1
|
Garrett S, Zhang Y, Xia Y, Sun J. Intestinal Epithelial Axin1 Deficiency Protects Against Colitis via Altered Gut Microbiota. ENGINEERING (BEIJING, CHINA) 2024; 35:241-256. [PMID: 38911180 PMCID: PMC11192507 DOI: 10.1016/j.eng.2023.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Intestinal homeostasis is maintained by specialized host cells and the gut microbiota. Wnt/β-catenin signaling is essential for gastrointestinal development and homeostasis, and its dysregulation has been implicated in inflammation and colorectal cancer. Axin1 negatively regulates activated Wnt/β-catenin signaling, but little is known regarding its role in regulating host-microbial interactions in health and disease. Here, we aim to demonstrate that intestinal Axin1 determines gut homeostasis and host response to inflammation. Axin1 expression was analyzed in human inflammatory bowel disease datasets. To explore the effects and mechanism of intestinal Axin1 in regulating intestinal homeostasis and colitis, we generated new mouse models with Axin1 conditional knockout in intestinal epithelial cell (IEC; Axin1 ΔIEC) and Paneth cell (PC; Axin1 ΔPC) to compare with control (Axin1 LoxP; LoxP: locus of X-over, P1) mice. We found increased Axin1 expression in the colonic epithelium of human inflammatory bowel disease (IBD). Axin1 ΔIEC mice exhibited altered goblet cell spatial distribution, PC morphology, reduced lysozyme expression, and enriched Akkermansia muciniphila (A. muciniphila). The absence of intestinal epithelial and PC Axin1 decreased susceptibility to dextran sulfate sodium (DSS)-induced colitis in vivo. Axin1 ΔIEC and Axin1 ΔPC mice became more susceptible to DSS-colitis after cohousing with control mice. Treatment with A. muciniphila reduced DSS-colitis severity. Antibiotic treatment did not change the IEC proliferation in the Axin1 Loxp mice. However, the intestinal proliferative cells in Axin1 ΔIEC mice with antibiotic treatment were reduced compared with those in Axin1 ΔIEC mice without treatment. These data suggest non-colitogenic effects driven by the gut microbiome. In conclusion, we found that the loss of intestinal Axin1 protects against colitis, likely driven by epithelial Axin1 and Axin1-associated A. muciniphila. Our study demonstrates a novel role of Axin1 in mediating intestinal homeostasis and the microbiota. Further mechanistic studies using specific Axin1 mutations elucidating how Axin1 modulates the microbiome and host inflammatory response will provide new therapeutic strategies for human IBD.
Collapse
Affiliation(s)
- Shari Garrett
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Yongguo Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Yinglin Xia
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Cancer Center, University of Illinois Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
2
|
Richter FC, Friedrich M, Kampschulte N, Piletic K, Alsaleh G, Zummach R, Hecker J, Pohin M, Ilott N, Guschina I, Wideman SK, Johnson E, Borsa M, Hahn P, Morriseau C, Hammock BD, Schipper HS, Edwards CM, Zechner R, Siegmund B, Weidinger C, Schebb NH, Powrie F, Simon AK. Adipocyte autophagy limits gut inflammation by controlling oxylipin and IL-10. EMBO J 2023; 42:e112202. [PMID: 36795015 PMCID: PMC10015370 DOI: 10.15252/embj.2022112202] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 02/17/2023] Open
Abstract
Lipids play a major role in inflammatory diseases by altering inflammatory cell functions, either through their function as energy substrates or as lipid mediators such as oxylipins. Autophagy, a lysosomal degradation pathway that limits inflammation, is known to impact on lipid availability, however, whether this controls inflammation remains unexplored. We found that upon intestinal inflammation visceral adipocytes upregulate autophagy and that adipocyte-specific loss of the autophagy gene Atg7 exacerbates inflammation. While autophagy decreased lipolytic release of free fatty acids, loss of the major lipolytic enzyme Pnpla2/Atgl in adipocytes did not alter intestinal inflammation, ruling out free fatty acids as anti-inflammatory energy substrates. Instead, Atg7-deficient adipose tissues exhibited an oxylipin imbalance, driven through an NRF2-mediated upregulation of Ephx1. This shift reduced secretion of IL-10 from adipose tissues, which was dependent on the cytochrome P450-EPHX pathway, and lowered circulating levels of IL-10 to exacerbate intestinal inflammation. These results suggest an underappreciated fat-gut crosstalk through an autophagy-dependent regulation of anti-inflammatory oxylipins via the cytochrome P450-EPHX pathway, indicating a protective effect of adipose tissues for distant inflammation.
Collapse
Affiliation(s)
| | - Matthias Friedrich
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Nadja Kampschulte
- Faculty of Mathematics and Natural SciencesUniversity of WuppertalWuppertalGermany
| | - Klara Piletic
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Ghada Alsaleh
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | | | - Julia Hecker
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
- Department of Gastroenterology, Infectious Diseases and RheumatologyCampus Benjamin FranklinBerlinGermany
| | - Mathilde Pohin
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Nicholas Ilott
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | | | - Sarah Karin Wideman
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Errin Johnson
- The Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Mariana Borsa
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Paula Hahn
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Christophe Morriseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer CenterUniversity of CaliforniaDavisCAUSA
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer CenterUniversity of CaliforniaDavisCAUSA
| | - Henk Simon Schipper
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
- Center for Translational ImmunologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Claire M Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research CentreUniversity of OxfordOxfordUK
- Nuffield Department of Surgical Sciences, Botnar Research CentreUniversity of OxfordOxfordUK
| | - Rudolf Zechner
- Institute of Molecular BiosciencesUniversity of GrazGrazAustria
| | - Britta Siegmund
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
- Department of Gastroenterology, Infectious Diseases and RheumatologyCampus Benjamin FranklinBerlinGermany
| | - Carl Weidinger
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
- Department of Gastroenterology, Infectious Diseases and RheumatologyCampus Benjamin FranklinBerlinGermany
| | - Nils Helge Schebb
- Faculty of Mathematics and Natural SciencesUniversity of WuppertalWuppertalGermany
| | - Fiona Powrie
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Anna Katharina Simon
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
- Max Delbrück CenterBerlinGermany
| |
Collapse
|
3
|
Peters LA, Friedman JR, Stojmirovic A, Hagen J, Houten S, Dodatko T, Amaro MP, Restrepo P, Chai Z, Rodrigo Mora J, Raymond HA, Curran M, Dobrin R, Das A, Xiong H, Schadt EE, Argmann C, Losic B. A temporal classifier predicts histopathology state and parses acute-chronic phasing in inflammatory bowel disease patients. Commun Biol 2023; 6:95. [PMID: 36694043 PMCID: PMC9873918 DOI: 10.1038/s42003-023-04469-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 01/12/2023] [Indexed: 01/25/2023] Open
Abstract
Previous studies have conducted time course characterization of murine colitis models through transcriptional profiling of differential expression. We characterize the transcriptional landscape of acute and chronic models of dextran sodium sulfate (DSS) and adoptive transfer (AT) colitis to derive temporal gene expression and splicing signatures in blood and colonic tissue in order to capture dynamics of colitis remission and relapse. We identify sub networks of patient-derived causal networks that are enriched in these temporal signatures to distinguish acute and chronic disease components within the broader molecular landscape of IBD. The interaction between the DSS phenotype and chronological time-point naturally defines parsimonious temporal gene expression and splicing signatures associated with acute and chronic phases disease (as opposed to ordinary time-specific differential expression/splicing). We show these expression and splicing signatures are largely orthogonal, i.e. affect different genetic bodies, and that using machine learning, signatures are predictive of histopathological measures from both blood and intestinal data in murine colitis models as well as an independent cohort of IBD patients. Through access to longitudinal multi-scale profiling from disease tissue in IBD patient cohorts, we can apply this machine learning pipeline to generation of direct patient temporal multimodal regulatory signatures for prediction of histopathological outcomes.
Collapse
Affiliation(s)
- Lauren A. Peters
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Joshua R. Friedman
- grid.476706.40000 0004 7647 0615Spark Therapeutics, Philadelphia, PA USA ,grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, Spring House, Philadelphia, PA USA
| | - Aleksandar Stojmirovic
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, Spring House, Philadelphia, PA USA
| | - Jacob Hagen
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Sander Houten
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Tetyana Dodatko
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Mariana P. Amaro
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Paula Restrepo
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Zhi Chai
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - J. Rodrigo Mora
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, Spring House, Philadelphia, PA USA ,grid.479574.c0000 0004 1791 3172Moderna, Cambridge, MA USA
| | - Holly A. Raymond
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, Spring House, Philadelphia, PA USA
| | - Mark Curran
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, Spring House, Philadelphia, PA USA
| | - Radu Dobrin
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, Spring House, Philadelphia, PA USA ,Pathos AI, Berwyn, PA USA
| | - Anuk Das
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, Spring House, Philadelphia, PA USA
| | - Huabao Xiong
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Eric E. Schadt
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Carmen Argmann
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Bojan Losic
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.511203.4Present Address: Guardant Health, Redwood City, CA USA
| |
Collapse
|
4
|
Hakeem AN, Kamal MM, Tawfiq RA, Abdelrahman BA, Hammam OA, Elmazar MM, El-Khatib AS, Attia YM. Elafibranor modulates ileal macrophage polarization to restore intestinal integrity in NASH: Potential crosstalk between ileal IL-10/STAT3 and hepatic TLR4/NF-κB axes. Biomed Pharmacother 2023; 157:114050. [PMID: 36462310 DOI: 10.1016/j.biopha.2022.114050] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/17/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
Experimental and clinical evidence implicate disrupted gut barrier integrity in provoking innate immune responses, specifically macrophages, towards the progression of non-alcoholic steatohepatitis (NASH). Peroxisome proliferator-activated receptors (PPARs), a subset of the nuclear receptor superfamily, act to fine-tune several metabolic and inflammatory processes implicated in NASH. As such, the current study was carried out to decipher the potential role of dual PPAR α/δ activation using elafibranor (ELA) on ileal macrophage polarization (MP) and its likely impact on the liver in a NASH setting. To achieve this aim, an in vitro NASH model using fat-laden HepG2 cells was first used to validate the impact of ELA on hepatic fat accumulation. Afterwards, ELA was used in a combined model of dietary NASH and chronic colitis analogous to the clinical presentation of NASH parallel with intestinal barrier dysfunction. ELA mitigated fat accumulation in vitro as evidenced by Oil Red-O staining and curbed triglyceride levels. Additionally, ELA restored the expression of tight junctional proteins, claudin-1 and occludin, along with decreasing intestinal permeability and inflammation skewing ileal macrophages towards the M2 phenotype, as indicated by boosted arginase-1 (Arg1) and curtailed inducible nitric oxide synthase (iNOS) expression levels. These changes were aligned with a modulation in hepatic toll-like receptor-4 (TLR4)/nuclear factor kappa B (NF-κB) along with ileal interleukin-10 (IL-10)/signal transducer and activator of transcription-3 (STAT3) axes. Overall, the present findings suggest that the dual PPAR α/δ agonist, ELA, may drive MP in the ileum towards the M2 phenotype improving intestinal integrity towards alleviating NASH.
