Huang
- Department of Neonatology, Shenzhen Bao’an Maternal and Child Health Hospital, Shenzhen, China
| | - Xiaodong Li
- Department of Neonatology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Hung-Chih Lin
- Department of Neonatology, Children’s Hospital of China Medical University, Taichung, Taiwan
| | - Jiaping Mei
- Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Paula P. Meier
- Department of Pediatrics, Section of Neonatology, Rush University Children’s Hospital, Chicago, United States of America
| | - Chuan Nie
- Department of Neonatology, Guangdong Provincial Women & Children’s Hospital, Guangzhou, China
| | - Aloka L. Patel
- Department of Pediatrics, Section of Neonatology, Rush University Children’s Hospital, Chicago, United States of America
| | - Per T. Sangild
- Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark,Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark,Hans Christian Andersen Children’s Hospital, Odense, Denmark
| | - Thomas Skeath
- Department of Neonatology, Newcastle Hospitals NHS Foundation Trust, Newcastle, UK
| | - Karen Simmer
- Centre for Neonatal Research and Education, University of Western Australia and King Edward Memorial Hospital, Perth, Australia
| | | | - Marita de Waard
- Department of Pediatrics, Amsterdam UMC, Vrije Universiteit, Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Sufen Ye
- Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Xuqiang Ye
- Foshan Woman and Children’s Hospital, Foshan, China
| | - Chunyi Zhang
- Department of Neonatology, Guangdong Provincial Women & Children’s Hospital, Guangzhou, China,Jinan University, Guangzhou, China
| | - Yanna Zhu
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ping Zhou
- Department of Neonatology, Shenzhen Bao’an Maternal and Child Health Hospital, Shenzhen, China
| |
Collapse
|
31
|
Mank E, van Harskamp D, van Toledo L, van Goudoever JB, Schierbeek H. Simultaneous assessment of intestinal permeability and lactase activity in human-milk-fed preterm infants by sugar absorption test: Clinical implementation and analytical method. Clin Nutr 2020; 40:1413-1419. [PMID: 32948350 DOI: 10.1016/j.clnu.2020.08.034] [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: 04/26/2020] [Revised: 05/25/2020] [Accepted: 08/27/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND & AIMS Experimental (nutritional) interventions in preterm infants frequently focus on intestinal maturation, as improving tolerance to enteral nutrition is a major goal. Intestinal permeability and lactase activity serve as markers for intestinal maturation. We aimed to develop a protocol for the simultaneous assessment of both markers in human-milk-fed preterm infants by a sugar absorption test. In addition, we developed a new gas chromatography-mass spectrometry (GC-MS) method for the analysis of lactulose, lactose, and mannitol in urine and milk collected during the sugar absorption test. METHODS The sugar absorption test was performed on days 4, 7, and 14 postpartum in 12 preterm infants (gestational age of 26-32 weeks). Human milk was collected, pooled, and divided into equal portions to provide a stable lactose intake for 24 h. Urine was collected in the last 6 h of this 24 h period, after administration of a bolus test sugar solution. Samples were analyzed by GC-MS after derivatization by oxime formation combined with acetylation. RESULTS The GC-MS method was validated and used for the accurate measurement of lactulose, lactose, and mannitol concentrations. The urinary lactulose/mannitol ratio declined with time, suggesting a decreased intestinal permeability. The urine-to-milk-lactulose/lactose ratio increased as a result of increased lactase activity with time. CONCLUSIONS The developed protocol for simultaneous assessment of intestinal permeability and lactase activity can be used to monitor the effect of experimental (nutritional) interventions in human-milk-fed preterm infants. Urine and milk samples obtained during the sugar absorption test can be accurately analyzed by GC-MS.
