1
|
Abstract
Human breast milk is the optimal nutrition for all infants and is comprised of many bioactive and immunomodulatory components. The components in human milk, such as probiotics, human milk oligosaccharides (HMOs), extracellular vesicles, peptides, immunoglobulins, growth factors, cytokines, and vitamins, play a critical role in guiding neonatal development beyond somatic growth. In this review, we will describe the bioactive factors in human milk and discuss how these factors shape neonatal immunity, the intestinal microbiome, intestinal development, and more from the inside out.
Collapse
Affiliation(s)
- Sarah F Andres
- Department of Pediatrics, Pediatric GI Division, School of Medicine, Oregon Health and Science University, Portland, OR 97229, United States
| | - Brian Scottoline
- Division of Neonatology, Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, United States
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, 101 Manning Drive, Campus Box 7596, Chapel Hill, NC 27599, United States.
| |
Collapse
|
2
|
Almonaem ERA, Almotaleb GSA, Alhameed MHA, El-Shimi OS. Utility of transforming growth factor beta-1 in diagnosis of neonatal necrotizing enterocolitis. J Neonatal Perinatal Med 2022; 15:795-801. [PMID: 35811541 DOI: 10.3233/npm-210973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Necrotizing enterocolitis (NEC) yet remains a leading cause of morbidity and mortality in premature infants. The developmental deficiency of transforming growth factor-Beta (TGF-β) in the intestine is a risk factor for NEC in premature infants.We aimed to investigate the potential utility of serum TGF-β1 in the early diagnosis and severity assessment of NEC. This prospective case-control study was conducted on 102 VLBW neonates aging less than 32 weeks and weighing less than 1500 gm. They were divided into NEC group of 52 preterm neonates with symptoms and signs of NEC and 50 age and sex-matched neonates without NEC as a control group. All neonates underwent full medical history taking, clinical examination, radiological and laboratory investigations including CBC, CRP, fecal occult blood, and serum TGF-β1. Serum TGF-β1 was tested in NEC patients at the onset of symptoms and signs and 7 days later. Serum TGF-β1 was significantly lower in NEC patients at the onset of symptoms than the control group (P = 0.004) while after 7 days of onset serum TGF-β1 was significantly higher than at the onset of symptoms (P < 0.001). In NEC patients with stage I, TGF-β1 was significantly higher than in NEC patients with stage ≥II (P = 0.027).In conclusion serum TGF-β1 is downregulated in neonatal necrotizing enterocolitis and can be used as a useful biomarker for early diagnosis of NEC and to assess disease severity.
Collapse
Affiliation(s)
| | | | | | - Ola Samir El-Shimi
- Clinical Pathology department, Faculty of Medicine, Benha University, Egypt
| |
Collapse
|
3
|
Maheshwari A, Traub TM, Garg PM, Ethawi Y, Buonocore G. Necrotizing Enterocolitis: Clinical Features, Histopathological Characteristics, and Genetic Associations. Curr Pediatr Rev 2022; 18:210-225. [PMID: 35125082 DOI: 10.2174/1573396318666220204113858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 11/22/2022]
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis seen in premature infants. Although the etiopathogenesis of NEC is unclear, genetic factors may alter a patient's susceptibility, clinical course, and outcomes. This review draws from existing studies focused on individual genes and others based on microarray-based high-throughput discovery techniques. We have included evidence from our own studies and from an extensive literature search in the databases PubMed, EMBASE, and Scopus. To avoid bias in the identification of studies, keywords were short-listed a priori from anecdotal experience and PubMed's Medical Subject Heading (MeSH) thesaurus.
Collapse
Affiliation(s)
| | - Terri M Traub
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Parvesh M Garg
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics, University of Mississippi, Jackson, Mississippi, USA
| | - Yahya Ethawi
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics, Saudi American Hospital, Ajman, United Arab Emirates
| | - Giuseppe Buonocore
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics/ Neonatology, University of Siena, Siena, Italy
| |
Collapse
|
4
|
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory disease affecting premature infants. Intestinal microbial composition may play a key role in determining which infants are predisposed to NEC and when infants are at highest risk of developing NEC. It is unclear how to optimize antibiotic therapy in preterm infants to prevent NEC and how to optimize antibiotic regimens to treat neonates with NEC. This article discusses risk factors for NEC, how dysbiosis in preterm infants plays a role in the pathogenesis of NEC, and how probiotic and antibiotic therapy may be used to prevent and/or treat NEC and its sequelae.
