1
|
Schaller ML, Sykes MM, Mecano J, Solanki S, Huang W, Rebernick RJ, Beydoun S, Wang E, Bugarin-Lapuz A, Shah YM, Leiser SF. Fmo5 plays a sex-specific role in goblet cell maturation and mucus barrier formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588360. [PMID: 38645243 PMCID: PMC11030302 DOI: 10.1101/2024.04.05.588360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background and Aims The intestine plays a key role in metabolism, nutrient and water absorption, and provides both physical and immunological defense against dietary and luminal antigens. The protective mucosal lining in the intestine is a critical component of intestinal barrier that when compromised, can lead to increased permeability, a defining characteristic of inflammatory bowel disease (IBD), among other intestinal diseases. Here, we define a new role for the flavin-containing monooxygenase (FMO) family of enzymes in maintaining a healthy intestinal epithelium. Methods Using Caenorhabditis elegans we measure intestinal barrier function, actin expression, and intestinal damage response. In mice, we utilize an intestine-specific, tamoxifen- inducible knockout model of the mammalian homolog of Cefmo-2 , Fmo5, and assess histology, mucus barrier thickness, and goblet cell physiology. We also treat mice with the ER chaperone Tauroursodeoxycholic acid (TUDCA). Results In nematodes, we find Cefmo-2 is necessary and sufficient for intestinal barrier function, intestinal actin expression, and is induced by intestinal damage. In mice, we find striking changes to the intestine within two weeks following Fmo5 disruption. Alterations include sex-dependent changes in colon epithelial histology, goblet cell localization, and mucus barrier formation. These changes are significantly more severe in female mice, mirroring differences observed in IBD patients. Furthermore, we find increased protein folding stress in Fmo5 knockout animals and successfully rescue the severe female phenotype with addition of a chemical ER chaperone. Conclusions Together, our results identify a highly conserved and novel role for Fmo5 in the mammalian intestine and support a key role for Fmo5 in maintenance of ER/protein homeostasis and proper mucus barrier formation.
Collapse
|
2
|
Cai C, Shen J. The roles of migrasomes in immunity, barriers, and diseases. Acta Biomater 2024:S1742-7061(24)00529-4. [PMID: 39284502 DOI: 10.1016/j.actbio.2024.09.013] [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: 03/19/2024] [Revised: 09/01/2024] [Accepted: 09/10/2024] [Indexed: 10/14/2024]
Abstract
Migrasomes are recently identified extracellular vesicles and organelles formed in conjunction with cell migration. They are situated at the rear of migrating cells, exhibit a circular or elliptical membrane-enclosed structure, and function as a new organelle. Migrasomes selectively sort intercellular components, mediating a cell migration-dependent release mechanism known as migracytosis and modulating cell-cell communication. Accumulated evidence clarifies migrasome formation processes and indicates their diverse functional roles. Migrasomes may also be potentially correlated with the occurrence, progression, and prognosis of certain diseases. Migrasomes' involvement in physiological and pathological processes highlights their potential for expanding our understanding of biological procedures and as a target in clinical therapy. However, the precise mechanisms and full extent of their involvement in immunity, barriers, and diseases remain unclear. This review aimed to provide a comprehensive overview of the roles of migrasomes in human immunity and barriers, in addition to providing insights into their impact on human diseases. STATEMENT OF SIGNIFICANCE: Migrasomes, newly identified extracellular vesicles and organelles, form during cell migration and are located at the rear of migrating cells. These circular or elliptical structures mediate migracytosis, selectively sorting intercellular components and modulating cell-cell communication. Evidence suggests diverse functional roles for migrasomes, including potential links to disease occurrence, progression, and prognosis. Their involvement in physiological and pathological processes highlights their significance in understanding biological procedures and potential clinical therapies. However, their exact mechanisms in immunity, barriers, and diseases remain unclear. This review provides an overview of migrasomes' roles in human immunity and barriers, and their impact on diseases.
Collapse
Affiliation(s)
- Changsheng Cai
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai 200127, China
| | - Jun Shen
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai 200127, China.
| |
Collapse
|
3
|
Gangaiah D, Gu M, Zaparte A, Will O, Dolan LC, Goering A, Pillai J, Mane SP, Plata G, Helmes EB, Welsh DA, Mahajan AK. Effects of Limosilactobacillus reuteri strains PTA-126787 and PTA-126788 on intestinal barrier integrity and immune homeostasis in an alcohol-induced leaky gut model. Sci Rep 2024; 14:19584. [PMID: 39179898 PMCID: PMC11344072 DOI: 10.1038/s41598-024-70549-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024] Open
Abstract
Intestinal barrier is a first line of defense that prevents entry of various harmful substances from the lumen into the systemic environment. Impaired barrier function with consequent translocation of harmful substances into systemic circulation ("leaky gut") is a central theme in many gastrointestinal, autoimmune, mental, and metabolic diseases. Probiotics have emerged as a promising strategy to maintain intestinal integrity and address "leaky gut". Using in silico, in vitro and avian in vivo analyses, we previously showed that two novel L. reuteri strains, PTA-126787 (L. reuteri 3630) and PTA-126788 (L. reuteri 3632), isolated from broiler chickens possess favorable safety profiles. Consistent with a recent study, here we show that L. reuteri 3630 and 3632 are phylogenetically similar to human L. reuteri strains. Daily administration of high doses of L. reuteri 3630 and 3632 to Sprague Dawley rats for 28 days was found to be safe with no adverse effects. More importantly, administration of L. reuteri 3630 and 3632 significantly reduced markers associated with alcohol-induced leaky gut, by downregulating inflammatory cytokines and upregulating anti-inflammatory cytokines in an alcohol model of leaky gut in mice. While L. reuteri 3630 cells and supernatant showed no activation, L. reuteri 3632 cells but not supernatant showed activation of AhR, a key transcription factor that regulates gut and immune homeostasis. L. reuteri 3630 is creamish white in morphology typical of Lactobacillus species and L. reuteri 3632 displays a unique orange pigmentation, which was stable even after passaging for 480 generations. We identified a rare polyketide biosynthetic gene cluster in L. reuteri 3632 that likely encodes for the orange-pigmented secondary metabolite. Similar to L. reuteri 3632 cells, the purified orange metabolite activated AhR. All together, these data provide evidence on the phylogenetic relatedness, safety, efficacy, and one of the likely mechanisms of action of L. reuteri 3630 and 3632 for potential probiotic applications to address "leaky gut" and associated pathologies in humans.
Collapse
Affiliation(s)
| | - Min Gu
- Department of Medicine, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | - Aline Zaparte
- Department of Medicine, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | - Olaf Will
- Elanco Animal Health, Inc., Alfred-Nobel-Strasse 50, 40789, Monheim Am Rhein, Germany
| | - Laurie C Dolan
- GRAS Associates, 1180 Grand Park Avenue, North Bethesda, MD, 20852, USA
| | | | - Jason Pillai
- MicroMGx, Inc., 3440 S Dearborn St, Chicago, IL, 60616, USA
| | | | - German Plata
- BiomEdit, LLC, 2710 Innovation Way, Greenfield, IN, 46140, USA
| | - Emily B Helmes
- BiomEdit, LLC, 2710 Innovation Way, Greenfield, IN, 46140, USA
| | - David A Welsh
- Department of Medicine, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | | |
Collapse
|
4
|
do Nascimento DDSM, Mota ACCC, Carvalho MCDC, Andrade EDDO, de Oliveira ÉPSF, Galvão LLP, Maciel BLL. Can Diet Alter the Intestinal Barrier Permeability in Healthy People? A Systematic Review. Nutrients 2024; 16:1871. [PMID: 38931225 PMCID: PMC11206284 DOI: 10.3390/nu16121871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Dietary factors can modify the function of the intestinal barrier, causing permeability changes. This systematic review analyzed evidence on the link between diet or dietary interventions and changes in intestinal barrier permeability (IBP) in healthy individuals. A systematic search for primary studies was conducted using the virtual databases EMBASE, PubMed, Web of Science, CINAHL, and Scopus. This review adhered to PRISMA 2020 guidelines, assessing the methodological quality using the Newcastle-Ottawa scale for observational studies and ROB 2.0 for randomized clinical trials. Out of 3725 studies recovered, 12 were eligible for review. Chicory inulin and probiotics reduced IBP in adults with a moderate GRADE level of evidence. The opposite result was obtained with fructose, which increased IBP in adults, with a very low GRADE level of evidence. Only intervention studies with different dietary components were found, and few studies evaluated the effect of specific diets on the IBP. Thus, there was no strong evidence that diet or dietary interventions increase or decrease IBP in healthy individuals. Studies on this topic are necessary, with a low risk of bias and good quality of evidence generated, as there is still little knowledge on healthy populations.
Collapse
Affiliation(s)
- Daniele de Souza Marinho do Nascimento
- Post Graduate Program in Health Science, Center for Health Science, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil; (D.d.S.M.d.N.); (M.C.d.C.C.); (E.D.d.O.A.); (L.L.P.G.)
| | - Ana Carolina Costa Campos Mota
- Post Graduate Program in Nutrition, Department of Nutrition, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil; (A.C.C.C.M.); (É.P.S.F.d.O.)
| | - Maria Clara da Cruz Carvalho
- Post Graduate Program in Health Science, Center for Health Science, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil; (D.d.S.M.d.N.); (M.C.d.C.C.); (E.D.d.O.A.); (L.L.P.G.)
| | - Eva Débora de Oliveira Andrade
- Post Graduate Program in Health Science, Center for Health Science, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil; (D.d.S.M.d.N.); (M.C.d.C.C.); (E.D.d.O.A.); (L.L.P.G.)
| | - Érika Paula Silva Freitas de Oliveira
- Post Graduate Program in Nutrition, Department of Nutrition, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil; (A.C.C.C.M.); (É.P.S.F.d.O.)
| | - Liana Letícia Paulino Galvão
- Post Graduate Program in Health Science, Center for Health Science, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil; (D.d.S.M.d.N.); (M.C.d.C.C.); (E.D.d.O.A.); (L.L.P.G.)
| | - Bruna Leal Lima Maciel
- Post Graduate Program in Health Science, Center for Health Science, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil; (D.d.S.M.d.N.); (M.C.d.C.C.); (E.D.d.O.A.); (L.L.P.G.)
- Post Graduate Program in Nutrition, Department of Nutrition, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil; (A.C.C.C.M.); (É.P.S.F.d.O.)
- Department of Nutrition, Center for Health Science, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| |
Collapse
|
5
|
Wang Y, Jia Z, Zheng M, Wang P, Gao J, Zhang X, Zhou T, Zu G. Inhibition of miR-142-3p promotes intestinal epithelial proliferation and barrier function after ischemia/reperfusion injury by targeting FoxM1. Mol Cell Biochem 2024:10.1007/s11010-024-05038-5. [PMID: 38819598 DOI: 10.1007/s11010-024-05038-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 05/14/2024] [Indexed: 06/01/2024]
Abstract
Damage of intestinal barrier function (BF) after ischemia/reperfusion (I/R) injury can induce serious complications and high mortality. MicroRNAs (miRNAs) are involved in intestinal mucosal BF and epithelial proliferation after I/R injury have been reported. We aimed to investigate the role and regulatory mechanism of miR-142-3p (miR-142) in intestinal epithelial proliferation and BF after I/R injury. We detected the proliferation, barrier function and miR-142 expression in clinical ischemic intestinal tissues. Furthermore, we induced an in vivo intestinal I/R injury mouse model and in vitro IEC-6 cells hypoxia/reoxygenation (H/R) injury model. After increasing and decreasing expression of miR-142, we detected the proliferation and barrier function of intestinal epithelial cells after I/R or H/R injury. We found that miR-142 expression was significantly increased in clinical ischemic intestinal mucosa and mouse intestinal mucosa exposed to I/R injury, and there was an inverse relationship between miR-142 and proliferation/BF. Inhibition of miR-142 significant promoted intestinal epithelial proliferation and BF after I/R injury. Furthermore, inhibition of miR-142 improved overall survival rate of mice after I/R injury. MiR-142 directly targeted FoxM1 which was identified by bioinformatics analysis and luciferase activity assay in IEC-6 cells. Inhibition of miR-142 promotes intestinal epithelial proliferation and BF after I/R injury in a FoxM1-mediated manner.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Gastroenterology Surgery, the Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), No. 826 of Southwest Road Shahekou District, Dalian, 116033, People's Republic of China
- Department of Graduate School, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Zirui Jia
- Department of Gastroenterology Surgery, the Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), No. 826 of Southwest Road Shahekou District, Dalian, 116033, People's Republic of China
- Department of Graduate School, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Mingcan Zheng
- Department of Gastroenterology Surgery, the Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), No. 826 of Southwest Road Shahekou District, Dalian, 116033, People's Republic of China
- Department of Graduate School, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Puxu Wang
- Department of Gastroenterology Surgery, the Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), No. 826 of Southwest Road Shahekou District, Dalian, 116033, People's Republic of China
- Department of Graduate School, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Jiacheng Gao
- Department of Gastroenterology Surgery, the Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), No. 826 of Southwest Road Shahekou District, Dalian, 116033, People's Republic of China
- Department of Graduate School, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Xiangwen Zhang
- Department of Gastroenterology Surgery, the Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), No. 826 of Southwest Road Shahekou District, Dalian, 116033, People's Republic of China
| | - Tingting Zhou
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Guo Zu
- Department of Gastroenterology Surgery, the Central Hospital of Dalian University of Technology (Dalian Municipal Central Hospital), No. 826 of Southwest Road Shahekou District, Dalian, 116033, People's Republic of China.
