1
|
Chaaban H, Burge K, McElroy SJ. Evolutionary bridges: how factors present in amniotic fluid and human milk help mature the gut. J Perinatol 2024; 44:1552-1559. [PMID: 38844520 PMCID: PMC11521761 DOI: 10.1038/s41372-024-02026-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 08/22/2024]
Abstract
Necrotizing enterocolitis (NEC) continues to be a leading cause of morbidity and mortality in preterm infants. As modern medicine significantly improves the survival of extremely premature infants, the persistence of NEC underscores our limited understanding of its pathogenesis. Due to early delivery, a preterm infant's exposure to amniotic fluid (AF) is abruptly truncated. Replete with bioactive molecules, AF plays an important role in fetal intestinal maturation and preparation for contact with the environment, thus its absence during development of the intestine may contribute to increased susceptibility to NEC. Human milk (HM), particularly during the initial phases of lactation, is a cornerstone of neonatal intestinal defense. The concentrations and activities of several bioactive factors in HM parallel those of AF, suggesting continuity of protection. In this review, we discuss the predominant overlapping bioactive components of HM and AF, with an emphasis on those associated with intestinal growth or reduction of NEC.
Collapse
Affiliation(s)
- Hala Chaaban
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Kathryn Burge
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Steven J McElroy
- Department of Pediatrics, Division of Neonatology, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
2
|
Grases-Pintó B, Torres-Castro P, Abril-Gil M, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ, Franch À. TGF-β2, EGF and FGF21 influence the suckling rat intestinal maturation. J Nutr Biochem 2024; 135:109778. [PMID: 39374742 DOI: 10.1016/j.jnutbio.2024.109778] [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: 05/24/2024] [Revised: 09/04/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Some of the growth factors present in breast milk, such as transforming growth factor-β (TGF-β), epidermal growth factor (EGF) and fibroblast growth factor 21 (FGF21), play important roles in the development of the intestinal tract. The aim of this study was to determine the effect of a supplementation with TGF-β2, EGF and FGF21 on suckling rats intestinal maturation. For this purpose, Wistar rats were supplemented daily with TGF-β2, EGF or FGF21 throughout the suckling period. We evaluated the functionality of the intestinal epithelial barrier through an in vivo dextran permeability assay, and by a histomorphometric and immunohistochemical study. In addition, the intestinal gene expression of tight junction-associated proteins, mucins, toll-like receptors, and maturation markers was analyzed. Moreover, the intraepithelial lymphocyte (IEL) phenotypical composition was established. During the suckling period, the supplementation with TGF-β2, EGF and FGF21 showed important signs of intestinal maturation. These results suggest that these molecules, present in breast milk, play a modulatory role in the maturation of the intestinal barrier function and the IEL composition during the suckling period.
Collapse
Affiliation(s)
- Blanca Grases-Pintó
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain; Nutrition and Food Safety Research Institute (INSA·UB), Santa Coloma de Gramenet, Spain
| | - Paulina Torres-Castro
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain; Nutrition and Food Safety Research Institute (INSA·UB), Santa Coloma de Gramenet, Spain
| | - Mar Abril-Gil
- Klinikum rechts der Isar, Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain; Nutrition and Food Safety Research Institute (INSA·UB), Santa Coloma de Gramenet, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - María J Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain; Nutrition and Food Safety Research Institute (INSA·UB), 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, Barcelona, Spain; Nutrition and Food Safety Research Institute (INSA·UB), Santa Coloma de Gramenet, Spain.
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain; Nutrition and Food Safety Research Institute (INSA·UB), Santa Coloma de Gramenet, Spain
| |
Collapse
|
3
|
Macchiaverni P, Rekima A, van den Elsen L, Renz H, Verhasselt V. Allergen shedding in human milk: Could it be key for immune system education and allergy prevention? J Allergy Clin Immunol 2021; 148:679-688. [PMID: 34310930 DOI: 10.1016/j.jaci.2021.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/19/2021] [Accepted: 07/19/2021] [Indexed: 02/02/2023]
Abstract
In addition to being a source of nutrients for the developing newborn, human milk contains thousands of bioactive compounds, which influence infant health in the short-term as exemplified by its major benefits on infectious disease prevention. Many of the human milk compounds also have the required characteristics to instruct immune development and guide long-term health. Prebiotics, probiotics, and varied antimicrobial molecules all have the potential to shape the composition and function of the establishing gut microbiota, which is known to be a major determinant of immune function. Another and less explored way human milk can instruct long-term immunity is through antigen shedding. Here, we will review the evidence that antigens from maternal environment and more specifically from allergen sources are found in human milk. We will discuss data from rodent models and birth cohorts showing that allergen shedding in breast milk may influence long-term allergy risk. We will uncover the variables that may underlie heterogeneity in oral tolerance induction and allergy prevention in children breast-fed by allergen-exposed mothers. We will focus on the parameters that control antigen transfer to breast milk, on the unique biological characteristics of allergens in breast milk, and on the milk bioactive compounds that were found to influence immune response in offspring. We propose this understanding is fundamental to guide maternal interventions leading to lifelong allergen tolerance.
Collapse
Affiliation(s)
- Patricia Macchiaverni
- School of Medicine and Biomedical Sciences, University of Western Australia, Perth, Australia; Telethon Kids Institute, Perth, Australia
| | - Akila Rekima
- School of Medicine and Biomedical Sciences, University of Western Australia, Perth, Australia; Telethon Kids Institute, Perth, Australia
| | - Lieke van den Elsen
- School of Medicine and Biomedical Sciences, University of Western Australia, Perth, Australia; Telethon Kids Institute, Perth, Australia
| | - Harald Renz
- Institute of Laboratory Medicine, Philipps Universität Marburg, Marburg, Germany; Member of the Universities of Giessen and Marburg Lung Center (UGMLC), Marburg, Germany; German Center for Lung Research (DZL), Marburg, Germany; In Vivo Planetary Health, Worldwide Universities Network (WUN), West New York
| | - Valerie Verhasselt
- School of Medicine and Biomedical Sciences, University of Western Australia, Perth, Australia; Telethon Kids Institute, Perth, Australia; In Vivo Planetary Health, Worldwide Universities Network (WUN), West New York.
| |
Collapse
|
4
|
Haque M, Koski KG, Scott ME. A gastrointestinal nematode in pregnant and lactating mice alters maternal and neonatal microbiomes. Int J Parasitol 2021; 51:945-957. [PMID: 34081970 DOI: 10.1016/j.ijpara.2021.03.008] [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: 08/19/2020] [Revised: 03/24/2021] [Accepted: 03/28/2021] [Indexed: 11/29/2022]
Abstract
The maternal microbiome is understood to be the principal source of the neonatal microbiome but the consequences of intestinal nematodes on pregnant and lactating mothers and implications for the neonatal microbiome are unknown. Using pregnant CD1 mice infected with Heligmosomoides bakeri, we investigated the microbiomes in maternal tissues (intestine, vagina, and milk) and in the neonatal stomach using MiSeq sequencing of bacterial 16S rRNA genes. Our first hypothesis was that maternal nematode infection altered the maternal intestinal, vaginal, and milk microbiomes and associated metabolic pathways. Maternal nematode infection was associated with increased beta-diversity and abundance of fermenting bacteria as well as Lactobacillus in the maternal caecum 2 days after parturition, together with down-regulated carbohydrate, amino acid and vitamin biosynthesis pathways. Maternal nematode infection did not alter the vaginal or milk microbiomes. Our second hypothesis was that maternal infection would shape colonization of the neonatal microbiome. Although the pup stomach microbiome was similar to that of the maternal vaginal microbiome, pups of infected dams had higher beta-diversity at day 2, and a dramatic expansion in the abundance of Lactobacillus between days 2 and 7 compared with pups nursing uninfected dams. Our third hypothesis that maternal nematode infection altered the composition of neonatal microbiomes was confirmed as we observed up-regulation of several putatively beneficial microbial pathways associated with synthesis of essential and branched-chain amino acids, vitamins, and short-chain fatty acids. We believe this is the first study to show that a nematode living in the maternal intestine is associated with altered composition and function of the neonatal microbiome.
Collapse
Affiliation(s)
- Manjurul Haque
- Institute of Parasitology, McGill University (Macdonald Campus), 21 111 Lakeshore Road, Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Kristine G Koski
- School of Human Nutrition, McGill University (Macdonald Campus), 21 111 Lakeshore Road, Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University (Macdonald Campus), 21 111 Lakeshore Road, Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada.
| |
Collapse
|
5
|
Grases-Pintó B, Abril-Gil M, Torres-Castro P, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ, Franch À. Rat Milk and Plasma Immunological Profile throughout Lactation. Nutrients 2021; 13:nu13041257. [PMID: 33920419 PMCID: PMC8070501 DOI: 10.3390/nu13041257] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 01/02/2023] Open
Abstract
The composition of bioactive factors with immune activity in human breast milk is widely studied. However, the knowledge on rat milk immune factors during the whole lactation period is still scarce. This study aimed to analyze rat breast milk’s immunoglobulin (Ig) content and some critical adipokines and growth factors throughout the lactation period, and to assess relationships with corresponding plasma levels. During lactation, milk concentration of the transforming growth factor (TGF)-β2 and -β3 showed a punctual increase in the first week, whereas adiponectin and leptin remained stable. In the second period of lactation (d14–21), despite the increase in the milk epidermal growth factor (EGF), a decrease in fibroblast growth factor 21 (FGF21) was detected at day 21. Milk IgA concentration had a progressive increase during lactation, while no significant changes were found in IgM and IgG. Regarding plasma levels, a decrease in all studied adipokines was observed in the second period of lactation, with the exception of IgA and TGF-β1, which reached their highest values at the end of the study. A positive correlation in IgM, IgG, and adipokine concentration was detected between milk and plasma compartments. In summary, the changes in the pattern of these bioactive compounds in rat milk and plasma and their relationships during lactation are established.
Collapse
Affiliation(s)
- Blanca Grases-Pintó
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- 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 (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- 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 (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- 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 (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- 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 (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence: ; Tel.: +34-934-024-505
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
6
|
Torres-Castro P, Grases-Pintó B, Abril-Gil M, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ, Franch À. Modulation of the Systemic Immune Response in Suckling Rats by Breast Milk TGF-β2, EGF and FGF21 Supplementation. Nutrients 2020; 12:nu12061888. [PMID: 32599899 PMCID: PMC7353385 DOI: 10.3390/nu12061888] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 12/17/2022] Open
Abstract
Breast milk is a rich fluid containing bioactive compounds such as specific growth factors (GF) that contribute to maturation of the immune system in early life. The aim of this study was to determine whether transforming growth factor-β2 (TGF-β2), epidermal growth factor (EGF) and fibroblast growth factor 21 (FGF21), compounds present in breast milk, could promote systemic immune maturation. For this purpose, newborn Wistar rats were daily supplemented with these GF by oral gavage during the suckling period (21 days of life). At day 14 and 21 of life, plasma for immunoglobulin (Ig) quantification was obtained and spleen lymphocytes were isolated, immunophenotyped and cultured to evaluate their ability to proliferate and release cytokines. The main result was obtained at day 14, when supplementation with EGF increased B cell proportion to reach levels observed at day 21. At the end of the suckling period, all GF increased the plasma levels of IgG1 and IgG2a isotypes, FGF21 balanced the Th1/Th2 cytokine response and both EGF and FGF21 modified splenic lymphocyte composition. These results suggested that the studied milk bioactive factors, mainly EGF and FGF21, may have modulatory roles in the systemic immune responses in early life, although their physiological roles remain to be established.