Collapse
Affiliation(s)
- Andrew N Hakeem
- Department of Pharmacology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt; The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Mohamed M Kamal
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Rasha A Tawfiq
- Department of Pharmacology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt; The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Basma A Abdelrahman
- Department of Pharmacology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt; The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Olfat A Hammam
- Department of Pathology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mohamed M Elmazar
- Department of Pharmacology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt; The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Aiman S El-Khatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Yasmeen M Attia
- Department of Pharmacology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt; The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt.
| |
Collapse
|
5
|
Melo FJ, Pinto-Lopes P, Estevinho MM, Magro F. The Role of Dipeptidyl Peptidase 4 as a Therapeutic Target and Serum Biomarker in Inflammatory Bowel Disease: A Systematic Review. Inflamm Bowel Dis 2021; 27:1153-1165. [PMID: 33295607 DOI: 10.1093/ibd/izaa324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The roles dipeptidyl peptidase 4 (DPP4), aminopeptidase N (APN), and their substrates in autoimmune diseases are being increasingly recognized. However, their significance in inflammatory bowel diseases (IBD) is not entirely understood. This systematic review aims to discuss the pathophysiological processes related to these ectopeptidases while comparing findings from preclinical and clinical settings. METHODS This review was conducted according to the PRISMA guidelines. We performed a literature search in PubMed, SCOPUS, and Web of Science to identify all reports from inception until February 2020. The search included validated animal models of intestinal inflammation and studies in IBD patients. Quality assessment was performed using SYRCLE's risk of bias tool and CASP qualitative and cohort checklists. RESULTS From the 45 included studies, 36 were performed in animal models and 12 in humans (3 reports included both). Overall, the methodological quality of preclinical studies was acceptable. In animal models, DPP4 and APN inhibition significantly improved intestinal inflammation.Glucagon-like peptide (GLP)-1 and GLP-2 analogs and GLP-2-relase-inducing drugs also showed significant benefits in recovery from inflammatory damage. A nonsignificant trend toward disease remission with the GLP-2 analog teduglutide was observed in the sole interventional human study. All human studies reported an inverse correlation between soluble DPP4/CD26 levels and disease severity, in accordance with the proposal of DPP4 as a biomarker for IBD. CONCLUSIONS The use of DPP4 inhibitors and analogs of its substrates has clear benefits in the treatment of experimentally induced intestinal inflammation. Further research is warranted to validate their potential diagnostic and therapeutic applications in IBD patients.
Collapse
Affiliation(s)
- Francisco Jorge Melo
- Department of Biomedicine, Unity of Pharmacology and Therapeutics, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Pedro Pinto-Lopes
- Department of Biomedicine, Unity of Pharmacology and Therapeutics, Faculty of Medicine of the University of Porto, Porto, Portugal.,Department of Internal Medicine, Tâmega e Sousa Hospital Center, Padre Américo Hospital, Penafiel, Portugal
| | - Maria Manuela Estevinho
- Department of Biomedicine, Unity of Pharmacology and Therapeutics, Faculty of Medicine of the University of Porto, Porto, Portugal.,Department of Gastroenterology, Vila Nova de Gaia/Espinho Hospital Center, Vila Nova de Gaia, Portugal
| | - Fernando Magro
- Department of Biomedicine, Unity of Pharmacology and Therapeutics, Faculty of Medicine of the University of Porto, Porto, Portugal.,Unit of Clinical Pharmacology, São João Hospital Center, Porto, Portugal
| |
Collapse
|
6
|
Yazbeck R, Jaenisch SE, Abbott CA. Dipeptidyl peptidase 4 inhibitors: Applications in innate immunity? Biochem Pharmacol 2021; 188:114517. [PMID: 33722535 PMCID: PMC7954778 DOI: 10.1016/j.bcp.2021.114517] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 12/25/2022]
Abstract
Dipeptidyl peptidase (DPP)-4 inhibitors are a class of orally available, small molecule inhibitors that prolong the insulinotropic activity of the incretin hormone glucagon-like peptide-1 (GLP-1) and are highly effective for the treatment of Type-2 diabetes. DPP4 can also cleave several immunoregulatory peptides including chemokines. Emerging evidence continues to implicate DPP4 inhibitors as immunomodulators, with recent findings suggesting DPP4 inhibitors modify specific aspects of innate immunity. This review summarises recent insights into how DPP4 inhibitors could be implicated in endothelial, neutrophil and monocyte/macrophage mediated immunity. Additionally, this review highlights additional avenues of research with DPP4 inhibitors in the context of the COVID-19 pandemic.
Collapse
Affiliation(s)
- R Yazbeck
- College of Medicine and Public Health & Flinders Health and Medical Research Institute, Flinders University, Adelaide, Australia; College of Science and Engineering, Flinders University, Adelaide, Australia.
| | - S E Jaenisch
- College of Medicine and Public Health & Flinders Health and Medical Research Institute, Flinders University, Adelaide, Australia; College of Science and Engineering, Flinders University, Adelaide, Australia.
| | - C A Abbott
- College of Medicine and Public Health & Flinders Health and Medical Research Institute, Flinders University, Adelaide, Australia; College of Science and Engineering, Flinders University, Adelaide, Australia.
| |
Collapse
|
7
|
Ai G, Huang Z, Cheng J, Xie J, Zeng H, Liu Y, Li Y, Huang X, Chen J, Su Z. Gut Microbiota-Mediated Transformation of Coptisine Into a Novel Metabolite 8-Oxocoptisine: Insight Into Its Superior Anti-Colitis Effect. Front Pharmacol 2021; 12:639020. [PMID: 33859564 PMCID: PMC8042337 DOI: 10.3389/fphar.2021.639020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/15/2021] [Indexed: 01/22/2023] Open
Abstract
Coptisine (COP) is a bioactive isoquinoline alkaloid derived from Coptis Chinemsis Franch, which is traditionally applied for the management of colitis. However, the blood concentration of COP was extremely low, and its gut microbiota-mediated metabolites were thought to contribute to its prominent bioactivities. To comparatively elucidate the protective effect and underlying mechanism of COP and its novel gut microbiota metabolite (8-oxocoptisine, OCOP) against colitis, we used dextran sulfate sodium (DSS) to induce colitis in mice. Clinical symptoms, microscopic alternation, immune-inflammatory parameters for colitis were estimated. The results indicated that OCOP dramatically ameliorated disease activity index (DAI), the shortening of colon length and colonic histopathological deteriorations. OCOP treatment also suppressed the mRNA expression and release of inflammatory mediators (TGF-β, TNF-α, IL-6, IL-18, IL-1β and IFN-γ) and elevated the transcriptional and translational levels of anti-inflammatory cytokine (IL-10) as well as the mRNA expression levels of adhesion molecules (ICAM-1 and VCAM-1). Besides, the activation of NF-κB pathway and NLRP3 inflammasome was markedly inhibited by OCOP. Furthermore, OCOP displayed superior anti-colitis effect to COP, and was similar to MSZ with much smaller dosage. Taken together, the protective effect of OCOP against DSS-induced colitis might be intimately related to inhibition of NF-κB pathway and NLRP3 inflammasome. And the findings indicated that OCOP might have greater potential than COP to be further exploited as a promising candidate in the treatment of colitis.
Collapse
Affiliation(s)
- Gaoxiang Ai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziwei Huang
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Juanjuan Cheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhui Xie
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huifang Zeng
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoqi Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
8
|
Wang D, Sun M, Zhang Y, Chen Z, Zang S, Li G, Li G, Clark AR, Huang J, Si L. Enhanced therapeutic efficacy of a novel colon-specific nanosystem loading emodin on DSS-induced experimental colitis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 78:153293. [PMID: 32777486 DOI: 10.1016/j.phymed.2020.153293] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) is an intricate enteric disease with a rising incidence that is closely related to mucosa-barrier destruction, gut dysbacteriosis, and immune disorders. Emodin (1,3,8-trihydroxy-6-methyl-9,10-anthraquinone, EMO) is a natural anthraquinone derivative that occurs in many Polygonaceae plants. Its multiple pharmacological effects, including antioxidant, immune-suppressive, and anti-bacteria activities, make it a promising treatment option for UC. However, its poor solubility, extensive absorption, and metabolism in the upper gastrointestinal tract may compromise its anti-colitis effects. PURPOSE EMO was loaded in a colon-targeted delivery system using multifunctional biomedical materials and the enhanced anti-colitis effect involving mucosa reconstruction was investigated in this study. METHODS EMO-loaded Poly (DL-lactide-co-glycolide)/EudragitⓇ S100/montmorillonite nanoparticles (EMO/PSM NPs) were prepared by a versatile single-step assembly approach. The colon-specific release behavior was characterized in vitro and in vivo, and the anti-colitis effect was evaluated in dextran sulfate sodium (DSS)-induced acute colitis in mice by weight loss, disease activity index (DAI) score, colon length, histological changes, and colitis biomarkers. The integrity of the intestinal mucosal barrier was evaluated through transwell co-culture model in vitro and serum zonulin-related tight junctions and mucin2 (MUC2) in vivo. RESULTS EMO/PSM NPs with a desirable hydrodynamic diameter (~ 235 nm) and negative zeta potential (~ -31 mV) could prevent the premature drug release (< 4% in the first 6 h in vitro) in the upper gastrointestinal tract (GIT) and boost retention in the lower GIT and inflamed colon mucosa in vivo. Compared to free EMO-treatment of different doses in UC mice, the NPs could enhance the remedial efficacy of EMO in DAI decline, histological remission, and regulation of colitis indicators, such as myeloperoxidase (MPO), nitric oxide (NO), and glutathione (GSH). The inflammatory factors including induced nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), TNF-α, and IL-1β were suppressed by EMO/PSM NPs at both mRNA and protein levels. The obtained NPs could also promote the regeneration of the mucosal barrier via reduced fluorescein isothiocyanate (FITC)-dextran leakage in the transwell co-culture model and decreased serum zonulin levels, which was demonstrated to be associated with the upregulated tight junctions (TJs)-related proteins (claudin-2, occludin, and zo-1) and MUC2 at mRNA level. Moreover, the NPs could contribute to attenuating the liver injury caused by free EMO under excessive immune inflammation. CONCLUSION Our results demonstrated that EMO/PSM NPs could specifically release EMO in the diseased colon, and effectively enhance the anti-colitis effects of EMO related to intestinal barrier improvement. It can be considered as a novel potential alternative for oral colon-targeted UC therapy by increasing therapeutic efficacy and reducing side-effects.