Collapse
Affiliation(s)
- Elise Mank
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Department of Neonatology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Dewi van Harskamp
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Department of Neonatology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Stable Isotope Research Laboratory, Endocrinology, Amsterdam Gastroenterology Endocrinology and Metabolism, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Letty van Toledo
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Department of Neonatology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Johannes B van Goudoever
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Department of Neonatology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| | - Henk Schierbeek
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Department of Neonatology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Stable Isotope Research Laboratory, Endocrinology, Amsterdam Gastroenterology Endocrinology and Metabolism, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| |
Collapse
|
32
|
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: 1.5] [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
|
33
|
Weström B, Arévalo Sureda E, Pierzynowska K, Pierzynowski SG, Pérez-Cano FJ. The Immature Gut Barrier and Its Importance in Establishing Immunity in Newborn Mammals. Front Immunol 2020; 11:1153. [PMID: 32582216 PMCID: PMC7296122 DOI: 10.3389/fimmu.2020.01153] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/11/2020] [Indexed: 12/16/2022] Open
Abstract
The gut is an efficient barrier which protects against the passage of pathogenic microorganisms and potential harmful macromolecules into the body, in addition to its primary function of nutrient digestion and absorption. Contrary to the restricted macromolecular passage in adulthood, enhanced transfer takes place across the intestines during early life, due to the high endocytic capacity of the immature intestinal epithelial cells during the fetal and/or neonatal periods. The timing and extent of this enhanced endocytic capacity is dependent on animal species, with a prominent non-selective intestinal macromolecular transfer in newborn ungulates, e.g., pigs, during the first few days of life, and a selective transfer of mainly immunoglobulin G (IgG), mediated by the FcRn receptor, in suckling rodents, e.g., rats and mice. In primates, maternal IgG is transferred during fetal life via the placenta, and intestinal macromolecular transfer is largely restricted in human neonates. The period of intestinal macromolecular transmission provides passive immune protection through the transfer of IgG antibodies from an immune competent mother; and may even have extra-immune beneficial effects on organ maturation in the offspring. Moreover, intestinal transfer during the fetal/neonatal periods results in increased exposure to microbial and food antigens which are then presented to the underlying immune system, which is both naïve and immature. This likely stimulates the maturation of the immune system and shifts the response toward tolerance induction instead of activation or inflammation, as usually seen in adulthood. Ingestion of mother's milk and the dietary transition to complex food at weaning, as well as the transient changes in the gut microbiota during the neonatal period, are also involved in the resulting immune response. Any disturbances in timing and/or balance of these parallel processes, i.e., intestinal epithelial maturation, luminal microbial colonization and mucosal immune maturation due to, e.g., preterm birth, infection, antibiotic use or nutrient changes during the neonatal period, might affect the establishment of the immune system in the infant. This review will focus on how differing developmental processes in the intestinal epithelium affect the macromolecular passage in different species and the possible impact of such passage on the establishment of immunity during the critical perinatal period in young mammals.
Collapse
Affiliation(s)
- Björn Weström
- Department of Biology, Lund University, Lund, Sweden
| | - Ester Arévalo Sureda
- Precision Livestock and Nutrition Unit, TERRA Teaching and Research Centre, Gembloux Agro-Biotech, University of Liège, Gembloux, Belgium
| | - Kateryna Pierzynowska
- Department of Biology, Lund University, Lund, Sweden
- Department of Animal Physiology, Kielanowski Institute of Animal Physiology and Nutrition, Jablonna, Poland
| | - Stefan G. Pierzynowski
- Department of Biology, Lund University, Lund, Sweden
- Department of Medical Biology, Institute of Rural Health, Lublin, Poland
| | - Francisco-José Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
- Research Institute of Nutrition and Food Safety of the University of Barcelona (INSA-UB), Santa Coloma de Gramenet, Spain
| |
Collapse
|
34
|
Early Effect of Supplemented Infant Formulae on Intestinal Biomarkers and Microbiota: A Randomized Clinical Trial. Nutrients 2020; 12:nu12051481. [PMID: 32443684 PMCID: PMC7284641 DOI: 10.3390/nu12051481] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/11/2020] [Accepted: 05/16/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Post-natal gut maturation in infants interrelates maturation of the morphology, digestive, and immunological functions and gut microbiota development. Here, we explored both microbiota development and markers of gut barrier and maturation in healthy term infants during their early life to assess the interconnection of gut functions during different infant formulae regimes. Methods: A total of 203 infants were enrolled in this randomized double-blind controlled trial including a breastfed reference group. Infants were fed starter formulae for the first four weeks of life, supplemented with different combination of nutrients (lactoferrin, probiotics (Bifidobacterium animal subsp. Lactis) and prebiotics (Bovine Milk-derived Oligosaccharides—BMOS)) and subsequently fed the control formula up to eight weeks of life. Stool microbiota profiles and biomarkers of early gut maturation, calprotectin (primary outcome), elastase, α-1 antitrypsin (AAT) and neopterin were measured in feces at one, two, four, and eight weeks. Results: Infants fed formula containing BMOS had lower mean calprotectin levels over the first two to four weeks compared to the other formula groups. Elastase and AAT levels were closer to levels observed in breastfed infants. No differences were observed for neopterin. Global differences between the bacterial communities of all groups were assessed by constrained multivariate analysis with hypothesis testing. The canonical correspondence analysis (CCA) at genus level showed overlap between microbiota profiles at one and four weeks of age in the BMOS supplemented formula group with the breastfed reference, dominated by bifidobacteria. Microbiota profiles of all groups at four weeks were significantly associated with the calprotectin levels at 4 (CCA, p = 0.018) and eight weeks of age (CCA, p = 0.026). Conclusion: A meaningful correlation was observed between changes in microbiota composition and gut maturation marker calprotectin. The supplementation with BMOS seems to favor gut maturation closer to that of breastfed infants.