Collapse
Affiliation(s)
- Jennifer Duchon
- Division of Newborn Medicine, Jack and Lucy Department of Pediatrics, Icahn School of Medicine at Mount Sinai, 1000 10th Avenue, New York, NY 10019, USA
| | - Maria E Barbian
- Division of Neonatal-Perinatal Medicine, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive Northeast, 3rd Floor, Atlanta, GA 30322, USA
| | - Patricia W Denning
- Division of Neonatal-Perinatal Medicine, Emory University School of Medicine, Children's Healthcare of Atlanta, Emory University Hospital Midtown, 550 Peachtree Street, 3rd Floor MOT, Atlanta, GA 30308, USA.
| |
Collapse
|
5
|
Demers-Mathieu V, Huston RK, Dallas DC. Cytokine Expression by Human Macrophage-Like Cells Derived from the Monocytic Cell Line THP-1 Differs Between Treatment With Milk from Preterm- and Term-Delivering Mothers and Pasteurized Donor Milk. Molecules 2020; 25:molecules25102376. [PMID: 32443898 PMCID: PMC7287623 DOI: 10.3390/molecules25102376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 11/25/2022] Open
Abstract
Immunomodulatory proteins from human milk may enhance the protection and development of the infant’s gut. This study compared the immunomodulatory effects of treatment with milk from preterm-(PM) and term-delivering (TM) mothers and pasteurized donor milk (DM) on cytokine gene expression in human macrophage-like cells derived from the monocytic cell line THP-1. The gene expression of tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-12 (p40), IL-10 and GAPDH in macrophages treated with PM, TM and DM at steady and activated (inflammatory) states were measured using real-time reverse transcription-polymerase chain reaction. TNF-α and IL-6 in macrophages (both states) with DM were higher than PM or TM. IL-10 in steady state macrophages with DM was higher than PM whereas DM increased IL-10 in activated macrophages compared with TM. TM increased IL-6 and IL-12 (p40) in steady state macrophages compared with PM. IL-12 (p40) in activated macrophages with TM was higher than PM. IL-10 in steady state macrophages with TM was higher than PM. These results suggest that DM induces higher gene expression of pro-inflammatory and anti-inflammatory cytokines in macrophages compared with PM or TM. PM reduced gene expression of pro-inflammatory cytokines compared with TM, which may decrease the development of necrotizing enterocolitis and systematic inflammation.
Collapse
Affiliation(s)
- Veronique Demers-Mathieu
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA
- Correspondence: ; Tel.: +1-541-286-8366
| | - Robert K. Huston
- Department of Pediatrics, Randall Children’s Hospital at Legacy Emanuel, Portland, OR 97227, USA; (R.K.H.); (D.C.D.)
| | - David C. Dallas
- Department of Pediatrics, Randall Children’s Hospital at Legacy Emanuel, Portland, OR 97227, USA; (R.K.H.); (D.C.D.)
| |
Collapse
|
6
|
Does Caesarean Section or Preterm Delivery Influence TGF-β2 Concentrations in Human Colostrum? Nutrients 2020; 12:nu12041095. [PMID: 32326558 PMCID: PMC7230194 DOI: 10.3390/nu12041095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 12/21/2022] Open
Abstract
Human colostrum (HC) is a rich source of immune mediators that play a role in immune defences of a newly born infant. The mediators include transforming growth factor β (TGF-β) which exists in three isoforms that regulate cellular homeostasis and inflammation, can induce or suppress immune responses, limit T helper 1 cells (Th1) reactions and stimulate secretory immunoglobulin A (IgA) production. Human milk TGF-β also decreases apoptosis of intestinal cells and suppresses macrophage cytokine expression. The aim of the study was to determine the concentration of TGF-β2 in HC obtained from the mothers who delivered vaginally (VD) or by caesarean section (CS), and to compare the concentrations in HC from mothers who delivered at term (TB) or preterm (PB). In this study, 56% of preterm pregnancies were delivered via CS. The concentrations of TGF-β2 were measured in HC from 299 women who delivered in the 1st Department of Obstetrics and Gynaecology, Medical University of Warsaw: 192 (VD), 107 (CS), 251 (TB), and 48 (PB). The colostrum samples were collected within 5 days post-partum. TGF-β2 levels in HC were measured by the enzyme-linked immunosorbent assay (ELISA) test with the Quantikine ELISA Kit-Human TGF-β2 (cat.no. SB250). Statistical significance between groups was calculated by the Student t-test using StatSoft Statistica 13 software. The mean TGF-β2 concentration in patients who delivered at term or preterm were comparable. The levels of TGF-β2 in HC were higher after preterm than term being 4648 vs. 3899 ng/mL (p = 0.1244). The delivery via CS was associated with higher HC concentrations of TGF-β2. The levels of TGF-β2 were significantly higher in HC after CS than VD (7429 vs. 5240 ng/mL; p = 0.0017). The data from this study suggest: caesarean section was associated with increased levels of TGF-β2 in HC. The increased levels of TGF-β2 in HC of women who delivered prematurely require further research. Early and exclusive breast-feeding by mothers after caesarean section and premature births with colostrum containing high TGF-β2 levels may prevent the negative impact of pathogens which often colonize the gastrointestinal tract and may reduce the risk of chronic diseases in this group of patients.