Collapse
Affiliation(s)
- Paulina Torres-Castro
- Section of Physiology, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (P.T.-C.); (B.G.-P.); (M.A.-G.); (M.C.); (M.J.R.-L.); (A.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Blanca Grases-Pintó
- Section of Physiology, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (P.T.-C.); (B.G.-P.); (M.A.-G.); (M.C.); (M.J.R.-L.); (A.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Mar Abril-Gil
- Section of Physiology, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (P.T.-C.); (B.G.-P.); (M.A.-G.); (M.C.); (M.J.R.-L.); (A.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Section of Physiology, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (P.T.-C.); (B.G.-P.); (M.A.-G.); (M.C.); (M.J.R.-L.); (A.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - María J. Rodríguez-Lagunas
- Section of Physiology, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (P.T.-C.); (B.G.-P.); (M.A.-G.); (M.C.); (M.J.R.-L.); (A.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Section of Physiology, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (P.T.-C.); (B.G.-P.); (M.A.-G.); (M.C.); (M.J.R.-L.); (A.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence: ; Tel.: +34-93-402-45-05
| | - Àngels Franch
- Section of Physiology, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; (P.T.-C.); (B.G.-P.); (M.A.-G.); (M.C.); (M.J.R.-L.); (A.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
7
|
Holvoet S, Perrot M, de Groot N, Prioult G, Mikogami T, Verhasselt V, Nutten S. Oral Tolerance Induction to Newly Introduced Allergen is Favored by a Transforming Growth Factor-β-Enriched Formula. Nutrients 2019; 11:E2210. [PMID: 31540231 PMCID: PMC6769637 DOI: 10.3390/nu11092210] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022] Open
Abstract
Food allergies have become a major healthcare concern, hence preventive efforts to ensure oral tolerance induction to newly introduced antigens are particularly relevant. Given that transforming growth factor-β (TGF-β) plays a key role in immune tolerance, we tested whether an infant formula enriched with TGF-β would improve oral tolerance induction. A partially hydrolyzed whey protein-based formula was enriched with cow's-milk-derived TGF-β (TGF-β-enriched formula) by adding a specific whey protein isolate (WPI). The manufacturing process was optimized to achieve a concentration of TGF-β within the range of human breast milk concentrations. Protection from allergic sensitization and immune response was assessed in a mouse model. Adult mice received the TGF-β-enriched formula, a control non-enriched formula, or water ad libitum for 13 days before sensitization and suboptimal tolerization to ovalbumin (OVA). When compared to non-tolerized mice, suboptimally-tolerized mice supplemented with the TGF-β-enriched formula showed significantly lower levels of total immunoglobulin-E (IgE) and OVA-specific (IgG1). Mouse mast-cell protease-1 (mMCP-1) and cytokine levels were also significantly decreased in suboptimally-tolerized mice fed the TGF-β-enriched formula. In conclusion, oral supplementation with cow's-milk-derived TGF-β decreased allergic responses to newly introduced allergens and thus reduced the risk of developing food allergy.
Collapse
Affiliation(s)
- Sébastien Holvoet
- Nestlé Institute of Health Science, Gastro Intestinal Health Department, 1000 Lausanne, Switzerland.
| | - Marie Perrot
- Nestlé Institute of Health Science, Gastro Intestinal Health Department, 1000 Lausanne, Switzerland.
| | | | - Guénolée Prioult
- Nestlé Product Technology Center Nutrition, 3510 Konolfingen, Switzerland.
| | | | - Valérie Verhasselt
- University Nice Sophia Antipolis, Hopital de l'Archet, 06200 Nice, France.
| | - Sophie Nutten
- Nestlé Institute of Health Science, Gastro Intestinal Health Department, 1000 Lausanne, Switzerland.
| |
Collapse
|
8
|
Alsharnoubi J, Ishaak M, Elsheikh S, Ezzat S. Transforming Growth Factor Beta-1 in Human Breast Milk and Its Correlation with Infants' Parameters. Breastfeed Med 2019; 14:404-407. [PMID: 30990330 DOI: 10.1089/bfm.2018.0214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background: Breastfeeding provides optimal nutrition and health protection for the infant; it contains many anti-inflammatory factors, including transforming growth factor beta-1 (TGF-β1). Our study aimed to measure the level of TGF-β1 in human milk and to find its correlation with some infant anthropometric characteristics. Subjects and Methods: A milk sample was collected from 84 mothers and the level of TGF-β1 was measured using enzyme-linked immunosorbent assay. Results: TGF-β1 was significantly higher in vegetarian mothers compared with nonvegetarian mothers (p = 0.044). Additionally, the mean value of breast milk TGF-β1 was significantly higher in mothers using contraceptive pills compared with those who do not (p = 0.021). Also, the mean value of TGF-β1 was significantly higher in infants 3-6 months than those <3 months (p = 0.010); also there was a significant difference regarding infants' weight and length with average weight and length (p = 0.042) and (p = 0.009), respectively. Conclusions: TGF-β1 in human milk may play a role in infants' growth and development; mothers' diet is known to influence TGF-β1 level and its relation to infants' age and weight. Contraceptive method could have an influence on TGF-β1 levels during breastfeeding.
Collapse
Affiliation(s)
- Jehan Alsharnoubi
- 1 Department of Pediatrics, National Institute of Laser Enhanced Sciences, Cairo University, Cairo, Egypt
| | - Marian Ishaak
- 2 Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | |
Collapse
|
9
|
TGF-β2, EGF, and FGF21 Growth Factors Present in Breast Milk Promote Mesenteric Lymph Node Lymphocytes Maturation in Suckling Rats. Nutrients 2018; 10:nu10091171. [PMID: 30150532 PMCID: PMC6163676 DOI: 10.3390/nu10091171] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/22/2018] [Accepted: 08/24/2018] [Indexed: 01/12/2023] Open
Abstract
Breast milk, due to its large number of nutrients and bioactive factors, contributes to optimal development and immune maturation in early life. In this study, we aimed to assess the influence of some growth factors present in breast milk, such as transforming growth factor-β2 (TGF-β2), epidermal growth factor (EGF), and fibroblast growth factor 21 (FGF21), on the immune response development. Newborn Wistar rats were supplemented daily with TGF-β2, EGF, or FGF21, throughout the suckling period. At day 14 and 21 of life, lymphocytes from mesenteric lymph nodes (MLNs) were isolated, immunophenotyped, and cultured to evaluate their ability to proliferate and release cytokines. The main results demonstrated that supplementation with TGF-β2, EGF, or FGF21 modified the lymphocyte composition in MLNs. At day 14, all supplementations were able to induce a lower percentage of natural killer (NK) cells with the immature phenotype (CD8+), and they reduced the CD8αα/CD8αβ ratio at day 21. Moreover, the cytokine pattern was modified by the three treatments, with a down regulation of interleukin (IL)-13 secretion. These results showed the contribution of these growth factors in the lymphocytes MLNs immune maturation during the neonatal period.
Collapse
|
10
|
Escuder-Vieco D, Espinosa-Martos I, Rodríguez JM, Fernández L, Pallás-Alonso CR. Effect of HTST and Holder Pasteurization on the Concentration of Immunoglobulins, Growth Factors, and Hormones in Donor Human Milk. Front Immunol 2018; 9:2222. [PMID: 30319659 PMCID: PMC6170621 DOI: 10.3389/fimmu.2018.02222] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/07/2018] [Indexed: 02/06/2023] Open
Abstract
Donor human milk (DHM) is submitted to Holder pasteurization (HoP) to ensure its microbiological safety in human milk banks but this treatment affects some of its bioactive compounds. The objective of this work was to compare the effects of HoP and high temperature short time (HTST) treatments on some bioactive compounds found in DHM. A total of 24 DHM batches were processed in a continuous HTST system (70, 72, and 75°C for 5-25 s) and by HoP (62.5°C for 30 min). The concentrations of immunoglobulins (Igs) A, G, and M, transforming growth factor-beta 2 (TGF-β2), adiponectine, ghrelin, and leptin were measured using a multiplex system, whereas the concentration of epidermal growth factor (EGF) was determined by ELISA. In relation to Igs, IgG showed the highest preservation rates (87-101%) after HTST treatments, followed by IgA (54-88%) and IgM (25-73%). Ig retention after any of the HTST treatments was higher than after HoP (p < 0.001). Treatment times required to reduce the concentration of IgM by 90% (D-value) were 130, 88, and 49 s at 70, 72, and 75°C, while the number of degrees Celsius required to change the D-value by one factor of 10 (z-value) was 11.79°C. None of the heat treatments had a significant effect on the concentrations of TGF-β2, EGF, adiponectin, and ghrelin. In contrast, leptin was detected only in 4 of the samples submitted to HoP, whereas it was present in all samples after the different HTST treatments, with retention rates ranging between 34 and 68%. Globally, the concentration of IgA, IgG, IgM, and leptin in DHM was significantly higher after HTST pasteurization performed in a continuous system designed to be used in human milk banks than after the HoP procedure that is routinely applied at present.
Collapse
Affiliation(s)
- Diana Escuder-Vieco
- Banco Regional de Leche Materna, Hospital Universitario 12 de Octubre, Instituto de Investigación i+12, Madrid, Spain
- *Correspondence: Diana Escuder-Vieco
| | | | - Juan M. Rodríguez
- Sección Departamental de Nutrición y Ciencia de los Alimentos (Veterinaria), Universidad Complutense de Madrid, Madrid, Spain
| | - Leónides Fernández
- Sección Departamental de Farmacia Galénica y Tecnología Alimentaria (Veterinaria), Universidad Complutense de Madrid, Madrid, Spain
| | - Carmen Rosa Pallás-Alonso
- Banco Regional de Leche Materna, Hospital Universitario 12 de Octubre, Instituto de Investigación i+12, Madrid, Spain
- Servicio de Neonatología, Hospital Universitario 12 de Octubre, Instituto de Investigación i+12, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
11
|
Abstract
Cytokines are required for normal growth and development of the mammary gland and TGF-β prominently represents an established effector of apoptosis, e.g., during involution of the mammary gland. By the control of intracellular signaling pathways, including JAK/STAT, MAPK, PI-3K, and NF-κB, cytokines efficiently regulate cell proliferation and inflammation in the breast. Therefore, cytokines are discussed also in a context of malignant mammary growth. As a group of tissue hormones produced by somatic cells or by cells from the immune system, cytokines are defined by their immunomodulatory potential. Over the past 40 years, multiple cytokines were identified in colostrum and milk. Importantly, cytokines derived from mammary secretions after birth are required for maturation of the immune system in the developing gastrointestinal tract from the suckling. Moreover, recent studies have further assessed the particular interactions between probiotic bacterial strains and cytokines. In light of the increasing prevalence of inflammatory diseases of the gastrointestinal system, the effects of probiotic microorganisms during milk fermentation may have immunotherapeutic potential in the future.