| |
Collapse
|
6
|
Qiu D, Xu S, Ji K, Tang C. Myeloid Cell-Derived IL-1 Signaling Damps Neuregulin-1 from Fibroblasts to Suppress Colitis-Induced Early Repair of the Intestinal Epithelium. Int J Mol Sci 2024; 25:4469. [PMID: 38674054 PMCID: PMC11050633 DOI: 10.3390/ijms25084469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Neuregulin-1 (Nrg1, gene symbol: Nrg1), a ligand of the ErbB receptor family, promotes intestinal epithelial cell proliferation and repair. However, the dynamics and accurate derivation of Nrg1 expression during colitis remain unclear. By analyzing the public single-cell RNA-sequencing datasets and employing a dextran sulfate sodium (DSS)-induced colitis model, we investigated the cell source of Nrg1 expression and its potential regulator in the process of epithelial healing. Nrg1 was majorly expressed in stem-like fibroblasts arising early in mouse colon after DSS administration, and Nrg1-Erbb3 signaling was identified as a potential mediator of interaction between stem-like fibroblasts and colonic epithelial cells. During the ongoing colitis phase, a significant infiltration of macrophages and neutrophils secreting IL-1β emerged, accompanied by the rise in stem-like fibroblasts that co-expressed Nrg1 and IL-1 receptor 1. By stimulating intestinal or lung fibroblasts with IL-1β in the context of inflammation, we observed a downregulation of Nrg1 expression. Patients with inflammatory bowel disease also exhibited an increase in NRG1+IL1R1+ fibroblasts and an interaction of NRG1-ERBB between IL1R1+ fibroblasts and colonic epithelial cells. This study reveals a novel potential mechanism for mucosal healing after inflammation-induced epithelial injury, in which inflammatory myeloid cell-derived IL-1β suppresses the early regeneration of intestinal tissue by interfering with the secretion of reparative neuregulin-1 by stem-like fibroblasts.
Collapse
Affiliation(s)
- Ding Qiu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, Guangzhou 510080, China;
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (S.X.); (K.J.)
| | - Shaoting Xu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (S.X.); (K.J.)
| | - Kaile Ji
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (S.X.); (K.J.)
| | - Ce Tang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, Guangzhou 510080, China;
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (S.X.); (K.J.)
- Animal Experiment Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
7
|
Kong L, Chen S, Huang S, Zheng A, Gao S, Ye J, Hua C. Challenges and opportunities in inflammatory bowel disease: from current therapeutic strategies to organoid-based models. Inflamm Res 2024; 73:541-562. [PMID: 38345635 DOI: 10.1007/s00011-024-01854-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/20/2024] [Accepted: 01/24/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is an increasingly prevalent global health concern that has garnered substantial attention. However, the underlying mechanisms are still unclear and the current treatments have significant limitations. Intestinal organoids provide an in vitro model to explore the pathogenesis, test the therapeutic effects, and develop regenerative treatments as well as offer the potential to transform drug discovery of IBD. METHODS To advance our understanding of the whole story of IBD spanning from the pathogenesis to the current therapeutic strategies and latest advancements, a comprehensive search of major databases including PubMed, Scopus, and Web of Science was conducted to retrieve original articles and reviews related to IBD, organoids, pathogenesis and therapy. RESULTS This review deciphers the etiopathogenesis and the current therapeutic approaches in the treatment of IBD. Notably, critical aspects of intestinal organoids in IBD, such as their potential applications, viability, cell renewal ability, and barrier functionality are highlighted. We also discuss the advances, limitations, and prospects of intestinal organoids for precision medicine. CONCLUSION The latest strides made in research about intestinal organoids help elucidate intricate aspects of IBD pathogenesis, and pave the prospective avenues for novel therapeutic interventions.
Collapse
Affiliation(s)
- Lingjie Kong
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Siyan Chen
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Shenghao Huang
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Anzhe Zheng
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Sheng Gao
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Jianzhong Ye
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
8
|
Jin C, Wu S, Liang Z, Zhang J, Lei X, Bai H, Liang G, Su X, Chen X, Wang P, Wang Y, Guan L, Yao J. Multi-omics reveal mechanisms of high enteral starch diet mediated colonic dysbiosis via microbiome-host interactions in young ruminant. MICROBIOME 2024; 12:38. [PMID: 38395946 PMCID: PMC10893732 DOI: 10.1186/s40168-024-01760-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 01/08/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Although rumen development is crucial, hindgut undertakes a significant role in young ruminants' physiological development. High-starch diet is usually used to accelerate rumen development for young ruminants, but always leading to the enteral starch overload and hindgut dysbiosis. However, the mechanism behind remains unclear. The combination of colonic transcriptome, colonic luminal metabolome, and metagenome together with histological analysis was conducted using a goat model, with the aim to identify the potential molecular mechanisms behind the disrupted hindgut homeostasis by overload starch in young ruminants. RESULT Compared with low enteral starch diet (LES), high enteral starch diet (HES)-fed goats had significantly higher colonic pathology scores, and serum diamine oxidase activity, and meanwhile significantly decreased colonic mucosal Mucin-2 (MUC2) protein expression and fecal scores, evidencing the HES-triggered colonic systemic inflammation. The bacterial taxa Prevotella sp. P4-67, Prevotella sp. PINT, and Bacteroides sp. CAG:927, together with fungal taxa Fusarium vanettenii, Neocallimastix californiae, Fusarium sp. AF-8, Hypoxylon sp. EC38, and Fusarium pseudograminearum, and the involved microbial immune pathways including the "T cell receptor signaling pathway" were higher in the colon of HES goats. The integrated metagenome and host transcriptome analysis revealed that these taxa were associated with enhanced pathogenic ability, antigen processing and presentation, and stimulated T helper 2 cell (TH2)-mediated cytokine secretion functions in the colon of HES goats. Further luminal metabolomics analysis showed increased relative content of chenodeoxycholic acid (CDCA) and deoxycholic acid (DCA), and decreased the relative content of hypoxanthine in colonic digesta of HES goats. These altered metabolites contributed to enhancing the expression of TH2-mediated inflammatory-related cytokine secretion including GATA Binding Protein 3 (GATA3), IL-5, and IL-13. Using the linear mixed effect model, the variation of MUC2 biosynthesis explained by the colonic bacteria, bacterial functions, fungi, fungal functions, and metabolites were 21.92, 20.76, 19.43, 12.08, and 44.22%, respectively. The variation of pathology scores explained by the colonic bacterial functions, fungal functions, and metabolites were 15.35, 17.61, and 57.06%. CONCLUSIONS Our findings revealed that enteral starch overload can trigger interrupted hindgut host-microbiome homeostasis that led to impaired mucosal, destroyed colonic water absorption, and TH2-mediated inflammatory process. Except for the colonic metabolites mostly contribute to the impaired mucosa, the nonnegligible contribution from fungi deserves more future studies focused on the fungal functions in hindgut dysbiosis of young ruminants. Video Abstract.
Collapse
Affiliation(s)
- Chunjia Jin
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Shengru Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China.
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Ziqi Liang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Jun Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xinjian Lei
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hanxun Bai
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Gaofeng Liang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xiaodong Su
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xiaodong Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Peiyue Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yue Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Leluo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 116 St. and 85 Ave., Edmonton, AB, T6G 2P5, Canada.
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China.
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
9
|
Vergne-Salle P, Salle L, Fressinaud-Marie AC, Descamps-Deplas A, Montestruc F, Bonnet C, Bertin P. Diet and Disease Activity in Patients with Axial Spondyloarthritis: SpondyloArthritis and NUTrition Study (SANUT). Nutrients 2022; 14:nu14224730. [PMID: 36432416 PMCID: PMC9695957 DOI: 10.3390/nu14224730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Axial Spondyloarthritis (axSpA) patients with inflamed intestines have higher SpA activity. Diets that modulate microbiota may influence inflammation and SpA activity. Today, data concerning the impact of diet on SpA activity are scarce. SANUT was a single-center, noninterventional, cohort study that assessed dietetic profiles associated with SpA activity in axSpA. Demographic, clinical, SpA-related, quality of life (QoL), fatigue, physical activity, and dietary data were collected. SpA activity was assessed by Ankylosing Spondylitis Disease Activity Score (ASDAS) and by Bath Ankylosing Spondylitis Disease Activity Index (BASDAI). We assessed whether high SpA activity was associated with nutriment consumption. Between 12 February 2018 and 12 February 2020, 278 patients participated. High SpA activity, as measured by ASDAS and BASDAI, was significantly associated with higher body mass index and waist circumference, negative HLA-B27, lower QoL, higher fatigue, and higher digestive-symptom scores. Furthermore, high SpA activity, as measured by BASDAI, was associated with female sex, smoking status, patients who were not actively employed, reduced physical activity, and high intake of ultra-transformed foods, while high SpA activity, as measured by ASDAS, was associated with low intake of omega-3 PUFAs and fiber. Therefore, low intakes of omega-3 PUFAs and fiber, and high intake of ultra-transformed foods, are associated with high SpA activity.
Collapse
Affiliation(s)
- Pascale Vergne-Salle
- Rheumatology Department, University Hospital of Limoges and Laboratory PEIRENE UR 22722 Institut OmegaHealth, 87042 Limoges, France
- Correspondence:
| | - Laurence Salle
- Endocrinology Department, University Hospital of Limoges and Inserm U1094, EpiMaCT—Epidemiology of Chronic Diseases in Tropical Zone, 87042 Limoges, France
| | - Anne Catherine Fressinaud-Marie
- Rheumatology Department, University Hospital of Limoges and Laboratory PEIRENE UR 22722 Institut OmegaHealth, 87042 Limoges, France
| | - Adeline Descamps-Deplas
- Rheumatology Department, University Hospital of Limoges and Laboratory PEIRENE UR 22722 Institut OmegaHealth, 87042 Limoges, France
| | | | - Christine Bonnet
- Rheumatology Department, University Hospital of Limoges and Laboratory PEIRENE UR 22722 Institut OmegaHealth, 87042 Limoges, France
| | - Philippe Bertin
- Rheumatology Department, University Hospital of Limoges and Laboratory PEIRENE UR 22722 Institut OmegaHealth, 87042 Limoges, France
| |
Collapse
|
10
|
Yan L, Liang B, Feng J, Zhang HY, Chang HS, Liu B, Chen YL. Safety and feasibility of irreversible electroporation for the pancreatic head in a porcine model. World J Gastrointest Oncol 2022; 14:1499-1509. [PMID: 36160734 PMCID: PMC9412922 DOI: 10.4251/wjgo.v14.i8.1499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/12/2022] [Accepted: 07/11/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Irreversible electroporation (IRE) is a local non-thermal ablative technique which has been suggested as a potential cancer therapy. However, the specific anatomic characteristics of the pancreatic head make it challenging to perform any local ablation in this region. Therefore, the safety and feasibility of IRE in the pancreatic head region should be further explored.
AIM To evaluate the safety of IRE in pancreatic head region including its effects on pancreatic ducts, vessels, and adjacent gastrointestinal organs.
METHODS Eight landrace miniature pigs underwent IRE of pancreatic head tissue successfully, with a total of 16 lesions created. Laboratory testing including white blood cell (WBC) count and serum amylase before IRE with follow-up laboratory analysis and pathological examination at 1, 7, 14, and 28 d postablation were performed.
RESULTS All pigs tolerated the ablation procedure without serious perioperative complications. Transiently elevated WBC count and amylase were observed at 24 h post-IRE, suggesting an acute pancreatic tissue damage which was confirmed by pathological observations. Vascular endothelial cells and pancreatic duct epithelial cells in ablation zone were also positive in terminal deoxynucleotidyl transferase dUTP nick end labeling staining. There was extensive duodenum mucosa damage with local hemorrhage 24 h after ablation, while regeneration of new villous structures were observed at 7 and 28 d post-IRE. Masson’s trichromatic staining showed that the extracellular matrix was still intact in vessels and pancreatic ducts, and even in the duodenum.
CONCLUSION IRE ablation to the pancreatic head may be safe and feasible without long-term damage to the surrounding vital structures. However, risks of stress injuries in acute phase should be taken into consideration to prevent severe perioperative complications.