Collapse
|
7
|
Panahipour L, Biasi MD, Bokor TS, Thajer A, Haiden N, Gruber R. Milk lactoperoxidase decreases ID1 and ID3 expression in human oral squamous cell carcinoma cell lines. Sci Rep 2020; 10:5836. [PMID: 32246075 PMCID: PMC7125221 DOI: 10.1038/s41598-020-62390-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 03/12/2020] [Indexed: 01/07/2023] Open
Abstract
Milk consumption may modify the risk of squamous cell carcinoma. The role of milk to modulate the gene expression in oral squamous cell carcinoma cells has not been investigated so far. Here, HSC2 oral squamous carcinoma cells were exposed to an aqueous fraction of human milk and a whole-genome array was performed. Among the genes that were significantly reduced by human and cow milk were the DNA-binding protein inhibitor 1 (ID1), ID3 and Distal-Less Homeobox 2 (DLX2) in HSC2 cells. Also, in TR146 oral squamous carcinoma cells, there was a tendency towards a decreased gene expression. Upon size fractionation, lactoperoxidase but not lactoferrin and osteopontin was identified to reduce ID1 and ID3 in HSC2 cells. Dairy products and hypoallergenic infant formula failed to decrease the respective genes. These data suggest that milk can reduce the expression of transcription factors in oral squamous carcinoma cells.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
| | - Maria De Biasi
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
| | - Theresa Sophia Bokor
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
| | - Alexandra Thajer
- Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Nadja Haiden
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria. .,Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010, Bern, Switzerland. .,Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1200, Vienna, Austria.
| |
Collapse
|
8
|
Prolyl hydroxylase 2 silencing enhances the paracrine effects of mesenchymal stem cells on necrotizing enterocolitis in an NF-κB-dependent mechanism. Cell Death Dis 2020; 11:188. [PMID: 32179740 PMCID: PMC7075868 DOI: 10.1038/s41419-020-2378-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Treatment options for necrotizing enterocolitis (NEC) remain inadequate. Here we examined if and how prolyl hydroxylase 2 (PHD2) silencing enhances the paracrine effects of bone-marrow-derived mesenchymal stem cells (BM-MSCs) on NEC. In this study, BM-MSCs were transduced with lentiviruses containing GFP (GFP-MSC) or shPHD2-GFP constructs (PHDMSC), followed by intraperitoneal injection of the PHDMSC-conditioned medium (PHDMSC-CM) or the GFP-MSC-conditioned medium (MSC-CM) into a rat pup model of NEC. Our results showed that systemic infusion of PHDMSC-CM, but not MSC-CM, significantly improved intestinal damage and survival of NEC rats. Such benefits may involve the modulation of epithelial regeneration and inflammation, as indicated by the regeneration of intestinal epithelial/stem cells, the regulation of Treg cells function and pro-/anti-inflammatory cytokine balance. The mechanism for the superior paracrine efficacy of PHDMSC is related to a higher release of pivotal factor IGF-1 and TGF-β2. NF-κB activation was induced by PHD2 silencing to induce IGF-1 and TGF-β2 secretion via binding to IGF-1 and TGF-β2 gene promoter. Our work indicated that PHD2 silencing enhanced the paracrine effect of BM-MSCs on NEC via the NF-κB-dependent mechanism which may be a novel strategy for stem cell therapy on NEC.
Collapse
|
9
|
Panahipour L, Kochergina E, Kreissl A, Haiden N, Gruber R. Milk modulates macrophage polarization in vitro. Cytokine X 2019; 1:100009. [PMID: 33604549 PMCID: PMC7885867 DOI: 10.1016/j.cytox.2019.100009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/17/2019] [Accepted: 05/17/2019] [Indexed: 02/01/2023] Open
Abstract
Objective Milk holds an anti-inflammatory response that is particularly important to protecting infants against necrotizing enterocolitis. Milk might also exert anti-inflammatory effects in adulthood, including the oral cavity where macrophages of the oral mucosal control innate immunity defense. It remains unknown, however, whether milk can modulate the local inflammatory response by affecting the polarization of macrophages. Material and Methods To determine whether pasteurized human milk and pasteurized cow milk can provoke macrophage polarization, murine bone marrow macrophages and RAW264.7 cells were exposed to human saliva or the inflammatory cytokines IL1β and TNFα. Activation of pro-(M1) inflammatory response is indicated by the expression of IL1 and IL8. To determine polarization towards a M2 phenotype, the expression of arginase 1 (ARG1) and chitinase-like 3 (Chil3) was determined by reverse transcriptase PCR and immunoassay. Western blot was done on phosphorylated p38 and JNK. Results Aqueous fractions of human milk and cow milk from different donors, respectively, significantly decreased the inflammatory response of primary macrophages and RAW264.7 cells when exposed to saliva or IL1 and TNFα. Similar to IL4, human milk and cow milk caused a robust expression of ARG1 and Chil3 in primary macrophages. The polarization of macrophages by pasteurized milk occurred independent of the phosphorylation of p38 and JNK. Conclusion These data suggest that pasteurized milk, independent of the origin, can cause the polarization of macrophages from a pro-inflammatory M1 towards a pro-resolving M2 phenotype. Thus, milk might have a protective role for the oral cavity by modulation of the macrophage-based innate immune system.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria
