1
|
Zhao H, Liu T, Yang CE, Hu YH, Niu Y, Lei SP, Chen L, Zhang MX. Poricoic acid A attenuates renal fibrosis by inhibiting endoplasmic reticulum stress-mediated apoptosis. Braz J Med Biol Res 2024; 57:e14249. [PMID: 39607209 DOI: 10.1590/1414-431x2024e14249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/11/2024] [Indexed: 11/29/2024] Open
Abstract
Renal fibrosis is a common manifestation in the progression of chronic kidney disease (CKD) to kidney failure. Currently, there is no available therapy to prevent the progression of renal fibrosis. Poricoic acid A (PAA) isolated from Poria cocos shows notable antifibrotic effects. However, its potential mechanism is still unclear. This study aimed to evaluate the effects and the potential mechanisms of PAA against renal fibrosis. A mouse model of renal fibrosis was established using unilateral ureteral obstruction (UUO). We showed that PAA administration significantly alleviated renal lesions and collagen deposition in UUO mice. Mice with UUO resulted in epithelial-to-mesenchymal transition (EMT) and the activation of endoplasmic reticulum stress (ERS) in the renal tissues, while PAA treatment significantly inhibited EMT and ERS activation. Additionally, PAA markedly alleviated ERS-mediated apoptosis in UUO mice. Molecular docking results indicated that PAA stably combined to GRP78 and ATF4. In conclusion, these results demonstrated that PAA possesses a significant bioactivity against renal fibrosis and its treatment mechanism might be the inhibition of ERS-mediated apoptosis.
Collapse
Affiliation(s)
- Hui Zhao
- Clinical Experimental Center, Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, Northwest University Affiliated Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Tian Liu
- Clinical Experimental Center, Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, Northwest University Affiliated Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Chang-E Yang
- Clinical Experimental Center, Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, Northwest University Affiliated Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Yue-Huan Hu
- Clinical Experimental Center, Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, Northwest University Affiliated Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Yan Niu
- Clinical Experimental Center, Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, Northwest University Affiliated Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Sheng-Ping Lei
- Clinical Experimental Center, Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, Northwest University Affiliated Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Lin Chen
- College of Biology, Pharmacy and Food Engineering, Shangluo University, Shangluo, Shaanxi, China
| | - Ming-Xia Zhang
- Clinical Experimental Center, Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, Northwest University Affiliated Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
2
|
van Gorp C, de Lange IH, Hütten MC, López-Iglesias C, Massy KRI, Kessels L, Knoops K, Cuijpers I, Sthijns MMJPE, Troost FJ, van Gemert WG, Spiller OB, Birchenough GMH, Zimmermann LJI, Wolfs TGAM. Antenatal Ureaplasma Infection Causes Colonic Mucus Barrier Defects: Implications for Intestinal Pathologies. Int J Mol Sci 2024; 25:4000. [PMID: 38612809 PMCID: PMC11011967 DOI: 10.3390/ijms25074000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
Chorioamnionitis is a risk factor for necrotizing enterocolitis (NEC). Ureaplasma parvum (UP) is clinically the most isolated microorganism in chorioamnionitis, but its pathogenicity remains debated. Chorioamnionitis is associated with ileal barrier changes, but colonic barrier alterations, including those of the mucus barrier, remain under-investigated, despite their importance in NEC pathophysiology. Therefore, in this study, the hypothesis that antenatal UP exposure disturbs colonic mucus barrier integrity, thereby potentially contributing to NEC pathogenesis, was investigated. In an established ovine chorioamnionitis model, lambs were intra-amniotically exposed to UP or saline for 7 d from 122 to 129 d gestational age. Thereafter, colonic mucus layer thickness and functional integrity, underlying mechanisms, including endoplasmic reticulum (ER) stress and redox status, and cellular morphology by transmission electron microscopy were studied. The clinical significance of the experimental findings was verified by examining colon samples from NEC patients and controls. UP-exposed lambs have a thicker but dysfunctional colonic mucus layer in which bacteria-sized beads reach the intestinal epithelium, indicating undesired bacterial contact with the epithelium. This is paralleled by disturbed goblet cell MUC2 folding, pro-apoptotic ER stress and signs of mitochondrial dysfunction in the colonic epithelium. Importantly, the colonic epithelium from human NEC patients showed comparable mitochondrial aberrations, indicating that NEC-associated intestinal barrier injury already occurs during chorioamnionitis. This study underlines the pathogenic potential of UP during pregnancy; it demonstrates that antenatal UP infection leads to severe colonic mucus barrier deficits, providing a mechanistic link between antenatal infections and postnatal NEC development.
Collapse
Affiliation(s)
- Charlotte van Gorp
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Ilse H. de Lange
- Department of Pediatrics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Matthias C. Hütten
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
- Neonatology, Department of Pediatrics, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Carmen López-Iglesias
- Microscopy CORE Lab, Maastricht Multimodal Molecular Imaging Institute (M4I), Maastricht University, 6211 LK Maastricht, The Netherlands; (C.L.-I.); (K.K.)
| | - Kimberly R. I. Massy
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Lilian Kessels
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Kèvin Knoops
- Microscopy CORE Lab, Maastricht Multimodal Molecular Imaging Institute (M4I), Maastricht University, 6211 LK Maastricht, The Netherlands; (C.L.-I.); (K.K.)