Collapse
Affiliation(s)
- Li Yan
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepetobiliary Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Bin Liang
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepetobiliary Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jian Feng
- Department of Hepatopancreatobiliary Surgery, Peking University Shougang Hospital, Beijing 100144, China
| | - Hang-Yu Zhang
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepetobiliary Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Hao-Sheng Chang
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepetobiliary Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Bing Liu
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepetobiliary Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yong-Liang Chen
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery of Chinese PLA, Key Laboratory of Digital Hepetobiliary Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
11
|
Alpha-Lipoic Acid Promotes Intestinal Epithelial Injury Repair by Regulating MAPK Signaling Pathways. Mediators Inflamm 2022; 2022:1894379. [PMID: 35712055 PMCID: PMC9197635 DOI: 10.1155/2022/1894379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/16/2022] [Indexed: 11/17/2022] Open
Abstract
Intestinal epithelial cells are an essential barrier in human gastrointestinal tract, and healing of epithelial wound is a key process in many intestinal diseases. α-Lipoic acid (ALA) was shown to have antioxidative and anti-inflammatory effects, which could be helpful in intestinal epithelial injury repair. The effects of ALA in human colonic epithelial cells NCM460 and human colorectal adenocarcinoma cells Caco-2 were studied. ALA significantly promoted NCM460 and Caco-2 migration, increased mucosal tight junction factors ZO-1 and OCLN expression, and ALA accelerated cell injury repair of both cells in wound healing assay. Western blot analysis indicated that ALA inhibited a variety of mitogen-activated protein kinase (MAPK) signaling pathways in the epithelial cells. In conclusion, ALA was beneficial to repair of intestinal epithelial injury by regulating MAPK signaling pathways.
Collapse
|
12
|
Exposomic determinants of immune-mediated diseases. Environ Epidemiol 2022; 6:e212. [PMID: 35702504 PMCID: PMC9187189 DOI: 10.1097/ee9.0000000000000212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/04/2022] [Indexed: 11/25/2022] Open
|
13
|
Probiotics Supplementation Improves Intestinal Permeability, Obesity Index and Metabolic Biomarkers in Elderly Thai Subjects: A Randomized Controlled Trial. Foods 2022; 11:foods11030268. [PMID: 35159419 PMCID: PMC8834517 DOI: 10.3390/foods11030268] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/06/2022] [Accepted: 01/14/2022] [Indexed: 02/07/2023] Open
Abstract
Intestinal integrity prevents the diffusion of allergens, toxins, and pathogens from the gastrointestinal lumen into the tissue and the circulatory system. Damage in intestinal integrity may cause mild to serious health issues, such as inflammation, gastrointestinal disorders, neurological diseases, and neurodegenerative disorders. Thus, maintaining a healthy intestinal barrier function is essential to sustain health. Probiotics are known for their ability to protect and restore intestinal permeability in vitro and in vivo. The multi-strain probiotics are more efficient than that of a single strain in terms of their protective efficacy. Therefore, the present study was planned and implemented to study the supplementation of probiotic mix (Lactobacillus paracasei HII01, Bifidobacteriumbreve, and Bifidobacterium longum) on intestinal permeability, lipid profile, obesity index and metabolic biomarkers in elderly Thai subjects. The results revealed that the supplementation of studied probiotics improved the intestinal barrier function (up to 48%), significantly increasing the high-density lipoprotein (HDL)-cholesterol. Moreover, the intervention improved obesity-related anthropometric biomarkers and short-chain fatty acid levels in human subjects. The current study strongly recommends further extended research to confirm the beneficial effect of probiotics, which may pave the way to formulate probiotic-based health supplements to adjuvant the treatment of several metabolic diseases.
Collapse
|
14
|
Zhang P, Jing C, Liang M, Jiang S, Huang L, Jiao N, Li Y, Yang W. Zearalenone Exposure Triggered Cecal Physical Barrier Injury through the TGF-β1/Smads Signaling Pathway in Weaned Piglets. Toxins (Basel) 2021; 13:toxins13120902. [PMID: 34941739 PMCID: PMC8708673 DOI: 10.3390/toxins13120902] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
This study aims to investigate the effects of exposure to different dosages of zearalenone (ZEA) on cecal physical barrier functions and its mechanisms based on the TGF-β1/Smads signaling pathway in weaned piglets. Thirty-two weaned piglets were allotted to four groups and fed a basal diet supplemented with ZEA at 0, 0.15, 1.5, and 3.0 mg/kg, respectively. The results showed that 1.5 and 3.0 mg/kg ZEA damaged cecum morphology and microvilli, and changed distribution and shape of M cells. Moreover, 1.5 and 3.0 mg/kg ZEA decreased numbers of goblet cells, the expressions of TFF3 and tight junction proteins, and inhibited the TGF-β1/Smads signaling pathway. Interestingly, the 0.15 mg/kg ZEA had no significant effect on cecal physical barrier functions but decreased the expressions of Smad3, p-Smad3 and Smad7. Our study suggests that high-dose ZEA exposure impairs cecal physical barrier functions through inhibiting the TGF-β1/Smads signaling pathway, but low-dose ZEA had no significant effect on cecum morphology and integrity through inhibiting the expression of smad7. These findings provide a scientific basis for helping people explore how to reduce the toxicity of ZEA in feeds.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
| | - Changwei Jing
- Technical Department, Shandong Chinwhiz Co., Ltd., Weifang 262400, China;
| | - Ming Liang
- Department of Feeding Microecology, Shandong Baolaililai Bioengineering Co., Ltd., Tai’an 271001, China;
| | - Shuzhen Jiang
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
| | - Libo Huang
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
| | - Ning Jiao
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
| | - Yang Li
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
- Correspondence: (Y.L.); (W.Y.)
| | - Weiren Yang
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
- Correspondence: (Y.L.); (W.Y.)
| |
Collapse
|
15
|
Boby N, Cao X, Ransom A, Pace BT, Mabee C, Shroyer MN, Das A, Didier PJ, Srivastav SK, Porter E, Sha Q, Pahar B. Identification, Characterization, and Transcriptional Reprogramming of Epithelial Stem Cells and Intestinal Enteroids in Simian Immunodeficiency Virus Infected Rhesus Macaques. Front Immunol 2021; 12:769990. [PMID: 34887863 PMCID: PMC8650114 DOI: 10.3389/fimmu.2021.769990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Epithelial cell injury and impaired epithelial regeneration are considered key features in HIV pathogenesis and contribute to HIV-induced generalized immune activation. Understanding the molecular mechanisms underlying the disrupted epithelial regeneration might provide an alternative approach for the treatment of HIV-mediated enteropathy and immune activation. We have observed a significant increased presence of α defensin5+ (HD5) Paneth cells and proliferating Ki67+ epithelial cells as well as decreased expression of E-cadherin expression in epithelial cells during SIV infection. SIV infection did not significantly influence the frequency of LGR5+ stem cells, but the frequency of HD5+ cells was significantly higher compared to uninfected controls in jejunum. Our global transcriptomics analysis of enteroids provided novel information about highly significant changes in several important pathways like metabolic, TCA cycle, and oxidative phosphorylation, where the majority of the differentially expressed genes were downregulated in enteroids grown from chronically SIV-infected macaques compared to the SIV-uninfected controls. Despite the lack of significant reduction in LGR5+ stem cell population, the dysregulation of several intestinal stem cell niche factors including Notch, mTOR, AMPK and Wnt pathways as well as persistence of inflammatory cytokines and chemokines and loss of epithelial barrier function in enteroids further supports that SIV infection impacts on epithelial cell proliferation and intestinal homeostasis.
Collapse
Affiliation(s)
- Nongthombam Boby
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Xuewei Cao
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI, United States
| | - Alyssa Ransom
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Barcley T Pace
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Christopher Mabee
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Monica N Shroyer
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Arpita Das
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | - Peter J Didier
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Sudesh K Srivastav
- Department of Biostatistics, Tulane University, New Orleans, LA, United States
| | - Edith Porter
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Qiuying Sha
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI, United States
| | - Bapi Pahar
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States.,Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States.,Department of Tropical Medicine, Tulane School of Public Health and Tropical Medicine, New Orleans, LA, United States
| |
Collapse
|
16
|
Wu L, Li X, Li P, Pan L, Ji Z, Feng Y, Shi C. Bioabsorbable flexible elastomer of
PTMC‐b‐PEG‐b‐PTMC
copolymer as intestinal anastomosis scaffold. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Lei Wu
- School of Chemical Engineering and Technology Tianjin University Tianjin China
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang China
| | - Xujian Li
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang China
| | - Pengpeng Li
- School of Biomedical Engineering Wenzhou Medical University Wenzhou Zhejiang China
| | - Luqi Pan
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang China
| | - Zhixiao Ji
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang China
| | - Yakai Feng
- School of Chemical Engineering and Technology Tianjin University Tianjin China
- Key Laboratory of Systems Bioengineering (Ministry of Education) Tianjin University Tianjin China
| | - Changcan Shi
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang China
| |
Collapse
|
17
|
Slifer ZM, Hernandez L, Pridgen TA, Carlson AR, Messenger KM, Madan J, Krishnan BR, Laumas S, Blikslager AT. Larazotide acetate induces recovery of ischemia-injured porcine jejunum via repair of tight junctions. PLoS One 2021; 16:e0250165. [PMID: 33886649 PMCID: PMC8061941 DOI: 10.1371/journal.pone.0250165] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022] Open
Abstract
Intestinal ischemia results in mucosal injury, including paracellular barrier loss due to disruption of tight junctions. Larazotide acetate (LA), a small peptide studied in Phase III clinical trials for treatment of celiac disease, regulates tight junctions (TJs). We hypothesized that LA would dose-dependently hasten recovery of intestinal ischemic injury via modulation of TJs. Ischemia-injured tissue from 6-8-week-old pigs was recovered in Ussing chambers for 240-minutes in the presence of LA. LA (1 μM but not 0.1 μM or 10 μM) significantly enhanced transepithelial electrical resistance (TER) above ischemic injured controls and significantly reduced serosal-to-mucosal flux LPS (P<0.05). LA (1 μM) enhanced localization of the sealing tight junction protein claudin-4 in repairing epithelium. To assess for the possibility of fragmentation of LA, an in vitro enzyme degradation assay using the brush border enzyme aminopeptidase M, revealed generation of peptide fragments. Western blot analysis of total protein isolated from uninjured and ischemia-injured porcine intestine showed aminopeptidase M enzyme presence in both tissue types, and mass spectrometry analysis of samples collected during ex vivo analysis confirmed formation of LA fragments. Treatment of tissues with LA fragments had no effect alone, but treatment with a fragment missing both amino-terminus glycines inhibited barrier recovery stimulated by 1 μM LA. To reduce potential LA inhibition by fragments, a D-amino acid analog of larazotide Analog #6, resulted in a significant recovery response at a 10-fold lower dose (0.1 μM) similar in magnitude to that of 1 μM LA. We conclude that LA stimulates repair of ischemic-injured epithelium at the level of the tight junctions, at an optimal dose of 1 μM LA. Higher doses were less effective because of inhibition by LA fragments, which could be subverted by chirally-modifying the molecule, or microdosing LA.
Collapse
Affiliation(s)
- Zachary M. Slifer
- Comparative Medicine Institute, Department of Clinical Sciences, North Carolina State University, Raleigh, NC, United States of America
| | - Liliana Hernandez
- Comparative Medicine Institute, Department of Clinical Sciences, North Carolina State University, Raleigh, NC, United States of America
| | - Tiffany A. Pridgen
- Comparative Medicine Institute, Department of Clinical Sciences, North Carolina State University, Raleigh, NC, United States of America
| | - Alexandra R. Carlson
- Comparative Medicine Institute, Department of Clinical Sciences, North Carolina State University, Raleigh, NC, United States of America
| | - Kristen M. Messenger
- Comparative Medicine Institute, Department of Clinical Sciences, North Carolina State University, Raleigh, NC, United States of America
| | - Jay Madan
- Innovate Biopharmaceuticals, Inc., Raleigh, NC, United States of America
| | - B. Radha Krishnan
- Innovate Biopharmaceuticals, Inc., Raleigh, NC, United States of America
| | - Sandeep Laumas
- 9 Meters Biopharma, Inc., Raleigh, NC, United States of America
| | - Anthony T. Blikslager
- Comparative Medicine Institute, Department of Clinical Sciences, North Carolina State University, Raleigh, NC, United States of America
- * E-mail:
| |
Collapse
|
18
|
Qian S, Li C, Liu X, Jia X, Xiao Y, Li Z. Activation of the JNK/MAPK Signaling Pathway by TGF-β1 Enhances Neonatal Fc Receptor Expression and IgG Transcytosis. Microorganisms 2021; 9:879. [PMID: 33923917 PMCID: PMC8073669 DOI: 10.3390/microorganisms9040879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 01/03/2023] Open
Abstract
The neonatal Fc receptor (FcRn) transports maternal immunoglobulin G (IgG) to the foetus or newborn and protects the IgG from degradation. FcRn is expressed in several porcine tissues and cell types and its expression levels are regulated by immune and inflammatory events. IPEC-J2 cells are porcine intestinal columnar epithelial cells that were isolated from neonatal piglet mid-jejunum. We hypothesized that transforming growth factor β1 (TGF-β1) upregulated pFcRn expression in IPEC-J2 cells. To test this hypothesis, we treated IPEC-J2 cells with TGF-β1 and demonstrated that porcine FcRn (pFcRn) expression was significantly increased. SP600125, a specific mitogen-activated protein kinase (MAPK) inhibitor, reduced TGF-β1-induced pFcRn expression in IPEC-J2 cells. We performed luciferase reporter assays and showed that the c-JUN sensitive region of the pFcRn promoter gene was located between positions -1215 and -140. The c-JUN sequence, in combination with the pFcRn promoter, regulated luciferase reporter activity in response to TGF-β1 stimulation. Chromatin immunoprecipitation confirmed that there were three c-JUN binding sites in the pFcRn promoter. Furthermore, in addition to increased pFcRn expression, TGF-β1 also enhanced IgG transcytosis in IPEC-J2 cells. In summary, our data showed that the modulation of JNK/MAPK signaling by TGF-β1 was sufficient to upregulate pFcRn expression.