| | - Evgeniya Kochergina
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria
| | - Alexandra Kreissl
- Department of Paediatrics and Adolescent Medicine, Division of Neonatology, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria
| | - Nadja Haiden
- Department of Clinical Pharmacology, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria.,Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland.,Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1200 Vienna, Austria
| |
Collapse
|
10
|
Bazacliu C, Neu J. Pathophysiology of Necrotizing Enterocolitis: An Update. Curr Pediatr Rev 2019; 15:68-87. [PMID: 30387398 DOI: 10.2174/1573396314666181102123030] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/12/2018] [Accepted: 09/15/2018] [Indexed: 12/12/2022]
Abstract
NEC is a devastating disease that, once present, is very difficult to treat. In the absence of an etiologic treatment, preventive measures are required. Advances in decoding the pathophysiology of NEC are being made but a more comprehensive understanding is needed for the targeting of preventative strategies. A better definition of the disease as well as diagnostic criteria are needed to be able to specifically label a disease as NEC. Multiple environmental factors combined with host susceptibility appear to contribute to enhanced risks for developing this disease. Several different proximal pathways are involved, all leading to a common undesired outcome: Intestinal necrosis. The most common form of this disease appears to involve inflammatory pathways that are closely meshed with the intestinal microbiota, where a dysbiosis may result in dysregulated inflammation. The organisms present in the intestinal tract prior to the onset of NEC along with their diversity and functional capabilities are just beginning to be understood. Fulfillment of postulates that support causality for particular microorganisms is needed if bacteriotherapies are to be intelligently applied for the prevention of NEC. Identification of molecular effector pathways that propagate inflammation, understanding of, even incipient role of genetic predisposition and of miRNAs may help solve the puzzle of this disease and may bring the researchers closer to finding a treatment. Despite recent progress, multiple limitations of the current animal models, difficulties related to studies in humans, along with the lack of a "clear" definition will continue to make it a very challenging disease to decipher.
Collapse
Affiliation(s)
- Catalina Bazacliu
- Department of Pediatrics, Division of Neonatology, University of Florida, FL, United States
| | - Josef Neu
- Department of Pediatrics, Division of Neonatology, University of Florida, FL, United States
| |
Collapse
|
11
|
Panahipour L, Nasserzare S, Amer Z, Brücke F, Stähli A, Kreissl A, Haiden N, Gruber R. The anti-inflammatory effect of milk and dairy products on periodontal cells: an in vitro approach. Clin Oral Investig 2018; 23:1959-1966. [DOI: 10.1007/s00784-018-2642-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023]
|
12
|
Panahipour L, Stähli A, Haiden N, Gruber R. TGF-β activity in cow milk and fermented milk products: An in vitro bioassay with oral fibroblasts. Arch Oral Biol 2018; 95:15-21. [PMID: 30036732 DOI: 10.1016/j.archoralbio.2018.07.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 06/19/2018] [Accepted: 07/08/2018] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Milk is a rich source of transforming growth factor (TGF)-β which supports intestinal mucosal homeostasis of infants. Milk may also have beneficial effects on the integrity of the oral cavity, its being part of the gastrointestinal tract. However, it is unclear if milk and fermented milk products provoke a TGF-β response in oral cells. MATERIAL AND METHODS Human gingival fibroblasts were exposed to pasteurized cow milk, yoghurt, sour milk, buttermilk and whey, followed by a reverse transcriptase polymerase chain reaction of the TGF-β target genes interleukin11 (IL11), proteoglycan4 (PRG4), and NADPH oxidase 4 (NOX4). Immunoassays were performed for IL11 and TGF-β in cell culture supernatant and milk products, respectively. Signaling was investigated with the TGF-β receptor type I kinase inhibitor SB431542. RESULTS We report here that pasteurized cow milk and the aqueous fractions of yoghurt, sour milk, buttermilk and whey enhanced the expression of IL11, NOX4 and PRG4 in gingival fibroblasts. Moreover, IL11 protein levels in the respective supernatant were significantly increased. Cow milk, yoghurt, sour milk and buttermilk contain approximately 1-2 ng TGF-β1, whereas active TGF-β1 is hardly detectable in whey. SB431542 reduced the response of gingival fibroblasts to pasteurized cow milk and fermented milk products based on IL11 release into the supernatant. CONCLUSIONS These results demonstrate that gingival fibroblasts respond to pasteurized cow milk and to fermented milk products with an increased expression of TGF-β target genes.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Austria
| | - Alexandra Stähli
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Austria; Department of Periodontology, School of Dental Medicine, University of Bern, Switzerland
| | - Nadja Haiden
- Department of Paediatrics and Adolescent Medicine, Division of Neonatology, Medical University of Vienna, Vienna, Austria
| | - Reinhard Gruber
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Austria; Department of Periodontology, School of Dental Medicine, University of Bern, Switzerland.