Collapse
|
35
|
Wang W, Zhong M, Yu T, Chen L, Shi L, Zong J, Wang S. Polysaccharide extracted from WuGuChong reduces high-fat diet-induced obesity in mice by regulating the composition of intestinal microbiota. Nutr Metab (Lond) 2020; 17:27. [PMID: 32256675 PMCID: PMC7106597 DOI: 10.1186/s12986-020-00442-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/12/2020] [Indexed: 01/06/2023] Open
Abstract
Background Obesity is a severe public health threat worldwide. Emerging evidence suggests that gut microbiota dysbiosis is closely associated with obesity and its related metabolic complications. The aim of the present study is to investigate the effects of polysaccharide extracted from WuGuChong (PEW) on high-fat diet (HFD)-induced obesity, and the potential mechanisms involving modulation of the gut microbiota composition. Methods Mice were fed a normal chow diet and a high-fat diet with or without PEW (300 mg/kg/day) by oral gavage for 8 weeks. Body weight, obesity-related metabolic disorders, and gut microbiota were examined at the end of the experiment. Results PEW supplementation reduces body weight, adipose hypertrophy, liver steatosis, insulin resistance and systemic inflammation in HFD-fed mice, as well as maintains intestinal epithelium integrity. High-throughput 16S rRNA analysis demonstrates that PEW supplementation alters the composition of gut microbiota. The Firmicutes to Bacteroidetes ratio and the relative abundance of Proteobacteria are increased in HFD-fed mice, which are reversed by PEW supplementation to approximately the control levels. Conclusions Our results suggest that PEW may be used as a bioactive ingredient to prevent obesity and its related metabolic disorders by modulating the composition of gut microbiota.
Collapse
Affiliation(s)
- Wendong Wang
- 1Department of Orthopedic Surgery, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, China.,2College of Integrative Medicine, Dalian Medical University, 9 South Lushun Road West, Dalian, China
| | - Mintao Zhong
- 3Department of Microbiology, Dalian Medical University, 9 South Lushun Road West, Dalian, China
| | - Tiantian Yu
- Department of Gynaecology and Obstetrics, Dalian Municipal Woman and Children's Medical Center, 1 and 3 Sports new town first Road, Dalian, China
| | - Lei Chen
- 3Department of Microbiology, Dalian Medical University, 9 South Lushun Road West, Dalian, China
| | - Lijun Shi
- 1Department of Orthopedic Surgery, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, China
| | - Junwei Zong
- 1Department of Orthopedic Surgery, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, China
| | - Shouyu Wang
- 1Department of Orthopedic Surgery, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, China.,2College of Integrative Medicine, Dalian Medical University, 9 South Lushun Road West, Dalian, China
| |
Collapse
|
36
|
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: 12] [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
|
37
|
Cowan CSM, Dinan TG, Cryan JF. Annual Research Review: Critical windows - the microbiota-gut-brain axis in neurocognitive development. J Child Psychol Psychiatry 2020; 61:353-371. [PMID: 31773737 DOI: 10.1111/jcpp.13156] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/26/2019] [Accepted: 10/09/2019] [Indexed: 02/06/2023]
Abstract
The gut microbiota is a vast, complex, and fascinating ecosystem of microorganisms that resides in the human gastrointestinal tract. As an integral part of the microbiota-gut-brain axis, it is now being recognized that the microbiota is a modulator of brain and behavior, across species. Intriguingly, periods of change in the microbiota coincide with the development of other body systems and particularly the brain. We hypothesize that these times of parallel development are biologically relevant, corresponding to 'sensitive periods' or 'critical windows' in the development of the microbiota-gut-brain axis. Specifically, signals from the microbiota during these periods are hypothesized to be crucial for establishing appropriate communication along the axis throughout the life span. In other words, the microbiota is hypothesized to act like an expected input to calibrate the development of the microbiota-gut-brain axis. The absence or disruption of the microbiota during specific developmental windows would therefore be expected to have a disproportionate effect on specific functions or potentially for regulation of the system as a whole. Evidence for microbial modulation of neurocognitive development and neurodevelopmental risk is discussed in light of this hypothesis, finishing with a focus on the challenges that lay ahead for the future study of the microbiota-gut-brain axis during development.