Collapse
Affiliation(s)
- Shaoju Qian
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.Q.); (C.L.); (X.L.); (X.J.); (Y.X.)
| | - Chenxi Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.Q.); (C.L.); (X.L.); (X.J.); (Y.X.)
| | - Xi Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.Q.); (C.L.); (X.L.); (X.J.); (Y.X.)
| | - Xiangchao Jia
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.Q.); (C.L.); (X.L.); (X.J.); (Y.X.)
| | - Yuncai Xiao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.Q.); (C.L.); (X.L.); (X.J.); (Y.X.)
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Zili Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.Q.); (C.L.); (X.L.); (X.J.); (Y.X.)
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People’s Republic of China, Wuhan 430070, China
| |
Collapse
|
19
|
Walia S, Kamal R, Kanwar SS, Dhawan DK. Hepato-protective role of chemo-preventive probiotics during DMH-induced CRC in rats. J Biochem Mol Toxicol 2021; 35:e22788. [PMID: 33866645 DOI: 10.1002/jbt.22788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 02/11/2021] [Accepted: 03/30/2021] [Indexed: 11/12/2022]
Abstract
The aim of the study was to assess the hepatotoxicity, and therefore pharmacological safety of probiotics Lactobacillus plantarum (AdF10) and Lactobacillus rhamnosus GG (LGG) for potential use in colorectal cancer (CRC) prevention. Thirty-six female Sprague Dawley (SD) rats were divided into six groups: normal control, AdF10-treated, LGG-treated, 1,2-Dimethyl hydrazine (DMH)-treated, AdF10 + DMH-treated, and LGG + DMH-treated groups. Antioxidant enzyme activity, lipid proxidation, and liver function were assessed. Administration of probiotics in both AdF10 + DMH-treated and LGG + DMH-treated groups downregulated DMH induced a rise in lipid peroxide (LPO), glutathione reductase (GR) activity, and increased the diminished glutathione reduced (GSH) content and catalase (CAT), glutathione-transferase (GST), superoxide dismutase (SOD), and glutathione peroxidase (GPx) activities. DMH-treated rats receiving the probiotic treatment suffered less liver damage when compared with rats that did not receive probiotics. In conclusion, the study identifies the use of probiotics as an effective and nontoxic chemo-preventive interventional in CRC.
Collapse
Affiliation(s)
- Sohini Walia
- Department of Microbiology, CSK Himachal Pradesh Agricultural University, Palampur, Himachal Pradesh, India
| | - Rozy Kamal
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sarbjit S Kanwar
- Department of Microbiology, CSK Himachal Pradesh Agricultural University, Palampur, Himachal Pradesh, India
| | | |
Collapse
|
20
|
Hunt JE, Holst JJ, Jeppesen PB, Kissow H. GLP-1 and Intestinal Diseases. Biomedicines 2021; 9:biomedicines9040383. [PMID: 33916501 PMCID: PMC8067135 DOI: 10.3390/biomedicines9040383] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 12/18/2022] Open
Abstract
Accumulating evidence implicates glucagon-like peptide-1 (GLP-1) to have, beyond glucose maintenance, a beneficial role in the gastrointestinal tract. Here, we review emerging data investigating GLP-1 as a novel treatment for intestinal diseases, including inflammatory bowel diseases, short-bowel syndrome, intestinal toxicities and coeliac disease. Possible beneficial mechanisms for these diseases include GLP-1′s influence on gastric emptying, its anti-inflammatory properties and its intestinotrophic effect. The current knowledge basis derives from the available GLP-1 agonist treatments in experimental animals and small clinical trials. However, new novel strategies including dual GLP-1/GLP-2 agonists are also in development for the treatment of intestinal diseases.
Collapse
Affiliation(s)
- Jenna Elizabeth Hunt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (J.E.H.); (J.J.H.)
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (J.E.H.); (J.J.H.)
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Palle Bekker Jeppesen
- Department of Medical Gastroenterology and Hepatology, Rigshospitalet, 2200 Copenhagen, Denmark;
| | - Hannelouise Kissow
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (J.E.H.); (J.J.H.)
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Correspondence:
| |
Collapse
|
21
|
Gagnon KB, Delpire E. Sodium Transporters in Human Health and Disease. Front Physiol 2021; 11:588664. [PMID: 33716756 PMCID: PMC7947867 DOI: 10.3389/fphys.2020.588664] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
Sodium (Na+) electrochemical gradients established by Na+/K+ ATPase activity drives the transport of ions, minerals, and sugars in both excitable and non-excitable cells. Na+-dependent transporters can move these solutes in the same direction (cotransport) or in opposite directions (exchanger) across both the apical and basolateral plasma membranes of polarized epithelia. In addition to maintaining physiological homeostasis of these solutes, increases and decreases in sodium may also initiate, directly or indirectly, signaling cascades that regulate a variety of intracellular post-translational events. In this review, we will describe how the Na+/K+ ATPase maintains a Na+ gradient utilized by multiple sodium-dependent transport mechanisms to regulate glucose uptake, excitatory neurotransmitters, calcium signaling, acid-base balance, salt-wasting disorders, fluid volume, and magnesium transport. We will discuss how several Na+-dependent cotransporters and Na+-dependent exchangers have significant roles in human health and disease. Finally, we will discuss how each of these Na+-dependent transport mechanisms have either been shown or have the potential to use Na+ in a secondary role as a signaling molecule.
Collapse
Affiliation(s)
- Kenneth B. Gagnon
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, United States
| | - Eric Delpire
- Department of Anesthesiology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
22
|
Ren Y, Li X, Wu L, Pan L, Ji Z, Shi C, Zhang X. Poly(trimethylene carbonate) flexible intestinal anastomosis scaffolds to reduce the probability of intestinal fistula and obstruction. J Mater Chem B 2021; 9:5340-5351. [PMID: 34152354 DOI: 10.1039/d1tb00759a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Biodegradable anastomat play an important role in the reconstruction process of the digestive tract. However, the biocompatibility and organizational compliance of anastomotic tubes still need to be improved. Electrospun tissue engineering scaffolds have excellent biomimetic extracellular matrix properties, biocompatibility and biodegradability. In the present study, electrospun poly(trimethylene carbonate) (PTMC) intestinal anastomosis scaffolds loaded with triclosan (TCS) were reported to reduce the probability of intestinal fistula and obstruction. When the viscosity average molecular weight of PTMC was 157 × 103, the elastic modulus and tensile strength of the anastomosis scaffolds could reach 20.11 MPa and 16.08 MPa, respectively, which indicated that the anastomosis scaffolds exhibited excellent tensile flexibility. The degradation of PTMC was accelerated with the increase of Mw. After 28 days, the weight and length of the anastomosis scaffolds reduced 40% and 50%, respectively. Furthermore, the application of PTMC anastomosis scaffolds could promote intestinal healing and reduce the probability of intestinal fistula and obstruction.
Collapse
Affiliation(s)
- Yuehan Ren
- Department of Colorectal anal surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China.
| | - Xujian Li
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China and Oujiang Laboratory, Wenzhou, Zhejiang 325011, China.
| | - Lei Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Luqi Pan
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China and Oujiang Laboratory, Wenzhou, Zhejiang 325011, China.
| | - Zhixiao Ji
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China and Oujiang Laboratory, Wenzhou, Zhejiang 325011, China.
| | - Changcan Shi
- Department of Colorectal anal surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China. and Wenzhou Institute of Biomaterials and Engineering, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China and Oujiang Laboratory, Wenzhou, Zhejiang 325011, China.
| | - Xiaodong Zhang
- Department of Colorectal anal surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China.
| |
Collapse
|
23
|
Couderc M, Pereira B, Schaeverbeke T, Thomas T, Chapurlat R, Gaudin P, Morel J, Dougados M, Soubrier M. GlutenSpA trial: protocol for a randomised double-blind placebo-controlled trial of the impact of a gluten-free diet on quality of life in patients with axial spondyloarthritis. BMJ Open 2020; 10:e038715. [PMID: 33444189 PMCID: PMC7682451 DOI: 10.1136/bmjopen-2020-038715] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Subclinical intestinal inflammation and gut dysbiosis have been reported in patients with spondyloarthritis (SpA). In common practice, rheumatologists are increasingly confronted with patients with inflammatory rheumatism who are on gluten-free diets (GFDs), despite the lack of reliable data from controlled studies. This study aims to determine the impact of a GFD on the quality of life of patients with axial SpA. METHODS AND ANALYSIS The GlutenSpA study is a 24-week, randomised, double-blinded, placebo-controlled, multicentre trial. Patients with axial SpA (n=200) will follow a 16-week GFD and be randomly assigned (1:1) to an experimental or control arm. In the experimental arm with receive at least 6 gluten-free breads per day + 200 g of gluten-free penne pasta per week + 6 rice flavour capsules per day. The control arm will receive at least 6 gluten-containing breads per day + 200 g of gluten-containing penne pasta per week + 6 vital gluten-containing capsules per day. The primary end-point is the variation in Assessment of SpondyloArthritis International Society-Health Index (ASAS-HI) questionnaire between week 16 and baseline. A second open-label period of 8 weeks will follow the intervention period, during which the patient will be free to decide whether they will follow the GFD. The secondary outcomes comprise several patient-reported outcomes (SpA activity (Bath Ankylosing Spondylitis Disease Activity Index)), fatigue (Functional Assessment of Chronic Illness Therapy), depression (Hospital Anxiety and Depression Scale), functional disability index (Bath Ankylosing Spondylitis Functional Index)), variations in body mass index and Homeostasis Model Assessment Index and variations in the abundance and type of bacterial species found in the gut microbiota for a subgroup of patients (n=40). The data will be analysed using the intention-to-treat principle.The regional ethics committee (CPP Nord-ouest IV) has approved the study (IDRCB 2018-A00309-46). The results of the trial will be submitted for publication in peer-reviewed journals. The authors have no relationship that may have influenced the submitted work. TRIAL REGISTRATION NUMBER NCT04274374.
Collapse
Affiliation(s)
- Marion Couderc
- Rheumatology, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Bruno Pereira
- Biostatistical Unit, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | | | | | | | - Philippe Gaudin
- Rheumatology, CHU Grenoble Alpes, Grenoble, Rhône-Alpes, France
| | - Jacques Morel
- Rheumatology, CHU Montpellier, Montpellier, Languedoc-Roussillon, France
| | - Maxime Dougados
- Rheumatology, Cochin Institute, Paris, Île-de-France, France
| | - Martin Soubrier
- Rheumatology, CHU Clermont-Ferrand, Clermont-Ferrand, France
| |
Collapse
|
24
|
Bonvicini F, Pagnotta E, Punzo A, Calabria D, Simoni P, Mirasoli M, Passerini N, Bertoni S, Ugolini L, Lazzeri L, Gentilomi GA, Caliceti C, Roda A. Effect of Lactobacillus acidophilus Fermented Broths Enriched with Eruca sativa Seed Extracts on Intestinal Barrier and Inflammation in a Co-Culture System of an Enterohemorrhagic Escherichia coli and Human Intestinal Cells. Nutrients 2020; 12:nu12103064. [PMID: 33036498 PMCID: PMC7600469 DOI: 10.3390/nu12103064] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 12/11/2022] Open
Abstract
Lactic acid bacteria (LAB) “fermentates” confer a beneficial effect on intestinal function. However, the ability of new fermentations to improve LAB broth activity in preventing pathogen-induced intestinal inflammation and barrier dysfunction has not yet been studied. The objective of this study was to determine if broths of LAB fermented with Eruca sativa or Barbarea verna seed extracts prevent gut barrier dysfunction and interleukin-8 (CXCL8) release in vitro in human intestinal Caco-2 cells infected with enterohemorrhagic Escherichia coli (EHEC) O157:H7. LAB broths were assayed for their effects on EHEC growth and on Caco-2 viability; thereafter, their biological properties were analysed in a co-culture system consisting of EHEC and Caco-2 cells. Caco-2 cells infected with EHEC significantly increased CXCL8 release, and decreased Trans-Epithelial Electrical Resistance (TEER), a barrier-integrity marker. Notably, when Caco-2 cells were treated with LAB broth enriched with E. sativa seed extract and thereafter infected, both CXCL8 expression and epithelial dysfunction reduced compared to in untreated cells. These results underline the beneficial effect of broths from LAB fermented with E. sativa seed extracts in gut barrier and inflammation after EHEC infection and reveal that these LAB broths can be used as functional bioactive compounds to regulate intestinal function.