| |
Collapse
|
13
|
Koyama N, Yamazaki T, Kanetsuki Y, Hirota J, Asai T, Mitsumoto Y, Mizuno M, Shima T, Kanbara Y, Arai S, Miyazaki T, Okanoue T. Activation of apoptosis inhibitor of macrophage is a sensitive diagnostic marker for NASH-associated hepatocellular carcinoma. J Gastroenterol 2018; 53:770-779. [PMID: 29086016 DOI: 10.1007/s00535-017-1398-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 09/30/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND A diagnostic marker is needed enabling early and specific diagnosis of hepatocellular carcinoma (HCC) associated with non-alcoholic steatohepatitis (NASH). Our recent findings have indicated that circulating apoptosis inhibitor of macrophage (AIM), which usually associates with IgM pentamer in the blood, is activated by its dissociation from IgM. We investigated the serum levels of IgM-free AIM for AIM activation and its possible relationship with development of HCC in NASH. METHODS Serum levels of IgM-associated and IgM-free AIM were evaluated in patients with non-alcoholic fatty liver, NASH, and NASH-HCC using enzyme-linked immunosorbent assays and immunoblots. Liver biopsy specimens were graded and staged using Brunt's classification. RESULTS Forty-two patients with fatty liver, 141 with NASH, and 26 with NASH-HCC were evaluated. Patients with stage 4 or grade 3 NASH (with or without HCC) exhibited significantly higher levels of both IgM-free and total AIM than those with fatty liver, whereas the ratio of IgM-free-to-total AIM was equivalent in these groups. Among patients with the same fibrosis stage of NASH, those with HCC had significantly higher IgM-free but not total AIM levels, resulting in a proportional increase in the IgM-free/total AIM ratio. Analysis of the areas under the receiver operating characteristic curves indicated the high sensitivity of the IgM-free AIM for NASH-HCC. CONCLUSIONS Our observations suggest the activation of AIM in blood in the presence of NASH-HCC, with a significant increase in IgM-free AIM levels. IgM-free AIM serum levels appear to be a sensitive diagnostic marker for NASH-HCC.
Collapse
Affiliation(s)
- Noriyuki Koyama
- Research and Development Division, Eidia Co Ltd, Tokyo, Japan
- Eisai Co Ltd, Tokyo, Japan
| | - Tomoko Yamazaki
- Laboratory of Molecular Biomedicine for Pathogenesis, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuka Kanetsuki
- Research and Development Division, Sekisui Medical Co Ltd, Tokyo, Japan
| | - Jiro Hirota
- Research and Development Division, Sekisui Medical Co Ltd, Tokyo, Japan
| | - Tomohide Asai
- Research and Development Division, Sekisui Medical Co Ltd, Tokyo, Japan
| | - Yasuhide Mitsumoto
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita City, Osaka, Japan
| | - Masayuki Mizuno
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita City, Osaka, Japan
| | - Toshihide Shima
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita City, Osaka, Japan
| | - Yoshihiro Kanbara
- Department of Digestive Surgery, Saiseikai Suita Hospital, Suita City, Osaka, Japan
| | - Satoko Arai
- Laboratory of Molecular Biomedicine for Pathogenesis, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toru Miyazaki
- Laboratory of Molecular Biomedicine for Pathogenesis, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
- Max Planck-The University of Tokyo Center for Integrative Inflammology, Tokyo, Japan
| | - Takeshi Okanoue
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita City, Osaka, Japan.
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease in premature infants with high case fatality and significant morbidity among survivors. Immaturity of intestinal host defenses predisposes the premature infant gut to injury. An abnormal bacterial colonization pattern with a deficiency of commensal bacteria may lead to a further breakdown of these host defense mechanisms, predisposing the infant to NEC. Here, we review the role of the innate and adaptive immune system in the pathophysiology of NEC.
Collapse
MESH Headings
- Adaptive Immunity
- Enterocolitis, Necrotizing/immunology
- Enterocolitis, Necrotizing/microbiology
- Enterocolitis, Necrotizing/physiopathology
- Evidence-Based Medicine
- Humans
- Immunity, Innate
- Infant, Premature
- Infant, Premature, Diseases/immunology
- Infant, Premature, Diseases/microbiology
- Infant, Premature, Diseases/physiopathology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/microbiology
- Intestinal Mucosa/physiopathology
- Intestines/blood supply
- Intestines/immunology
- Intestines/physiopathology
- Milk, Human/immunology
Collapse
Affiliation(s)
- Timothy L Denning
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA
| | - Amina M Bhatia
- Department of Surgery, Emory University School of Medicine, Atlanta, GA
| | - Andrea F Kane
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Ravi M Patel
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Patricia W Denning
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA.
| |
Collapse
|
17
|
MohanKumar K, Namachivayam K, Ho TT, Torres BA, Ohls RK, Maheshwari A. Cytokines and growth factors in the developing intestine and during necrotizing enterocolitis. Semin Perinatol 2017; 41:52-60. [PMID: 27832931 PMCID: PMC5334139 DOI: 10.1053/j.semperi.2016.09.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cytokines and growth factors play diverse roles in the uninflamed fetal/neonatal intestinal mucosa and in the development of inflammatory bowel injury during necrotizing enterocolitis (NEC). During gestational development and the early neonatal period, the fetal/premature intestine is exposed to high levels of many "inflammatory" cytokines and growth factors, first via swallowed amniotic fluid in utero and then, after birth, in colostrum and mother's milk. This article reviews the dual, seemingly counter-intuitive roles of cytokines, where these agents play a "trophic" role and promote maturation of the uninflamed mucosa, but can also cause inflammation and promote intestinal injury during NEC.