Collapse
Affiliation(s)
- Dan Wang
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College of Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, PR China
| | - Minghui Sun
- Department of Pharmaceutics, Affiliated Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, PR China
| | - Ying Zhang
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College of Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, PR China
| | - Zehong Chen
- Department of Pharmacy, Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, 1227 Jiefang Road, Wuhan 430030, PR China
| | - Shuya Zang
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College of Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, PR China
| | - Genyun Li
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College of Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, PR China
| | - Gao Li
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College of Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, PR China
| | - Andrew R Clark
- School of Medicine, Indiana University, 975W. Walnut St, IB 008, Indianapolis, IN 46202, USA
| | - Jiangeng Huang
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College of Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, PR China.
| | - Luqin Si
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College of Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, PR China.
| |
Collapse
|
9
|
Dipeptidyl peptidase 4 inhibitor sitagliptin protected against dextran sulfate sodium-induced experimental colitis by potentiating the action of GLP-2. Acta Pharmacol Sin 2020; 41:1446-1456. [PMID: 32398684 DOI: 10.1038/s41401-020-0413-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
Dipeptidyl peptidase 4 (DPP4), a ubiquitously expressed protease that cleaves off the N-terminal dipeptide from proline and alanine on the penultimate position, has important roles in many physiological processes. In the present study, experimental colitis was induced in mice receiving 3% dextran sulfate sodium (DSS) in drinking water. We found that mice with DSS-induced colitis had significantly increased intestinal DPP activity and decreased serum DPP activity, suggesting a probable correlation of DPP4 with experimental colitis. Then, we investigated whether sitagliptin, a specific DPP4 inhibitor could protect against DSS-induced colitis. We showed that oral administration of single dose of sitagliptin (30 mg/kg) on D7 remarkably inhibited DPP enzyme activity in both serum and intestine of DSS-induced colitic mice. Repeated administration of sitagliptin (10, 30 mg/kg, bid, from D0 to D8) significantly ameliorated DSS-induced colitis, including reduction of disease activity index (DAI) and body weight loss, improvement of histological score and colon length. Sitagliptin administration dose-dependently increased plasma concentrations of active form of GLP-1 and colonic expression of GLP-2R. Co-administration of GLP-2R antagonist GLP-23-33 (500 μg/kg, bid, sc) abolished the protective effects of sitagliptin in DSS-induced colitic mice. Moreover, sitagliptin administration significantly decreased the ratio of apoptotic cells and increased the ratio of proliferative cells in colon epithelium of DSS-induced colitic mice, and this effect was also blocked by GLP-23-33. Taken together, our results demonstrate that sitagliptin could attenuate DSS-induced experimental colitis and the effects can be attributed to the enhancement of GLP-2 action and the subsequent protective effects on intestinal barrier by inhibiting epithelial cells apoptosis and promoting their proliferation. These findings suggest sitagliptin as a novel therapeutic approach for the treatment of ulcerative colitis.
Collapse
|
10
|
Kong X, Wang X, Qin Y, Han J. Effects of sunset yellow on proliferation and differentiation of intestinal epithelial cells in murine intestinal organoids. J Appl Toxicol 2020; 41:953-963. [DOI: 10.1002/jat.4080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 08/14/2020] [Accepted: 09/02/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Xiunan Kong
- School of Food Science and Biotechnology Zhejiang Gongshang University Hangzhou China
| | - Xiu Wang
- School of Food Science and Biotechnology Zhejiang Gongshang University Hangzhou China
| | - Yumei Qin
- School of Food Science and Biotechnology Zhejiang Gongshang University Hangzhou China
| | - Jianzhong Han
- School of Food Science and Biotechnology Zhejiang Gongshang University Hangzhou China
| |
Collapse
|
11
|
Cortez V, Boyd DF, Crawford JC, Sharp B, Livingston B, Rowe HM, Davis A, Alsallaq R, Robinson CG, Vogel P, Rosch JW, Margolis E, Thomas PG, Schultz-Cherry S. Astrovirus infects actively secreting goblet cells and alters the gut mucus barrier. Nat Commun 2020; 11:2097. [PMID: 32350281 PMCID: PMC7190700 DOI: 10.1038/s41467-020-15999-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 04/08/2020] [Indexed: 12/18/2022] Open
Abstract
Astroviruses are a global cause of pediatric diarrhea, but they are largely understudied, and it is unclear how and where they replicate in the gut. Using an in vivo model, here we report that murine astrovirus preferentially infects actively secreting small intestinal goblet cells, specialized epithelial cells that maintain the mucus barrier. Consequently, virus infection alters mucus production, leading to an increase in mucus-associated bacteria and resistance to enteropathogenic E. coli colonization. These studies establish the main target cell type and region of the gut for productive murine astrovirus infection. They further define a mechanism by which an enteric virus can regulate the mucus barrier, induce functional changes to commensal microbial communities, and alter host susceptibility to pathogenic bacteria.
Collapse
Affiliation(s)
- Valerie Cortez
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - David F Boyd
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Bridgett Sharp
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Brandi Livingston
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hannah M Rowe
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Amy Davis
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ramzi Alsallaq
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Camenzind G Robinson
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peter Vogel
- Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jason W Rosch
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Elisa Margolis
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
12
|
Kopecki Z, Yang G, Treloar S, Mashtoub S, Howarth GS, Cummins AG, Cowin AJ. Flightless I exacerbation of inflammatory responses contributes to increased colonic damage in a mouse model of dextran sulphate sodium-induced ulcerative colitis. Sci Rep 2019; 9:12792. [PMID: 31488864 PMCID: PMC6728368 DOI: 10.1038/s41598-019-49129-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/15/2019] [Indexed: 12/13/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by cytokine driven inflammation that disrupts the mucosa and impedes intestinal structure and functions. Flightless I (Flii) is an immuno-modulatory protein is a member of the gelsolin family of actin-remodelling proteins that regulates cellular and inflammatory processes critical in tissue repair. Here we investigated its involvement in UC and show that Flii is significantly elevated in colonic tissues of patients with inflammatory bowel disease. Using an acute murine model of colitis, we characterised the contribution of Flii to UC using mice with low (Flii+/-), normal (Flii+/+) and high Flii (FliiTg/Tg). High levels of Flii resulted in significantly elevated disease severity index scores, increased rectal bleeding and degree of colon shortening whereas, low Flii expression decreased disease severity, reduced tissue inflammation and improved clinical indicators of UC. Mice with high levels of Flii had significantly increased histological disease severity and elevated mucosal damage with significantly increased inflammatory cell infiltrate and significantly higher levels of TNF-α, IFN-γ, IL-5 and IL-13 pro-inflammatory cytokines. Additionally, Flii overexpression resulted in decreased β-catenin levels, inhibited Wnt/β-catenin signalling and impaired regeneration of colonic crypts. These studies suggest that high levels of Flii, as is observed in patients with UC, may adversely affect mucosal healing via mechanisms involving Th1 and Th2 mediated tissue inflammation and Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Z Kopecki
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia.
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia.
| | - G Yang
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia
| | - S Treloar
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, Adelaide, South Australia, Australia
| | - S Mashtoub
- Department of Gastroenterology, Women's and Children's Hospital, North Adelaide, South Australia, Australia
- Discipline of Physiology, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - G S Howarth
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - A G Cummins
- Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville South, Adelaide, South Australia, Australia
| | - A J Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
13
|
Valero MS, González M, Ramón-Gimenez M, Andrade PB, Moreo E, Les F, Fernandes F, Gómez-Rincón C, Berzosa C, García de Jalón JA, Arruebo MP, Plaza MÁ, Köhler R, López V, Valentão P, Castro M. Jasonia glutinosa (L.) DC., a traditional herbal medicine, reduces inflammation, oxidative stress and protects the intestinal barrier in a murine model of colitis. Inflammopharmacology 2019; 28:1717-1734. [PMID: 31410747 DOI: 10.1007/s10787-019-00626-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/29/2019] [Indexed: 01/03/2023]
Abstract
Jasonia glutinosa (L.) DC., known as rock tea (RT), is traditionally used in Spain as a digestive due to its beneficial properties in bowel disorders. The pharmacological nature of these properties has not been established yet. The aim of this work was to evaluate the therapeutic utility of RT in experimental colitis and to identify chemical constituents with anti-inflammatory and/or anti-oxidative properties. RT extract was prepared with ethanol in a Soxhlet apparatus and analysed by HPLC-DAD. Superoxide radical scavenging properties, xanthine oxidase and lipoxygenase (5-LOX) inhibitory activity, and capability to lower nitric oxide (NO) and tumor necrosis factor α (TNF-α) levels were measured in cell-free and cell-based assays. In the 2.5%-dextran-sodium sulphate (DSS) injury-repair model of ulcerative colitis (UC), mice were daily treated with sulfasalazine (SSZ, as reference drug, 100 mg/kg bw), RT (5, 25 and 50 mg/kg bw, p.o.), or vehicle over 20 days. Colitis was scored daily. Colon samples were examined macroscopically and histopathologically. Protein levels of myeloperoxidase (MPO), interleukins 6, and 10 (IL-6, IL-10), inducible NO synthase (iNOS), and cyclooxygenase-2 (COX-2) were studied as markers of oxidative stress and inflammatory activity. The integrity of the apical epithelial layer was assessed by immunofluorescence staining of zonula ocludens-1 (ZO-1). Finally, intestinal contractility was also evaluated by isometric myography. Fifteen phenolic compounds and three pigments were identified and quantified, of which caffeoylquinic acids, and the flavonoid, quercetin-3-O-galactoside, were the most abundant. RT extract significantly scavenged superoxide radicals, inhibited 5-LOX activity, and lowered NO and TNF-α levels. DSS-treated mice receiving RT scored clinically lower than controls during the first 3 days of DSS treatment and during the recovery period. SSZ was less effective than RT. Anatomical and histological examination of colon samples revealed that RT significantly prevented colon shortening, increased colon thickness, and lowered the macroscopic damage score. RT also significantly prevented the increase of MPO activity, IL-6 levels, iNOS and COX-2 expression, the loss of ZO-1 apical expression, and normalized contractility disturbances. In conclusion, daily administration of RT showed therapeutic properties in the DSS-model of UC. The benefits of RT can likely be attributed to its anti-inflammatory and antioxidant phenolic and flavonoid constituents.