Collapse
Affiliation(s)
- Francesca Bonvicini
- Department of Pharmacy and Biotechnology—FABIT, University of Bologna, 40126 Bologna, Italy; (F.B.); (N.P.); (S.B.); (G.A.G.)
| | - Eleonora Pagnotta
- CREA-Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, via di Corticella 133, 40128 Bologna, Italy; (E.P.); (L.U.); (L.L.)
| | - Angela Punzo
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, 40126 Bologna, Italy; (A.P.); (D.C.); (M.M.); (A.R.)
| | - Donato Calabria
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, 40126 Bologna, Italy; (A.P.); (D.C.); (M.M.); (A.R.)
| | - Patrizia Simoni
- Department of Medical and Surgical Sciences—DIMEC, University of Bologna, 40126 Bologna, Italy;
| | - Mara Mirasoli
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, 40126 Bologna, Italy; (A.P.); (D.C.); (M.M.); (A.R.)
| | - Nadia Passerini
- Department of Pharmacy and Biotechnology—FABIT, University of Bologna, 40126 Bologna, Italy; (F.B.); (N.P.); (S.B.); (G.A.G.)
| | - Serena Bertoni
- Department of Pharmacy and Biotechnology—FABIT, University of Bologna, 40126 Bologna, Italy; (F.B.); (N.P.); (S.B.); (G.A.G.)
| | - Luisa Ugolini
- CREA-Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, via di Corticella 133, 40128 Bologna, Italy; (E.P.); (L.U.); (L.L.)
| | - Luca Lazzeri
- CREA-Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, via di Corticella 133, 40128 Bologna, Italy; (E.P.); (L.U.); (L.L.)
| | - Giovanna Angela Gentilomi
- Department of Pharmacy and Biotechnology—FABIT, University of Bologna, 40126 Bologna, Italy; (F.B.); (N.P.); (S.B.); (G.A.G.)
| | - Cristiana Caliceti
- Department of Biomedical and Neuromotor Sciences—DIBINEM, University of Bologna, 40126 Bologna, Italy
- Istituto Nazionale Biosistemi e Biostrutture—INBB, 00136 Rome, Italy
- Correspondence:
| | - Aldo Roda
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, 40126 Bologna, Italy; (A.P.); (D.C.); (M.M.); (A.R.)
- Istituto Nazionale Biosistemi e Biostrutture—INBB, 00136 Rome, Italy
| |
Collapse
|
25
|
Braga Emidio N, Brierley SM, Schroeder CI, Muttenthaler M. Structure, Function, and Therapeutic Potential of the Trefoil Factor Family in the Gastrointestinal Tract. ACS Pharmacol Transl Sci 2020; 3:583-597. [PMID: 32832864 DOI: 10.1021/acsptsci.0c00023] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Indexed: 12/20/2022]
Abstract
Trefoil factor family peptides (TFF1, TFF2, and TFF3) are key players in protecting, maintaining, and repairing the gastrointestinal tract. Accordingly, they have the therapeutic potential to treat and prevent a variety of gastrointestinal disorders associated with mucosal damage. TFF peptides share a conserved motif, including three disulfide bonds that stabilize a well-defined three-loop-structure reminiscent of a trefoil. Although multiple functions have been described for TFF peptides, their mechanisms at the molecular level remain poorly understood. This review presents the status quo of TFF research relating to gastrointestinal disorders. Putative TFF receptors and protein partners are described and critically evaluated. The therapeutic potential of these peptides in gastrointestinal disorders where altered mucosal biology plays a crucial role in the underlying etiology is discussed. Finally, areas of investigation that require further research are addressed. Thus, this review provides a comprehensive update on TFF literature as well as guidance toward future research to better understand this peptide family and its therapeutic potential for the treatment of gastrointestinal disorders.
Collapse
Affiliation(s)
- Nayara Braga Emidio
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medicial Research Insittitue (FHMRI), Flinders University, Bedford Park, South Australia 5042, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Christina I Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.,National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Markus Muttenthaler
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
26
|
Slifer ZM, Blikslager AT. The Integral Role of Tight Junction Proteins in the Repair of Injured Intestinal Epithelium. Int J Mol Sci 2020; 21:ijms21030972. [PMID: 32024112 PMCID: PMC7036844 DOI: 10.3390/ijms21030972] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/10/2019] [Accepted: 10/14/2019] [Indexed: 12/29/2022] Open
Abstract
The intestinal epithelial monolayer forms a transcellular and paracellular barrier that separates luminal contents from the interstitium. The paracellular barrier consists of a highly organized complex of intercellular junctions that is primarily regulated by apical tight junction proteins and tight junction-associated proteins. This homeostatic barrier can be lost through a multitude of injurious events that cause the disruption of the tight junction complex. Acute repair after injury leading to the reestablishment of the tight junction barrier is crucial for the return of both barrier function as well as other cellular functions, including water regulation and nutrient absorption. This review provides an overview of the tight junction complex components and how they link to other plasmalemmal proteins, such as ion channels and transporters, to induce tight junction closure during repair of acute injury. Understanding the components of interepithelial tight junctions and the mechanisms of tight junction regulation after injury is crucial for developing future therapeutic targets for patients experiencing dysregulated intestinal permeability.
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Probiotics are promising remedial treatments for symptoms of small intestine (SI) diseases and promoters of overall good health. Probiotics play an important role in supporting a healthy SI microbiome (eubiosis), and in preventing establishment of unhealthy microbiota. SI eubiosis promotes optimal nutrient uptake, and optimal nutritional status maintains a healthy SI, reducing the likelihood of SI diseases. It is important to understand the advantages and limitations of probiotic therapies. RECENT FINDINGS Microbial dysbiosis decreases the capacity of the small bowel to utilize and absorb dietary compounds. In some studies, probiotic supplements containing lactic acid bacteria and Bifidobacterium have been demonstrated effective in supporting beneficial microbes in the SI while improving barrier integrity and reducing nutrient malabsorption and SI disease-related pathology. Strain-specific probiotic therapy may be a natural and effective approach to restoring SI barrier integrity and eubiosis, resulting in improved nutrient absorption and better health, including reducing the incidence of and severity of SI diseases.
Collapse
Affiliation(s)
- Taylor C Judkins
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr., Gainesville, FL, 32611, USA
| | - Douglas L Archer
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr., Gainesville, FL, 32611, USA
| | | | - Rebecca J Solch
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr., Gainesville, FL, 32611, USA.
| |
Collapse
|
28
|
Wagar LE, Bolen CR, Sigal N, Lopez Angel CJ, Guan L, Kirkpatrick BD, Haque R, Tibshirani RJ, Parsonnet J, Petri WA, Davis MM. Increased T Cell Differentiation and Cytolytic Function in Bangladeshi Compared to American Children. Front Immunol 2019; 10:2239. [PMID: 31620139 PMCID: PMC6763580 DOI: 10.3389/fimmu.2019.02239] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 09/04/2019] [Indexed: 01/03/2023] Open
Abstract
During the first 5 years of life, children are especially vulnerable to infection-related morbidity and mortality. Conversely, the Hygiene Hypothesis suggests that a lack of exposure to infectious agents early in life could explain the increasing incidence of allergies and autoimmunity in high-income countries. Understanding these phenomena, however, is hampered by a lack of comprehensive, direct immune monitoring in children with differing degrees of microbial exposure. Using mass cytometry, we provide an in-depth profile of the peripheral blood mononuclear cells (PBMCs) of children in regions at the extremes of exposure: the San Francisco Bay Area, USA and an economically poor district of Dhaka, Bangladesh. Despite variability in clinical health, functional characteristics of PBMCs were similar in Bangladeshi and American children at 1 year of age. However, by 2–3 years of age, Bangladeshi children's immune cells often demonstrated altered activation and cytokine production profiles upon stimulation with PMA-ionomycin, with an overall immune trajectory more in line with American adults. Conversely, immune responses in children from the US remained steady. Using principal component analysis, donor location, ethnic background, and cytomegalovirus infection status were found to account for some of the variation identified among samples. Within Bangladeshi 1-year-olds, stunting (as measured by height-for-age z-scores) was found to be associated with IL-8 and TGFβ expression in PMA-ionomycin stimulated samples. Combined, these findings provide important insights into the immune systems of children in high vs. low microbial exposure environments and suggest an important role for IL-8 and TGFβ in mitigating the microbial challenges faced by the Bangladeshi children.
Collapse
Affiliation(s)
- Lisa E Wagar
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Christopher R Bolen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Natalia Sigal
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, United States
| | - Cesar J Lopez Angel
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Leying Guan
- Data Sciences and Statistics, Stanford University, Stanford, CA, United States
| | - Beth D Kirkpatrick
- Department of Microbiology and Molecular Genetics, University of Vermont College of Medicine and Vaccine Testing Center, Burlington, VT, United States
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Robert J Tibshirani
- Data Sciences and Statistics, Stanford University, Stanford, CA, United States
| | - Julie Parsonnet
- Departments of Medicine and of Health Research and Policy, Stanford University, Stanford, CA, United States
| | - William A Petri
- Department of Medicine, Pathology, and Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States.,Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, United States.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
29
|
Vidal-Lletjós S, Andriamihaja M, Blais A, Grauso M, Lepage P, Davila AM, Gaudichon C, Leclerc M, Blachier F, Lan A. Mucosal healing progression after acute colitis in mice. World J Gastroenterol 2019; 25:3572-3589. [PMID: 31367158 PMCID: PMC6658396 DOI: 10.3748/wjg.v25.i27.3572] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/27/2019] [Accepted: 06/08/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mucosal healing has become a therapeutic goal to achieve stable remission in patients with inflammatory bowel diseases. To achieve this objective, overlapping actions of complex cellular processes, such as migration, proliferation, and differentiation, are required. These events are longitudinally and tightly controlled by numerous factors including a wide range of distinct regulatory proteins. However, the sequence of events associated with colon mucosal repair after colitis and the evolution of the luminal content characteristics during this process have been little studied.
AIM To document the evolution of colon mucosal characteristics during mucosal healing using a mouse model with chemically-induced colitis.
METHODS C57BL/6 male mice were given 3.5% dextran sodium sulfate (DSS) in drinking water for 5 d. They were euthanized 2 (day 7), 5 (day 10), 8 (day 13), and 23 (day 28) d after DSS removal. The colonic luminal environment and epithelial repair processes during the inflammatory flare and colitis resolution were analyzed with reference to a non-DSS treated control group, euthanized at day 0. Epithelial repair events were assessed histo-morphologically in combination with functional permeability tests, expression of key inflammatory and repairing factors, and evaluation of colon mucosa-adherent microbiota composition by 16S rRNA sequencing.
RESULTS The maximal intensity of colitis was concomitant with maximal alterations of intestinal barrier function and histological damage associated with goblet cell depletion in colon mucosa. It was recorded 2 d after termination of the DSS-treatment, followed by a progressive return to values similar to those of control mice. Although signs of colitis were severe (inflammatory cell infiltrate, crypt disarray, increased permeability) and associated with colonic luminal alterations (hyperosmolarity, dysbiosis, decrease in short-chain fatty acid content), epithelial healing processes were launched early during the inflammatory flare with increased gene expression of certain key epithelial repair modulators, including transforming growth factor-β, interleukin (Il)-15, Il-22, Il-33, and serum amyloid A. Whereas signs of inflammation progressively diminished, luminal colonic environment alterations and microscopic abnormalities of colon mucosa persisted long after colitis induction.
CONCLUSION This study shows that colon repair can be initiated in the context of inflamed mucosa associated with alterations of the luminal environment and highlights the longitudinal involvement of key modulators.