Collapse
Affiliation(s)
| | | | - Thao T.B. Ho
- Department of Pediatrics, Morsani College of Medicine, Tampa, FL
| | | | - Robin K. Ohls
- Department of Pediatrics, University of New Mexico, Albuquerque, NM
| | - Akhil Maheshwari
- Department of Pediatrics, Morsani College of Medicine, Tampa, FL; Departments of Molecular Medicine, Morsani College of Medicine, Tampa, FL; Department of Community and Family Health, College of Public Health, University of South Florida, 1 Tampa General Circle, Suite F170, Tampa, FL.
| |
Collapse
|
18
|
Simpson MR, Rø ADB, Grimstad Ø, Johnsen R, Storrø O, Øien T. Atopic dermatitis prevention in children following maternal probiotic supplementation does not appear to be mediated by breast milk TSLP or TGF-β. Clin Transl Allergy 2016; 6:27. [PMID: 27453775 PMCID: PMC4957279 DOI: 10.1186/s13601-016-0119-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/29/2016] [Indexed: 12/21/2022] Open
Abstract
Background The Probiotics in Prevention of Allergy among Children in Trondheim (ProPACT) study, a randomised, placebo controlled trial, demonstrated that maternal supplementation with probiotic milk reduced the incidence of atopic dermatitis (AD) in infancy. The mechanisms behind this effect are incompletely understood and breast milk cytokines have been postulated as possible mediating factors. In this study we aimed to assess whether breast milk TLSP and TGF-β are affected by a maternal probiotic supplementation regime, and their contribution to the preventive effect of this regime on AD in the offspring. Methods TSLP and TGF-β isoforms (TGF-β1, TGF-β2 and TGF-β3) were measured using ELISA and multiplex assays, respectively, in breast milk samples collected at 10 days and 3 months postpartum from women participating in the ProPACT trial (n = 259). The natural indirect and direct effects of maternal probiotics on AD, due to changes in breast milk cytokines, were estimated using causal mediation techniques. Results Probiotic supplementation tend to lead to high levels of breast milk TSLP at 10 days postpartum (p = 0.062), but this change did not contribute to the prevention of AD according to the mediation analysis. Probiotics had no apparent effect on TSLP at 3 months or TGF-βs at either time points. Thus, these are unlikely to be mediators of the effect of maternal probiotics on AD in offspring. Conclusions Whilst maternal probiotic supplementation resulted in higher breast milk concentrations of TLSP at 10 days postpartum, this does not appear to be a mechanism for prevention of AD by maternal probiotics. Trial registration The original trial protocol is registered in ClinicalTrials.gov (identifier NCT00159523)
Collapse
Affiliation(s)
- Melanie Rae Simpson
- Department of Public Health and General Practice, Norwegian University of Science and Technology, Trondheim, Norway ; Det medisinske fakultet, Instiutt for samfunnsmedisin, MTFS, NTNU, Postboks 8905, 7491 Trondheim, Norway
| | - Anne Dorthea Bjerkenes Rø
- Department of Public Health and General Practice, Norwegian University of Science and Technology, Trondheim, Norway ; Department of Immunology and Transfusion Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Øystein Grimstad
- Department of Dermatology, University Hospital of North Norway, Tromsö, Norway
| | - Roar Johnsen
- Department of Public Health and General Practice, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ola Storrø
- Department of Public Health and General Practice, Norwegian University of Science and Technology, Trondheim, Norway
| | - Torbjørn Øien
- Department of Public Health and General Practice, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
19
|
MohanKumar K, Namachivayam K, Chapalamadugu K, Garzon SA, Premkumar MH, Tipparaju S, Maheshwari A. Smad7 interrupts TGF-β signaling in intestinal macrophages and promotes inflammatory activation of these cells during necrotizing enterocolitis. Pediatr Res 2016; 79:951-61. [PMID: 26859364 PMCID: PMC4899224 DOI: 10.1038/pr.2016.18] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/18/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants. Based on our recent findings of increased Smad7 expression in surgically resected bowel affected by NEC, we hypothesized that NEC macrophages undergo inflammatory activation because increased Smad7 expression renders these cells resistant to normal, gut-specific, transforming growth factor (TGF)-β-mediated suppression of inflammatory pathways. METHODS We used surgically resected human NEC tissue, murine models of NEC-like injury, bone marrow-derived and intestinal macrophages, and RAW264.7 cells. Smad7 and IκB kinase-beta (IKK-β) were measured by quantitative PCR, western blots, and immunohistochemistry. Promoter activation was confirmed in luciferase reporter and chromatin immunoprecipitation assays. RESULTS NEC macrophages showed increased Smad7 expression, particularly in areas with severe tissue damage and high bacterial load. Lipopolysaccharide-induced Smad7 expression suppressed TGF-β signaling and augmented nuclear factor-kappa B (NF-κB) activation and cytokine production in macrophages. Smad7-mediated NF-κB activation was likely mediated via increased expression of IKK-β, which, further increased Smad7 expression in a feed-forward loop. We show that Smad7 induced IKK-β expression through direct binding to the IKK-β promoter and its transcriptional activation. CONCLUSION Smad7 expression in NEC macrophages interrupts TGF-β signaling and promotes NF-κB-mediated inflammatory signaling in these cells through increased expression of IKK-β.