Collapse
Affiliation(s)
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
38
|
Probiotic strategies to prevent necrotizing enterocolitis in preterm infants: a meta-analysis. Pediatr Surg Int 2019; 35:1143-1162. [PMID: 31420743 DOI: 10.1007/s00383-019-04547-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/09/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE We aimed to compare probiotics with placebo for necrotizing enterocolitis in preterm infants and to evaluate the safety and effect and strict effect of specific probiotic genera. METHODS Data recorded until January 2019 were searched, and relevant academic articles from PubMed, MEDLINE, EMBASE, and the Cochrane Central Register of Controlled Trials were selected by two independent reviewers. Two reviewers independently included randomized controlled trials that compared probiotics and placebo in preterm infants. The outcomes included more than one of the following outcomes: incidence of necrotizing enterocolitis, necrotizing enterocolitis-related mortality, incidence of sepsis, and all-cause mortality. Two reviewers independently extracted data and assessed the risk of bias and quality of evidence. RESULTS We identified 34 eligible studies of 9161 participants. This meta-analysis showed an overall advantage of probiotics to prevent the incidence of necrotizing enterocolitis (3.54%) and gut-associated sepsis (15.59%), and decrease mortality (5.23%) in preterm infants. A probiotic mixture showed a huge advantage and vitality in preventing necrotizing enterocolitis (2.48%) and gut-associated sepsis (18.39%), and in reducing mortality (5.57%) in preterm infants. CONCLUSION The probiotic mixture showed advantages over the single strains to decrease the incidences of necrotizing enterocolitis and gut-associated sepsis, and mortality in preterm infants.
Collapse
|
39
|
Ajayi OO, Davis NL, Saleem B, Kapoor S, Okogbule-Wonodi AC, Viscardi RM, Sundararajan S. Impact of red blood cell transfusions on intestinal barrier function in preterm infants. J Neonatal Perinatal Med 2019; 12:95-101. [PMID: 30412507 DOI: 10.3233/npm-1828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To determine the relationships of red blood cell (RBC) transfusion and enteral feeding to changes in intestinal permeability (IP) measured by the relative intestinal uptake of lactulose (La) and rhamnose (Rh) in preterm infants <33 wk gestation. DESIGN/METHODS Infants 240-326wk gestation received La/Rh solution enterally on study days 1, 8 and 15.Urinary La/Rh ratio was measured by HPLC. Hematocrit preceding transfusion, total RBC transfusion volume, volume/kg, and feeding status during each study interval (birth-d1; d1-d8, and d8-d15) were determined. RESULTS Of the seventeen (40.5%) subjects who received≥1 transfusion during the study period, 12 (70.6%) infants were <28 wk gestation and 5 (29.4%) infants were≥28 wk gestation, p < 0.0001. Lower pre-transfusion hematocrit was observed in intervals preceding high IP (La/Rh > 0.05) than in intervals preceding low IP (La/Rh≤0.05) measurements (33 vs 35.8, p = 0.1051). RBC transfusions occurred more frequently in intervals preceding high IP than in intervals preceding low IP (26.8%; vs 8.3%, p = 0.0275) with 5-fold higher total RBC volume and volume/kg in intervals preceding any time point with high IP. RBC transfusion during an interval was associated with a three-fold increased risk of high IP (aOR 2.7; 95% C.I 0.564-12.814; p = 0.2143). Exclusive breast milk exposure and post-menstrual age reduced the risk for high IP following RBC transfusion. CONCLUSIONS Both RBC transfusion number and volume was associated with subsequent high IP measurements in preterm infants <33 weeks gestation and potentially may contribute to impairment of the preterm intestinal barrier.