Collapse
Affiliation(s)
| | | | - Anne Blais
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris 75005, France
| | - Marta Grauso
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris 75005, France
| | - Patricia Lepage
- UMR MICALIS, INRA, Université Paris-Saclay, Jouy-en-Josas 78350, France
| | - Anne-Marie Davila
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris 75005, France
| | - Claire Gaudichon
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris 75005, France
| | - Marion Leclerc
- UMR MICALIS, INRA, Université Paris-Saclay, Jouy-en-Josas 78350, France
| | - François Blachier
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris 75005, France
| | - Annaïg Lan
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, Paris 75005, France
| |
Collapse
|
30
|
Izuddin WI, Loh TC, Foo HL, Samsudin AA, Humam AM. Postbiotic L. plantarum RG14 improves ruminal epithelium growth, immune status and upregulates the intestinal barrier function in post-weaning lambs. Sci Rep 2019; 9:9938. [PMID: 31289291 PMCID: PMC6616331 DOI: 10.1038/s41598-019-46076-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 06/21/2019] [Indexed: 12/18/2022] Open
Abstract
We investigate the effects of postbiotic Lactobacillus plantarum RG14 on gastrointestinal histology, haematology, mucosal IgA concentration, microbial population and mRNA expression related to intestinal mucosal immunity and barrier function. Twelve newly weaned lambs were randomly allocated to two treatment groups; the control group without postbiotic supplementation and postbiotic group with supplementation of 0.9% postbiotic in the diet over a 60-day trial. The improvement of rumen papillae height and width were observed in lambs fed with postbiotics. In contrast, no difference was shown in villi height of duodenum, jejunum and ileum between the two groups. Lambs received postbiotics had a lower concentration of IgA in jejunum but no difference in IgA concentration in serum and mucosal of the rumen, duodenum and ileum. In respect of haematology, postbiotics lowered leukocyte, lymphocyte, basophil, neutrophil and platelets, no significant differences in eosinophil. The increase in of IL-6 mRNA and decrease of IL-1β, IL-10, TNF mRNA were observed in the jejunum of lambs receiving postbiotics. Postbiotics also improved the integrity of the intestinal barrier by the upregulation of TJP-1, CLDN-1 and CLDN-4 mRNA. Postbiotic supplementation derived from L. plantarum RG14 in post-weaning lambs enhance the ruminal papillae growth, immune status and gastrointestinal health.
Collapse
Affiliation(s)
- Wan Ibrahim Izuddin
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Teck Chwen Loh
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
- Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
| | - Hooi Ling Foo
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
| | - Anjas Asmara Samsudin
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Ali Merzza Humam
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Department of Animal Resources, Faculty of Agriculture, University of Baghdad, Baghdad, Iraq
| |
Collapse
|
31
|
Jee J, Jeong SY, Kim HK, Choi SY, Jeong S, Lee J, Ko JS, Kim MS, Kwon M, Yoo J. In vivo
evaluation of scaffolds compatible for colonoid engraftments onto injured mouse colon epithelium. FASEB J 2019; 33:10116-10125. [DOI: 10.1096/fj.201802692rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Joohyun Jee
- Department of MicrobiologyCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
- CHA Organoid Research CenterCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
| | - Sang Yun Jeong
- Department of MicrobiologyCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
- CHA Organoid Research CenterCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
| | - Han Kyung Kim
- Department of MicrobiologyCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
- CHA Organoid Research CenterCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
| | - Seon Young Choi
- Department of MicrobiologyCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
- CHA Organoid Research CenterCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
| | - Sukin Jeong
- Department of MicrobiologyCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
- CHA Organoid Research CenterCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
| | - Joongwoon Lee
- Department of MicrobiologyCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
- CHA Organoid Research CenterCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
| | - Ji Su Ko
- Department of MicrobiologyCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
- CHA Organoid Research CenterCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
| | - Mi Sun Kim
- Department of MicrobiologyCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
- CHA Organoid Research CenterCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
| | - Min‐Soo Kwon
- CHA Organoid Research CenterCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
- Department of PharmacologySchool of MedicineCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
| | - Jongman Yoo
- Department of MicrobiologyCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
- CHA Organoid Research CenterCHA University Seongnam‐si Gyeonggi‐do Republic of Korea
| |
Collapse
|
32
|
Kotla NG, Rana S, Sivaraman G, Sunnapu O, Vemula PK, Pandit A, Rochev Y. Bioresponsive drug delivery systems in intestinal inflammation: State-of-the-art and future perspectives. Adv Drug Deliv Rev 2019; 146:248-266. [PMID: 29966684 DOI: 10.1016/j.addr.2018.06.021] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/27/2018] [Accepted: 06/25/2018] [Indexed: 02/07/2023]
Abstract
Oral colon-specific delivery systems emerged as the main therapeutic cargos by making a significant impact in the field of modern medicine for local drug delivery in intestinal inflammation. The site-specific delivery of therapeutics (aminosalicylates, glucocorticoids, biologics) to the ulcerative mucus tissue can provide prominent advantages in mucosal healing (MH). Attaining gut mucosal healing and anti-fibrosis are main treatment outcomes in inflammatory bowel disease (IBD). The pharmaceutical strategies that are commonly used to achieve a colon-specific drug delivery system include time, pH-dependent polymer coating, prodrug, colonic microbiota-activated delivery systems and a combination of these approaches. Amongst the different approaches reported, the use of biodegradable polysaccharide coated systems holds great promise in delivering drugs to the ulcerative regions. The present review focuses on major physiological gastro-intestinal tract challenges involved in altering the pharmacokinetics of delivery systems, pathophysiology of MH and fibrosis, reported drug-polysaccharide cargos and focusing on conventional to advanced disease responsive delivery strategies, highlighting their limitations and future perspectives in intestinal inflammation therapy.
Collapse
Affiliation(s)
- Niranjan G Kotla
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Newcastle, Galway, Ireland.
| | - Shubhasmin Rana
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Newcastle, Galway, Ireland
| | - Gandhi Sivaraman
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bengaluru 560062, India
| | - Omprakash Sunnapu
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bengaluru 560062, India
| | - Praveen K Vemula
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bengaluru 560062, India
| | - Abhay Pandit
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Newcastle, Galway, Ireland
| | - Yury Rochev
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Newcastle, Galway, Ireland; Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, Moscow, Russian Federation.
| |
Collapse
|
33
|
Histidine and arginine modulate intestinal cell restitution via transforming growth factor-β1. Eur J Pharmacol 2019; 850:35-42. [DOI: 10.1016/j.ejphar.2019.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 02/04/2019] [Accepted: 02/08/2019] [Indexed: 12/18/2022]
|
34
|
Stronati L, Palone F, Negroni A, Colantoni E, Mancuso AB, Cucchiara S, Cesi V, Isoldi S, Vitali R. Dipotassium Glycyrrhizate Improves Intestinal Mucosal Healing by Modulating Extracellular Matrix Remodeling Genes and Restoring Epithelial Barrier Functions. Front Immunol 2019; 10:939. [PMID: 31105713 PMCID: PMC6498413 DOI: 10.3389/fimmu.2019.00939] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/11/2019] [Indexed: 12/16/2022] Open
Abstract
Gut mucosal healing (MH) is considered a key therapeutic target and prognostic parameter in the management of inflammatory bowel disease (IBD). The dipotassium glycyrrhizate (DPG), a salt of the glycoconjugated triterpene glycyrrhizin, has been shown to inhibit the High Mobility Group Box 1 (HMGB1) protein, an allarmin strongly implicated in the pathogenesis of most inflammatory and auto-immune disorders. Here we discuss new insights on how DPG acts on MH comparing the acute phase and the recovery phase from experimental colitis in mice. We found that DPG strongly accelerates MH by differently regulating pro-inflammatory (CXCL1, CXCL3, CXCL5, PTGS2, IL-1β, IL-6, CCL12, CCL7) and wound healing (COL3A1, MMP9, VTN, PLAUR, SERPINE, CSF3, FGF2, FGF7, PLAT, TIMP1) genes as observed only during the recovery phase of colitis. Relevant issue is the identification of extracellular matrix (ECM) remodeling genes, VTN, and PLAUR, as crucial genes to achieve MH during DPG treatment. Furthermore, a noticeable recovery of intestinal epithelial barrier structural organization, wound repair ability, and functionality is observed in two human colorectal adenocarcinoma cell lines exposed to DPG during inflammation. Thus, our study identifies DPG as a potent tool for controlling intestinal inflammation and improving MH.
Collapse
Affiliation(s)
- Laura Stronati
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesca Palone
- Pediatric Gastroenterology and Liver Unit, Department of Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Anna Negroni
- Division of Health Protection Technologies, Territorial and Production Systems Sustainability Department, ENEA, Rome, Italy
| | - Eleonora Colantoni
- Pediatric Gastroenterology and Liver Unit, Department of Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Anna Barbara Mancuso
- Pediatric Gastroenterology and Liver Unit, Department of Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Salvatore Cucchiara
- Pediatric Gastroenterology and Liver Unit, Department of Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Vincenzo Cesi
- Division of Health Protection Technologies, Territorial and Production Systems Sustainability Department, ENEA, Rome, Italy
| | - Sara Isoldi
- Pediatric Gastroenterology and Liver Unit, Department of Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Roberta Vitali
- Division of Health Protection Technologies, Territorial and Production Systems Sustainability Department, ENEA, Rome, Italy
| |
Collapse
|
35
|
Nielsen SD, Purup S, Larsen LB. Effect of Casein Hydrolysates on Intestinal Cell Migration and Their Peptide Profiles by LC-ESI/MS/MS. Foods 2019; 8:foods8030091. [PMID: 30845637 PMCID: PMC6462906 DOI: 10.3390/foods8030091] [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: 01/27/2019] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/18/2022] Open
Abstract
Potential beneficial effects of bioactive peptides derived from casein on epithelial cellular wound healing in the gastrointestinal tract were studied. Bovine casein was digested by a combination of pepsin and pancreatic proteases at different time intervals to represent ranges of duration of gastrointestinal digestion. Intestinal epithelial cells were used as an in vitro model of the small intestine. The effect of casein hydrolysates on cell migration was studied by scratch assay as a model of wound healing. Casein digested by pepsin and pancreatin for 10 to 30 min were found to have a significant stimulatory effect of >40% on cell migration relative to the control. A potential effect of casein gastrointestinal digests on gastro-intestinal wound healing has not previously been reported. The peptide profiles of active as well as inactive casein hydrolysates were characterised by liquid chromatography coupled to ion trap tandem mass spectrometry. By comparison of identified peptides in active and inactive casein hydrolysates, a pool of 11 peptides derived from casein were identified as potential candidates for effects on cell migration. Searching the milk bioactive peptide database (MBPDB) showed that 15 of the identified peptides had known biological functions such as antimicrobial, antioxidant, and immunomodulatory activity.
Collapse
Affiliation(s)
- Søren D Nielsen
- Department of Food Science, Aarhus University, Blichers Allé 20, P.O. Box 50, DK-8830 Tjele, Denmark.
| | - Stig Purup
- Department of Animal Science, Aarhus University, Blichers Allé 20, P.O. Box 50, DK-8830 Tjele, Denmark.
| | - Lotte B Larsen
- Department of Food Science, Aarhus University, Blichers Allé 20, P.O. Box 50, DK-8830 Tjele, Denmark.
| |
Collapse
|
36
|
Benzoic Acid Used as Food and Feed Additives Can Regulate Gut Functions. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5721585. [PMID: 30931328 PMCID: PMC6413358 DOI: 10.1155/2019/5721585] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/27/2019] [Accepted: 02/06/2019] [Indexed: 12/26/2022]
Abstract
As a kind of antibacterial and antifungal preservative, benzoic acid is widely used in foods and feeds. Recently, many studies showed that it could improve the growth and health, which should, at least partially, be derived from the promotion of gut functions, including digestion, absorption, and barrier. Based on the similarity of gut physiology between human and pigs, many relative studies in which piglets and porcine intestinal epithelial cells were used as the models have been done. And the results showed that using appropriate benzoic acid levels might improve gut functions via regulating enzyme activity, redox status, immunity, and microbiota, but excess administration would lead to the damage of gut health through redox status. However, the further mechanisms that some intestinal physiological functions might be regulated are not well understood. The present review will, in detail, summarize the effect of benzoic acid on gut functions.
Collapse
|
37
|
Gershuni VM, Friedman ES. The Microbiome-Host Interaction as a Potential Driver of Anastomotic Leak. Curr Gastroenterol Rep 2019; 21:4. [PMID: 30684121 DOI: 10.1007/s11894-019-0668-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The goal of this paper is to review current literature on the gut microbiome within the context of host response to surgery and subsequent risk of developing complications, particularly anastomotic leak. We provide background on the relationship between host and gut microbiota with description of the role of the intestinal mucus layer as an important regulator of host health. RECENT FINDINGS Despite improvements in surgical technique and adherence to the tenets of creating a tension-free anastomosis with adequate blood flow, the surgical community has been unable to decrease rates of anastomotic leak using the current paradigm. Rather than adhere to empirical strategies of decontamination, it is imperative to focus on the interaction between the human host and the gut microbiota that live within us. The gut microbiome has been found to play a potential role in development of post-operative complications, including but not limited to anastomotic leak. Evidence suggests that peri-operative interventions may have a role in instigating or mitigating the impact of the gut microbiota via disruption of the protective mucus layer, use of multiple medications, and activation of virulence factors. The microbiome plays a potential role in the development of surgical complications and can be modulated by peri-operative interventions. As such, further research into this relationship is urgently needed.