Collapse
Affiliation(s)
- Marta Sofía Valero
- Departamento de Farmacología y Fisiología, Universidad de Zaragoza, Saragossa, Spain.
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Saragossa, Spain.
- Instituto Agroalimentario de Aragón, IA2, Universidad de Zaragoza-CITA, Saragossa, Spain.
| | - Mateo González
- Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Zaragoza, Spain
| | - Mariano Ramón-Gimenez
- Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Zaragoza, Spain
| | - Paula B Andrade
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira no. 228, 4050-313, Porto, Portugal
| | - Eduardo Moreo
- Grupo de genética de micobacterias. Dpto. Microbiología, Medicina Preventiva y Salud Pública, Universidad de Zaragoza, Saragossa, Spain
| | - Francisco Les
- Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Zaragoza, Spain
| | - Fátima Fernandes
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira no. 228, 4050-313, Porto, Portugal
| | - Carlota Gómez-Rincón
- Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Zaragoza, Spain
| | - César Berzosa
- Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Zaragoza, Spain
| | | | - Mª Pilar Arruebo
- Departamento de Farmacología y Fisiología, Universidad de Zaragoza, Saragossa, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Saragossa, Spain
- Instituto Agroalimentario de Aragón, IA2, Universidad de Zaragoza-CITA, Saragossa, Spain
| | - Miguel Ángel Plaza
- Departamento de Farmacología y Fisiología, Universidad de Zaragoza, Saragossa, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Saragossa, Spain
- Instituto Agroalimentario de Aragón, IA2, Universidad de Zaragoza-CITA, Saragossa, Spain
| | - Ralf Köhler
- Aragon Agency for Research and Development (ARAID), Saragossa, Spain
| | - Víctor López
- Instituto Agroalimentario de Aragón, IA2, Universidad de Zaragoza-CITA, Saragossa, Spain.
- Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Zaragoza, Spain.
| | - Patricia Valentão
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira no. 228, 4050-313, Porto, Portugal
| | - Marta Castro
- Departamento de Farmacología y Fisiología, Universidad de Zaragoza, Saragossa, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Saragossa, Spain
- Instituto Agroalimentario de Aragón, IA2, Universidad de Zaragoza-CITA, Saragossa, Spain
| |
Collapse
|
14
|
Vázquez-Arreguín K, Bensard C, Schell JC, Swanson E, Chen X, Rutter J, Tantin D. Oct1/Pou2f1 is selectively required for colon regeneration and regulates colon malignancy. PLoS Genet 2019; 15:e1007687. [PMID: 31059499 PMCID: PMC6522070 DOI: 10.1371/journal.pgen.1007687] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 05/16/2019] [Accepted: 04/16/2019] [Indexed: 12/22/2022] Open
Abstract
The transcription factor Oct1/Pou2f1 promotes poised gene expression states, mitotic stability, glycolytic metabolism and other characteristics of stem cell potency. To determine the effect of Oct1 loss on stem cell maintenance and malignancy, we deleted Oct1 in two different mouse gut stem cell compartments. Oct1 deletion preserved homeostasis in vivo and the ability to establish organoids in vitro, but blocked the ability to recover from treatment with dextran sodium sulfate, and the ability to maintain organoids after passage. In a chemical model of colon cancer, loss of Oct1 in the colon severely restricted tumorigenicity. In contrast, loss of one or both Oct1 alleles progressively increased tumor burden in a colon cancer model driven by loss-of-heterozygosity of the tumor suppressor gene Apc. The different outcomes are consistent with prior findings that Oct1 promotes mitotic stability, and consistent with differentially expressed genes between the two models. Oct1 ChIPseq using HCT116 colon carcinoma cells identifies target genes associated with mitotic stability, metabolism, stress response and malignancy. This set of gene targets overlaps significantly with genes differentially expressed in the two tumor models. These results reveal that Oct1 is selectively required for recovery after colon damage, and that Oct1 has potent effects in colon malignancy, with outcome (pro-oncogenic or tumor suppressive) dictated by tumor etiology.
Collapse
Affiliation(s)
- Karina Vázquez-Arreguín
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Claire Bensard
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - John C. Schell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Eric Swanson
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Xinjian Chen
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Jared Rutter
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Howard Hughes Medical Institute, Salt Lake City, Utah, United States of America
| | - Dean Tantin
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| |
Collapse
|
15
|
Omidi-Ardali H, Lorigooini Z, Soltani A, Balali-Dehkordi S, Amini-Khoei H. Inflammatory responses bridge comorbid cardiac disorder in experimental model of IBD induced by DSS: protective effect of the trigonelline. Inflammopharmacology 2019; 27:1265-1273. [PMID: 30924005 DOI: 10.1007/s10787-019-00581-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 03/06/2019] [Indexed: 12/11/2022]
Abstract
Pathogenesis of the inflammatory bowel disease (IBD) involves the combination of immunological and inflammatory factors. IBD is associated with several extra-intestinal manifestations. The exact underlying bridge between the probable cardiac diseases in IBD patients is undetermined. Trigonelline is an alkaloid with several therapeutic potential properties. In this study, we aimed to assess the probable underlying mechanisms of this comorbidity as well as protective effect of trigonelline focusing inflammatory response and oxidative state in mouse model of colitis. Dextran sodium sulfate (DSS) was used for induction of colitis in mice. Trigonelline (10, 50 and 100 mg/kg) was administrated via intraperitoneal rout (i.p.) for 14 continuous days. Heart, intestine and serum samples were taken for assessment of total antioxidant capacity, malondialdehyde (MDA), gene expressions of inflammatory markers including tumor necrosis factor alpha (Tnf-α), interleukin 1-beta (Il/1β), toll- like receptor 4 (Tlr4) as well as for evaluation of histopathological alterations. Results demonstrated that trigonelline effectively attenuated the cellular/molecular and histopathological adverse effects of colitis in the intestine and heart tissues. In this regards, we found that trigonelline decreased the MDA level, attenuated the expression of Tnf-α, Il/1β and, Tlr4 as well as modulated the histopathological alterations in the intestine. Furthermore, trigonelline increased the antioxidant capacity in the related experimental groups. We concluded that IBD (colitis) is associated with comorbid cellular/molecular modifications in the heart and for the first time, we found that trigonelline has potential therapeutic effects (at least partially) to attenuate the cardiac manifestations of the colitis.
Collapse
Affiliation(s)
- Hossein Omidi-Ardali
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amin Soltani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shima Balali-Dehkordi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
16
|
Yazbeck R, Jaenisch S, Squire M, Abbott CA, Parkinson-Lawrence E, Brooks DA, Butler RN. Development of a 13C Stable Isotope Assay for Dipeptidyl Peptidase-4 Enzyme Activity A New Breath Test for Dipeptidyl Peptidase Activity. Sci Rep 2019; 9:4906. [PMID: 30894647 PMCID: PMC6427020 DOI: 10.1038/s41598-019-41375-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 03/07/2019] [Indexed: 01/15/2023] Open
Abstract
Dipeptidyl peptidase-4 inhibitors (DPP4i) are a class of orally available, small molecule inhibitors for the management of Type-II diabetes. A rapid, real-time, functional breath test for DPP4 enzyme activity could help to define DPP4i efficacy in patients that are refractory to treatment. We aimed to develop a selective, non-invasive, stable-isotope 13C-breath test for DPP4. In vitro experiments were performed using high (Caco-2) and low (HeLa) DPP4 expressing cells. DPP gene expression was determined in cell lines by qRT-PCR. A DPP4 selective 13C-tripeptide was added to cells in the presence and absence of the DPP4 inhibitor Sitagliptin. Gas samples were collected from the cell headspace and 13CO2 content quantified by isotope ratio mass spectrometry (IRMS). DPP4 was highly expressed in Caco-2 cells compared to HeLa cells and using the 13C-tripeptide, we detected a high 13CO2 signal from Caco2 cells. Addition of Sitaglitpin to Caco2 cells significantly inhibited this 13CO2 signal. 13C-assay DPP4 activity correlated positively with the enzyme activity detected using a colorimetric substrate. We have developed a selective, non-invasive, 13C-assay for DPP4 that could have broad translational applications in diabetes and gastrointestinal disease.
Collapse
Affiliation(s)
- Roger Yazbeck
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia. .,Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, South Australia, Australia.
| | - Simone Jaenisch
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.,Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, South Australia, Australia
| | - Michelle Squire
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Catherine A Abbott
- Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, South Australia, Australia.,College of Science and Engineering, Flinders University, Adelaide, South Australia, Australia
| | - Emma Parkinson-Lawrence
- School of Pharmacy and Medical Science, University of South Australia Cancer Research Institute, Adelaide, South Australia, Australia
| | - Douglas A Brooks
- School of Pharmacy and Medical Science, University of South Australia Cancer Research Institute, Adelaide, South Australia, Australia.,School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Ross N Butler
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.,School of Pharmacy and Medical Science, University of South Australia Cancer Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
17
|
Fuchslocher Chico J, Falk-Paulsen M, Luzius A, Saggau C, Ruder B, Bolik J, Schmidt-Arras D, Linkermann A, Becker C, Rosenstiel P, Rose-John S, Adam D. The enhanced susceptibility of ADAM-17 hypomorphic mice to DSS-induced colitis is not ameliorated by loss of RIPK3, revealing an unexpected function of ADAM-17 in necroptosis. Oncotarget 2018; 9:12941-12958. [PMID: 29560122 PMCID: PMC5849186 DOI: 10.18632/oncotarget.24410] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 01/25/2018] [Indexed: 12/27/2022] Open
Abstract
The disintegrin metalloprotease ADAM17 has a critical role in intestinal inflammation and regeneration in mice, as illustrated by the dramatically increased susceptibility of ADAM17 hypomorphic (ADAM17ex/ex) mice to dextran sulfate sodium (DSS)-induced colitis. Similarly, necroptosis has been implicated in inflammatory responses in the intestine. In this study, we have investigated the contribution of necroptosis to ADAM17-regulated intestinal inflammation in vivo by crossing ADAM17ex/ex mice with mice that lack the necroptotic core protein RIPK3. Despite the loss of RIPK3, ADAM17ex/ex/RIPK3−/− mice showed the same increased susceptibility as ADAM17ex/ex mice in both acute and chronic models of DSS-induced colitis. Mice of both genotypes revealed comparable results with regard to weight loss, disease activity index and colitis-associated changes of inner organs. Histopathological analyses confirmed similar tissue destruction, loss of barrier integrity, immune cell infiltration, and cell death; serum analyses revealed similar levels of the pro-inflammatory cytokine KC. Resolving these unexpected findings, ADAM17ex/ex mice did not show phosphorylation of RIPK3 and its necroptotic interaction partner MLKL during DSS-induced colitis, although both proteins were clearly expressed. Consistent with these findings, murine embryonic fibroblasts derived from ADAM17ex/ex mice were protected from tumor necrosis factor (TNF)-induced necroptosis and failed to show phosphorylation of MLKL and RIPK3 after induction of necroptosis by TNF, revealing a novel, undescribed role of the protease ADAM17 in necroptosis.