Collapse
Affiliation(s)
- Julia Brenmoehl
- Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Daniela Ohde
- Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Elisa Wirthgen
- Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Andreas Hoeflich
- Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany.
| |
Collapse
|
12
|
Coletta R, Roberts NA, Randles MJ, Morabito A, Woolf AS. Exogenous transforming growth factor-β1 enhances smooth muscle differentiation in embryonic mouse jejunal explants. J Tissue Eng Regen Med 2018; 12:252-264. [PMID: 28084682 PMCID: PMC6485323 DOI: 10.1002/term.2409] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/13/2016] [Accepted: 01/10/2017] [Indexed: 12/14/2022]
Abstract
An ex vivo experimental strategy that replicates in vivo intestinal development would in theory provide an accessible setting with which to study normal and dysmorphic gut biology. The current authors recently described a system in which mouse embryonic jejunal segments were explanted onto semipermeable platforms and fed with chemically defined serum-free media. Over 3 days in organ culture, explants formed villi and they began to undergo spontaneous peristalsis. As defined in the current study, the wall of the explanted gut failed to form a robust longitudinal smooth muscle (SM) layer as it would do in vivo over the same time period. Given the role of transforming growth factor β1 (TGFβ1) in SM differentiation in other organs, it was hypothesized that exogenous TGFβ1 would enhance SM differentiation in these explants. In vivo, TGFβ receptors I and II were both detected in embryonic longitudinal jejunal SM cells and, in organ culture, exogenous TGFβ1 induced robust differentiation of longitudinal SM. Microarray profiling showed that TGFβ1 increased SM specific transcripts in a dose dependent manner. TGFβ1 proteins were detected in amniotic fluid at a time when the intestine was physiologically herniated. By analogy with the requirement for exogenous TGFβ1 for SM differentiation in organ culture, the TGFβ1 protein that was demonstrated to be present in the amniotic fluid may enhance intestinal development when it is physiologically herniated in early gestation. Future studies of embryonic intestinal cultures should include TGFβ1 in the defined media to produce a more faithful model of in vivo muscle differentiation. Copyright © 2017 The Authors Journal of Tissue Engineering and Regenerative Medicine Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Riccardo Coletta
- Institute of Human Development, Faculty of Medical and Human SciencesUniversity of ManchesterUK
- Paediatric Autologous Bowel Reconstruction and Rehabilitation Unit, Department of Paediatric Surgery, Royal Manchester Children's HospitalCentral Manchester Foundation TrustManchesterUK
| | - Neil A. Roberts
- Institute of Human Development, Faculty of Medical and Human SciencesUniversity of ManchesterUK
| | - Michael J. Randles
- Institute of Human Development, Faculty of Medical and Human SciencesUniversity of ManchesterUK
- Wellcome Trust Centre for Cell‐Matrix Research, Faculty of Life SciencesUniversity of ManchesterManchesterUK
| | - Antonino Morabito
- Paediatric Autologous Bowel Reconstruction and Rehabilitation Unit, Department of Paediatric Surgery, Royal Manchester Children's HospitalCentral Manchester Foundation TrustManchesterUK
- Institute of Inflammation and Repair, Faculty of Medical and Human SciencesUniversity of ManchesterManchesterUK
| | - Adrian S. Woolf
- Institute of Human Development, Faculty of Medical and Human SciencesUniversity of ManchesterUK
- Department of Paediatric Nephrology, Royal Manchester Children's HospitalCentral Manchester Foundation TrustManchesterUK
| |
Collapse
|
13
|
Allergy prevention by breastfeeding: possible mechanisms and evidence from human cohorts. Curr Opin Allergy Clin Immunol 2017; 16:427-33. [PMID: 27518839 DOI: 10.1097/aci.0000000000000303] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Allergy is a modern disease which does not seem to benefit from breast milk preventive effects. We propose that maternal milk composition has not adapted to the needs of allergy prevention because of the recent and rapid increase of allergy. Modulation of breast milk composition may be the best strategy to counteract allergy development. We will review recent advances in understanding of allergy physiopathology and how breast milk factors may be specifically appropriate to interfere with allergy development in early life. RECENT FINDINGS There is strong evidence both from rodent and human studies that breast milk factors may impact on parameters which are now recognized to be essential for allergy physiopathology: infant gut barrier function, microbiota metabolites production, and oral tolerance induction. Data from human cohorts support the possibility to modify breast milk composition by selected interventions and to impact health outcomes in offspring. SUMMARY Nutritional intervention in lactating mothers should endow breast milk with the capacity to combat allergy epidemics in addition to infectious disease.
Collapse
|
14
|
Ruiz L, Espinosa-Martos I, García-Carral C, Manzano S, McGuire MK, Meehan CL, McGuire MA, Williams JE, Foster J, Sellen DW, Kamau-Mbuthia EW, Kamundia EW, Mbugua S, Moore SE, Kvist LJ, Otoo GE, Lackey KA, Flores K, Pareja RG, Bode L, Rodríguez JM. What's Normal? Immune Profiling of Human Milk from Healthy Women Living in Different Geographical and Socioeconomic Settings. Front Immunol 2017; 8:696. [PMID: 28713365 PMCID: PMC5492702 DOI: 10.3389/fimmu.2017.00696] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/29/2017] [Indexed: 12/31/2022] Open
Abstract
Human milk provides a very wide range of nutrients and bioactive components, including immune factors, human milk oligosaccharides, and a commensal microbiota. These factors are essential for interconnected processes including immunity programming and the development of a normal infant gastrointestinal microbiome. Newborn immune protection mostly relies on maternal immune factors provided through milk. However, studies dealing with an in-depth profiling of the different immune compounds present in human milk and with the assessment of their natural variation in healthy women from different populations are scarce. In this context, the objective of this work was the detection and quantification of a wide array of immune compounds, including innate immunity factors (IL1β, IL6, IL12, INFγ, TNFα), acquired immunity factors (IL2, IL4, IL10, IL13, IL17), chemokines (IL8, Groα, MCP1, MIP1β), growth factors [IL5, IL7, epidermal growth factor (EGF), granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, TGFβ2], and immunoglobulins (IgA, IgG, IgM), in milk produced by healthy women of different ethnicities living in different geographic, dietary, socioeconomic, and environmental settings. Among the analyzed factors, IgA, IgG, IgM, EGF, TGFβ2, IL7, IL8, Groα, and MIP1β were detected in all or most of the samples collected in each population and, therefore, this specific set of compounds might be considered as the "core" soluble immune factors in milk produced by healthy women worldwide. This approach may help define which immune factors are (or are not) common in milk produced by women living in various conditions, and to identify host, lifestyle, and environmental factors that affect the immunological composition of this complex biological fluid. Clinical Trial Registration: www.ClinicalTrials.gov, identifier NCT02670278.
Collapse
Affiliation(s)
- Lorena Ruiz
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Irene Espinosa-Martos
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
- Probisearch S.L., C/Santiago Grisolía, Tres Cantos, Spain
| | - Cristina García-Carral
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Susana Manzano
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Michelle K. McGuire
- School of Biological Sciences, Washington State University, Pullman, WA, United States
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA, United States
| | - Courtney L. Meehan
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | - Mark A. McGuire
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, United States
| | - Janet E. Williams
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, United States
| | - James Foster
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Daniel W. Sellen
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | | | | | - Samwel Mbugua
- Department of Human Nutrition, Egerton University, Nakuru, Kenya
| | - Sophie E. Moore
- Division of Women’s Health, King’s College London, London, United Kingdom
- MRC Unit, Serekunda, Gambia
| | | | - Gloria E. Otoo
- Department of Nutrition and Food Science, University of Ghana, Accra, Ghana
| | - Kimberly A. Lackey
- School of Biological Sciences, Washington State University, Pullman, WA, United States
| | - Katherine Flores
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | | | - Lars Bode
- Department of Pediatrics, and Mother Milk Infant Center of Research Excellence (MoMICoRE), University of California, San Diego, La Jolla, CA, United States
| | - Juan M. Rodríguez
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
15
|
Sanjabi S, Oh SA, Li MO. Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022236. [PMID: 28108486 DOI: 10.1101/cshperspect.a022236] [Citation(s) in RCA: 388] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine involved in both suppressive and inflammatory immune responses. After 30 years of intense study, we have only begun to elucidate how TGF-β alters immunity under various conditions. Under steady-state conditions, TGF-β regulates thymic T-cell selection and maintains homeostasis of the naïve T-cell pool. TGF-β inhibits cytotoxic T lymphocyte (CTL), Th1-, and Th2-cell differentiation while promoting peripheral (p)Treg-, Th17-, Th9-, and Tfh-cell generation, and T-cell tissue residence in response to immune challenges. Similarly, TGF-β controls the proliferation, survival, activation, and differentiation of B cells, as well as the development and functions of innate cells, including natural killer (NK) cells, macrophages, dendritic cells, and granulocytes. Collectively, TGF-β plays a pivotal role in maintaining peripheral tolerance against self- and innocuous antigens, such as food, commensal bacteria, and fetal alloantigens, and in controlling immune responses to pathogens.