Collapse
Affiliation(s)
- O O Ajayi
- Department of Pediatrics, University of Maryland School of Medicine, Division of Neonatology Baltimore, MD, USA
| | - N L Davis
- Department of Pediatrics, University of Maryland School of Medicine, Division of Neonatology Baltimore, MD, USA
| | - B Saleem
- Department of Pediatrics, University of Maryland School of Medicine, Division of Neonatology Baltimore, MD, USA
| | - S Kapoor
- Department of Pediatrics, University of Maryland School of Medicine, Division of Neonatology Baltimore, MD, USA
| | - A C Okogbule-Wonodi
- Department of Pediatrics, Howard University College of Medicine, Division of Neonatology, Washington, DC, USA
| | - R M Viscardi
- Department of Pediatrics, University of Maryland School of Medicine, Division of Neonatology Baltimore, MD, USA
| | - Sripriya Sundararajan
- Department of Pediatrics, University of Maryland School of Medicine, Division of Neonatology Baltimore, MD, USA
| |
Collapse
|
40
|
Castenmiller J, de Henauw S, Hirsch-Ernst KI, Kearney J, Knutsen HK, Maciuk A, Mangelsdorf I, McArdle HJ, Naska A, Pelaez C, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Bresson JL, Fewtrell M, Kersting M, Przyrembel H, Dumas C, Titz A, Turck D. Appropriate age range for introduction of complementary feeding into an infant's diet. EFSA J 2019; 17:e05780. [PMID: 32626427 PMCID: PMC7009265 DOI: 10.2903/j.efsa.2019.5780] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Following a request from the European Commission, the Panel on Nutrition, Novel Foods and Food Allergens (NDA) revised its 2009 Opinion on the appropriate age for introduction of complementary feeding of infants. This age has been evaluated considering the effects on health outcomes, nutritional aspects and infant development, and depends on the individual's characteristics and development. As long as foods have an age-appropriate texture, are nutritionally appropriate and prepared following good hygiene practices, there is no convincing evidence that at any age investigated in the included studies (< 1 to < 6 months), the introduction of complementary foods (CFs) is associated with adverse health effects or benefits (except for infants at risk of iron depletion). For nutritional reasons, the majority of infants need CFs from around 6 months of age. Infants at risk of iron depletion (exclusively breastfed infants born to mothers with low iron status, or with early umbilical cord clamping (< 1 min after birth), or born preterm, or born small-for-gestational age or with high growth velocity) may benefit from earlier introduction of CFs that are a source of iron. The earliest developmental skills relevant for consuming pureed CFs can be observed between 3 and 4 months of age. Skills for consuming finger foods can be observed in some infants at 4 months, but more commonly at 5-7 months. The fact that an infant may be ready from a neurodevelopmental perspective to progress to a more diversified diet before 6 months of age does not imply that there is a need to introduce CFs. There is no reason to postpone the introduction of potentially allergenic foods (egg, cereals, fish and peanut) to a later age than that of other CFs as far as the risk of developing atopic diseases is concerned. Regarding the risk of coeliac disease, gluten can be introduced with other CFs.
Collapse
|
41
|
Abstract
More than 10,000 preterm babies worldwide have been enrolled in trials evaluating probiotics administration for the prevention of necrotising enterocolitis, with very few adverse events reported. Despite this, probiotic safety is frequently cited as a concern when using this intervention. This review addresses why a preterm baby may be at risk when administered a live microbial product, short- and longer-term safety data in relation to probiotic use and regulatory aspects around probiotic manufacture and preparations.
Collapse
Affiliation(s)
- Paul F Fleming
- Homerton University Hospital NHS Foundation Trust, London, UK; Barts and the London School of Medicine and Dentistry, London, UK.
| | - Janet E Berrington
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Richardson Road, Newcastle, UK
| | - Susan E Jacobs
- Neonatal Services, The Royal Women's Hospital, Melbourne, Australia; Clinical Sciences, Murdoch Childrens Research Institute, Melbourne, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
42
|
A wave of Foxp3 + regulatory T cell accumulation in the neonatal liver plays unique roles in maintaining self-tolerance. Cell Mol Immunol 2019; 17:507-518. [PMID: 31171863 DOI: 10.1038/s41423-019-0246-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/15/2019] [Indexed: 12/24/2022] Open
Abstract
Newborn animals require tightly regulated local and systemic immune environments to govern the development and maturation of multiple organs/tissues even though the immune system itself is far from mature during the neonatal period. Regulatory T cells (Tregs) are essential for maintaining immune tolerance/homeostasis and modulating inflammatory responses. The features of Tregs in the neonatal liver under steady-state conditions are not well understood. The present study aimed to investigate the phenotype, functions, and significance of neonatal Tregs in the liver. We found a wave of thymus-derived Treg influx into the liver during 1-2 weeks of age. Depletion of these Tregs between days 7 and 11 after birth rapidly resulted in Th1-type liver inflammation and metabolic disorder. More Tregs in the neonatal liver than in the spleen underwent MHC II-dependent activation and proliferation, and the liver Tregs acquired stronger suppressive functions. The transcriptomic profile of these neonatal liver Tregs showed elevated expression of PPARγ and T-bet and features of Tregs that utilize lipid metabolic machinery and are capable of regulating Th1 responses. The accumulation of Tregs with unique features in the neonatal liver is critical to ensure self-tolerance and liver maturation.