| | - Iris Cuijpers
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Mireille M. J. P. E. Sthijns
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Freddy J. Troost
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Wim G. van Gemert
- Department of Surgery, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Owen B. Spiller
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XW, UK;
| | - George M. H. Birchenough
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, 40530 Gothenburg, Sweden;
| | - Luc J. I. Zimmermann
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Tim G. A. M. Wolfs
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
- Department of Biomedical Engineering (BMT), School for Cardiovascular Diseases (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
3
|
Gong ZZ, Li T, Yan H, Xu MH, Lian Y, Yang YX, Wei W, Liu T. Exploring the autophagy-related pathogenesis of active ulcerative colitis. World J Clin Cases 2024; 12:1622-1633. [PMID: 38576744 PMCID: PMC10989433 DOI: 10.12998/wjcc.v12.i9.1622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 01/23/2024] [Accepted: 02/27/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND The pathogenesis of ulcerative colitis (UC) is complex, and recent therapeutic advances remain unable to fully alleviate the condition. AIM To inform the development of novel UC treatments, bioinformatics was used to explore the autophagy-related pathogenesis associated with the active phase of UC. METHODS The GEO database was searched for UC-related datasets that included healthy controls who met the screening criteria. Differential analysis was conducted to obtain differentially expressed genes (DEGs). Autophagy-related targets were collected and intersected with the DEGs to identiy differentially expressed autophagy-related genes (DEARGs) associated with active UC. DEARGs were then subjected to KEGG, GO, and DisGeNET disease enrichment analyses using R software. Differential analysis of immune infiltrating cells was performed using the CiberSort algorithm. The least absolute shrinkage and selection operator algorithm and protein-protein interaction network were used to narrow down the DEARGs, and the top five targets in the Dgree ranking were designated as core targets. RESULTS A total of 4822 DEGs were obtained, of which 58 were classified as DEARGs. SERPINA1, BAG3, HSPA5, CASP1, and CX3CL1 were identified as core targets. GO enrichment analysis revealed that DEARGs were primarily enriched in processes related to autophagy regulation and macroautophagy. KEGG enrichment analysis showed that DEARGs were predominantly associated with NOD-like receptor signaling and other signaling pathways. Disease enrichment analysis indicated that DEARGs were significantly linked to diseases such as malignant glioma and middle cerebral artery occlusion. Immune infiltration analysis demonstrated a higher presence of immune cells like activated memory CD4 T cells and follicular helper T cells in active UC patients than in healthy controls. CONCLUSION Autophagy is closely related to the active phase of UC and the potential targets obtained from the analysis in this study may provide new insight into the treatment of active UC patients.
Collapse
Affiliation(s)
- Zhuo-Zhi Gong
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Teng Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - He Yan
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Min-Hao Xu
- College of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Beijing 100102, China
| | - Yue Lian
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Yi-Xuan Yang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Wei Wei
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Tao Liu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| |
Collapse
|
4
|
Tak J, An Q, Lee SG, Lee CH, Kim SG. Gα12 and endoplasmic reticulum stress-mediated pyroptosis in a single cycle of dextran sulfate-induced mouse colitis. Sci Rep 2024; 14:6335. [PMID: 38491049 PMCID: PMC10943197 DOI: 10.1038/s41598-024-56685-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/09/2024] [Indexed: 03/18/2024] Open
Abstract
Inflammatory bowel disease (IBD) pathogenesis involves complex inflammatory events and cell death. Although IBD involves mainly necrosis in the digestive tract, pyroptosis has also been recognized. Nonetheless, the underlying basis is elusive. Gα12/13 overexpression may affect endoplasmic reticulum (ER) stress. This study examined how Gα12/13 and ER stress affect pyroptosis using dextran sulfate sodium (DSS)-induced colitis models. Gα12/13 levels were increased in the distal and proximal colons of mice exposed to a single cycle of DSS, as accompanied by increases of IRE1α, ATF6, and p-PERK. Moreover, Il-6, Il-1β, Ym1, and Arg1 mRNA levels were increased with caspase-1 and IL-1β activation, supportive of pyroptosis. In the distal colon, RIPK1/3 levels were enhanced to a greater degree, confirming necroptosis. By contrast, the mice subjected to three cycles of DSS treatments showed decreases of Gα12/13, as accompanied by IRE1α and ATF6 suppression, but increases of RIPK1/3 and c-Cas3. AZ2 treatment, which inhibited Gα12, has an anti-pyroptotic effect against a single cycle of colitis. These results show that a single cycle of DSS-induced colitis may cause ER stress-induced pyroptosis as mediated by Gα12 overexpression in addition to necroptosis, but three cycles model induces only necroptosis, and that AZ2 may have an anti-pyroptotic effect.