Collapse
Affiliation(s)
- Shomyseh Sanjabi
- Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California 94158.,Department of Microbiology and Immunology, University of California, San Francisco, California 94143
| | - Soyoung A Oh
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
16
|
Rekima A, Macchiaverni P, Turfkruyer M, Holvoet S, Dupuis L, Baiz N, Annesi-Maesano I, Mercenier A, Nutten S, Verhasselt V. Long-term reduction in food allergy susceptibility in mice by combining breastfeeding-induced tolerance and TGF-β-enriched formula after weaning. Clin Exp Allergy 2017; 47:565-576. [PMID: 27883236 DOI: 10.1111/cea.12864] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/17/2016] [Accepted: 10/20/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Oral tolerance induction in early life is a promising approach for food allergy prevention. Its success requires the identification of factors necessary for its persistence. OBJECTIVES We aimed to assess in mice duration of allergy prevention by breastfeeding-induced oral tolerance and whether oral TGF-β supplementation after weaning would prolong it. METHODS We quantified ovalbumin (OVA) and OVA-specific immunoglobulin levels by ELISA in milk from the EDEN birth cohort. As OVA-specific Ig was found in all samples, we assessed whether OVA-immunized mice exposed to OVA during lactation could prevent allergic diarrhoea in their 6- and 13-week-old progeny. In some experiments, a TGF-β-enriched formula was given after weaning. RESULTS At 6 weeks, only 13% and 34% of mice breastfed by OVA-exposed mothers exhibited diarrhoea after six and seven OVA challenges vs. 44% and 72% in mice breastfed by naïve mothers (P = 0.02 and 0.01). Protection was associated with decreased levels of MMCP1 and OVA-specific IgE (P < 0.0001). At 13 weeks, although OVA-specific IgE remained low (P = 0.001), diarrhoea occurrence increased to 32% and 46% after six and seven OVA challenges in mice breastfed by OVA-exposed mothers. MMCP1 levels were not significantly inhibited. Supplementation with TGF-β after weaning induced a strong protection in 13-week-old mice breastfed by OVA-exposed mothers compared with mice breastfed by naive mothers (0%, 13% and 32% of diarrhoea at the fifth, sixth and seventh challenges vs. 17, 42 and 78%; P = 0.05, 0.0043 and 0.0017). MMCP1 levels decreased by half compared with control mice (P = 0.02). Prolonged protection was only observed in mice rendered tolerant by breastfeeding and was associated with an improved gut barrier. CONCLUSIONS In mice, prevention of food allergy by breastfeeding-induced tolerance is of limited duration. Nutritional intervention by TGF-β supplementation after weaning could prolong beneficial effects of breast milk on food allergy prevention.
Collapse
Affiliation(s)
- A Rekima
- University of Nice Sophia Antipolis, TIM, EA 6302, Nice, France
| | - P Macchiaverni
- Institute of Biomedical Sciences - University of São Paulo, São Paulo, Brazil
| | - M Turfkruyer
- University of Nice Sophia Antipolis, TIM, EA 6302, Nice, France
| | - S Holvoet
- Nestle Research Center, Lausanne, Switzerland
| | - L Dupuis
- Nestle Research Center, Lausanne, Switzerland
| | - N Baiz
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP UMRS 1136), Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Medical School Saint-Antoine, Paris, France
| | - I Annesi-Maesano
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP UMRS 1136), Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Medical School Saint-Antoine, Paris, France
| | - A Mercenier
- Nestle Research Center, Lausanne, Switzerland
| | - S Nutten
- Nestle Research Center, Lausanne, Switzerland
| | - V Verhasselt
- University of Nice Sophia Antipolis, TIM, EA 6302, Nice, France.,The International Inflammation 'in-FLAME' Network, Worldwide Universities Network, Perth, Australia
| |
Collapse
|
17
|
Milk growth factors and expression of small intestinal growth factor receptors during the perinatal period in mice. Pediatr Res 2016; 80:759-765. [PMID: 27603563 DOI: 10.1038/pr.2016.150] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/26/2016] [Indexed: 01/23/2023]
Abstract
BACKGROUND Growth factors (GFs) are milk bioactive components contributing to the regulation of neonatal small intestinal maturation, and their receptors on the small intestinal epithelium play essential roles in mediating the functions of GFs. There is limited data correlating milk GFs and their receptors in the neonatal small intestine during the perinatal period. METHODS Small intestines of C57BL/6N mouse pups were collected at regular intervals during fetal life and up to postnatal day (PD) 60. Gene expression of GF receptors was determined by real-time qPCR. Milk GF concentrations up to PD21 were analyzed by enzyme-linked immunosorbent assay. RESULTS The majority of GF receptors showed significantly greater expression in the fetus than in postnatal life, and a sharp decrease occurred from PD14 extending to PD60; solid food restriction (PD14 and PD18) did not affect this decrease. Concentrations of five detected milk GFs demonstrated that GFs and the corresponding small intestinal receptors exhibited different correlations, with only milk transforming growth factor β1 (TGF-β1) having a significant positive correlation with TGF-β receptor 1 mRNA. CONCLUSION Gene expression of small intestinal GF receptors is likely a process of neonatal intestinal maturation that is affected concurrently by milk GFs and additional endogenous factors.
Collapse
|
18
|
Whey protein concentrate enhances intestinal integrity and influences transforming growth factor-β1 and mitogen-activated protein kinase signalling pathways in piglets after lipopolysaccharide challenge. Br J Nutr 2016; 115:984-93. [PMID: 26810899 DOI: 10.1017/s0007114515005085] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Whey protein concentrate (WPC) has been reported to have protective effects on the intestinal barrier. However, the molecular mechanisms involved are not fully elucidated. Transforming growth factor-β1 (TGF-β1) is an important component in the WPC, but whether TGF-β1 plays a role in these processes is not clear. The aim of this study was to investigate the protective effects of WPC on the intestinal epithelial barrier as well as whether TGF-β1 is involved in these protection processes in a piglet model after lipopolysaccharide (LPS) challenge. In total, eighteen weanling pigs were randomly allocated to one of the following three treatment groups: (1) non-challenged control and control diet; (2) LPS-challenged control and control diet; (3) LPS+5 %WPC diet. After 19 d of feeding with control or 5 %WPC diets, pigs were injected with LPS or saline. At 4 h after injection, pigs were killed to harvest jejunal samples. The results showed that WPC improved (P<0·05) intestinal morphology, as indicated by greater villus height and villus height:crypt depth ratio, and intestinal barrier function, which was reflected by increased transepithelial electrical resistance and decreased mucosal-to-serosal paracellular flux of dextran (4 kDa), compared with the LPS group. Moreover, WPC prevented the LPS-induced decrease (P<0·05) in claudin-1, occludin and zonula occludens-1 expressions in the jejunal mucosae. WPC also attenuated intestinal inflammation, indicated by decreased (P<0·05) mRNA expressions of TNF-α, IL-6, IL-8 and IL-1β. Supplementation with WPC also increased (P<0·05) TGF-β1 protein, phosphorylated-Smad2 expression and Smad4 and Smad7 mRNA expressions and decreased (P<0·05) the ratios of the phosphorylated to total c-jun N-terminal kinase (JNK) and p38 (phospho-JNK:JNK and p-p38:p38), whereas it increased (P<0·05) the ratio of extracellular signal-regulated kinase (ERK) (phospho-ERK:ERK). Collectively, these results suggest that dietary inclusion of WPC attenuates the LPS-induced intestinal injury by improving mucosal barrier function, alleviating intestinal inflammation and influencing TGF-β1 canonical Smad and mitogen-activated protein kinase signalling pathways.
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW This article aims to review the evidence that breast milk can actively shape neonate gut immune system development toward a mature immune system capable of responding appropriately to encountered antigens. RECENT FINDINGS Recent findings in the adult have demonstrated the critical role of the interaction between diet, gut microbiota, gut epithelial cells and gut-associated lymphoid tissue in the development of immune responses. Here, we will review what is known in this field in the neonate, compare these data to those obtained in the adult and review how milk factors impact gut immune function in the short and long term. SUMMARY We propose that the neonate immune system and maternal milk represent an entity necessary to ensure not only appropriate function in early life but also long term immune homeostasis.
Collapse
|
20
|
Ozen A, Gulcan EM, Ercan Saricoban H, Ozkan F, Cengizlier R. Food Protein-Induced Non-Immunoglobulin E-Mediated Allergic Colitis in Infants and Older Children: What Cytokines Are Involved? Int Arch Allergy Immunol 2015; 168:61-8. [PMID: 26588792 DOI: 10.1159/000441471] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 09/29/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Food protein-induced allergic proctocolitis (FPIAP) is mostly a non-immunoglobulin E-mediated disease where a T-cell-mediated reaction to cow's milk protein has been suggested. We determined the expression of transforming growth factor (TGF)-β, TGF-β receptor-1, tumor necrosis factor (TNF)-α, CD86, and CD23 on the colon mucosa to investigate their roles in the pathogenesis of the two subtypes of FPIAP, i.e. infantile FPIAP and FPIAP in older children. METHODS Group 1 comprised children with infantile FPIAP (age <6 months, n = 21), group 2 referred to FPIAP in older children (age >1.5 years, n = 7), and group 3 included children with juvenile hyperplastic polyps (n = 22). Immunohistochemical staining of colonic biopsy specimens was performed. RESULTS The expression of TNF-α was significantly higher in groups 1 and 2 compared to group 3. Group 2 patients had a significantly lower TGF-β expression compared to the other groups. The expression of CD86 was higher in group 1 than in group 3 (p = 0.012). Eosinophil counts per high-power field in the lamina propria were significantly correlated with CD86 expression (p = 0.026, r = 0.388). CONCLUSION Our results suggest that TNF-α is implicated in the pathogenesis of both types of FPIAP. The decreased activity of TGF-β receptor-1 accompanied by the increased expression of CD86 in infants and the decreased activity of TGF-β in older children appear to play a role in the development of FPIAP.
Collapse
Affiliation(s)
- Ahmet Ozen
- Division of Pediatric Allergy, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | | | | | | | | |
Collapse
|
21
|
Bittar NMVR, Zulian JG, Ogias D, Gama P. Ghrelin and GHS-R in the rat gastric mucosa: Are they involved in regulation of growth during early weaning? Nutrition 2015; 32:101-7. [PMID: 26520918 DOI: 10.1016/j.nut.2015.06.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/10/2015] [Accepted: 06/28/2015] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Based on previous evidence showing that early weaning disturbs the ontogenesis of rat gastric glands, which are the major site of ghrelin synthesis, we investigated the distribution of ghrelin and its receptor (GHS-R) in the rat gastric epithelium during postnatal development and evaluated the effects of early weaning on their levels. Additionally, we studied the contribution of ghrelin to gastric growth during the abrupt nutrient transition. METHODS Wistar rats were submitted to early weaning at 15 d and suckling counterparts were taken as controls. RESULTS By running quantitative reverse transcription polymerase chain reaction, immunoblots, and immunohistochemistry, we detected a variation of ghrelin levels and an increase of expression and number of immunolabeled cells, 3 d after treatment (P < 0.05). Through confocal microscopy, we identified GHS-R in the neck region of the gland and did not observe changes in protein levels. Growth was evaluated after ghrelin antagonist ([D-Lys-3]-GHRP-6) administration, which reduced DNA synthesis index in early-weaned rats (P < 0.05) as determined by bromodeoxyuridine incorporation. CONCLUSION The present study demonstrated that ghrelin and GHS-R are distributed in gastric mucosa during the postnatal development, indicating that they can signal and function in epithelial cells. We concluded that early weaning increased ghrelin levels in the stomach, and it takes part of cell proliferation control that is essential for stomach growth. Therefore, among the many effects previously described for early weaning, this abrupt nutrient transition also changed ghrelin levels, which might represent an additional element in the complex mechanism that coordinates stomach development.