Collapse
|
43
|
Grases-Pintó B, Torres-Castro P, Abril-Gil M, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ, Franch À. A Preterm Rat Model for Immunonutritional Studies. Nutrients 2019; 11:nu11050999. [PMID: 31052461 PMCID: PMC6566403 DOI: 10.3390/nu11050999] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/14/2022] Open
Abstract
Neonates are born with an immature immune system, which develops during the first stages of life. This early immaturity is more acute in preterm newborns. The aim of the present study was to set up a preterm rat model, in which representative biomarkers of innate and adaptive immunity maturation that could be promoted by certain dietary interventions are established. Throughout the study, the body weight was registered. To evaluate the functionality of the intestinal epithelial barrier, in vivo permeability to dextrans was measured and a histomorphometric study was performed. Furthermore, the blood cell count, phagocytic activity of blood leukocytes and plasmatic immunoglobulins (Ig) were determined. Preterm rats showed lower erythrocyte and platelet concentration but a higher count of leukocytes than the term rats. Although there were no changes in the granulocytes’ ability to phagocytize, preterm monocytes had lower phagocytic activity. Moreover, lower plasma IgG and IgM concentrations were detected in preterm rats compared to full-term rats, without affecting IgA. Finally, the intestinal study revealed lower permeability in preterm rats and reduced goblet cell size. Here, we characterized a premature rat model, with differential immune system biomarkers, as a useful tool for immunonutritional studies aimed at boosting the development of the immune system.
Collapse
Affiliation(s)
- Blanca Grases-Pintó
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain.
| | - Paulina Torres-Castro
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain.
| | - Mar Abril-Gil
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain.
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain.
| | - María J Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain.
| | - Francisco J Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain.
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain.
| |
Collapse
|
44
|
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 2018; 47:296-313. [DOI: 10.1124/dmd.118.084418] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022] Open
|
45
|
Navis M, Martins Garcia T, Renes IB, Vermeulen JL, Meisner S, Wildenberg ME, van den Brink GR, van Elburg RM, Muncan V. Mouse fetal intestinal organoids: new model to study epithelial maturation from suckling to weaning. EMBO Rep 2018; 20:embr.201846221. [PMID: 30530633 PMCID: PMC6362357 DOI: 10.15252/embr.201846221] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 11/02/2018] [Accepted: 11/19/2018] [Indexed: 11/09/2022] Open
Abstract
During the suckling-to-weaning transition, the intestinal epithelium matures, allowing digestion of solid food. Transplantation experiments with rodent fetal epithelium into subcutaneous tissue of adult animals suggest that this transition is intrinsically programmed and occurs in the absence of dietary or hormonal signals. Here, we show that organoids derived from mouse primary fetal intestinal epithelial cells express markers of late fetal and neonatal development. In a stable culture medium, these fetal epithelium-derived organoids lose all markers of neonatal epithelium and start expressing hallmarks of adult epithelium in a time frame that mirrors epithelial maturation in vivo In vitro postnatal development of the fetal-derived organoids accelerates by dexamethasone, a drug used to accelerate intestinal maturation in vivo Together, our data show that organoids derived from fetal epithelium undergo suckling-to-weaning transition, that the speed of maturation can be modulated, and that fetal organoids can be used to model the molecular mechanisms of postnatal epithelial maturation.
Collapse
Affiliation(s)
- Marit Navis
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, AG&M, University of Amsterdam, Amsterdam, The Netherlands
| | - Tânia Martins Garcia
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, AG&M, University of Amsterdam, Amsterdam, The Netherlands
| | - Ingrid B Renes
- Department of Pediatrics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Danone Nutricia Research, Utrecht, The Netherlands
| | - Jacqueline Lm Vermeulen
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, AG&M, University of Amsterdam, Amsterdam, The Netherlands
| | - Sander Meisner
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, AG&M, University of Amsterdam, Amsterdam, The Netherlands
| | - Manon E Wildenberg
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, AG&M, University of Amsterdam, Amsterdam, The Netherlands
| | - Gijs R van den Brink
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, AG&M, University of Amsterdam, Amsterdam, The Netherlands.,GlaxoSmithKline, Medicines Research Center, London, UK
| | - Ruurd M van Elburg
- Department of Pediatrics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Danone Nutricia Research, Utrecht, The Netherlands
| | - Vanesa Muncan
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, AG&M, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Guo B, Liu B, Wei H, Cheng KW, Chen F. Extract of the Microalga Nitzschia laevis Prevents High-Fat-Diet-Induced Obesity in Mice by Modulating the Composition of Gut Microbiota. Mol Nutr Food Res 2018; 63:e1800808. [PMID: 30475446 DOI: 10.1002/mnfr.201800808] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/01/2018] [Indexed: 12/18/2022]
Abstract
SCOPE The aim of the present study is to investigate the efficacy of Nitzschia laevis extract (NLE) in preventing obesity in mice fed with a high-fat diet (HFD), and the potential underlying mechanisms focusing on modulation of the gut microbiota profile. METHODS AND RESULTS Physiological, histological, and biochemical parameters and gut microbiota compositions are compared among four experimental groups fed respectively with the following diets for 8 weeks: Normal chow diet, HFD, HFD + low concentration of NLE, and HFD + high concentration of NLE. The results demonstrate that NLE supplementation significantly reduces body weight gain and effectively prevents lipid accumulation in the white adipose tissue and liver of the mice. Mechanistic analysis reveals that NLE promotes the expression of uncoupling protein 1 and peroxisome proliferators-activated receptor-γ coactivator-1 mRNA in brown adipose tissue. Furthermore, NLE protects the gut epithelium and positively reshapes the gut microbiota composition against the damaging effect of HFD. CONCLUSIONS NLE supplementation demonstrates a protective effect against HFD-induced obesity in mice, which is associated with reshaping the profile of gut microbiota. To the best of our knowledge, this has been the first report on the potential of microalgal extract to prevent obesity by modulating gut microbiota.