Collapse
Affiliation(s)
- Jihoon Tak
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Quanxi An
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Sang Gil Lee
- Research and Development Institute, A Pharma Inc, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Chang Hoon Lee
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
5
|
Martín-Adrados B, Wculek SK, Fernández-Bravo S, Torres-Ruiz R, Valle-Noguera A, Gomez-Sánchez MJ, Hernández-Walias JC, Ferreira FM, Corraliza AM, Sancho D, Esteban V, Rodriguez-Perales S, Cruz-Adalia A, Nakaya HI, Salas A, Bernardo D, Campos-Martín Y, Martínez-Zamorano E, Muñoz-López D, Gómez del Moral M, Cubero FJ, Blumberg RS, Martínez-Naves E. Expression of HMGCS2 in intestinal epithelial cells is downregulated in inflammatory bowel disease associated with endoplasmic reticulum stress. Front Immunol 2023; 14:1185517. [PMID: 37457727 PMCID: PMC10348483 DOI: 10.3389/fimmu.2023.1185517] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction The Unfolded Protein Response, a mechanism triggered by the cell in response to Endoplasmic reticulum stress, is linked to inflammatory responses. Our aim was to identify novel Unfolded Protein Response-mechanisms that might be involved in triggering or perpetuating the inflammatory response carried out by the Intestinal Epithelial Cells in the context of Inflammatory Bowel Disease. Methods We analyzed the transcriptional profile of human Intestinal Epithelial Cell lines treated with an Endoplasmic Reticulum stress inducer (thapsigargin) and/or proinflammatory stimuli. Several genes were further analyzed in colonic biopsies from Ulcerative Colitis patients and healthy controls. Lastly, we generated Caco-2 cells lacking HMGCS2 by CRISPR Cas-9 and analyzed the functional implications of its absence in Intestinal Epithelial Cells. Results Exposure to a TLR ligand after thapsigargin treatment resulted in a powerful synergistic modulation of gene expression, which led us to identify new genes and pathways that could be involved in inflammatory responses linked to the Unfolded Protein Response. Key differentially expressed genes in the array also exhibited transcriptional alterations in colonic biopsies from active Ulcerative Colitis patients, including NKG2D ligands and the enzyme HMGCS2. Moreover, functional studies showed altered metabolic responses and epithelial barrier integrity in HMGCS2 deficient cell lines. Conclusion We have identified new genes and pathways that are regulated by the Unfolded Protein Response in the context of Inflammatory Bowel Disease including HMGCS2, a gene involved in the metabolism of Short Chain Fatty Acids that may have an important role in intestinal inflammation linked to Endoplasmic Reticulum stress and the resolution of the epithelial damage.
Collapse
Affiliation(s)
- Beatriz Martín-Adrados
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de octubre (imas12), Madrid, Spain
| | - Stefanie K. Wculek
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sergio Fernández-Bravo
- Department of Allergy and Immunology, IIS-Fundación Jiménez Díaz, Universidad Autónoma of Madrid, Madrid, Spain
| | - Raúl Torres-Ruiz
- Molecular Cytogenetics & Genome Editing Unit, Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro, Madrid, Spain
- Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Advanced Therapies Unit, Hematopoietic Innovative Therapies Division, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Ana Valle-Noguera
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de octubre (imas12), Madrid, Spain
| | - Maria José Gomez-Sánchez
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de octubre (imas12), Madrid, Spain
| | - José Carlos Hernández-Walias
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de octubre (imas12), Madrid, Spain
| | | | - Ana María Corraliza
- Department of Gastroenterology, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Hospital Clinic of Barcelona, Centro de Investigación Biomédica en Red-Enfermedades Hepáticas y Digestivas (CIBER-EHD), Barcelona, Spain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Vanesa Esteban
- Department of Allergy and Immunology, IIS-Fundación Jiménez Díaz, Universidad Autónoma of Madrid, Madrid, Spain
| | - Sandra Rodriguez-Perales
- Molecular Cytogenetics & Genome Editing Unit, Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro, Madrid, Spain
| | - Aránzazu Cruz-Adalia
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de octubre (imas12), Madrid, Spain
| | - Helder I. Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Azucena Salas
- Department of Gastroenterology, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Hospital Clinic of Barcelona, Centro de Investigación Biomédica en Red-Enfermedades Hepáticas y Digestivas (CIBER-EHD), Barcelona, Spain
| | - David Bernardo
- Gut Immunology Research Group, Instituto de Investigación del Hospital Universitario de la Princesa, Madrid, Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM, Universidad de Valladolid-Consejo Superior de Investigaciones Científicas (CSIC)), Valladolid, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Diego Muñoz-López
- Department of Pathology, Hospital Universitario de Toledo, Toledo, Spain
| | - Manuel Gómez del Moral
- Department of Cellular Biology, School of Medicine, Universidad Complutense of Madrid (UCM), Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de octubre (imas12), Madrid, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermeddes Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Richard S. Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Eduardo Martínez-Naves
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de octubre (imas12), Madrid, Spain
| |
Collapse
|
6
|
Chen J, Shen B, Jiang Z. Traditional Chinese medicine prescription Shenling BaiZhu powder to treat ulcerative colitis: Clinical evidence and potential mechanisms. Front Pharmacol 2022; 13:978558. [PMID: 36160392 PMCID: PMC9494158 DOI: 10.3389/fphar.2022.978558] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Ulcerative colitis (UC), characterized by syndromes including abdominal pain, bloody stool, diarrhea, weight loss, and repeated relapse, is a non-specific inflammatory intestinal disease. In recent years, with the changing dietary habits in China, the incidence of UC has shown an upward trend. UC belongs to the category of recorded as "diarrhea," "chronic dysentery," and "hematochezia" in traditional Chinese medicine (TCM), and Shenling BaiZhu powder (SLBZP) is one of the most effective and commonly used prescriptions. In this review, we aim to systematically summarize the clinical application and pharmacological mechanism of SLBZP in the treatment of UC to provide a theoretical basis for its clinical use and experimental evaluation of SLBZP. Our results showed that both SLBZP and SLBZP in combination with chemical drugs, have a significant therapeutic effect against UC with few adverse reactions. Furthermore, combined therapy was better than western medicine. Further, pathophysiological studies indicated that SLBZP has anti-inflammatory, immunomodulatory, antioxidant effects, regulation relative cell signal transduction and regulation of gut microbiota. Although evidence suggests superior therapeutic efficacy of SLBZP for treating UC and the relative mechanism has been studied extensively, various shortcomings limit the existing research on the topic. There is a lack of UC animal models, especially UC with TCM syndromes, with no uniform standard and certain differences between the animal model and clinical syndrome. The dosage, dosage form, and therapeutic time of SLBZP are inconsistent and lack pharmacological verification, and clinical trial data are not detailed or sufficiently rigorous. In addition, SLSZP is composed of multiple Chinese drugs that contain massive numbers of ingredients and which or several components contribute to therapeutic effects. How they work synergistically together remains unknown. Therefore, on the one hand, large sample prospective cohort studies to clarify the clinical efficacy and safety of SLBZP in the treatment of UC are needed. In contrast, researchers should strengthen the study of the molecular biological mechanism of active ingredients and its synergistic actions, clarifying the mechanism of SLBZP in treating UC by multi-component, multi-target, and multi-pathway.