Collapse
Affiliation(s)
| | - Maren Falk-Paulsen
- Institut für Klinische Molekularbiologie, Christian-Albrechts-Universität zu Kiel, 24105 Kiel, Germany
| | - Anne Luzius
- Institut für Klinische Molekularbiologie, Christian-Albrechts-Universität zu Kiel, 24105 Kiel, Germany
| | - Carina Saggau
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, 24105 Kiel, Germany
| | - Barbara Ruder
- Medizinische Klinik 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Julia Bolik
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24118 Kiel, Germany
| | - Dirk Schmidt-Arras
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24118 Kiel, Germany
| | - Andreas Linkermann
- Medizinische Klinik und Poliklinik III, Universitätsklinikum Carl Gustav Carus, 01307 Dresden, Germany
| | - Christoph Becker
- Medizinische Klinik 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Philip Rosenstiel
- Institut für Klinische Molekularbiologie, Christian-Albrechts-Universität zu Kiel, 24105 Kiel, Germany
| | - Stefan Rose-John
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24118 Kiel, Germany
| | - Dieter Adam
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, 24105 Kiel, Germany
| |
Collapse
|
18
|
Rabbi MF, Eissa N, Munyaka PM, Kermarrec L, Elgazzar O, Khafipour E, Bernstein CN, Ghia JE. Reactivation of Intestinal Inflammation Is Suppressed by Catestatin in a Murine Model of Colitis via M1 Macrophages and Not the Gut Microbiota. Front Immunol 2017; 8:985. [PMID: 28871257 PMCID: PMC5566981 DOI: 10.3389/fimmu.2017.00985] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/02/2017] [Indexed: 12/22/2022] Open
Abstract
While there is growing awareness of a relationship between chromogranin-A (CHGA) and susceptibility to inflammatory conditions, the role of human catestatin [(hCTS); CHGA352–67] in the natural history of established inflammatory bowel disease is not known. Recently, using two different experimental models, we demonstrated that hCTS-treated mice develop less severe acute colitis. We have also shown the implication of the macrophages in this effect. The aims of this study were to determine (1) whether hCTS treatment could attenuate the reactivation of inflammation in adult mice with previously established chronic colitis; (2) whether this effect is mediated through macrophages or the gut microbiota. Quiescent colitis was induced in 7–8-week-old C57BL6 mice using four cycles (2–4%) of dextran sulfate sodium. hCTS (1.5 mg/kg/day) treatment or vehicle started 2 days before the last induction of colitis and continuing for 7 days. At sacrifice, macro- and microscopic scores were determined. Colonic pro-inflammatory cytokines [interleukin (IL)-6, IL-1β, and TNF- α], anti-inflammatory cytokines (IL-10, TGF- β), classically activated (M1) (iNOS, Mcp1), and alternatively activated (M2) (Ym1, Arg1) macrophages markers were studied using ELISA and/or RT-qPCR. In vitro, peritoneal macrophages isolated from naïve mice and treated with hCTS (10−5 M, 12 h) were exposed to either lipopolysaccharide (100 ng/ml, 12 h) to polarize M1 macrophages or to IL-4/IL-13 (20 ng/ml) to polarize M2 macrophages. M1/M2 macrophage markers along with cytokine gene expression were determined using RT-qPCR. Feces and mucosa-associated microbiota (MAM) samples were collected, and the V4 region of 16 s rRNA was sequenced. Micro- and macroscopic scores, colonic IL-6, IL-1β, TNF- α, and M1 macrophages markers were significantly decreased in the hCTS-treated group. Treatment did not have any effect on colonic IL-10, TGF-β, and M2 markers nor modified the bacterial richness, diversity, or the major phyla in colitic fecal and MAM samples. In vitro, pro-inflammatory cytokines levels, as well as their gene expression, were significantly reduced in hCTS-treated M1 macrophages. hCTS treatment did not affect M2 macrophage markers. These findings suggest that hCTS treatment attenuates the severity of inflammatory relapse through the modulation of the M1 macrophages and the release of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Mohammad F Rabbi
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada.,The Children Research Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Nour Eissa
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada.,The Children Research Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Peris M Munyaka
- Department of Animal Sciences, University of Manitoba, Winnipeg, MB, Canada
| | | | - Omar Elgazzar
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Ehsan Khafipour
- Department of Animal Sciences, University of Manitoba, Winnipeg, MB, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Charles N Bernstein
- Department of Internal Medicine, Section of Gastroenterology, University of Manitoba, Winnipeg, MB, Canada.,Inflammatory Bowel Disease Clinical and Research Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Jean Eric Ghia
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada.,The Children Research Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada.,Department of Internal Medicine, Section of Gastroenterology, University of Manitoba, Winnipeg, MB, Canada.,Inflammatory Bowel Disease Clinical and Research Centre, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
19
|
The Anti-Inflammatory Effect and Intestinal Barrier Protection of HU210 Differentially Depend on TLR4 Signaling in Dextran Sulfate Sodium-Induced Murine Colitis. Dig Dis Sci 2017; 62:372-386. [PMID: 27995407 DOI: 10.1007/s10620-016-4404-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 11/30/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) is strongly associated with inflammation and intestinal barrier disorder. The nonselective cannabinoid receptor agonist HU210 has been shown to ameliorate inflamed colon in colitis, but its effects on intestinal barrier function and extraintestinal inflammation are unclear. AIMS To investigate the effects and the underlying mechanism of HU210 action on the UC in relation to a role of TLR4 and MAP kinase signaling. METHODS Wild-type (WT) and TLR4 knockout (Tlr4 -/-) mice were exposed to 4% dextran sulfate sodium (DSS) for 7 days. The effects of HU210 on inflammation and intestinal barrier were explored. RESULTS Upon DSS challenge, mice suffered from bloody stool, colon shortening, intestinal mucosa edema, pro-inflammatory cytokine increase and intestinal barrier destruction with goblet cell depletion, increased intestinal microflora accompanied with elevated plasma lipopolysaccharide, reduced mRNA expression of the intestinal tight junction proteins, and abnormal ratio of CD4+/CD8+ T cells in the intestinal Peyer's patches. Pro-inflammatory cytokines in the plasma and the lung, as well as pulmonary myeloperoxidase activity, indicators of extraintestinal inflammation were increased. Protein expression of p38α and pp38 was up-regulated in the colon of WT mice. Tlr4 -/- mice showed milder colitis. HU210 reversed the intestinal barrier changes in both strains of mice, but alleviated inflammation only in WT mice. CONCLUSIONS Our study indicates that in experimental colitis, HU210 displays a protective effect on the intestinal barrier function independently of the TLR4 signaling pathway; however, in the extraintestinal tissues, the anti-inflammatory action seems through affecting TLR4-mediated p38 mitogen-activated protein kinase pathway.
Collapse
|
20
|
Schewe M, Franken PF, Sacchetti A, Schmitt M, Joosten R, Böttcher R, van Royen ME, Jeammet L, Payré C, Scott PM, Webb NR, Gelb M, Cormier RT, Lambeau G, Fodde R. Secreted Phospholipases A2 Are Intestinal Stem Cell Niche Factors with Distinct Roles in Homeostasis, Inflammation, and Cancer. Cell Stem Cell 2016; 19:38-51. [PMID: 27292189 DOI: 10.1016/j.stem.2016.05.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 02/10/2016] [Accepted: 05/19/2016] [Indexed: 01/04/2023]
Abstract
The intestinal stem cell niche provides cues that actively maintain gut homeostasis. Dysregulation of these cues may compromise intestinal regeneration upon tissue insult and/or promote tumor growth. Here, we identify secreted phospholipases A2 (sPLA2s) as stem cell niche factors with context-dependent functions in the digestive tract. We show that group IIA sPLA2, a known genetic modifier of mouse intestinal tumorigenesis, is expressed by Paneth cells in the small intestine, while group X sPLA2 is expressed by Paneth/goblet-like cells in the colon. During homeostasis, group IIA/X sPLA2s inhibit Wnt signaling through intracellular activation of Yap1. However, upon inflammation they are secreted into the intestinal lumen, where they promote prostaglandin synthesis and Wnt signaling. Genetic ablation of both sPLA2s improves recovery from inflammation but increases colon cancer susceptibility due to release of their homeostatic Wnt-inhibitory role. This "trade-off" effect suggests sPLA2s have important functions as genetic modifiers of inflammation and colon cancer.