Collapse
Affiliation(s)
- Bingbing Guo
- Institute for Food and Bioresource Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Bin Liu
- Institute for Food and Bioresource Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Hehong Wei
- Institute for Food and Bioresource Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Ka-Wing Cheng
- Institute for Advanced Study, Shenzhen University, Nanshan District, Shenzhen, Guangdong, 518060, China
| | - Feng Chen
- Institute for Food and Bioresource Engineering, College of Engineering, Peking University, Beijing, 100871, China.,Institute for Advanced Study, Shenzhen University, Nanshan District, Shenzhen, Guangdong, 518060, China
| |
Collapse
|
47
|
van Sadelhoff JHJ, Mastorakou D, Weenen H, Stahl B, Garssen J, Hartog A. Short Communication: Differences in Levels of Free Amino Acids and Total Protein in Human Foremilk and Hindmilk. Nutrients 2018; 10:nu10121828. [PMID: 30486227 PMCID: PMC6315640 DOI: 10.3390/nu10121828] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/15/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022] Open
Abstract
Free amino acids (FAAs) in human milk are indicated to have specific functional roles in infant development. Studies have shown differences between human milk that is expressed at the beginning of a feed (i.e., foremilk) and the remainder of the milk expressed (i.e., hindmilk). For example, it is well established that human hindmilk is richer in fat and energy than foremilk. Hence, exclusively feeding hindmilk is used to enhance weight gain of preterm, low birthweight infants. Whether FAAs occur differently between foremilk and hindmilk has never been reported, but given their bioactive capacities, this is relevant to consider especially in situations where hindmilk is fed exclusively. Therefore, this study analyzed and compared the FAA and total protein content in human foremilk and hindmilk samples donated by 30 healthy lactating women. The total protein content was found to be significantly higher in hindmilk (p < 0.001), whereas foremilk contained a significantly higher total content of FAAs (p = 0.015). With regards to individual FAAs, foremilk contained significantly higher levels of phenylalanine (p = 0.009), threonine (p = 0.003), valine (p = 0.018), alanine (p = 0.004), glutamine (p < 0.001), and serine (p = 0.012) than hindmilk. Although statistical significance was reached, effect size analysis of the milk fraction on FAA levels in milk revealed that the observed differences were only small. To what extent these differences are of physiological importance for infant development remains to be examined in future research.
Collapse
Affiliation(s)
- Joris H J van Sadelhoff
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands.
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
| | - Dimitra Mastorakou
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands.
| | - Hugo Weenen
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands.
| | - Bernd Stahl
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands.
| | - Johan Garssen
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands.
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
| | - Anita Hartog
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, The Netherlands.
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
| |
Collapse
|
48
|
Supeck DJ, Assefa S, Meek W, Curtis JT, Köhler GA. Postnatal maturation of the intestinal epithelial barrier in prairie voles. Tissue Cell 2018; 54:30-37. [PMID: 30309507 DOI: 10.1016/j.tice.2018.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 08/01/2018] [Accepted: 08/01/2018] [Indexed: 11/15/2022]
Abstract
Intestinal epithelium develops during gestation and continues to mature post-natally into a selective barrier that will protect the individual while still allowing passage of nutrients. Until fully mature, the risk of translocation of microorganisms, toxins or antigens into the sub-epithelial tissue is high and could result in pathologies with life-altering consequences, or even premature death. Because of their monogamous mating system, prairie voles are an emerging model for studying the role of the intestinal microbiota in modulating social behavior via the microbiota-gut-brain-behavior axis. However, knowledge about the voles' intestinal barrier maturation is lacking. Understanding the maturation of the intestine epithelial barrier can complement the extensive behavioral literature for future studies involving the vole gut-brain axis. In this study, we characterized intestinal barrier function by demonstrating that two-week-old prairie voles have high paracellular absorption of FITC-dextran molecules prior to markedly decreased permeability at three weeks of age. In light of the fundamental role of tight junctions in maintaining epithelial integrity regulating intestinal permeability, we examined tight junction gene expression profiles. Transmission electron microscopy was used to visualize tight junction structure. Our results provide a timeline for intestinal barrier maturation and point to tight junction proteins involved in this process in prairie voles.