Collapse
Affiliation(s)
- Jing Chen
- Department of Pharmacy, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Lin Hai, China
| | - Bixin Shen
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhengli Jiang
- Department of Pharmacy, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Lin Hai, China
| |
Collapse
|
7
|
HSPA5 Inhibitor Meliorate DSS-Induced Colitis through HSPA1A/CHIP. DISEASE MARKERS 2022; 2022:7115181. [PMID: 35872700 PMCID: PMC9300310 DOI: 10.1155/2022/7115181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/27/2022] [Indexed: 12/01/2022]
Abstract
Objective Ulcerative colitis (UC) is closely related to immune response, in which Treg cells (Tregs) suppress the autoimmune response of effector T cells to maintain homeostasis. As a marker of endoplasmic reticulum stress (ERS), HSPA5 was highly expressed in the colon tissue of UC patients. This study is aimed at evaluating the therapeutic effect of HSPA5 inhibitor (HA15) on dextran sulfate sodium- (DSS-) induced ulcerative colitis in mice and explored the effect and related mechanism of HSPA5 inhibitor on the differentiation and function of Tregs. Methods Thirty-two C57BL/6 mice were randomly divided into four groups (8 mice per group): normal control group, DSS model group, HSPA5 inhibitor (HA15) group (intraperitoneal injection), and dexamethasone (DXM) group (intraperitoneal injection). Except for the blank control group, the other groups were induced with 3% DSS for 7 days and then given corresponding intervention therapy for 7 days. Results The disease activity index (DAI) score, colon length, histopathological changes, and scores of DSS-induced mice show that HA15 could significantly improve the degree of inflammation in ulcerative colitis. Moreover, HA15 can better inhibit the expression of HSPA5, HSPA1A, and CHIP in the colon and increase the level of FOXP3 mRNA. Finally, the content of Treg cells and the levels of IL-10 and TGF-β1 were significantly increased, and the levels of IL-6 were significantly reduced. Conclusions HA15 can improve the differentiation and function of Treg cells by inhibiting the HSPA1A/CHIP pathway, thereby improving ulcerative colitis. Therefore, inhibiting the expression of HSPA5 may serve as a new approach to treat ulcerative colitis.
Collapse
|
8
|
Lin S, Que Y, Que C, Li F, Deng M, Xu D. Exosome miR-3184-5p inhibits gastric cancer growth by targeting XBP1 to regulate the AKT, STAT3, and IRE1 signalling pathways. Asia Pac J Clin Oncol 2022; 19:e27-e38. [PMID: 35394683 DOI: 10.1111/ajco.13663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/12/2021] [Accepted: 08/09/2021] [Indexed: 11/30/2022]
Abstract
MicroRNAs can regulate the transcription of protein-coding genes associated with the development and progression of cancer. In this study, we explored the potential diagnostic function of exosome miR-3184-5p in gastric cancer. This exosome was isolated from the blood samples of 150 patients with gastric cancer and 60 healthy participants. The mean particle size and concentration of serum exosome in the patients with gastric cancer were 104.6 nm (93.97-115.84) and 6.21e+009 particles/ml (5.15e+009-7.12e+009), respectively. miR-3184-5p expression was substantially downregulated in the patients with gastric cancer compared with that in the healthy participants. The gastric cancer cell line HGC-27 was cultured and transfected with the mimic and an inhibitor to overexpress and inhibit miR-3184-5p expression. miR-3184-5p strongly suppressed cell proliferation, migration, and invasion but induced cell apoptosis. Luciferase reporter assay revealed that XBP1 was the target of miR-3184-5p. miR-3184-5p substantially downregulated the expression of CD44, cyclin D1, MMP2, p65, p-AKT, and p-STAT3 but upregulated that of GRP78, IRE1, p-JNK, and CHOP. Moreover, miR-3184-5p cleaved caspase-12 and inhibited BCL-2 expression. These results suggested that the downregulation of miR-3184-5p in patients with gastric cancer might regulate the AKT, STAT3, and IRE1 pathways to promote the vitality of gastric cancer cells.