Collapse
Affiliation(s)
- Matthias Schewe
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam 3000CA, The Netherlands
| | - Patrick F Franken
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam 3000CA, The Netherlands
| | - Andrea Sacchetti
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam 3000CA, The Netherlands
| | - Mark Schmitt
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam 3000CA, The Netherlands
| | - Rosalie Joosten
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam 3000CA, The Netherlands
| | - René Böttcher
- Department of Urology, Erasmus MC Cancer Institute, Rotterdam 3000CA, The Netherlands
| | - Martin E van Royen
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam 3000CA, The Netherlands; Erasmus Optical Imaging Centre, Erasmus MC Cancer Institute, Rotterdam 3000CA, The Netherlands
| | - Louise Jeammet
- Institute of Molecular and Cellular Pharmacology, Centre National de la Recherche Scientifique and University of Nice Sophia Antipolis, Valbonne 06560, France
| | - Christine Payré
- Institute of Molecular and Cellular Pharmacology, Centre National de la Recherche Scientifique and University of Nice Sophia Antipolis, Valbonne 06560, France
| | - Patricia M Scott
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN 55812-3031, USA
| | - Nancy R Webb
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40506-9983, USA
| | - Michael Gelb
- Department of Chemistry, University of Washington, Seattle, WA 98195-1700, USA
| | - Robert T Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN 55812-3031, USA
| | - Gérard Lambeau
- Institute of Molecular and Cellular Pharmacology, Centre National de la Recherche Scientifique and University of Nice Sophia Antipolis, Valbonne 06560, France
| | - Riccardo Fodde
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam 3000CA, The Netherlands.
| |
Collapse
|
21
|
Wagner L, Klemann C, Stephan M, von Hörsten S. Unravelling the immunological roles of dipeptidyl peptidase 4 (DPP4) activity and/or structure homologue (DASH) proteins. Clin Exp Immunol 2016; 184:265-83. [PMID: 26671446 DOI: 10.1111/cei.12757] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/01/2015] [Accepted: 12/14/2015] [Indexed: 12/31/2022] Open
Abstract
Dipeptidyl peptidase (DPP) 4 (CD26, DPP4) is a multi-functional protein involved in T cell activation by co-stimulation via its association with adenosine deaminase (ADA), caveolin-1, CARMA-1, CD45, mannose-6-phosphate/insulin growth factor-II receptor (M6P/IGFII-R) and C-X-C motif receptor 4 (CXC-R4). The proline-specific dipeptidyl peptidase also modulates the bioactivity of several chemokines. However, a number of enzymes displaying either DPP4-like activities or representing structural homologues have been discovered in the past two decades and are referred to as DPP4 activity and/or structure homologue (DASH) proteins. Apart from DPP4, DASH proteins include fibroblast activation protein alpha (FAP), DPP8, DPP9, DPP4-like protein 1 (DPL1, DPP6, DPPX L, DPPX S), DPP4-like protein 2 (DPL2, DPP10) from the DPP4-gene family S9b and structurally unrelated enzyme DPP2, displaying DPP4-like activity. In contrast, DPP6 and DPP10 lack enzymatic DPP4-like activity. These DASH proteins play important roles in the immune system involving quiescence (DPP2), proliferation (DPP8/DPP9), antigen-presenting (DPP9), co-stimulation (DPP4), T cell activation (DPP4), signal transduction (DPP4, DPP8 and DPP9), differentiation (DPP4, DPP8) and tissue remodelling (DPP4, FAP). Thus, they are involved in many pathophysiological processes and have therefore been proposed for potential biomarkers or even drug targets in various cancers (DPP4 and FAP) and inflammatory diseases (DPP4, DPP8/DPP9). However, they also pose the challenge of drug selectivity concerning other DASH members for better efficacy and/or avoidance of unwanted side effects. Therefore, this review unravels the complex roles of DASH proteins in immunology.
Collapse
Affiliation(s)
- L Wagner
- Deutschsprachige Selbsthilfegruppe für Alkaptonurie (DSAKU) e.V, Stuttgart.,Department for Experimental Therapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - C Klemann
- Centre of Paediatric Surgery.,Centre for Paediatrics and Adolescent Medicine
| | - M Stephan
- Clinic for Psychosomatics and Psychotherapy, Hannover Medical School, Hannover
| | - S von Hörsten
- Department for Experimental Therapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
22
|
Runtsch MC, Hu R, Alexander M, Wallace J, Kagele D, Petersen C, Valentine JF, Welker NC, Bronner MP, Chen X, Smith DP, Ajami NJ, Petrosino JF, Round JL, O'Connell RM. MicroRNA-146a constrains multiple parameters of intestinal immunity and increases susceptibility to DSS colitis. Oncotarget 2015; 6:28556-72. [PMID: 26456940 PMCID: PMC4745677 DOI: 10.18632/oncotarget.5597] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 08/22/2015] [Indexed: 12/12/2022] Open
Abstract
Host-microbial interactions within the mammalian intestines must be properly regulated in order to promote host health and limit disease. Because the microbiota provide constant immunological signals to intestinal tissues, a variety of regulatory mechanisms have evolved to ensure proper immune responses to maintain homeostasis. However, many of the genes that comprise these regulatory pathways, including immune-modulating microRNAs (miRNAs), have not yet been identified or studied in the context of intestinal homeostasis. Here, we investigated the role of microRNA-146a (miR-146a) in regulating intestinal immunity and barrier function and found that this miRNA is expressed in a variety of gut tissues in adult mice. By comparing intestinal gene expression in WT and miR-146a-/- mice, we demonstrate that miR-146a represses a subset of gut barrier and inflammatory genes all within a network of immune-related signaling pathways. We also found that miR-146a restricts the expansion of intestinal T cell populations, including Th17, Tregs, and Tfh cells. GC B cells, Tfh ICOS expression, and the production of luminal IgA were also reduced by miR-146a in the gut. Consistent with an enhanced intestinal barrier, we found that miR-146a-/- mice are resistant to DSS-induced colitis, a model of Ulcerative Colitis (UC), and this correlated with elevated colonic miR-146a expression in human UC patients. Taken together, our data describe a role for miR-146a in constraining intestinal barrier function, a process that alters gut homeostasis and enhances at least some forms of intestinal disease in mice.
Collapse
Affiliation(s)
- Marah C. Runtsch
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Ruozhen Hu
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | | | - Jared Wallace
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Dominique Kagele
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Charisse Petersen
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - John F. Valentine
- Department of Medicine, Division of Gastroenterology, University of Utah, Salt Lake City, UT, USA
| | - Noah C. Welker
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Mary P. Bronner
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Xinjian Chen
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Daniel P. Smith
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Nadim J. Ajami
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Joseph F. Petrosino
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - June L. Round
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Ryan M. O'Connell
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
23
|
Salmenkari H, Issakainen T, Vapaatalo H, Korpela R. Local corticosterone production and angiotensin-I converting enzyme shedding in a mouse model of intestinal inflammation. World J Gastroenterol 2015; 21:10072-10079. [PMID: 26401072 PMCID: PMC4572788 DOI: 10.3748/wjg.v21.i35.10072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 04/30/2015] [Accepted: 07/03/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate local corticosterone production and angiotensin-I converting enzyme (ACE) protein expression and their interaction in healthy and inflamed intestine.
METHODS: Acute intestinal inflammation was induced to six weeks old male Balb/c mice by administration of either 3% or 5% dextran sodium sulfate (DSS) in drinking water for 7 d (n = 12 in each group). Healthy controls (n = 12) were given tap water. Corticosterone production and ACE protein shedding were measured from ex vivo incubates of the small and large intestine using EIA and ELISA, respectively. Morphological changes of the intestinal wall were assessed in hematoxylin-eosin stained tissue preparations of jejunum and distal colon. Effects of angiotensin II, captopril and metyrapone on corticosterone production was assessed by incubating pieces of small intestine of healthy mice in the presence of 0.1, 1 or 10 μmol/L angiotensin II, 1, 10 or 100 μmol/L captopril or 1, 10 or 100 μmol/L metyrapone solutions and measuring corticosterone released to the incubation buffer after 90 min (n = 5 in each group).
RESULTS: Both concentrations of DSS induced inflammation and morphological changes in large intestines but not in small intestines. Changes were observed as distortions of the crypt structure, mucosal erosion, immune cell infiltration to the mucosa and submucosal edema. Ex vivo corticosterone production (2.9 ± 1.0 ng/mL vs 2.0 ± 0.8 ng/mL, P = 0.034) and ACE shedding (269.2 ± 97.1 ng/mL vs 175.7 ± 52.2 ng/mL, P = 0.016) were increased in small intestines in 3% DSS group compared to the controls. In large intestine, corticosterone production was increased compared to the controls in both 3% DSS (229 ± 81 pg/mL vs 158 ± 30 pg/mL, P = 0.017) and 5% DSS groups (366 ± 163 pg/mL vs 158 ± 30 pg/mL, P = 0.002). Large intestine ACE shedding was increased in 5% DSS group (41.5 ± 9.0 ng/mL vs 20.9 ± 5.2 ng/mL, P = 0.034). Angiotensin II treatment augmented corticosterone production in small intestine at concentration of 10 μmol/L (0.97 ± 0.21 ng/mg protein vs 0.40 ± 0.09 ng/mg protein, P = 0.036).
CONCLUSION: Intestinal ACE shedding is increased by DSS-induced intestinal inflammation and parallels local corticosterone production. ACE product angiotensin II stimulates corticosterone formation in healthy intestine.
Collapse
|
24
|
Waumans Y, Baerts L, Kehoe K, Lambeir AM, De Meester I. The Dipeptidyl Peptidase Family, Prolyl Oligopeptidase, and Prolyl Carboxypeptidase in the Immune System and Inflammatory Disease, Including Atherosclerosis. Front Immunol 2015; 6:387. [PMID: 26300881 PMCID: PMC4528296 DOI: 10.3389/fimmu.2015.00387] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/13/2015] [Indexed: 12/19/2022] Open
Abstract
Research from over the past 20 years has implicated dipeptidyl peptidase (DPP) IV and its family members in many processes and different pathologies of the immune system. Most research has been focused on either DPPIV or just a few of its family members. It is, however, essential to consider the entire DPP family when discussing any one of its members. There is a substantial overlap between family members in their substrate specificity, inhibitors, and functions. In this review, we provide a comprehensive discussion on the role of prolyl-specific peptidases DPPIV, FAP, DPP8, DPP9, dipeptidyl peptidase II, prolyl carboxypeptidase, and prolyl oligopeptidase in the immune system and its diseases. We highlight possible therapeutic targets for the prevention and treatment of atherosclerosis, a condition that lies at the frontier between inflammation and cardiovascular disease.