Collapse
Affiliation(s)
| | - Juliana Guimarães Zulian
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Daniela Ogias
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Patrícia Gama
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
22
|
All-Trans Retinoic Acid Induces TGF-β2 in Intestinal Epithelial Cells via RhoA- and p38α MAPK-Mediated Activation of the Transcription Factor ATF2. PLoS One 2015. [PMID: 26225425 PMCID: PMC4520553 DOI: 10.1371/journal.pone.0134003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Objective We have shown previously that preterm infants are at risk of necrotizing enterocolitis (NEC), an inflammatory bowel necrosis typically seen in infants born prior to 32 weeks’ gestation, because of the developmental deficiency of transforming growth factor (TGF)-β2 in the intestine. The present study was designed to investigate all-trans retinoic acid (atRA) as an inducer of TGF-β2 in intestinal epithelial cells (IECs) and to elucidate the involved signaling mechanisms. Methods AtRA effects on intestinal epithelium were investigated using IEC6 cells. TGF-β2 expression was measured using reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) and Western blots. Signaling pathways were investigated using Western blots, transiently-transfected/transduced cells, kinase arrays, chromatin immunoprecipitation, and selective small molecule inhibitors. Results AtRA-treatment of IEC6 cells selectively increased TGF-β2 mRNA and protein expression in a time- and dose-dependent fashion, and increased the activity of the TGF-β2 promoter. AtRA effects were mediated via RhoA GTPase, Rho-associated, coiled-coil-containing protein kinase 1 (ROCK1), p38α MAPK, and activating transcription factor (ATF)-2. AtRA increased phospho-ATF2 binding to the TGF-β2 promoter and increased histone H2B acetylation in the TGF-β2 nucleosome, which is typically associated with transcriptional activation. Conclusions AtRA induces TGF-β2 expression in IECs via RhoA- and p38α MAPK-mediated activation of the transcription factor ATF2. Further studies are needed to investigate the role of atRA as a protective/therapeutic agent in gut mucosal inflammation.
Collapse
|
23
|
Palm F, Kuhl J, Walter I, Budik S, Nagel C, Hirt R, Auer U, Eberspächer E, Aurich C, Aurich J. Colostrum Withdrawal is Without Effect on Duodenal Development in Newborn Foals. J Equine Vet Sci 2015. [DOI: 10.1016/j.jevs.2015.03.195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
24
|
Abstract
The significance of contact with microbes in early life for subsequent health has been the subject of intense research during the last 2 decades. Disturbances in the establishment of the indigenous intestinal microbiome caused by cesarean section delivery or antibiotic exposure in early life have been linked to the risk of immune-mediated and inflammatory conditions such as atopic disorders, inflammatory bowel disease and obesity later in life. Distinct microbial populations have recently been discovered at maternal sites including the amniotic cavity and breast milk, as well as meconium, which have previously been thought to be sterile. Our understanding of the impact of fetal microbial contact on health outcomes is still rudimentary. Breast milk is known to modulate immune and metabolic programming. The breast milk microbiome is hypothesized to guide infant gut colonization and is affected by maternal health status and mode of delivery. Immunomodulatory factors in breast milk interact with the maternal and infant gut microbiome and may mediate some of the health benefits associated with breastfeeding. The intimate connection between the mother and the fetus or the infant is a potential target for microbial therapeutic interventions aiming to support healthy microbial contact and protect against disease.
Collapse
|
25
|
Takahashi N, Takahashi K, Kobayashi M, Yada Y, Koike Y, Kono Y. Constitutively high-level expression of TGFβ isoforms in cord blood and its relationship to perinatal findings. Cytokine 2015; 73:101-7. [PMID: 25743244 DOI: 10.1016/j.cyto.2015.01.024] [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: 08/14/2014] [Revised: 01/17/2015] [Accepted: 01/23/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND The clinical significance of TGFβ isoforms in cord blood is not well understood. METHODS We obtained cord blood samples from 37 term infants and 85 preterm infants who were born in several clinical settings. The serum levels of 3 TGFβ isoforms and of the other 17 cytokines in cord blood were investigated using cytometric bead array technology. RESULTS Very high levels of TGFβ1 and TGFβ2 isoforms compared to the level of other cytokines were found; mean levels were 44,180 and 1871pg/mL, respectively. The levels of all 3 isoforms of TGFβ were significantly correlated with birth weight, and the levels of TGFβ1 and TGFβ3 were correlated with gestational age. The levels of TGFβ1 and β2 isoforms were strongly correlated with each other, but not with levels of other cytokines. The levels of TGFβ1 and TGFβ2 were significantly higher in male infants and significantly lower in infants with fetal growth restriction. The prevalence of chronic lung disease was related to a low level of TGFβ1, and that of patent ductus arteriosus was related to a high level of TGFβ1 in preterm infants. CONCLUSIONS TGFβ1 and TGFβ2 appeared to play a significant role in physiological and pathological conditions in the fetus. TGFβ isoform levels appear to be regulated independently of those of other cytokines and do not appear to be influenced by inflammation in the fetal period. The role of TGFβ3 in cord blood and the postnatal chronological changes of the TGFβ isoforms should be investigated in the future.
Collapse
Affiliation(s)
- Naoto Takahashi
- Department of Pediatrics, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Pediatrics, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi-ken 329-0498, Japan.
| | - Kayo Takahashi
- Department of Obstetrics and Gynecology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi-ken 329-0498, Japan
| | - Mami Kobayashi
- Department of Pediatrics, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi-ken 329-0498, Japan
| | - Yukari Yada
- Department of Pediatrics, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi-ken 329-0498, Japan
| | - Yasunori Koike
- Department of Pediatrics, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi-ken 329-0498, Japan
| | - Yumi Kono
- Department of Pediatrics, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi-ken 329-0498, Japan
| |
Collapse
|
26
|
Xiao K, Song ZH, Jiao LF, Ke YL, Hu CH. Developmental changes of TGF-β1 and Smads signaling pathway in intestinal adaption of weaned pigs. PLoS One 2014; 9:e104589. [PMID: 25170924 PMCID: PMC4149345 DOI: 10.1371/journal.pone.0104589] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 07/10/2014] [Indexed: 12/18/2022] Open
Abstract
Weaning stress caused marked changes in intestinal structure and function. Transforming growth factor-β1 (TGF-β1) and canonical Smads signaling pathway are suspected to play an important regulatory role in post-weaning adaptation of the small intestine. In the present study, the intestinal morphology and permeability, developmental expressions of tight junction proteins and TGF-β1 in the intestine of piglets during the 2 weeks after weaning were assessed. The expressions of TGF-β receptor I/II (TβRI, TβRII), smad2/3, smad4 and smad7 were determined to investigate whether canonical smads signaling pathways were involved in early weaning adaption process. The results showed that a shorter villus and deeper crypt were observed on d 3 and d 7 postweaning and intestinal morphology recovered to preweaning values on d 14 postweaning. Early weaning increased (P<0.05) plasma level of diamine oxidase (DAO) and decreased DAO activities (P<0.05) in intestinal mucosa on d 3 and d 7 post-weaning. Compared with the pre-weaning stage (d 0), tight junction proteins level of occludin and claudin-1 were reduced (P<0.05) on d 3, 7 and 14 post-weaning, and ZO-1 protein was reduced (P<0.05) on d 3 and d 7 post-weaning. An increase (P<0.05) of TGF-β1 in intestinal mucosa was observed on d 3 and d 7 and then level down on d 14 post-weaning. Although there was an increase (P<0.05) of TβR II protein expression in the intestinal mucosa on d3 and d 7, no significant increase of mRNA of TβRI, TβRII, smad2/3, smad4 and smad7 was observed during postweaning. The results indicated that TGF-β1 was associated with the restoration of intestinal morphology and barrier function following weaning stress. The increased intestinal endogenous TGF-β1 didn't activate the canonical Smads signaling pathway.
Collapse
Affiliation(s)
- Kan Xiao
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Ze-He Song
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Le-Fei Jiao
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Ya-Lu Ke
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Cai-Hong Hu
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| |
Collapse
|
27
|
Fiore APZP, Osaki LH, Gama P. Transforming growth factor β1 increases p27 levels via synthesis and degradation mechanisms in the hyperproliferative gastric epithelium in rats. PLoS One 2014; 9:e101965. [PMID: 25000203 PMCID: PMC4085006 DOI: 10.1371/journal.pone.0101965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 06/13/2014] [Indexed: 12/31/2022] Open
Abstract
Throughout postnatal development, the gastric epithelium expresses Transforming Growth Factor beta1 (TGFβ1), but it is also exposed to luminal peptides that are part of milk. During suckling period, fasting promotes the withdrawal of milk-born molecules while it stimulates gastric epithelial cell proliferation. Such response can be reversed by exogenous TGFβ1, as it directly affects cell cycle through the regulation of p27 levels. We used fasting condition to induce the hyperproliferation of gastric epithelial cells in 14-day-old Wistar rats, and evaluated the effects of TGFβ1 gavage on p27 expression, phosphorylation at threonine 187 (phospho-p27Thr187) and degradation. p27 protein level was reduced during fasting when compared to suckling counterparts, while phospho-p27Thr187/p27 ratio was increased. TGFβ1 gavage reversed this response, which was confirmed through immunostaining. By using a neutralizing antibody against TGFβ1, we found that it restored the p27 and phosphorylation levels detected during fasting, indicating the specific role of the growth factor. We noted that neither fasting nor TGFβ1 changed p27 expression, but after cycloheximide administration, we observed that protein synthesis was influenced by TGFβ1. Next, we evaluated the capacity of the gastric mucosa to degrade p27 and we recorded a higher concentration of the remaining protein in pups treated with TGFβ1, suggesting augmented stability under this condition. Thus, we showed for the first time that luminal TGFβ1 increased p27 levels in the rat gastric mucosa by up- regulating translation and reducing protein degradation. We concluded that such mechanisms might be used by rapidly proliferating cells to respond to milk-born TGFβ1 and food restriction.