Collapse
Affiliation(s)
- Shuangming Lin
- Department of Gastrointestinal and Anal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| | - Yonggu Que
- Department of Gastrointestinal and Anal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| | - Changrong Que
- Department of Gastrointestinal and Anal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| | - Fudi Li
- Department of Gastrointestinal and Anal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| | - Maoqing Deng
- Department of laboratory, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| | - Dongbo Xu
- Department of Gastrointestinal and Anal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| |
Collapse
|
9
|
Jianpi Qingchang Decoction Ameliorates Chronic Colitis in Piroxicam-Induced IL-10 Knockout Mice by Inhibiting Endoplasmic Reticulum Stress. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7378807. [PMID: 35186102 PMCID: PMC8849791 DOI: 10.1155/2022/7378807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/06/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
Abstract
Background Excessive endoplasmic reticulum (ER) stress in intestinal epithelial cells (IEC) may lead to impaired intestinal mucosal barrier function and then participate in the pathogenesis of ulcerative colitis (UC). Jianpi Qingchang decoction (JPQCD) has been shown to have protective effects on UC. However, further studies are needed to determine whether JPQCD regulates PERK/eIF2α/ATF4/CHOP pathways to play a role in treating UC. Methods IL-10−/− mice were randomly assigned into five groups: control, model, low-dose JPQCD (JPQCD L), middle-dose JPQCD (JPQCD M), and high-dose JPQCD (JPQCD H). All groups except for the control group were given model feed containing 200 ppm piroxicam for 10 d to induce colitis. As a comparison, we used wild-type mice that were the progeny of IL-10+/− matings, bred in the same facility. The control group and wild-type mice were fed with common feed. At the same time, mice in each group were given corresponding drugs by gavage for 14 d. The disease activity index of mice in each group was evaluated daily. Colon tissues of mice were collected, colon length was measured, and pathological changes and ultrastructure of colon epithelial cells were observed. The effects of JPQCD on the PERK/eIF2α/ATF4/CHOP pathways were evaluated by western blotting and reverse transcription-polymerase chain reaction (RT-PCR). The expression of CHOP in colon tissue was detected by tissue immunofluorescence assay. The expression of NF-κB, p-NF-κB p65 protein was analyzed by western blotting; the level of IL-17 in colon tissue was detected by enzyme-linked immunosorbent assay (ELISA) and verified by examining NF-κB and IL-17 mRNA levels by RT-PCR. Results Compared with the control group, the model group showed significant colitis symptoms and severe colonic tissue damage. The results showed that JPQCD significantly reduced body weight loss, ameliorated disease activity index, and restored colon length in IL-10−/− mice with piroxicam-induced colitis. Western blotting and RT-PCR showed that the PERK/eIF2α/ATF4/CHOP pathway was activated in colon tissue of model mice, suggesting that the pathway is involved in the pathogenesis of ulcerative colitis (UC) and could become a potential therapeutic target. The JPQCD treatment inhibited the activation of the PERK/eIF2α/ATF4/CHOP pathway, alleviated the ER stress, and played a role in preventing and treating UC. In addition, JPQCD can also downregulate the protein of NF-κB, p-NF-κB p65, downregulate the mRNA expression of NF-κB, and reduce the content of IL-17 and its mRNA expression in colon tissues. Conclusion JPQCD may play a protective role in UC by regulating the PERK/eIF2α/ATF4/CHOP signaling pathway and relieving endoplasmic reticulum stress.
Collapse
|
10
|
Zhang B, Su X, Xie Z, Ding H, Wang T, Xie R, Wen Z. Inositol-Requiring Kinase 1 Regulates Apoptosis via Inducing Endoplasmic Reticulum Stress in Colitis Epithelial Cells. Dig Dis Sci 2021; 66:3015-3025. [PMID: 33043405 DOI: 10.1007/s10620-020-06622-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 09/16/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Endoplasmic reticulum stress (ERS) has been studied as critical factor during occurrence and development of ulcerative colitis (UC). However, the role of ERS in inflamed UC remains unclear. AIMS The purpose of this study was to analyze the role of inositol-requiring kinase 1 (IRE-1), a major regulator of ER, in regulating ERS and cell viability. METHODS In UC mucosa tissue, IRE-1, BiP, XBP-1s, CHOP caspase-12 and GADD34 mRNA were assayed by qRT-PCR. Then, human normal colon epithelial cell line (NCM-460) and colon fibroblast cell line (CCD-33Co) were cultured, and downregulated or upregulated IRE-1 expression. ERS was induced with 100 ng/mL of Interleukin 6 (IL-6). CCK8 assay was performed to analyze cell proliferation. Flow cytometry analysis was conducted to detect the apoptosis. Western blot assay was used to examine ERS markers. RESULTS IRE-1, BiP, XBP-1s, caspase-12 and CHOP mRNA were highly expressed in UC mucosa tissue, and the expression of GADD34 mRNA significantly decreased. These results show that ERS-induced unfolded protein response was enhanced in UC mucosa tissue. In cells, silencing the expression of IRE-1 could suppress cell proliferation and promote apoptosis through activating unfolded protein response, while the over-expression of IRE-1 had the opposite effect. IL-6 could induce ERS and cells apoptosis. Furthermore, we demonstrated that shRNA IRE-1 could enhance the inhibition of IL-6 on cells viability. CONCLUSIONS Inhibition of IRE-1 enhanced unfolded protein response and cells apoptosis and IL-6-induced ERS and suggested that IRE-1 might be a potential target of UC.