Collapse
Affiliation(s)
- Victoria M Gershuni
- Department of Surgery, Perelman School of Medicine, Hospital of the University of Pennsylvania, 3400 Spruce Street, 4 Maloney, Philadelphia, PA, 19104, USA. .,Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Elliot S Friedman
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
38
|
Zheng XB, Liu HS, Zhang LJ, Liu XH, Zhong XL, Zhou C, Hu T, Wu XR, Hu JC, Lian L, Deng QL, Chen YF, Ke J, He XW, Wu XJ, He XS, Lan P. Engulfment and Cell Motility Protein 1 Protects Against DSS-induced Colonic Injury in Mice via Rac1 Activation. J Crohns Colitis 2019; 13:100-114. [PMID: 30219846 DOI: 10.1093/ecco-jcc/jjy133] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Mucosal healing is an emerging therapeutic goal that could result in clinical remission of inflammatory bowel disease [IBD]. We sought to determine the role of engulfment and cell motility protein 1 [ELMO1] in wound healing in vitro and in vivo and to investigate the underlying pathways. METHODS RNA transcriptome sequencing was performed to detect the expression profiles of mRNA between inflamed tissues and corresponding non-inflamed tissues of IBD patients, followed by Gene Expression Omnibus [GEO] datasets and western blot analysis. The effects of ELMO1 overexpression or knockdown on cell migration and proliferation were determined. The dependence of these effects on Rac1 was assessed using a Rac1 inhibitor [NSC23766] and a Rac1 pull-down assay. We identified the underlying pathways involved by Gene Ontology [GO] analysis. A dextran sulphate sodium [DSS]-induced colitis model was established to evaluate the role of ELMO1 in colonic mucosal healing. RESULTS ELMO1 was upregulated in inflamed tissues compared with corresponding non-inflamed tissues. ELMO1 overexpression increased cell migration in a Rac1-dependent manner. Depletion of ELMO1, or NSC23766 administration, abolished this effect. GO analysis revealed that ELMO1 overexpression preferentially affected pathways involved in cytoskeletal regulation and wound healing, which was demonstrated by enhanced F-actin staining and increased numbers of extending lamellipodia in cells overexpressing ELMO1. In DSS-induced colitis, systemic delivery of pSin-EF2-ELMO1-Pur attenuated colonic inflammation and promoted recovery from colonic injury. The protective effect of ELMO1 was dependent on Rac1 activation. CONCLUSIONS ELMO1 protects against DSS-induced colonic injury in mice through its effect on epithelial migration via Rac1 activation.
Collapse
Affiliation(s)
- Xiao-Bin Zheng
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hua-Shan Liu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Long-Juan Zhang
- Laboratory of Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xuan-Hui Liu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Li Zhong
- Joint Cardiac Surgery Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Chi Zhou
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tuo Hu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xian-Rui Wu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian-Cong Hu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lei Lian
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qi-Ling Deng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu-Feng Chen
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jia Ke
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Wen He
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Jian Wu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Sheng He
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ping Lan
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
39
|
Polysaccharide extracts of Astragalus membranaceus and Atractylodes macrocephala promote intestinal epithelial cell migration by activating the polyamine-mediated K+ channel. Chin J Nat Med 2018; 16:674-682. [DOI: 10.1016/s1875-5364(18)30107-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Indexed: 12/22/2022]
|
40
|
Abstract
The intestinal epithelium is a multicellular interface in close proximity to a dense microbial milieu that is completely renewed every 3-5 days. Pluripotent stem cells reside at the crypt, giving rise to transient amplifying cells that go through continuous steps of proliferation, differentiation and finally anoikis (a form of programmed cell death) while migrating upwards to the villus tip. During these cellular transitions, intestinal epithelial cells (IECs) possess distinct metabolic identities reflected by changes in mitochondrial activity. Mitochondrial function emerges as a key player in cell fate decisions and in coordinating cellular metabolism, immunity, stress responses and apoptosis. Mediators of mitochondrial signalling include molecules such as ATP and reactive oxygen species and interrelate with pathways such as the mitochondrial unfolded protein response (MT-UPR) and AMP kinase signalling, in turn affecting cell cycle progression and stemness. Alterations in mitochondrial function and MT-UPR activation are integral aspects of pathologies, including IBD and cancer. Mitochondrial signalling and concomitant changes in metabolism contribute to intestinal homeostasis and regulate IEC dedifferentiation-differentiation programmes in the context of diseases, suggesting that mitochondrial function as a cellular checkpoint critically contributes to disease outcome. This Review highlights mitochondrial function and MT-UPR signalling in epithelial cell stemness, differentiation and lineage commitment and illustrates mitochondrial function in intestinal diseases.
Collapse
|
41
|
Bibi S, Du M, Zhu MJ. Dietary Red Raspberry Reduces Colorectal Inflammation and Carcinogenic Risk in Mice with Dextran Sulfate Sodium-Induced Colitis. J Nutr 2018; 148:667-674. [PMID: 29897487 PMCID: PMC7263837 DOI: 10.1093/jn/nxy007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/29/2017] [Accepted: 01/09/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Ulcerative colitis causes recurring intestinal mucosal injury and sustained inflammation, increasing the likelihood of colorectal cancer (CRC) development. Dietary red raspberry (RB) is a rich source of phytonutrients known to have anti-inflammatory activity; however, the role of RB on CRC prevention in chronic colitis has not been examined. OBJECTIVE This study examined the effects of dietary RB supplementation on inflammation, epithelium repair, and oncogenic signaling in dextran sulfate sodium (DSS)-induced chronic colitis in mice. METHODS Six-week-old male C57BL/6J mice were fed a control or RB (5% of dry feed weight; n = 12/group) diet for 10 wk. Starting from the fourth week, mice were administered 2 repeated cycles of 1% DSS (7-d DSS treatment plus 14-d recovery) and were monitored daily for disease activity index (DAI) score. Colonic tissues were collected at the end of the study for histochemical, immunohistochemical, and biochemical analysis of inflammation, differentiation and proliferation markers. RESULTS RB supplementation reduced the DAI score and histologic damage (by 38.9%; P ≤ 0.01), expression of inflammatory mediators (by 20-70%; P ≤ 0.01), infiltration of CD4 T cells (by 50%; P ≤ 0.05), and α4β7 integrin and related adhesion molecules (by 33.3%; P ≤ 0.01). Furthermore, RB supplementation facilitated epithelium repair, as evidenced by enhanced goblet cell density, expression of transcription factors including Kruppel-like factor 4 (Klf4) and Hairy and enhancer of split 1 (Hes1), terminal differentiation markers, mucin 2 (Muc2), and intestinal alkaline phosphatase (by 20-200%; P ≤ 0.01). Conversely, proliferating cell nuclear antigen (by 70%; P ≤ 0.01), β-catenin, and signal transducer and activator of transcription 3 (STAT3) signaling (by 19-33%; P ≤ 0.05) were reduced by RB supplementation. In addition, RB supplementation enhanced p53 stability (by 53%) and reduced oncogenic gene expression (by 50-60%). CONCLUSION RB supplementation reduced DAI score and the risk of CRC development during recurring colitis in mice, suggesting that RB is a possible dietary supplement for patients with ulcerative colitis and related gut inflammatory diseases.
Collapse
Affiliation(s)
| | - Min Du
- Department of Animal Science, Washington State University, Pullman, WA
| | - Mei-Jun Zhu
- School of Food Science,Address correspondence to MJZ (e-mail:)
| |
Collapse
|
42
|
|
43
|
Perdijk O, van Splunter M, Savelkoul HFJ, Brugman S, van Neerven RJJ. Cow's Milk and Immune Function in the Respiratory Tract: Potential Mechanisms. Front Immunol 2018; 9:143. [PMID: 29483908 PMCID: PMC5816034 DOI: 10.3389/fimmu.2018.00143] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/17/2018] [Indexed: 12/12/2022] Open
Abstract
During the last decades, the world has witnessed a dramatic increase in allergy prevalence. Epidemiological evidence shows that growing up on a farm is a protective factor, which is partly explained by the consumption of raw cow’s milk. Indeed, recent studies show inverse associations between raw cow’s milk consumption in early life and asthma, hay fever, and rhinitis. A similar association of raw cow’s milk consumption with respiratory tract infections is recently found. In line with these findings, controlled studies in infants with milk components such as lactoferrin, milk fat globule membrane, and colostrum IgG have shown to reduce respiratory infections. However, for ethical reasons, it is not possible to conduct controlled studies with raw cow’s milk in infants, so formal proof is lacking to date. Because viral respiratory tract infections and aeroallergen exposure in children may be causally linked to the development of asthma, it is of interest to investigate whether cow’s milk components can modulate human immune function in the respiratory tract and via which mechanisms. Inhaled allergens and viruses trigger local immune responses in the upper airways in both nasal and oral lymphoid tissue. The components present in raw cow’s milk are able to promote a local microenvironment in which mucosal immune responses are modified and the epithelial barrier is enforced. In addition, such responses may also be triggered in the gut after exposure to allergens and viruses in the nasal cavity that become available in the GI tract after swallowing. However, these immune cells that come into contact with cow’s milk components in the gut must recirculate into the blood and home to the (upper and lower) respiratory tract to regulate immune responses locally. Expression of the tissue homing-associated markers α4β7 and CCR9 or CCR10 on lymphocytes can be influenced by vitamin A and vitamin D3, respectively. Since both vitamins are present in milk, we speculate that raw milk may influence homing of lymphocytes to the upper respiratory tract. This review focuses on potential mechanisms via which cow’s milk or its components can influence immune function in the intestine and the upper respiratory tract. Unraveling these complex mechanisms may contribute to the development of novel dietary approaches in allergy and asthma prevention.
Collapse
Affiliation(s)
- Olaf Perdijk
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Marloes van Splunter
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Huub F J Savelkoul
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Sylvia Brugman
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - R J Joost van Neerven
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands.,FrieslandCampina, Amersfoort, Netherlands
| |
Collapse
|
44
|
Zhang M, Xu C, Liu D, Han MK, Wang L, Merlin D. Oral Delivery of Nanoparticles Loaded With Ginger Active Compound, 6-Shogaol, Attenuates Ulcerative Colitis and Promotes Wound Healing in a Murine Model of Ulcerative Colitis. J Crohns Colitis 2018; 12:217-229. [PMID: 28961808 PMCID: PMC5881712 DOI: 10.1093/ecco-jcc/jjx115] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/15/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Oral drug delivery is the most attractive pathway for ulcerative colitis [UC] therapy, since it has many advantages. However, this strategy has encountered many challenges, including the instability of drugs in the gastrointestinal tract [GT], low targeting of disease tissues, and severe adverse effects. Nanoparticles capable of colitis tissue-targeted delivery and site-specific drug release may offer a unique and therapeutically effective system that addresses these formidable challenges. METHODS We used a versatile single-step surface-functionalising technique to prepare PLGA/PLA-PEG-FA nanoparticles loaded with the ginger active compound, 6-shogaol [NPs-PEG-FA/6-shogaol]. The therapeutic efficacy of NPs-PEG-FA/6-shogaol was evaluated in the well-established mouse model of dextran sulphate sodium [DSS]-induced colitis. RESULTS NPs-PEG-FA exhibited very good biocompatibility both in vitro and in vivo. Subsequent cellular uptake experiments demonstrated that NPs-PEG-FA could undergo efficient receptor-mediated uptake by colon-26 cells and activated Raw 264.7 macrophage cells. In vivo, oral administration of NPs-PEG-FA/6-shogaol encapsulated in a hydrogel system [chitosan/alginate] significantly alleviated colitis symptoms and accelerated colitis wound repair in DSS-treated mice by regulating the expression levels of pro-inflammatory [TNF-α, IL-6, IL-1β, and iNOS] and anti-inflammatory [Nrf-2 and HO-1] factors. CONCLUSIONS Our study demonstrates a convenient, orally administered 6-shogaol drug delivery system that effectively targets colitis tissue, alleviates colitis symptoms, and accelerates colitis wound repair. This system may represent a promising therapeutic approach for treating inflammatory bowel disease [IBD].