Collapse
Affiliation(s)
- David J Supeck
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Senait Assefa
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - William Meek
- Department of Anatomy and Cell Biology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - J Thomas Curtis
- Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Gerwald A Köhler
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA.
| |
Collapse
|
49
|
Bein A, Eventov-Friedman S, Arbell D, Schwartz B. Intestinal tight junctions are severely altered in NEC preterm neonates. Pediatr Neonatol 2018; 59:464-473. [PMID: 29276042 DOI: 10.1016/j.pedneo.2017.11.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 07/09/2017] [Accepted: 11/30/2017] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND & AIMS Necrotizing Enterocolitis (NEC) is a severe inflammatory disorder of the intestine endangering the health and survival of preterm infants. It is well established that the gut barrier is severely damaged in NEC patients, nonetheless an in depth investigation of modifications at the transcriptional and translational levels of tight junction genes and proteins during NEC are still missing. The aim of this study was to investigate changes in the expression of tight junctions and other associated proteins during NEC and determine their correlation to the disease severity. METHODS We examined intestinal specimens from six NEC patients and compared them with six control specimens from patients that underwent surgeries for reasons other than NEC. The expression of genes was analyzed by real time PCR and protein expression by immunohistochemistry. RESULTS The tight junction genes ZO-1, occludin, cingulin and claudin-4 were significantly down regulated in NEC. Furthermore TLR4, BAX and SIRT1 genes were found to be significantly down regulated while HIF-1A showed a trend of up regulation in NEC patients. These changes were found to correlate with the severity of the disease. Additionally we demonstrated in an ex-vivo model that hypoxic conditions initiated a destructive process of the epithelial barrier. We also showed that the expression of the tight junction proteins ZO-1 and occludin were significantly down regulated in NEC specimens. CONCLUSIONS The expression of tight junction proteins and their encoding genes are significantly altered in NEC. We surmise that SIRT1 and HIF-1A may play a role in controlling these effects.
Collapse
Affiliation(s)
- Amir Bein
- The Hebrew University of Jerusalem, School of Nutritional Sciences, Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, Rehovot, Israel
| | | | - Dan Arbell
- Pediatric Surgery, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Betty Schwartz
- The Hebrew University of Jerusalem, School of Nutritional Sciences, Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, Rehovot, Israel.
| |
Collapse
|
50
|
Yrjänä JMS, Koski T, Törölä H, Valkama M, Kulmala P. Very early introduction of semisolid foods in preterm infants does not increase food allergies or atopic dermatitis. Ann Allergy Asthma Immunol 2018; 121:353-359. [PMID: 29981439 DOI: 10.1016/j.anai.2018.06.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/21/2018] [Accepted: 06/28/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND The optimal age for the introduction of solid foods for infants has long been a controversial issue. OBJECTIVE To determine whether the early introduction of semisolid foods influences the incidence of food allergy or atopic dermatitis among preterm infants. METHODS Retrospective data from 464 preterm infants born in Oulu University Hospital between 2008 and 2012 were analyzed. Age- and sex-matched full-term control children from the general population were identified. The primary outcome of the study was the difference in timing of the introduction of complementary feeding between preterm and full-term infants. The secondary outcomes were the incidences of food allergies and atopic dermatitis by the ages of 1 and 2 years. RESULTS Semisolid food was introduced at the median corrected age of 1.4 months for all preterm infants, at 1.9 months for late preterm, at 0.9 months for very preterm, and at 0.1 months for extremely preterm infants. The cumulative incidence, either of food allergies or of atopic dermatitis, did not differ significantly between preterm infants and controls by the ages of 1 and 2 years. CONCLUSION The very early introduction of complementary foods into the diet of preterm babies did not increase the incidence of food allergies or atopic dermatitis even among the most preterm infants. This finding supports the hypothesis that the gut-associated lymphoid tissue of preterm infants is ready to encounter food proteins and to begin the maturation process within 3 to 6 months of birth, regardless of gestational age.
Collapse
Affiliation(s)
- Jaakko M S Yrjänä
- PEDEGO Research Unit and MRC Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.
| | - Teppo Koski
- PEDEGO Research Unit and MRC Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Helena Törölä
- PEDEGO Research Unit and MRC Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Marita Valkama
- PEDEGO Research Unit and MRC Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Petri Kulmala
- PEDEGO Research Unit and MRC Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland; Biomedicine Research Unit, Medical Microbiology and Immunology, University of Oulu, Oulu, Finland
| |
Collapse
|