Collapse
Affiliation(s)
- Ana P. Z. P. Fiore
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, SP Brazil
| | - Luciana H. Osaki
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, SP Brazil
| | - Patricia Gama
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, SP Brazil
- * E-mail:
| |
Collapse
|
28
|
|
29
|
Namachivayam K, Blanco CL, Frost BL, Reeves AA, Jagadeeswaran R, MohanKumar K, Safarulla A, Mandal P, Garzon SA, Raj JU, Maheshwari A. Preterm human milk contains a large pool of latent TGF-β, which can be activated by exogenous neuraminidase. Am J Physiol Gastrointest Liver Physiol 2013; 304:G1055-65. [PMID: 23558011 PMCID: PMC3680715 DOI: 10.1152/ajpgi.00039.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human milk contains substantial amounts of transforming growth factor (TGF)-β, particularly the isoform TGF-β2. We previously showed in preclinical models that enterally administered TGF-β2 can protect against necrotizing enterocolitis (NEC), an inflammatory bowel necrosis of premature infants. In this study we hypothesized that premature infants remain at higher risk of NEC than full-term infants, even when they receive their own mother's milk, because preterm human milk contains less bioactive TGF-β than full-term milk. Our objective was to compare TGF-β bioactivity in preterm vs. full-term milk and identify factors that activate milk-borne TGF-β. Mothers who delivered between 23 0/7 and 31 6/7 wk or at ≥37 wk of gestation provided milk samples at serial time points. TGF-β bioactivity and NF-κB signaling were measured using specific reporter cells and in murine intestinal tissue explants. TGF-β1, TGF-β2, TGF-β3, and various TGF-β activators were measured by real-time PCR, enzyme immunoassays, or established enzymatic activity assays. Preterm human milk showed minimal TGF-β bioactivity in the native state but contained a large pool of latent TGF-β. TGF-β2 was the predominant isoform of TGF-β in preterm milk. Using a combination of several in vitro and ex vivo models, we show that neuraminidase is a key regulator of TGF-β bioactivity in human milk. Finally, we show that addition of bacterial neuraminidase to preterm human milk increased TGF-β bioactivity. Preterm milk contains large quantities of TGF-β, but most of it is in an inactive state. Addition of neuraminidase can increase TGF-β bioactivity in preterm milk and enhance its anti-inflammatory effects.
Collapse
Affiliation(s)
- Kopperuncholan Namachivayam
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Cynthia L. Blanco
- 3Division of Neonatology, Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas;
| | - Brandy L. Frost
- 4Department of Pathology, University of Illinois at Chicago, Chicago, Illinois; ,5Department of Pediatrics, NorthShore University Health System, Evanston, Illinois;
| | - Aaron A. Reeves
- 3Division of Neonatology, Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas;
| | - Ramasamy Jagadeeswaran
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Krishnan MohanKumar
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Azif Safarulla
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Partha Mandal
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Steven A. Garzon
- 2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,6University of Chicago Pritzker School of Medicine, Chicago, Illinois; and
| | - J. Usha Raj
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Akhil Maheshwari
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,7Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
30
|
Osaki LH, Gama P. MAPKs and signal transduction in the control of gastrointestinal epithelial cell proliferation and differentiation. Int J Mol Sci 2013; 14:10143-61. [PMID: 23670595 PMCID: PMC3676833 DOI: 10.3390/ijms140510143] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 04/19/2013] [Accepted: 04/22/2013] [Indexed: 02/06/2023] Open
Abstract
Mitogen-activated protein kinase (MAPK) pathways are activated by several stimuli and transduce the signal inside cells, generating diverse responses including cell proliferation, differentiation, migration and apoptosis. Each MAPK cascade comprises a series of molecules, and regulation takes place at different levels. They communicate with each other and with additional pathways, creating a signaling network that is important for cell fate determination. In this review, we focus on ERK, JNK, p38 and ERK5, the major MAPKs, and their interactions with PI3K-Akt, TGFβ/Smad and Wnt/β-catenin pathways. More importantly, we describe how MAPKs regulate cell proliferation and differentiation in the rapidly renewing epithelia that lines the gastrointestinal tract and, finally, we highlight the recent findings on nutritional aspects that affect MAPK transduction cascades.
Collapse
Affiliation(s)
- Luciana H Osaki
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, SP 05508-000, Brazil.
| | | |
Collapse
|
31
|
The suckling rat as a model for immunonutrition studies in early life. Clin Dev Immunol 2012; 2012:537310. [PMID: 22899949 PMCID: PMC3415261 DOI: 10.1155/2012/537310] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 06/18/2012] [Accepted: 06/19/2012] [Indexed: 12/17/2022]
Abstract
Diet plays a crucial role in maintaining optimal immune function. Research demonstrates the immunomodulatory properties and mechanisms of particular nutrients; however, these aspects are studied less in early life, when diet may exert an important role in the immune development of the neonate. Besides the limited data from epidemiological and human interventional trials in early life, animal models hold the key to increase the current knowledge about this interaction in this particular period. This paper reports the potential of the suckling rat as a model for immunonutrition studies in early life. In particular, it describes the main changes in the systemic and mucosal immune system development during rat suckling and allows some of these elements to be established as target biomarkers for studying the influence of particular nutrients. Different approaches to evaluate these immune effects, including the manipulation of the maternal diet during gestation and/or lactation or feeding the nutrient directly to the pups, are also described in detail. In summary, this paper provides investigators with useful tools for better designing experimental approaches focused on nutrition in early life for programming and immune development by using the suckling rat as a model.
Collapse
|
32
|
TGF-β2 induces maturation of immature human intestinal epithelial cells and inhibits inflammatory cytokine responses induced via the NF-κB pathway. J Pediatr Gastroenterol Nutr 2012; 54:630-8. [PMID: 22067113 PMCID: PMC3319014 DOI: 10.1097/mpg.0b013e31823e7c29] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Breast milk transforming growth factor (TGF)-β2 is associated with healthy immune maturation and reduced risk of immune-mediated disease in infants. We sought to investigate whether conditioning with TGF-β2 may result in a more mature immune responder phenotype in immature human intestinal epithelial cells (IECs). METHODS Primary human fetal IECs (hFIECs) and the human fetal small intestinal epithelial cell line (H4 cells) were conditioned with breast milk levels of TGF-β2, and an inflammatory response was subsequently induced. Inflammatory cytokine secretion and mRNA expression were measured by enzyme-linked immunosorbent assay and quantitative real-time polymerase chain reaction, respectively. Alterations in activation of inflammatory signaling pathways were detected from IECs by immunoblotting and immunofluorescence. The effects of TGF-β2 conditioning on gene expression patterns in hFIECs were assessed by cDNA microarray analysis and quantitative PCR. RESULTS Conditioning with TGF-β2 significantly attenuated subsequent interleukin (IL)-1β-, TNF-α-, and poly I:C-induced IL-8 and IL-6 responses in immature human IECs. Conditioning with TGF-β2 inhibited IL-1β-induced IκB-α degradation and NF-κB p65 nuclear translocation, which may partially result from TGF-β2-induced changes in the expression of genes in the NF-κB signaling pathway detected by cDNA microarray and qPCR. CONCLUSIONS Conditioning with TGF-β2 attenuates the subsequent inflammatory cytokine response in immature human IECs by inhibiting signaling in the NF-κB pathway. The immunomodulatory potential of breast milk may in part be mediated by TGF-β2, which may provide a novel means of supporting intestinal immune maturation in neonates.
Collapse
|
33
|
Early oral ovalbumin exposure during maternal milk feeding prevents spontaneous allergic sensitization in allergy-prone rat pups. Clin Dev Immunol 2011; 2012:396232. [PMID: 22203855 PMCID: PMC3235444 DOI: 10.1155/2012/396232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 08/09/2011] [Accepted: 09/08/2011] [Indexed: 12/27/2022]
Abstract
There are conflicting data to support the practice of delaying the introduction of allergenic foods into the infant diet to prevent allergy development. This study investigated immune response development after early oral egg antigen (Ovalbumin; OVA) exposure in a rat pup model. Brown Norway (BN) rat pups were randomly allocated into groups: dam reared (DR), DR pups challenged daily (days 4–13) with oral OVA (DR + OVAc), DR pups challenged intermittently (on day 4, 10, 12, and 13) with oral OVA (DR + OVAi), formula-fed pups (FF), and FF pups challenged daily with oral OVA (FF + OVA). Immune parameters assessed included OVA-specific serum IgE, IgG1, and IgA. Ileal and splenic messenger ribonucleic acid (mRNA) expression of transforming growth factor-beta (TGF-β1), mothers against decapentaplegic (Smad) 2/4/7, and forkhead box P3 (Foxp3) were determined. Ileum was stained for TGF-β1 and Smad4. Results. Feeding OVA daily to DR pups maintained systemic and local gut antibody and immunoregulatory marker mRNA responses. Systemic TGF-β1 was lower in DR + OVAi pups compared to DR and DR + OVAc pups. Feeding OVA to FF pups resulted in significantly greater OVA-specific IgE and IgG1, and lower IgA and TGF-β1 and Smad expression compared to DR pups. Conclusions. Early daily OVA exposure in the presence of maternal milk maintains immune markers associated with a regulated immune response, preventing early allergic sensitization.
Collapse
|
34
|
Caubet JC, Nowak-Węgrzyn A. Current understanding of the immune mechanisms of food protein-induced enterocolitis syndrome. Expert Rev Clin Immunol 2011; 7:317-27. [PMID: 21595598 DOI: 10.1586/eci.11.13] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Food protein-induced enterocolitis syndrome (FPIES) is an under-recognized and frequently misdiagnosed non-IgE-mediated food hypersensitivity disorder, characterized by severe vomiting and/or diarrhea. Despite the potential severity of acute reactions, FPIES can be considered self-limiting as avoidance of the incriminating allergen(s) leads to resolution of symptoms. Symptoms typically begin in the first month of life in association with failure to thrive and may progress to acidemia and shock. Although FPIES is well established as a distinct clinical entity, its pathophysiology has not yet been clearly defined and requires further characterization. Several immunologic alterations have been reported in FPIES, suggesting the involvement of antigen-specific T cells and their production of proinflammatory cytokines that regulate the permeability of the intestinal barrier. Humoral immune responses may also be involved in the pathomechanism of FPIES. The aim of this article is to delineate the immunological characteristics of this disorder based on the existing reports and to review the possible pathophysiologic basis of this disease.
Collapse
Affiliation(s)
- Jean-Christoph Caubet
- Jaffe Food Allergy Institute, Department of Pediatrics, Division of Allergy and Immunology, Mount Sinai School of Medicine, New York, NY, USA
| | | |
Collapse
|
35
|
Oddy WH, McMahon RJ. Milk-derived or recombinant transforming growth factor-beta has effects on immunological outcomes: a review of evidence from animal experimental studies. Clin Exp Allergy 2011; 41:783-93. [DOI: 10.1111/j.1365-2222.2011.03762.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
36
|
Rautava S, Nanthakumar NN, Dubert-Ferrandon A, Lu L, Rautava J, Walker WA. Breast milk-transforming growth factor-β₂ specifically attenuates IL-1β-induced inflammatory responses in the immature human intestine via an SMAD6- and ERK-dependent mechanism. Neonatology 2011; 99:192-201. [PMID: 20881435 PMCID: PMC3214931 DOI: 10.1159/000314109] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 04/20/2010] [Indexed: 01/26/2023]
Abstract
BACKGROUND Breast milk is known to protect the infant against infectious and immuno-inflammatory diseases, but the mechanisms of this protection are poorly understood. OBJECTIVES We hypothesized that transforming growth factor-β₂ (TGF-β₂), an immunoregulatory cytokine abundant in breast milk, may have a direct anti-inflammatory effect on immature human intestinal epithelial cells (IECs). METHODS Human fetal ileal organ culture, primary human fetal IECs, and the human fetal small intestinal epithelial cell line H4 were stimulated with interleukin 1β (IL-1β) with or without TGF-β₂. Pro-inflammatory cytokine secretion and mRNA expression were measured by ELISA and quantitative real-time polymerase chain reaction, respectively. Alterations in ERK signalling were detected from IECs by immunoblotting and in fetal ileal tissue culture by immunohistochemistry. SMAD6 knockdown was performed by transfecting the cells with SMAD6 siRNA. RESULTS TGF-β₂ significantly attenuated IL-1β-induced pro-inflammatory cytokine production in fetal intestinal organ culture and the cell culture models. In addition, TGF-β₂ reduced the IL-1β-induced IL-8 and IL-6 mRNA response in H4 cells. TGF-β₂ markedly inhibited IL-1β-induced phosphorylation of ERK, which was necessary for the cytokine response. The inhibitory effect of TGF-β₂ on IL-1β-induced cytokine production was completely abrogated by SMAD6 siRNA knockdown. CONCLUSIONS TGF-β₂ attenuates IL-1β-induced pro-inflammatory cytokine production in immature human IECs by inhibiting ERK signalling. The anti-inflammatory effect of TGF-β₂ is dependent on SMAD6. Breast milk TGF-β₂ may provide the neonate with important immunoregulatory support. TGF-β₂ might provide a novel means to improve intestinal immunophysiology in premature neonates.