Collapse
Affiliation(s)
- Yannick Waumans
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp , Antwerp , Belgium
| | - Lesley Baerts
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp , Antwerp , Belgium
| | - Kaat Kehoe
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp , Antwerp , Belgium
| | - Anne-Marie Lambeir
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp , Antwerp , Belgium
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp , Antwerp , Belgium
| |
Collapse
|
25
|
Cho JY, Kim HY, Kim SK, Park JHY, Lee HJ, Chun HS. β-Caryophyllene attenuates dextran sulfate sodium-induced colitis in mice via modulation of gene expression associated mainly with colon inflammation. Toxicol Rep 2015; 2:1039-1045. [PMID: 28962446 PMCID: PMC5598479 DOI: 10.1016/j.toxrep.2015.07.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/20/2015] [Accepted: 07/23/2015] [Indexed: 12/15/2022] Open
Abstract
We examined the modulatory activity of β-caryophyllene (CA) and gene expression in colitic colon tissues in a dextran sulfate sodium (DSS)-induced colitis model. Experimental colitis was induced by exposing male BALB/c mice to 5% DSS in drinking water for 7 days. CA (30 or 300 mg/kg) was administered orally once a day together with DSS. CA administration attenuated the increases in the disease activity index, colon weight/length ratio, inflammation score, and myeloperoxidase activity in DSS-treated mice. Microarray analysis showed that CA administration regulated the expression in colon tissue of inflammation-related genes including those for cytokines and chemokines (Ccl2, Ccl7, Ccl11, Ifitm3, IL-1β, IL-28, Tnfrsf1b, Tnfrsf12a); acute-phase proteins (S100a8, Saa3, Hp); adhesion molecules (Cd14, Cd55, Cd68, Mmp3, Mmp10, Sema6b, Sema7a, Anax13); and signal regulatory proteins induced by DSS. CA significantly suppressed NF-κB activity, which mediates the expression of a different set of genes. These results suggest that CA attenuates DSS-induced colitis, possibly by modulating the expression of genes associated mainly with colon inflammation through inhibition of DSS-induced NF-κB activity.
Collapse
Key Words
- CA, β-caryophyllene
- CD, crohn disease
- Cebpb, CCAAT/enhancer-binding protein &beta
- Colitis
- DSS, dextran sulfate sodium
- Dextran sulfate sodium
- Gene expression
- Hp, haptoglobin
- IBD, inflammatory bowel disease
- IL, interleukin
- Inflammation
- IκB, inhibitor κB
- MPO, myeloperoxidase
- NF-κB, nuclear factor-kappa B
- S100a8, S100 calcium binding protein a8
- SAL, sulfasalazine
- Saa3, serum amyloid A3
- TNF-α, tumor necrosis factor-α
- UC, ulcerative colitis
- β-Caryophyllene (PubChem CID5281515)
Collapse
Affiliation(s)
- Jae Young Cho
- CKD Research Institute, Dongbaekjukjeon-daero 315-20, Yungin, Kyonggi 446-916, Republic of Korea
| | - Hwa Yeon Kim
- Department of Food Science and Technology, Chung-Ang University, Naeri 72-1, Ansung, Kyonggi 456-756, Republic of Korea
| | - Sung-Kyu Kim
- Nutra R&BT Inc., 371-47 Gasan, Geumcheon-gu, Seoul 153-788, Republic of Korea
| | - Jung Han Yoon Park
- Department of Food Science and Nutrition, Hallym University, Hallymdaehak-gil 39, Chuncheon, Kangwon 200-702, Republic of Korea
| | - Hong Jin Lee
- Department of Food Science and Technology, Chung-Ang University, Naeri 72-1, Ansung, Kyonggi 456-756, Republic of Korea
| | - Hyang Sook Chun
- Department of Food Science and Technology, Chung-Ang University, Naeri 72-1, Ansung, Kyonggi 456-756, Republic of Korea
| |
Collapse
|
26
|
Menconi A, Hernandez-Velasco X, Vicuña E, Kuttappan V, Faulkner O, Tellez G, Hargis B, Bielke L. Histopathological and morphometric changes induced by a dextran sodium sulfate (DSS) model in broilers. Poult Sci 2015; 94:906-11. [DOI: 10.3382/ps/pev054] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2014] [Indexed: 01/03/2023] Open
|
27
|
Pai MH, Liu JJ, Hou YC, Yeh CL. Soybean and Fish Oil Mixture With Different ω-6/ω-3 Polyunsaturated Fatty Acid Ratios Modulates Dextran Sulfate Sodium–Induced Changes in Small Intestinal Intraepithelial γδT-Lymphocyte Expression in Mice. JPEN J Parenter Enteral Nutr 2014; 40:383-91. [DOI: 10.1177/0148607114559429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 10/15/2014] [Indexed: 01/28/2023]
Affiliation(s)
- Man-Hui Pai
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jun-Jen Liu
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen Hou
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chiu-Li Yeh
- Department of Nutrition and Health Sciences, Chinese Culture University, Taipei, Taiwan
| |
Collapse
|
28
|
Effect of surface chemistry on nanoparticle interaction with gastrointestinal mucus and distribution in the gastrointestinal tract following oral and rectal administration in the mouse. J Control Release 2014; 197:48-57. [PMID: 25449804 DOI: 10.1016/j.jconrel.2014.10.026] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/13/2014] [Accepted: 10/27/2014] [Indexed: 02/07/2023]
Abstract
It is believed that mucoadhesive surface properties on particles delivered to the gastrointestinal (GI) tract improve oral absorption or local targeting of various difficult-to-deliver drug classes. To test the effect of nanoparticle mucoadhesion on distribution of nanoparticles in the GI tract, we orally and rectally administered nano- and microparticles that we confirmed possessed surfaces that were either strongly mucoadhesive or non-mucoadhesive. We found that mucoadhesive particles (MAP) aggregated in mucus in the center of the GI lumen, far away from the absorptive epithelium, both in healthy mice and in a mouse model of ulcerative colitis (UC). In striking contrast, water absorption by the GI tract rapidly and uniformly transported non-mucoadhesive mucus-penetrating particles (MPP) to epithelial surfaces, including reaching the surfaces between villi in the small intestine. When using high gavage fluid volumes or injection into ligated intestinal loops, common methods for assessing oral drug and nanoparticle absorption, we found that both MAP and MPP became well-distributed throughout the intestine, indicating that the barrier properties of GI mucus were compromised. In the mouse colorectum, MPP penetrated into mucus in the deeply in-folded surfaces to evenly coat the entire epithelial surface. Moreover, in a mouse model of UC, MPP were transported preferentially into the disrupted, ulcerated tissue. Our results suggest that delivering drugs in non-mucoadhesive MPP is likely to provide enhanced particle distribution, and thus drug delivery, in the GI tract, including to ulcerated tissues.
Collapse
|
29
|
Mashukova A, Kozhekbaeva Z, Forteza R, Dulam V, Figueroa Y, Warren R, Salas PJ. The BAG-1 isoform BAG-1M regulates keratin-associated Hsp70 chaperoning of aPKC in intestinal cells during activation of inflammatory signaling. J Cell Sci 2014; 127:3568-77. [PMID: 24876225 DOI: 10.1242/jcs.151084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Atypical PKC (ι/λ and ζ; hereafter referred to as aPKC) is a key player in the acquisition of epithelial polarity and participates in other signaling cascades including the control of NF-κB signaling. This kinase is post-translationally regulated through Hsp70-mediated refolding. Previous work has shown that such a chaperoning activity is specifically localized to keratin intermediate filaments. Our work was performed with the goal of identifying the molecule(s) that block Hsp70 activity on keratin filaments during inflammation. A transcriptional screen allowed us to focus on BAG-1, a multi-functional protein that assists Hsp70 in nucleotide exchange but also blocks its activity at higher concentrations. We found the BAG-1 isoform BAG-1M upregulated threefold in human Caco-2 cells following stimulation with tumor necrosis factor receptor α (TNFα) to induce a pro-inflammatory response, and up to sixfold in mouse enterocytes following treatment with dextran sodium sulfate (DSS) to induce colitis. BAG-1M, but no other isoform, was found to co-purify with intermediate filaments and block Hsp70 activity in the keratin fraction but not in the soluble fraction within the range of concentrations found in epithelial cells cultured under control and inflammation conditions. Constitutive expression of BAG-1M decreased levels of phosphorylated aPKC. By contrast, knockdown of BAG-1, blocked the TNFα-induced decrease of phosphorylated aPKC. We conclude that BAG-1M mediates Hsp70 inhibition downstream of NF-κB.
Collapse
Affiliation(s)
- Anastasia Mashukova
- Nova Southeastern University, Department of Physiology, Fort Lauderdale, FL 33314, USA University of Miami Miller School of Medicine, Department of Cell Biology, Miami, FL 33136, USA
| | - Zhanna Kozhekbaeva
- University of Miami Miller School of Medicine, Department of Cell Biology, Miami, FL 33136, USA
| | - Radia Forteza
- University of Miami Miller School of Medicine, Department of Cell Biology, Miami, FL 33136, USA
| | - Vipin Dulam
- University of Miami Miller School of Medicine, Department of Cell Biology, Miami, FL 33136, USA
| | - Yolanda Figueroa
- University of Miami Miller School of Medicine, Department of Cell Biology, Miami, FL 33136, USA
| | - Robert Warren
- University of Miami Miller School of Medicine, Department of Cell Biology, Miami, FL 33136, USA
| | - Pedro J Salas
- University of Miami Miller School of Medicine, Department of Cell Biology, Miami, FL 33136, USA
| |
Collapse
|
30
|
Glutamine supplementation attenuates expressions of adhesion molecules and chemokine receptors on T cells in a murine model of acute colitis. Mediators Inflamm 2014; 2014:837107. [PMID: 24891768 PMCID: PMC4033481 DOI: 10.1155/2014/837107] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/21/2014] [Accepted: 04/07/2014] [Indexed: 01/11/2023] Open
Abstract
Background. Migration of T cells into the colon plays a major role in the pathogenesis in inflammatory bowel disease. This study investigated the effects of glutamine (Gln) supplementation on chemokine receptors and adhesion molecules expressed by T cells in mice with dextran sulfate sodium- (DSS-) induced colitis. Methods. C57BL/6 mice were fed either a standard diet or a Gln diet replacing 25% of the total nitrogen. After being fed the diets for 5 days, half of the mice from both groups were given 1.5% DSS in drinking water to induce colitis. Mice were killed after 5 days of DSS exposure. Results. DSS colitis resulted in higher expression levels of P-selectin glycoprotein ligand- (PSGL-) 1, leukocyte function-associated antigen- (LFA-) 1, and C-C chemokine receptor type 9 (CCR9) by T helper (Th) and cytotoxic T (Tc) cells, and mRNA levels of endothelial adhesion molecules in colons were upregulated. Gln supplementation decreased expressions of PSGL-1, LFA-1, and CCR9 by Th cells. Colonic gene expressions of endothelial adhesion molecules were also lower in Gln-colitis mice. Histological finding showed that colon infiltrating Th cells were less in the DSS group with Gln administration. Conclusions. Gln supplementation may ameliorate the inflammation of colitis possibly via suppression of T cell migration.