Collapse
Affiliation(s)
- Bei Zhang
- The Department of Gastroenterology, Second Hospital Affiliated to Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - XiaoYan Su
- The Department of Pathology, Second Hospital Affiliated to Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - ZhengYuan Xie
- The Department of Gastroenterology, Second Hospital Affiliated to Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Hao Ding
- The Department of Gastroenterology, Second Hospital Affiliated to Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Ting Wang
- The Department of Gastroenterology, First Hospital Affiliated to Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - RuYi Xie
- The Department of Gastroenterology, Second Hospital Affiliated to Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - ZhiLi Wen
- The Department of Gastroenterology, Second Hospital Affiliated to Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
11
|
A Gut-Ex-Vivo System to Study Gut Inflammation Associated to Inflammatory Bowel Disease (IBD). BIOLOGY 2021; 10:biology10070605. [PMID: 34209277 PMCID: PMC8301106 DOI: 10.3390/biology10070605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/17/2022]
Abstract
Simple Summary Inflammatory Bowel Disease (IBD) is a complex and multifactorial systemic disease of the gastrointestinal tract, characterized by chronic inflammation, thus resulting in tissue damage and, occasionally, in cancer development. Although the precise origin is still elusive, it is widely considered a disease of modern society, caused by a complex interaction between environment, genetic, immune system, and gut microflora (microbiota). Potentially affected by all the above-mentioned variables, which interplay are highly heterogeneous, the disease appears to be patient-specific. The latter phenomenon, together with the uncertain origin, also contributes to the lack of optimal clinical treatment of these patients. Therefore, the development of appropriate models is crucial to push the research forward and to define new valuable therapeutic approaches. Although tissue biopsies and/or animal models represent the best models to study IBD onset, progression, and clinical interventions, they are both affected by limitations such as invasiveness, cost- and time-consuming, and ethical issues such as animal suffering. Here we propose a novel approach based on the cultivation of mouse tissues (colon) in an ex vivo microfluidic device (Gut-Ex-Vivo System, GEVS) to study IBD. We demonstrate that explanted mouse tissues cultivated in our GEVS can be appropriately stimulated to recapitulate the onset of the disease, in a time- and cost- effective manner. Abstract Inflammatory bowel disease (IBD) is a complex, chronic, and dysregulated inflammatory condition which etiology is still largely unknown. Its prognosis and disease progression are highly variable and unpredictable. IBD comprises several heterogeneous inflammatory conditions ranging from Ulcerative Colitis (UC) to Crohn’s Disease (CD). Importantly, a definite, well-established, and effective clinical treatment for these pathologies is still lacking. The urgent need for treatment is further supported by the notion that patients affected by UC or CD are also at risk of developing cancer. Therefore, a deeper understanding of the molecular mechanisms at the basis of IBD development and progression is strictly required to design new and efficient therapeutic regimens. Although the development of animal models has undoubtedly facilitated the study of IBD, such in vivo approaches are often expensive and time-consuming. Here we propose an organ ex vivo culture (Gut-Ex-Vivo system, GEVS) based on colon from Balb/c mice cultivated in a dynamic condition, able to model the biochemical and morphological features of the mouse models exposed to DNBS (5–12 days), in 5 h. Indeed, upon DNBS exposure, we observed a dose-dependent: (i) up-regulation of the stress-related protein transglutaminase 2 (TG2); (ii) increased intestinal permeability associated with deregulated tight junction protein expression; (iii) increased expression of pro-inflammatory cytokines, such as TNFα, IFNγ, IL1β, IL6, IL17A, and IL15; (iv) down-regulation of the anti-inflammatory IL10; and (v) induction of Endoplasmic Reticulum stress (ER stress), all markers of IBD. Altogether, these data indicate that the proposed model can be efficiently used to study the pathogenesis of IBD, in a time- and cost-effective manner.
Collapse
|
12
|
Sittipo P, Kim HK, Han J, Lee MR, Lee YK. Vitamin D 3 suppresses intestinal epithelial stemness via ER stress induction in intestinal organoids. Stem Cell Res Ther 2021; 12:285. [PMID: 33985576 PMCID: PMC8117327 DOI: 10.1186/s13287-021-02361-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/28/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Vitamin D3 is important for normal function of the intestinal epithelial cells (IECs). In this study, we aimed to investigate the effects of vitamin D3 on the differentiation, stemness, and viability of healthy IECs in intestinal organoids. METHODS Intestinal organoids derived from mouse small intestine were treated with vitamin D3, and the effects on intestinal stemness and differentiation were evaluated using real-time PCR and immunofluorescence staining of the distinct lineage markers. Cell viability was analyzed using viability and apoptosis assays. RESULTS Vitamin D3 enhanced IEC differentiation into the distinct lineages of specialized IECs, including Paneth, goblet, and enteroendocrine cells and absorptive enterocytes. Decreased expression levels of leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5) and the presence of several LGR5-green fluorescent protein (GFP)-positive cells were observed in vitamin D3-treated organoids derived from LGR5-GFP mice. The formation of the crypt-villus structure was also inhibited by vitamin D3, suggesting that vitamin D3 suppresses intestinal cell stemness. Furthermore, the expression levels of unfolded protein response genes, C/EBP homologous protein (CHOP), and activating transcription factor 6 (ATF6) were upregulated in vitamin D3-treated organoids. Moreover, vitamin D3 promoted apoptotic cell death in intestinal cells, which may be associated with the decrease in intestinal stemness. LGR5 gene expression, ISC number, and apoptotic cell death were partially recovered in the presence of the ER stress inhibitor tauroursodeoxycholic acid (TUDCA), suggesting that intestinal stemness suppression and intestinal apoptosis occurred via ER stress activation. CONCLUSIONS Our study provides important insights into the effects of vitamin D3 on the induction of IEC differentiation and apoptotic cell death, and inhibition of intestinal stemness accompanied by ER stress augmentation.