Collapse
Affiliation(s)
- Krishnan MohanKumar
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, USA, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Kopperuncholan Namachivayam
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, USA, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Kalyan Chapalamadugu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida, USA
| | - Steven A. Garzon
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | - Srinivas Tipparaju
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida, USA
| | - Akhil Maheshwari
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, USA, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA, Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA, Department of Community and Family Health, College of Public Health, University of South Florida, Tampa, Florida, USA,Address for correspondence: Akhil Maheshwari, 1 Tampa General Circle, Suite F170, Tampa, FL 33606, USA; Phone: 813-844-3437; Fax: 813-844-1671;
| |
Collapse
|
20
|
Namachivayam K, Coffing HP, Sankaranarayanan NV, Jin Y, MohanKumar K, Frost BL, Blanco CL, Patel AL, Meier PP, Garzon SA, Desai UR, Maheshwari A. Transforming growth factor-β2 is sequestered in preterm human milk by chondroitin sulfate proteoglycans. Am J Physiol Gastrointest Liver Physiol 2015; 309:G171-80. [PMID: 26045614 PMCID: PMC4525106 DOI: 10.1152/ajpgi.00126.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/22/2015] [Indexed: 01/31/2023]
Abstract
Human milk contains biologically important amounts of transforming growth factor-β2 isoform (TGF-β2), which is presumed to protect against inflammatory gut mucosal injury in the neonate. In preclinical models, enterally administered TGF-β2 can protect against experimental necrotizing enterocolitis, an inflammatory bowel necrosis of premature infants. In this study, we investigated whether TGF-β bioactivity in human preterm milk could be enhanced for therapeutic purposes by adding recombinant TGF-β2 (rTGF-β2) to milk prior to feeding. Milk-borne TGF-β bioactivity was measured by established luciferase reporter assays. Molecular interactions of TGF-β2 were investigated by nondenaturing gel electrophoresis and immunoblots, computational molecular modeling, and affinity capillary electrophoresis. Addition of rTGF-β2 (20-40 nM) to human preterm milk samples failed to increase TGF-β bioactivity in milk. Milk-borne TGF-β2 was bound to chondroitin sulfate (CS) containing proteoglycan(s) such as biglycan, which are expressed in high concentrations in milk. Chondroitinase treatment of milk increased the bioactivity of both endogenous and rTGF-β2, and consequently, enhanced the ability of preterm milk to suppress LPS-induced NF-κB activation in macrophages. These findings provide a mechanism for the normally low bioavailability of milk-borne TGF-β2 and identify chondroitinase digestion of milk as a potential therapeutic strategy to enhance the anti-inflammatory effects of preterm milk.
Collapse
Affiliation(s)
- Kopperuncholan Namachivayam
- 1Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Department of Pediatrics, Morsani College of Medicine, University of South Florida Health, Tampa, Florida;
| | - Hayley P. Coffing
- 1Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,3Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois;
| | - Nehru Viji Sankaranarayanan
- 4Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia; ,5Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia;
| | - Yingzi Jin
- 4Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia; ,5Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia;
| | - Krishnan MohanKumar
- 1Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Department of Pediatrics, Morsani College of Medicine, University of South Florida Health, Tampa, Florida;
| | - Brandy L. Frost
- 6Department of Pediatrics, NorthShore University Health System, Evanston, Illinois;
| | - Cynthia L. Blanco
- 7Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas;
| | - Aloka L. Patel
- 8Department of Pediatrics, Rush University Medical Center, Chicago, Illinois; and
| | - Paula P. Meier
- 8Department of Pediatrics, Rush University Medical Center, Chicago, Illinois; and ,9Department of Women Children and Family Nursing, Rush University Medical Center, Chicago, Illinois
| | - Steven A. Garzon
- 10Department of Pathology, University of Illinois at Chicago, Chicago, Illinois;
| | - Umesh R. Desai
- 4Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia; ,5Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia;
| | - Akhil Maheshwari
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; Department of Pediatrics, Morsani College of Medicine, University of South Florida Health, Tampa, Florida; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida Health, Tampa, Florida;
| |
Collapse
|
21
|
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
|
22
|
Yang HP, Schneider SS, Chisholm CM, Browne EP, Mahmood S, Gierach GL, Lenington S, Anderton DL, Sherman ME, Arcaro KF. Association of TGF-β2 levels in breast milk with severity of breast biopsy diagnosis. Cancer Causes Control 2015; 26:345-54. [PMID: 25604865 DOI: 10.1007/s10552-014-0498-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 11/13/2014] [Indexed: 12/18/2022]