Collapse
Affiliation(s)
- Mingzhen Zhang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA,Corresponding author: Mingzhen Zhang, Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30302, USA. Tel.: +1 [404] 413 3597; fax: +1 [404] 413 3580;
| | - Changlong Xu
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA,Department of Gastroenterology, 2nd Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, P. R. China
| | - Dandan Liu
- Department of Chemistry, Georgia State University, Atlanta, GA, USA
| | - Moon Kwon Han
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Lixin Wang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA,Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Didier Merlin
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA,Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| |
Collapse
|
45
|
Kangawa Y, Yoshida T, Yonezawa Y, Maruyama K, Hayashi SM, Shibutani M. Suppression of epithelial restitution using an inhibitor against Rho-associated coiled-coil containing protein kinase aggravates colitis through reduced epithelial expression of A-kinase anchor protein 13. ACTA ACUST UNITED AC 2017; 69:557-563. [PMID: 28535907 DOI: 10.1016/j.etp.2017.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 04/30/2017] [Accepted: 05/02/2017] [Indexed: 12/11/2022]
Abstract
In the gastrointestinal tract, the immediate healing response to mucosal damage is critical to sustain mucosal homeostasis. The migration of surrounding epithelial cells to cover the denuded area without proliferation is termed restitution, followed by early reparation of the damage. In this study, we determined the role of A-kinase anchor protein 13 (AKAP13) in mice with dextran sulphate sodium (DSS)-induced colitis upon mucosal injury and restitution, and investigated whether inhibition of Rho-associated coiled-coil containing protein kinase (ROCK), downstream effector of AKAP13, affects these mucosal responses. BALB/c mice were challenged with 4% or 2% DSS in their drinking water for up to 8 or 16days, respectively. During this period, mice received subcutaneous injections of fasudil hydrochloride hydrate (FH, 10mg/kg, twice per day), an inhibitor of phosphorylation of ROCK. In immunohistochemistry, AKAP13 was highly expressed in the mucosal epithelium prior to DSS-induced mucosal injury, and also expressed in ulcer-covering non-proliferative epithelium, which corresponded to restituted epithelial cells. Coadministration of FH increased serum amyloid A levels and histopathological scores for mucosal injury, as compared with the DSS group. The effects were associated with a decrease in gene expression of Akap13 in the mucosal tissue and the inhibition of restitution rata (the length of restituted epithelial cells per ulcer). These results suggested that AKAP13 and ROCK are involved in mucosal response at early injury and restitution during healing in DSS-induced colitis in mice.
Collapse
Affiliation(s)
- Yumi Kangawa
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Pharmacokinetics and Safety Department, Drug Research Center, Kaken Pharmaceutical Co., Ltd., 301 Gensuke, Fujieda, Shizuoka 426-8646, Japan
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Yutaka Yonezawa
- Pharmacokinetics and Safety Department, Drug Research Center, Kaken Pharmaceutical Co., Ltd., 301 Gensuke, Fujieda, Shizuoka 426-8646, Japan; United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Kiyoshi Maruyama
- Pharmacokinetics and Safety Department, Drug Research Center, Kaken Pharmaceutical Co., Ltd., 301 Gensuke, Fujieda, Shizuoka 426-8646, Japan
| | - Shim-Mo Hayashi
- Global Scientific and Regulatory Affairs, San-Ei Gen F. F. I., Inc., 1-1-11 Sanwa-cho, Toyonaka, Osaka 561-8588, Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| |
Collapse
|
46
|
Matsui T, Ichikawa H, Fujita T, Takagi T, Osada-Oka M, Minamiyama Y. Histidine deficiency attenuates cell viability in rat intestinal epithelial cells by apoptosis via mitochondrial dysfunction. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2017. [DOI: 10.1016/j.jnim.2017.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
47
|
Cinque B, La Torre C, Lombardi F, Palumbo P, Evtoski Z, Jr Santini S, Falone S, Cimini A, Amicarelli F, Cifone MG. VSL#3 probiotic differently influences IEC-6 intestinal epithelial cell status and function. J Cell Physiol 2017; 232:3530-3539. [PMID: 28109129 DOI: 10.1002/jcp.25814] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 01/12/2017] [Accepted: 01/19/2017] [Indexed: 12/17/2022]
Abstract
The data here reported introduce the wound-healing assay as a tool for testing probiotics aimed at protecting gastrointestinal mucosal surfaces and to verify the consistency of their manufacturing. At the scope, we compared the in vitro effects of two multi-strain high concentration formulations both commercialized under the same brand VSL#3 but sourced from different production sites (USA and Italy) on a non-transformed small-intestinal epithelial cell line, IEC-6. The effects on cellular morphology, viability, migration, and H2 O2 -induced damage, were assessed before and after the treatment with both VSL#3 formulations. While the USA-sourced product ("USA-made") VSL#3 did not affect monolayer morphology and cellular density, the addition of bacteria from the Italy-derived product ("Italy-made") VSL#3 caused clear morphological cell damage and strongly reduced cellularity. The treatment with "USA-made" lysate led to a higher rate of wounded monolayer healing, while the addition of "Italy-made" bacterial lysate did not influence the closure rate as compared to untreated cells. While lysates from "USA-made" VSL#3 clearly enhanced the formation of elongated and aligned stress fibers, "Italy-made" lysates had not similar effect. "USA-made" lysate was able to cause a total inhibition of H2 O2 -induced cytotoxic effect whereas "Italy-made" VSL#3 lysate was unable to protect IEC-6 cells from H2 O2 -induced damage. ROS generation was also differently influenced, thus supporting the hypotesis of a protective action of "USA-made" VSL#3 lysates, as well as the idea that "Italy-made" formulation was unable to prevent significantly the H2 O2 -induced oxidative stress.
Collapse
Affiliation(s)
- Benedetta Cinque
- Division of Public Health, Department of Life, Health and Environmental Sciences, University of L'Aquila, Coppito, L'Aquila, Italy
| | - Cristina La Torre
- Division of Public Health, Department of Life, Health and Environmental Sciences, University of L'Aquila, Coppito, L'Aquila, Italy
| | - Francesca Lombardi
- Division of Public Health, Department of Life, Health and Environmental Sciences, University of L'Aquila, Coppito, L'Aquila, Italy
| | - Paola Palumbo
- Division of Molecular Medicine, Department of Life, Health and Environmental Sciences, University of L'Aquila, Coppito, L'Aquila, Italy
| | - Zoran Evtoski
- Division of Molecular Medicine, Department of Life, Health and Environmental Sciences, University of L'Aquila, Coppito, L'Aquila, Italy
| | - Silvano Jr Santini
- Division of Biological and Biotechnological Sciences, Department of Life, Health and Environmental Sciences, University of L'Aquila, Coppito, L'Aquila, Italy
| | - Stefano Falone
- Division of Biological and Biotechnological Sciences, Department of Life, Health and Environmental Sciences, University of L'Aquila, Coppito, L'Aquila, Italy
| | - Annamaria Cimini
- Division of Biological and Biotechnological Sciences, Department of Life, Health and Environmental Sciences, University of L'Aquila, Coppito, L'Aquila, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, Pennsylvania
- National Institute for Nuclear Physics (INFN), Gran Sasso National Laboratory (LNGS), Assergi, Italy
| | - Fernanda Amicarelli
- Division of Biological and Biotechnological Sciences, Department of Life, Health and Environmental Sciences, University of L'Aquila, Coppito, L'Aquila, Italy
- Institute of Translational Pharmacology (IFT) -National Research Council (CNR), L'Aquila, Italy
| | - Maria Grazia Cifone
- Division of Molecular Medicine, Department of Life, Health and Environmental Sciences, University of L'Aquila, Coppito, L'Aquila, Italy
| |
Collapse
|
48
|
Suppression of calpain expression by NSAIDs is associated with inhibition of cell migration in rat duodenum. Toxicology 2017; 383:1-12. [PMID: 28342779 DOI: 10.1016/j.tox.2017.03.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/20/2017] [Accepted: 03/20/2017] [Indexed: 12/27/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used for the alleviation of pain and inflammation, but these drugs are also associated with a suite of negative side effects. Gastrointestinal (GI) toxicity is particularly concerning since it affects an estimated 70% of individuals taking NSAIDs routinely, and evidence suggests the majority of toxicity is occurring in the small intestine. Traditionally, NSAID-induced GI toxicity has been associated with indiscriminate inhibition of cyclooxygenase isoforms, but other mechanisms, including inhibition of cell migration, intestinal restitution, and wound healing, are likely to contribute to toxicity. Previous efforts demonstrated that treatment of cultured intestinal epithelial cells (IEC) with NSAIDs inhibits expression and activity of calpain proteases, but the effects of specific inhibition of calpain expression in vitro or the effects of NSAIDs on intestinal cell migration in vivo remain to be determined. Accordingly, we examined the effect of suppression of calpain protease expression with siRNA on cell migration in cultured IECs and evaluated the effects of NSAID treatment on epithelial cell migration and calpain protease expression in rat duodenum. Our results show that calpain siRNA inhibits protease expression and slows migration in cultured IECs. Additionally, NSAID treatment of rats slowed migration up the villus axis and suppressed calpain expression in duodenal epithelial cells. Our results are supportive of the hypothesis that suppression of calpain expression leading to slowing of cell migration is a potential mechanism through which NSAIDs cause GI toxicity.
Collapse
|
49
|
Xiao K, Cao S, Jiao L, Song Z, Lu J, Hu C. TGF-β1 protects intestinal integrity and influences Smads and MAPK signal pathways in IPEC-J2 after TNF-α challenge. Innate Immun 2017; 23:276-284. [PMID: 28142299 DOI: 10.1177/1753425917690815] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The aim of this study was to investigate the protective effects of TGF-β1 on intestinal epithelial barrier, as well as canonical Smad and MAPK signal pathways involved in these protection processes by a IPEC-J2 model stimulated with TNF-α. IPEC-J2 monolayers were treated without or with TNF-α in the absence or presence of TGF-β1. The results showed that TGF-β1 pretreatment ameliorated TNF-α-induced intestinal epithelial barrier disturbances as indicated by decrease of transepithelial electrical resistance (TER) and increase of paracellular permeability. TGF-β1 also dramatically alleviated TNF-α-induced alteration of TJ proteins ZO-1 and occludin. Moreover, TGF-β1 pretreatment increased TβRII protein expression in IPEC-J2 monolayers challenged with TNF-α. In addition, a significant increase of Smad4 and Smad7 mRNA was also observed in the TGF-β1 pretreatment after TNF-α challenge compared with the control group. Furthermore, TGF-β1 pretreatment enhanced smad2 protein activation. These results indicated that the canonical Smad signaling pathway was activated by TGF-β1 pretreatment. Finally, TGF-β1 pretreatment decreased the ratios of the phosphorylated to total JNK and p38 (p-JNK/JNK and p-p38/p38) and increased the ratio of ERK (p-ERK/ERK). Anti-TGF-β1 Abs reduced these TGF-β1 effects. These results indicated that TGF-β1 protects intestinal integrity and influences Smad and MAPK signal pathways in IPEC-J2 after TNF-α challenge.
Collapse
Affiliation(s)
- Kan Xiao
- Animal Science College, Zhejiang University; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
| | - Shuting Cao
- Animal Science College, Zhejiang University; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
| | - Lefei Jiao
- Animal Science College, Zhejiang University; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
| | - Zehe Song
- Animal Science College, Zhejiang University; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
| | - Jianjun Lu
- Animal Science College, Zhejiang University; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
| | - Caihong Hu
- Animal Science College, Zhejiang University; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
| |
Collapse
|
50
|
Miyoshi H, VanDussen KL, Malvin NP, Ryu SH, Wang Y, Sonnek NM, Lai CW, Stappenbeck TS. Prostaglandin E2 promotes intestinal repair through an adaptive cellular response of the epithelium. EMBO J 2016; 36:5-24. [PMID: 27797821 DOI: 10.15252/embj.201694660] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 12/18/2022] Open
Abstract
Adaptive cellular responses are often required during wound repair. Following disruption of the intestinal epithelium, wound-associated epithelial (WAE) cells form the initial barrier over the wound. Our goal was to determine the critical factor that promotes WAE cell differentiation. Using an adaptation of our in vitro primary epithelial cell culture system, we found that prostaglandin E2 (PGE2) signaling through one of its receptors, Ptger4, was sufficient to drive a differentiation state morphologically and transcriptionally similar to in vivo WAE cells. WAE cell differentiation was a permanent state and dominant over enterocyte differentiation in plasticity experiments. WAE cell differentiation was triggered by nuclear β-catenin signaling independent of canonical Wnt signaling. Creation of WAE cells via the PGE2-Ptger4 pathway was required in vivo, as mice with loss of Ptger4 in the intestinal epithelium did not produce WAE cells and exhibited impaired wound repair. Our results demonstrate a mechanism by which WAE cells are formed by PGE2 and suggest a process of adaptive cellular reprogramming of the intestinal epithelium that occurs to ensure proper repair to injury.
Collapse
Affiliation(s)
- Hiroyuki Miyoshi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kelli L VanDussen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicole P Malvin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Stacy H Ryu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yi Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Naomi M Sonnek
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chin-Wen Lai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|