Collapse
Affiliation(s)
- Panida Sittipo
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Hyun Kyu Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Jaeseok Han
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Man Ryul Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| | - Yun Kyung Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| |
Collapse
|
13
|
Yin S, Li L, Tao Y, Yu J, Wei S, Liu M, Li J. The Inhibitory Effect of Artesunate on Excessive Endoplasmic Reticulum Stress Alleviates Experimental Colitis in Mice. Front Pharmacol 2021; 12:629798. [PMID: 33767628 PMCID: PMC7985062 DOI: 10.3389/fphar.2021.629798] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
Endoplasmic reticulum (ER) stress may contribute to the pathogenesis and perpetuation of ulcerative colitis (UC). Previous studies have shown artesuante (ARS) has the protective effect on experimental UC. Therefore, it can be assumed that ARS can regulate ER stress and its related reactions. Dextran sulfate sodium (DSS) induced UC model in mice was used to testify this hypothesis. The results clearly showed that DSS exposure caused excessive ER stress evidenced by a markedly increase of GRP78 and CHOP expression, and then activated the ER stress sensors PERK, IRE1, ATF6 and their respective signaling pathways, followed by upregulated caspases12 and lowered Bcl-2/Bax ratio. However, ARS treatment significantly inhibited the occurrence of ER stress via preventing the activation of PERK-eIF2α-ATF4-CHOP and IRE1α-XBP1 signaling pathways, concurrently ER-stress-associated apoptosis in colon tissues. Moreover, ARS treatment remarkably inhibited the activation of NF-κB and the expression levels of pro-inflammatory cytokines, improved the clinical and histopathological alterations as well as maintained the expression of claudin-1 and Muc2 in mucosal layer of colon. Notably, the classic ER stress inhibitor 4-phenyhlbutyric acid enhanced the beneficial effects of ARS; in contrast, the ER stress inducer 2-deoxy-d-glucose substantially abrogated the above-mentioned effects, uncovering the involvement of ER stress in the response. These findings indicated the protection of ARS on UC is associated with its suppressing excessive ER stress mediated intestinal barrier damage and inflammatory response. This study provides a novel aspect to understand the mechanism of ARS against UC.
Collapse
Affiliation(s)
- Shaojie Yin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Liuhui Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Ya Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jie Yu
- The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Simin Wei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Mingjiang Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jingui Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
14
|
Wang Q, Li Z, Liu K, Liu J, Chai S, Chen G, Wen S, Ming T, Wang J, Ma Y, Zeng H, Liu C, Xue B. Activation of the G Protein-Coupled Estrogen Receptor Prevented the Development of Acute Colitis by Protecting the Crypt Cell. J Pharmacol Exp Ther 2020; 376:281-293. [PMID: 33318078 DOI: 10.1124/jpet.120.000216] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
G protein-coupled estrogen receptor (GPER) might be involved in ulcerative colitis (UC), but the direct effect of GPER on UC is still unclear. We used male C57BL/6 mice to establish the acute colitis model with administration of dextran sulfate sodium and explored the effect of GPER on acute colitis and its possible mechanism. The selective GPER agonist G-1 inhibited weight loss and colon shortening and decreased the disease activity index for colitis and histologic damage in mice with colitis. All of these effects were prevented by a selective GPER blocker. G-1 administration prevented the dysfunction of tight junction protein expression and goblet cells in colitis model and thus inhibited the increase of mucosal permeability in colitis-suffering mice significantly. GPER activation reduced expression of glucose-regulating peptide-78 and anti-CCAAT/enhancer-binding protein homologous protein and attenuated the three arms of the unfolded protein response in colitis. G-1 therapy inhibited the increase of cleavage caspase-3- and TUNEL-positive cells in colonic crypts in the colitis model, increased the number of Ki67- and bromodeoxyuridine-positive cells in crypts, and reversed the decrease of cyclin D1 and cyclin B1 expression in colitis, indicating its protective effect on crypt cells. In cultured CCD841 cells, G-1 treatment fought against cell injury induced by endoplasmic reticulum stress. These findings demonstrate that GPER activation prevents colitis by protecting the colonic crypt cells, which are associated with inhibition of endoplasmic reticulum stress. SIGNIFICANCE STATEMENT: We demonstrate that G protein-coupled estrogen receptor (GPER) activation prevents dextran sulfate sodium-induced acute colitis by protecting the crypt cells, showing that it inhibited the crypt cell apoptosis and protected proliferation of crypt cells, which resulted in protection of the intestinal mucosal barrier. This protective effect was achieved (at least in part) by inhibiting endoplasmic reticulum stress. Mucosal healing is regarded as a key therapeutic target for colitis, and GPER is expected to become a new therapeutic target for colitis.