Collapse
|
31
|
Zheng B, van Bergenhenegouwen J, Overbeek S, van de Kant HJG, Garssen J, Folkerts G, Vos P, Morgan ME, Kraneveld AD. Bifidobacterium breve attenuates murine dextran sodium sulfate-induced colitis and increases regulatory T cell responses. PLoS One 2014; 9:e95441. [PMID: 24787575 PMCID: PMC4008378 DOI: 10.1371/journal.pone.0095441] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 03/27/2014] [Indexed: 12/19/2022] Open
Abstract
While some probiotics have shown beneficial effects on preventing or treating colitis development, others have shown no effects. In this study, we have assessed the immunomodulating effects of two probiotic strains, Lactobacillus rhamnosus (L. rhamnosus) and Bifidobacterium breve (B. breve) on T cell polarization in vitro, using human peripheral blood mononuclear cells (PBMC), and in vivo, using murine dextran sodium sulfate (DSS) colitis model. With respect to the latter, the mRNA expression of T cell subset-associated transcription factors and cytokines in the colon was measured and the T helper type (Th) 17 and regulatory T cell (Treg) subsets were determined in the Peyer's patches. Both L. rhamnosus and B. breve incubations in vitro reduced Th17 and increased Th2 cell subsets in human PBMCs. In addition, B. breve incubation was also able to reduce Th1 and increase Treg cell subsets in contrast to L. rhamnosus. In vivo intervention with B. breve, but not L. rhamnosus, significantly attenuated the severity of DSS-induced colitis. In DSS-treated C57BL/6 mice, intervention with B. breve increased the expression of mRNA encoding for Th2- and Treg-associated cytokines in the distal colon. In addition, intervention with B. breve led to increases of Treg and decreases of Th17 cell subsets in Peyer's patches of DSS-treated mice. B. breve modulates T cell polarization towards Th2 and Treg cell-associated responses in vitro and in vivo. In vivo B. breve intervention ameliorates DSS-induced colitis symptoms and this protective effect may mediated by its effects on the T-cell composition.
Collapse
Affiliation(s)
- Bin Zheng
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Science, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Jeroen van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Science, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Nutricia Research, Utrecht, The Netherlands
| | - Saskia Overbeek
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Science, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Hendrik J. G. van de Kant
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Science, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Science, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Nutricia Research, Utrecht, The Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Science, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Paul Vos
- Nutricia Research, Utrecht, The Netherlands
| | - Mary E. Morgan
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Science, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Science, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
32
|
Glutamine modulates acute dextran sulphate sodium-induced changes in small-intestinal intraepithelial γδ-T-lymphocyte expression in mice. Br J Nutr 2013; 111:1032-9. [PMID: 24229607 DOI: 10.1017/s0007114513003425] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The present study investigated the effect of glutamine (Gln) on dextran sulphate sodium (DSS)-induced changes in the expression of small-intestinal intraepithelial lymphocyte (IEL) γδ-T cells in mice. Mice were randomly assigned to a normal control (NC) group and two DSS-treated groups. The NC group and one of the DSS-treated groups (DSS-C) were fed a common semi-purified diet, while the other DSS-treated group (DSS-G) was fed an identical diet, except that part of casein was replaced by Gln, which provided 25 % of total amino acid nitrogen. After being fed the diets for 10 d, mice in the NC group were given distilled water, while the DSS-treated groups were given distilled water containing 2·5 % DSS for 5 d. At the end of the experiment, the mice were killed. The small-intestinal IEL γδ-T-cell subset was isolated for further analysis. The results indicated that DSS treatment resulted in a lower percentage of small-intestinal IEL γδ-T cells and higher mRNA expressions of interferon-γ, TNF-α, IL-17, complement 5a receptor and keratinocyte growth factor in IEL γδ-T cells. Gln administration increased the proportion of small-intestinal IEL γδ-T cells, and the expression levels of immunomodulatory mediator genes in IEL γδ-T cells were lower in the DSS-treated mice. The histological findings indicated that the immunoreactive intensity of the tight junction protein ZO-1 in the small-intestinal mucosa was higher in the DSS-G group than in the DSS-C group. These results indicate that pretreatment with Gln increases the proportion of small-intestinal IEL γδ-T cells and down-regulates γδ-T-cell-expressed inflammatory mediators, which may consequently ameliorate the severity of DSS-induced small-intestinal epithelial injury.
Collapse
|
33
|
Pott J, Hornef M. Innate immune signalling at the intestinal epithelium in homeostasis and disease. EMBO Rep 2012; 13:684-98. [PMID: 22801555 DOI: 10.1038/embor.2012.96] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 06/07/2012] [Indexed: 02/06/2023] Open
Abstract
The intestinal epithelium--which constitutes the interface between the enteric microbiota and host tissues--actively contributes to the maintenance of mucosal homeostasis and defends against pathogenic microbes. The recognition of conserved microbial products by cytosolic or transmembrane pattern recognition receptors in epithelial cells initiates signal transduction and influences effector cell function. However, the signalling pathways, effector molecules and regulatory mechanisms involved are not yet fully understood, and the functional outcome is poorly defined. This review analyses the complex and dynamic role of intestinal epithelial innate immune recognition and signalling, on the basis of results in intestinal epithelial cell-specific transgene or gene-deficient animals. This approach identifies specific epithelial cell functions within the diverse cellular composition of the mucosal tissue, in the presence of the complex and dynamic gut microbiota. These insights have thus provided a more comprehensive understanding of the role of the intestinal epithelium in innate immunity during homeostasis and disease.
Collapse
Affiliation(s)
- Johanna Pott
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | | |
Collapse
|
34
|
BERCIK P, PARK AJ, SINCLAIR D, KHOSHDEL A, LU J, HUANG X, DENG Y, BLENNERHASSETT PA, FAHNESTOCK M, MOINE D, BERGER B, HUIZINGA JD, KUNZE W, MCLEAN PG, BERGONZELLI GE, COLLINS SM, VERDU EF. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut-brain communication. Neurogastroenterol Motil 2011; 23:1132-9. [PMID: 21988661 PMCID: PMC3413724 DOI: 10.1111/j.1365-2982.2011.01796.x] [Citation(s) in RCA: 665] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The probiotic Bifidobacterium longum NCC3001 normalizes anxiety-like behavior and hippocampal brain derived neurotrophic factor (BDNF) in mice with infectious colitis. Using a model of chemical colitis we test whether the anxiolytic effect of B. longum involves vagal integrity, and changes in neural cell function. Methods Mice received dextran sodium sulfate (DSS, 3%) in drinking water during three 1-week cycles. Bifidobacterium longum or placebo were gavaged daily during the last cycle. Some mice underwent subdiaphragmatic vagotomy. Behavior was assessed by step-down test, inflammation by myeloperoxidase (MPO) activity and histology. BDNF mRNA was measured in neuroblastoma SH-SY5Y cells after incubation with sera from B. longum- or placebo-treated mice. The effect of B. longum on myenteric neuron excitability was measured using intracellular microelectrodes. KEY RESULTS Chronic colitis was associated with anxiety-like behavior, which was absent in previously vagotomized mice. B. longum normalized behavior but had no effect on MPO activity or histological scores. Its anxiolytic effect was absent in mice with established anxiety that were vagotomized before the third DSS cycle. B. longum metabolites did not affect BDNF mRNA expression in SH-SY5Y cells but decreased excitability of enteric neurons. CONCLUSIONS & INFERENCES In this colitis model, anxiety-like behavior is vagally mediated. The anxiolytic effect of B. longum requires vagal integrity but does not involve gut immuno-modulation or production of BDNF by neuronal cells. As B. longum decreases excitability of enteric neurons, it may signal to the central nervous system by activating vagal pathways at the level of the enteric nervous system.
Collapse
Affiliation(s)
- P. BERCIK
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - A. J. PARK
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - D. SINCLAIR
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - A. KHOSHDEL
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - J. LU
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - X. HUANG
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Y. DENG
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - P. A. BLENNERHASSETT
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - M. FAHNESTOCK
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - D. MOINE
- Bioanalytical Science Department, Nestlé Research Center, Lausanne, Switzerland
| | - B. BERGER
- Bioanalytical Science Department, Nestlé Research Center, Lausanne, Switzerland
| | - J. D. HUIZINGA
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - W. KUNZE
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - P. G. MCLEAN
- Digestive Health, Nutrition and Health Department, Nestlé Research Center, Lausanne, Switzerland
| | - G. E. BERGONZELLI
- Digestive Health, Nutrition and Health Department, Nestlé Research Center, Lausanne, Switzerland
| | - S. M. COLLINS
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - E. F. VERDU
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
35
|
Bercik P, Park AJ, Sinclair D, Khoshdel A, Lu J, Huang X, Deng Y, Blennerhassett PA, Fahnestock M, Moine D, Berger B, Huizinga JD, Kunze W, McLean PG, Bergonzelli GE, Collins SM, Verdu EF. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut-brain communication. NEUROGASTROENTEROLOGY AND MOTILITY : THE OFFICIAL JOURNAL OF THE EUROPEAN GASTROINTESTINAL MOTILITY SOCIETY 2011. [PMID: 21988661 DOI: 10.1111/j.1365-2982.2011.01796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND The probiotic Bifidobacterium longum NCC3001 normalizes anxiety-like behavior and hippocampal brain derived neurotrophic factor (BDNF) in mice with infectious colitis. Using a model of chemical colitis we test whether the anxiolytic effect of B. longum involves vagal integrity, and changes in neural cell function. Methods Mice received dextran sodium sulfate (DSS, 3%) in drinking water during three 1-week cycles. Bifidobacterium longum or placebo were gavaged daily during the last cycle. Some mice underwent subdiaphragmatic vagotomy. Behavior was assessed by step-down test, inflammation by myeloperoxidase (MPO) activity and histology. BDNF mRNA was measured in neuroblastoma SH-SY5Y cells after incubation with sera from B. longum- or placebo-treated mice. The effect of B. longum on myenteric neuron excitability was measured using intracellular microelectrodes. KEY RESULTS Chronic colitis was associated with anxiety-like behavior, which was absent in previously vagotomized mice. B. longum normalized behavior but had no effect on MPO activity or histological scores. Its anxiolytic effect was absent in mice with established anxiety that were vagotomized before the third DSS cycle. B. longum metabolites did not affect BDNF mRNA expression in SH-SY5Y cells but decreased excitability of enteric neurons. CONCLUSIONS & INFERENCES In this colitis model, anxiety-like behavior is vagally mediated. The anxiolytic effect of B. longum requires vagal integrity but does not involve gut immuno-modulation or production of BDNF by neuronal cells. As B. longum decreases excitability of enteric neurons, it may signal to the central nervous system by activating vagal pathways at the level of the enteric nervous system.
Collapse
Affiliation(s)
- P Bercik
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|