Collapse
Affiliation(s)
- Samuli Rautava
- Developmental Gastroenterology Laboratory, Division of Pediatric Gastroenterology, Massachusetts General Hospital for Children, Charlestown, Mass., USA.
| | | | | | | | | | | |
Collapse
|
37
|
Maheshwari A, Kelly DR, Nicola T, Ambalavanan N, Jain SK, Murphy-Ullrich J, Athar M, Shimamura M, Bhandari V, Aprahamian C, Dimmitt RA, Serra R, Ohls RK. TGF-β2 suppresses macrophage cytokine production and mucosal inflammatory responses in the developing intestine. Gastroenterology 2011; 140:242-53. [PMID: 20875417 PMCID: PMC3008335 DOI: 10.1053/j.gastro.2010.09.043] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Revised: 08/30/2010] [Accepted: 09/14/2010] [Indexed: 01/02/2023]
Abstract
BACKGROUND & AIMS Premature neonates are predisposed to necrotizing enterocolitis (NEC), an idiopathic, inflammatory bowel necrosis. We investigated whether NEC occurs in the preterm intestine due to incomplete noninflammatory differentiation of intestinal macrophages, which increases the risk of a severe mucosal inflammatory response to bacterial products. METHODS We compared inflammatory properties of human/murine fetal, neonatal, and adult intestinal macrophages. To investigate gut-specific macrophage differentiation, we next treated monocyte-derived macrophages with conditioned media from explanted human fetal and adult intestinal tissues. Transforming growth factor-β (TGF-β) expression and bioactivity were measured in fetal/adult intestine and in NEC. Finally, we used wild-type and transgenic mice to investigate the effects of deficient TGF-β signaling on NEC-like inflammatory mucosal injury. RESULTS Intestinal macrophages in the human preterm intestine (fetus/premature neonate), but not in full-term neonates and adults, expressed inflammatory cytokines. Macrophage cytokine production was suppressed in the developing intestine by TGF-β, particularly the TGF-β(2) isoform. NEC was associated with decreased tissue expression of TGF-β(2) and decreased TGF-β bioactivity. In mice, disruption of TGF-β signaling worsened NEC-like inflammatory mucosal injury, whereas enteral supplementation with recombinant TGF-β(2) was protective. CONCLUSIONS Intestinal macrophages progressively acquire a noninflammatory profile during gestational development. TGF-β, particularly the TGF-β(2) isoform, suppresses macrophage inflammatory responses in the developing intestine and protects against inflammatory mucosal injury. Enterally administered TGF-β(2) protected mice from experimental NEC-like injury.
Collapse
Affiliation(s)
- Akhil Maheshwari
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Verhasselt V. Neonatal tolerance under breastfeeding influence. Curr Opin Immunol 2010; 22:623-30. [PMID: 20851590 DOI: 10.1016/j.coi.2010.08.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 08/12/2010] [Indexed: 12/20/2022]
Abstract
Diseases due to defect in tolerance induction such as allergy, celiac disease, or Type 1 Diabetes develop mostly in childhood indicating the necessity of early intervention for primary prevention. Epidemiological studies report that breastfeeding could protect from these diseases. However, data are controversial and the mechanisms unclear. Experimental data suggest that breastfeeding-induced protection might rely on tolerance induction as long as some criteria are fulfilled. Thus, the tolerogenic potential of breast milk would depend on maternal exposure to common environmental and dietary antigens and the efficiency of antigen transfer across mammary epithelium. Induction of tolerance upon breast milk-mediated antigen transfer will also depend on the presence of immunomodulatory factors in breast milk and of its impact on neonatal gut and immune system maturation. The better understanding of maternal influence on tolerance induction through breastfeeding should allow the development of new strategies to prevent immune-mediated diseases.
Collapse
|
39
|
Peroni DG, Pescollderungg L, Piacentini GL, Rigotti E, Maselli M, Watschinger K, Piazza M, Pigozzi R, Boner AL. Immune regulatory cytokines in the milk of lactating women from farming and urban environments. Pediatr Allergy Immunol 2010; 21:977-82. [PMID: 20718928 DOI: 10.1111/j.1399-3038.2010.00995.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Children living on farms have fewer allergies. It is unclear whether breastfeeding in different environments contributes to preventing allergies by exposing offspring to different cytokines that can modulate immune responses. The aim of this study was to quantify and compare levels of Transforming Growth Factor-beta1 (TGF-beta1) and Interleukin-10 (IL-10) in the colostrum and mature milk of mothers living in towns at sea level (references) and mothers on farms. Milk samples were collected within 3 days postpartum (colostrum) and at the first month of the baby's life (mature milk). Sixty-nine reference mothers and 45 farm mothers participated in the study. TGF-beta1 concentrations were significantly higher both in the colostrum (p < 0.05) and in mature milk (p < 0.05) of farm mothers. In the reference mothers, a significant decrease in TGF-beta1 concentrations was observed between colostrum (650, range 0-8000 pg/ml) and mature milk (250, range 0-8000 pg/ml) (p < 0.05). In farm mothers, TGF-beta1 concentrations were 1102 pg/ml (range 0-14,500) in colostrum and remained high in mature milk (821 pg/ml, range 0-14,650). IL-10 concentrations were higher in the mature milk of farm mothers (p < 0.05). No significant differences in IL-10 were observed between colostrum and mature milk in the control group (15 pg/ml, range 0-1800, and 0 pg/ml, range 0-230) or in farm mothers (9.5 pg/ml, range 0-1775, and 14.2 pg/ml, range 0-930), respectively. Exposure to a farm environment is associated with higher concentrations of TGF-beta1 and IL-10 in breast milk when compared to exposure to an urban environment. Higher cytokine concentrations in breast milk may influence early modulation of the development of an immune response, leading to a reduced prevalence of allergy-related diseases in farm children.
Collapse
Affiliation(s)
- Diego G Peroni
- Paediatric Department, University of Verona, Verona, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Verhasselt V. Oral tolerance in neonates: from basics to potential prevention of allergic disease. Mucosal Immunol 2010; 3:326-33. [PMID: 20485330 DOI: 10.1038/mi.2010.25] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Oral tolerance refers to the observation that prior feeding of an antigen induces local and systemic immune tolerance to that antigen. Physiologically, this process is probably of central importance for preventing inflammatory responses to the numerous dietary and microbial antigens present in the gut. Defective oral tolerance can lead to gut inflammatory disease, food allergies, and celiac disease. In the last two cases, the diseases develop early in life, stressing the necessity of understanding how oral tolerance is set up in neonates. This article reviews the parameters that have been outlined in adult animal models as necessary for tolerance induction and assesses whether these factors operate in neonates. In addition, we highlight the factors that are specific for this period of life and discuss how they could have an impact on oral tolerance. We pay particular attention to maternal influence on early oral tolerance induction through breast-feeding and outline the major parameters that could be modified to optimize tolerance induction in early life and possibly prevent allergic diseases.
Collapse
|
41
|
Emami CN, Petrosyan M, Giuliani S, Williams M, Hunter C, Prasadarao NV, Ford HR. Role of the host defense system and intestinal microbial flora in the pathogenesis of necrotizing enterocolitis. Surg Infect (Larchmt) 2010; 10:407-17. [PMID: 19943775 DOI: 10.1089/sur.2009.054] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a devastating disease that affects primarily the intestine of premature infants. Despite recent advances in neonatology, NEC remains a major cause of morbidity and mortality in neonates. Neonatal mucosal defenses and adherence of bacterial pathogens may play an important role in the pathogenesis of NEC. METHODS Review and synthesis of pertinent literature. RESULTS Putative factors that have been implicated in the pathogenesis of NEC include abnormal patterns of gut colonization by bacteria, immaturity of the host immune system and mucosal defense mechanisms, intestinal ischemia, formula feeding, and loss of intestinal epithelial barrier integrity. CONCLUSION Host defenses and intestinal microbial ecology are believed to play important roles in the pathogenesis of NEC. Commensal bacteria and probiotic therapy may be of therapeutic utility in the maintenance of the gut epithelial barrier.
Collapse
Affiliation(s)
- Claudia N Emami
- Department of Surgery, Childrens Hospital Los Angeles, Keck School of Medicine of University of Southern California, Los Angeles, California 90027, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Verhasselt V. Neonatal tolerance under breastfeeding influence: the presence of allergen and transforming growth factor-beta in breast milk protects the progeny from allergic asthma. J Pediatr 2010; 156:S16-20. [PMID: 20105659 DOI: 10.1016/j.jpeds.2009.11.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Once the umbilical cord has been cut, immunologists have often looked at the neonate as an entity that develops on its own. For years, breast milk was considered mainly as a source of nutrients for the developing child. The extensive observations that breastfeeding affords protection toward infectious diseases and could reduce by more than the half the mortality rate because of common infections have added another key role to breastfeeding. This protection relies in great part on the passive transfer through breast milk of high amounts of microbe-specific immunoglobulins that compensate for the deficiency of immunoglobulins synthesis during the first year of life. Here, we will present and discuss our data showing how breast milk can actively shape the immune response of the progeny, particularly in the context of allergic disease. Indeed, our data obtained in a mouse model suggest that the protection attributed to breastfeeding toward asthma development might rely on immune tolerance induction. For this to occur, the mother mice needed to be exposed to the allergen by aerosol or oral route during the lactation period, which resulted into the transfer of the allergen to breast milk. The presence of the allergen together with transforming growth factor-beta in breast milk was necessary and sufficient to induce the development of regulatory T lymphocytes in the progeny and their protection from asthma development. If confirmed in human beings, this study may suggest new strategies for asthma prevention such as deliberate exposure of mother to allergens during breastfeeding and qualitative modification of artificial milks.