Abstract
PURPOSE TGF-β plays a dual role in breast carcinogenesis, acting at early stages as tumor-suppressors and later as tumor-promoters. TGF-β isoforms are expressed in breast tissues and secreted in milk, suggesting that analysis of levels in milk might be informative for breast cancer risk. Accordingly, we assessed TGF-β2 levels in milk from women who had undergone a breast biopsy and related the concentrations to diagnosis. METHODS Milk donated by women who had undergone or were scheduled for a breast biopsy was shipped on ice for processing and testing. Breast cancer risk factors were obtained through a self-administered questionnaire, and biopsy diagnoses were extracted from pathology reports. TGF-β2 levels in milk, assessed as absolute levels and in relation to total protein, were analyzed in bilateral samples donated by 182 women. Linear regression was used to estimate relationships of log-transformed TGF-β2 levels and TGF-β2/ total protein ratios to biopsy category. RESULTS Milk TGF-β2 levels from biopsied and non-biopsied breasts within women were highly correlated (r (2) = 0.77). Higher mean TGF-β2 milk levels (based on average of bilateral samples) were marginally associated with more severe breast pathological diagnosis, after adjusting for duration of nursing current child (adjusted p trend = 0.07). CONCLUSIONS Our exploratory analysis suggests a borderline significant association between higher mean TGF-β2 levels in breast milk and more severe pathologic diagnoses. Further analysis of TGF-β signaling in milk may increase understanding of postpartum remodeling and advance efforts to analyze milk as a means of assessing risk of breast pathology.
Collapse
Affiliation(s)
- Hannah P Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Room 7E-238 , Bethesda, MD, 20892, USA,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Tanner SM, Berryhill TF, Ellenburg JL, Jilling T, Cleveland DS, Lorenz RG, Martin CA. Pathogenesis of necrotizing enterocolitis: modeling the innate immune response. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:4-16. [PMID: 25447054 DOI: 10.1016/j.ajpath.2014.08.028] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 08/21/2014] [Accepted: 08/27/2014] [Indexed: 12/23/2022]
Abstract
Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants. The pathophysiology is likely secondary to innate immune responses to intestinal microbiota by the premature infant's intestinal tract, leading to inflammation and injury. This review provides an updated summary of the components of the innate immune system involved in NEC pathogenesis. In addition, we evaluate the animal models that have been used to study NEC with regard to the involvement of innate immune factors and histopathological changes as compared to those seen in infants with NEC. Finally, we discuss new approaches to studying NEC, including mathematical models of intestinal injury and the use of humanized mice.
Collapse
Affiliation(s)
- Scott M Tanner
- Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama; Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Taylor F Berryhill
- Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - James L Ellenburg
- Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tamas Jilling
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Dava S Cleveland
- Department of Pediatric Pathology, Children's Hospital of Alabama, Birmingham, Alabama
| | - Robin G Lorenz
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama.
| | - Colin A Martin
- Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
24
|
Maternal breast milk transforming growth factor-beta and feeding intolerance in preterm infants. Pediatr Res 2014; 76:386-93. [PMID: 24995914 PMCID: PMC4467901 DOI: 10.1038/pr.2014.96] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 04/07/2014] [Indexed: 11/08/2022]
Abstract
BACKGROUND Feeding intolerance (FI) occurs commonly in the neonatal intensive care unit. Breast milk contains a large pool of transforming growth factor-beta (TGF-β). Few studies describe TGF-β levels in preterm milk, and the relationship to FI remains unexplored. We measured TGF-β levels in preterm breast milk to investigate a correlation with FI in preterm infants. METHODS Prospective observational trial of 100 mother-infant pairs, enrolling infants born below 32 wk gestation and less than 1,500 g, and mothers who planned to provide breast milk. TGF-β levels were measured using enzyme-linked immunosorbent assay. Infant charts were reviewed for outcomes. RESULTS TGF-β declined postnatally, most elevated in colostrum (P < 0.01). TGF-β2 levels were higher than TGF-β1 at all time points (P < 0.01). Colostrum TGF-β levels correlated inversely with birth weight (P < 0.01) and gestational age (P < 0.05). One-week TGF-β2 levels were reduced in growth-restricted infants with FI (P < 0.01). Of infants with necrotizing enterocolitis (NEC), TGF-β2 levels appeared to be low, but small sample size precluded meaningful statistical comparisons. CONCLUSION TGF-β levels decline temporally in preterm milk. TGF-β1 colostrum levels correlate inversely with birth weight and gestational age. TGF-β2 may play a role in FI in growth-restricted infants. The relationship of TGF-β2 and NEC merits future investigation.
Collapse
|