Collapse
|
43
|
Abstract
Breast milk cytokines have the potential to regulate the immune response to food antigens in infants. Cytokines are present in all mammalian milks and are capable of inhibiting excess inflammation and modulating epithelial proliferation. There are a range of candidate cytokines in milk such as transforming growth factor-beta (TGF-beta), the major cytokine present, and interleukin-10, which play a role in immune regulation in the developing infant. This article will be a review of the current literature with regard to TGF-beta in infant immune development. Our data on supplementation of formula with rTGF-beta2 will be discussed in view of the current literature. Oral antigen exposure also plays an important role in priming the developing immune response. The influence of early introduction of oral beta-lactoglobulin in allergy prone rat pups will also be discussed.
Collapse
Affiliation(s)
- Irmeli A Penttila
- Women's and Children's Health Research Institute, Women's and Children's Hospital, North Adelaide, South Australia.
| |
Collapse
|
44
|
Oddy WH, Rosales F. A systematic review of the importance of milk TGF-beta on immunological outcomes in the infant and young child. Pediatr Allergy Immunol 2010; 21:47-59. [PMID: 19594862 DOI: 10.1111/j.1399-3038.2009.00913.x] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cytokines in milk like transforming growth factor-beta (TGF-beta) have been shown to induce oral tolerance in experimental animal studies. However, human studies are less consistent with these findings. The primary objective of this review was to conduct a systematic review of published studies on the association between TGF-beta identified in human milk and immunological outcomes in infancy and early childhood. Human prospective clinical studies were identified through MEDLINE, CAB Abstracts, Biological Abstracts and Scopus. Selection criteria included: well described populations of mothers and infants, time of milk sampling, immunological outcome measures and analytical methods of TGF-beta determination. We considered a wide range of immunological outcomes in infancy and early childhood, such as wheeze, atopy, eczema and the immunoglobulin switch. Twelve human studies were included in the review and 67% showed a positive association with TGF-beta1 or TGF-beta2 demonstrating protection against allergy-related outcomes in infancy and early childhood. High variability in concentrations of TGF-beta was noted between and within studies, some of it explained by maternal history of atopy or by consumption of probiotics. Human milk TGF-beta appears to be essential in developing and maintaining appropriate immune responses in infants and may provide protection against adverse immunological outcomes, corroborating findings from experimental animal studies. Further large clinical studies in diverse human populations are indicated to confirm these results.
Collapse
Affiliation(s)
- Wendy H Oddy
- Telethon Institute for Child Health Research, University of Western Australia, Perth, WA, Australia.
| | | |
Collapse
|
45
|
Nakao A. The role and potential use of oral transforming growth factor-beta in the prevention of infant allergy. Clin Exp Allergy 2010; 40:725-30. [PMID: 20067481 DOI: 10.1111/j.1365-2222.2009.03445.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The increasing prevalence of allergic diseases in infants and children as well as adults has become an important issue in public health in industrial countries. However, few preventive measures are available to reduce the risk of allergic diseases in infants; e.g. the avoidance of smoking and alcohol consumption during pregnancy and lactation. Therefore, there is an enthusiasm to identify certain factors in foods, nutrients, and environment responsible for the primary prevention of allergic diseases during infancy. In the last decade, TGF-beta in maternal milk has been implicated in the prevention of allergic diseases in infants and young children. This review summarizes the relevant epidemiological reports and highlights the recent animal studies to support the preventive role of orally administered TGF-beta, such as TGF-beta in human milk, in the development of allergic diseases in infants. We also provide suggestions for the potential use of dietary (oral) TGF-beta for the primary prevention of allergic diseases. Further studies to address the scientific validity and mechanistic insight to this Mother Nature-inspired concept are clearly required and will be important to develop new approaches to prevent allergic diseases.
Collapse
Affiliation(s)
- A Nakao
- Department of Immunology, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Yamanashi, Japan.
| |
Collapse
|
46
|
Khurana S, Mills JC. The gastric mucosa development and differentiation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 96:93-115. [PMID: 21075341 DOI: 10.1016/b978-0-12-381280-3.00004-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The development and differentiation of the gastric mucosa are controlled by a complex interplay of signaling proteins and transcriptional regulators. This process is complicated by the fact that the stomach is derived from two germ layers, the endoderm and the mesoderm, with the first giving rise to the mature epithelium and the latter contributing the smooth muscle required for peristalsis. Reciprocal epithelial-mesenchymal interactions dictate the formation of the stomach during fetal development, and also contribute to its continuous regeneration and differentiation throughout adult life. In this chapter, we discuss the discoveries that have been made in different model systems, from zebrafish to human, which show that the Hedgehog, Wnt, Notch, bone morphogenetic protein, and fibroblast growth factor (FGF) signaling systems play essential roles during various stages of stomach development.
Collapse
Affiliation(s)
- Shradha Khurana
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
47
|
The effects of formula feeding on physiological and immunological parameters in the gut of neonatal rats. Dig Dis Sci 2009; 54:1432-9. [PMID: 18975079 DOI: 10.1007/s10620-008-0513-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Accepted: 08/22/2008] [Indexed: 12/14/2022]
Abstract
A unique model of formula feeding in the neonatal rat was utilized to investigate the effects of an enterally delivered artificial milk formula on clinically relevant immunological and biological characteristics in the gut, compared to naturally reared pups. Hooded Wistar rat pups were randomly allocated to two treatment groups: formula-fed (FF) or naturally suckled (NS). A flexible silastic intra-gastric cannula was surgically implanted into the FF pups, through which an artificial rat milk supplement was continuously delivered from day 4 to day 10 of life. Rat pups were sacrificed at 10 days of age. Body weight, small intestinal weight, mucosal CD8(+) cell numbers, and ileal lactase activity in FF animals were significantly decreased compared to their NS counterparts (P < 0.05). Numbers of eosinophils, mucosal mast cells, CD4(+) T-cells, ileal villus height and gastric emptying times were significantly increased in FF pups (P < 0.05). We have developed a new rat model of artificial feeding which possesses important immunological and biological similarities to the premature human infant.
Collapse
|
48
|
Ogias D, de Andrade Sá ER, Alvares EP, Gama P. Opposite effects of fasting on TGF-beta3 and TbetaRI distribution in the gastric mucosa of suckling and early weanling rats. Nutrition 2009; 26:224-9. [PMID: 19524404 DOI: 10.1016/j.nut.2009.03.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Revised: 03/09/2009] [Accepted: 03/27/2009] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Our aim was to evaluate the effects of a dietary regimen (suckling or early weaning) and feeding status (fed or fasted) on the distribution of transforming growth factor-beta3 (TGF-beta3) and TGF receptor-I (TbetaRI) in the gastric epithelium of pups. METHODS Wistar rats were used. At 15 d, half of the pups were separated from dams and fed with hydrated powered chow. On day 17, suckling and early weanling rats were subjected to fasting (17h). Four different conditions were established: suckling fed and fasted and early weanling fed and fasted. At 18 d stomachs were collected under anesthesia and were fixed in 4% formaldehyde for immunohistochemistry. The number of immunostained epithelial cells per microscopic field was determined for TGF-beta3 and TbetaRI in longitudinal sections from the gastric mucosa. RESULTS We found that during suckling, fasting reduced the number of immunolabeled cells per field of both molecules when compared with the fed group (P<0.05), whereas in early weaning, food restriction increased TGF-beta3 and TbetaRI distributions (P<0.05). We also observed that TGF-beta3 and TbetaRI were more concentrated in parietal cells in the upper gland in suckling pups, whereas after early weaning these were displaced to parietal and chief cells at the bottom of the gland. CONCLUSION Suckling and early weaning directly influence TGF-beta3 and TbetaRI distributions in the gastric epithelium in response to fasting, such that early weaning anticipates the effects observed in adult rats. Furthermore, the differential concentrations of TGF-beta3 and TbetaRI indicate that they might be important for cell proliferation events in growth control.
Collapse
Affiliation(s)
- Daniela Ogias
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
49
|
Sangild PT, Mei J, Fowden AL, Xu RJ. The prenatal porcine intestine has low transforming growth factor-beta ligand and receptor density and shows reduced trophic response to enteral diets. Am J Physiol Regul Integr Comp Physiol 2009; 296:R1053-62. [DOI: 10.1152/ajpregu.90790.2008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Transforming growth factor-beta (TGF-β) plays a role in enterocyte proliferation control, cell differentiation, and immune regulation via binding to specific TGF-β receptors (TGF-β R) in the intestinal epithelium. Endogenous TGF-β production is low in the intestine during the perinatal period, but some exogenous TGF-β ligands are supplied by amniotic fluid intake in the fetus and by colostrum ingestion in the neonate. It is not clear, however, whether luminal TGF-β receptors are present and functional at this critical time. We studied intestinal TGF-β receptors by immunohistochemistry during the last 20% of gestation in pigs and in chronically catheterized fetuses following exposure to colostrum, milk, and amniotic fluid (control). In fetal pigs, the TGF-β Rs were predominantly localized to the crypt epithelium, but staining intensity increased markedly just before term and shifted to the villous epithelium in newborn pigs, concurrently with marked increases in villous heights and crypt depths (+100–200%, P < 0.05). In contrast to previous observations in term newborn pigs, fetal pigs did not show any milk-induced change in TGF-β receptor densities or localization, although a moderate increase in villous height was observed, relative to control (+25–50%, P < 0.05). We conclude that intestinal TGF-β receptor density and localization are immature and unresponsive to TGF-β containing milk diets in prenatal pigs. Immaturity of TGF-β-mediated immune regulation may play a role in the increased sensitivity of preterm neonates to diet-induced intestinal inflammatory disorders.
Collapse
|
50
|
Rautava S, Walker WA. Academy of Breastfeeding Medicine founder's lecture 2008: breastfeeding--an extrauterine link between mother and child. Breastfeed Med 2009; 4:3-10. [PMID: 19292608 PMCID: PMC2932545 DOI: 10.1089/bfm.2009.0004] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In addition to a near-optimal combination of nutrients for the growing infant, breastmilk contains a wide array of bioactive molecules that are known to protect the infant against infectious disease and modulate the composition of the indigenous intestinal microbiota. A growing number of factors that modulate the infant's immunophysiology have also been identified in breastmilk. We suggest that this early immunomodulation via breastmilk is vital for infant health and may explain the epidemiological data indicating that breastmilk reduces the risk of immunoinflammatory conditions in infancy and also later in life. The body of scientific data regarding the role of transforming growth factor-beta in breastmilk in enhancing healthy immune maturation and reducing the risk of disease is reviewed in this article.
Collapse
Affiliation(s)
- Samuli Rautava
- Developmental Gastroenterology Laboratory, Division of Pediatric Gastroenterology, Massachusetts General Hospital for Children, Charlestown, Massachusetts, USA.
| | | |
Collapse
|