Collapse
Affiliation(s)
- Qian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Zhao Li
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Kaixuan Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Jianbo Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Shiquan Chai
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Guanyu Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Shuyu Wen
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Tian Ming
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Jiayi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Yuntao Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Honghui Zeng
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Chuanyong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| | - Bing Xue
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China (Q.W., Z.L., K.L., J.L., S.C., G.C., S.W., T.M., H.Z., C.L., B.X.) and Second Clinical Medical College, Lanzhou University, Lanzhou, China (Y.M.)
| |
Collapse
|
15
|
Gao S, Cheng QC, Hu YG, Tan ZZ, Chen L, Liu SW, Kang QY, Wei T. LncRNA AK148321 alleviates neuroinflammation in LPS-stimulated BV2 microglial cell through regulating microRNA-1199-5p/HSPA5 axis. Life Sci 2020; 266:118863. [PMID: 33301806 DOI: 10.1016/j.lfs.2020.118863] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023]
Abstract
AIMS Dysregulated long non-coding RNA (lncRNA) expression is closely related to neuroinflammation, leading to multiple neurodegenerative diseases. In this study, we investigated the function and regulation of lncRNA AK148321 in neuroinflammation using an in vitro lipopolysaccharide (LPS)-stimulated BV2 microglial cell system. METHODS Expression of AK148321 was analyzed by qPCR. Inflammatory cytokine expression levels were determined by ELISA assay. The interaction between AK148321, microRNA (miRNA), and its target gene was validated by luciferase reporter assay and RNA immunoprecipitation (RIP). Cell apoptosis was analyzed by Annexin V/PI staining. RESULTS LPS treatment suppressed AK148321 expression in BV2 cells. Overexpression of AK148321 inhibited LPS-induced BV2 microglial cell activation and decreased the expression of inflammatory cytokine TNF-α and IL-1β. AK148321 function as a competing endogenous RNA (ceRNA) by sponging microRNA-1199-5p (MiR-1199-5p). In LPS-stimulated BV2 cells, AK148321 exerted its inhibitory function via negatively modulating miR-1199-5p expression. Moreover, we identified that Heat Shock Protein Family A Member 5 (HSPA5) was a direct target of miR-1199-5p. RIP assay using the anti-Ago2 antibody further validated the relationship among AK148321, miR-1199-5p and HSPA5. The AK148321/miR-1199-5p/HSPA5 axis regulated the neuroinflammation in LPS-induced BV2 microglial cells. Microglial cell culture supernatant from LPS-stimulated, AK148321-overexpressing BV2 cells suppressed the cell apoptosis of mouse hippocampal neuronal cell HT22, while HSPA5 knockdown abrogated the suppression effect. CONCLUSION Our findings suggest that AK148321 alleviates neuroinflammation in LPS-stimulated BV2 microglial cells through miR-1199-5p/HSPA5 axis.
Collapse
Affiliation(s)
- Shan Gao
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Qiao-Chu Cheng
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Ya-Guang Hu
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Zi-Zhu Tan
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Li Chen
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Si-Wei Liu
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Qian-Yan Kang
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Ting Wei
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
16
|
Song C, Chen J, Li X, Yang R, Cao X, Zhou L, Zhou Y, Ying H, Zhang Q, Sun Y. Limonin ameliorates dextran sulfate sodium-induced chronic colitis in mice by inhibiting PERK-ATF4-CHOP pathway of ER stress and NF-κB signaling. Int Immunopharmacol 2020; 90:107161. [PMID: 33168409 DOI: 10.1016/j.intimp.2020.107161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/22/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal inflammation regulated by intricate mechanisms. Limonin, a natural tetracyclic triterpenoid compound, possesses multiple bioactivities including anti-inflammation, anti-cancer and so on. However, the therapeutic potential and the underlying mechanism of limonin on IBD remain unclear. Here, we probe into the effect of limonin on chronic colitis induced by dextran sulfate sodium (DSS) and illustrated the potential mechanisms. We found that limonin relieved the risk and severity of DSS-induced chronic colitis in mice through various aspects including increasing body weight and colon length, decreasing the mortality rate, inhibiting MPO activity and improving colon pathology. Limonin also decreased the production of proinflammatory cytokines TNF-α, IL-1β, IL-6 and the expression of inflammatory proteins COX-2, iNOS in colon tissues from DSS-induced colitis mice. Moreover, limonin attenuated DSS-induced chronic colitis by inhibiting PERK-ATF4-CHOP pathway of endoplasmic reticulum (ER) stress and NF-κB signaling. In vitro, limonin not only decreased LPS-induced higher production of pro-inflammatory cytokines and inflammatory proteins mentioned above by inhibiting NF-κB signaling in macrophage cells RAW264.7, but also suppressed PERK-ATF4-CHOP pathway of ER stress. In summary, our study demonstrated that limonin mitigated DSS-induced chronic colitis via inhibiting PERK-ATF4-CHOP pathway of ER stress and NF-κB signaling. All of this study provides the possibility for limonin as an effective drug for chronic colitis of IBD in the future.
Collapse
Affiliation(s)
- Changqin Song
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, People's Republic of China; College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, People's Republic of China
| | - Jiaxi Chen
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, People's Republic of China
| | - Xiaotian Li
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, People's Republic of China
| | - Runyu Yang
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, People's Republic of China
| | - Xiaomei Cao
- Department of Pharmacology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, People's Republic of China
| | - Lvqi Zhou
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, People's Republic of China
| | - Yanfen Zhou
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, People's Republic of China
| | - Hanjie Ying
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, People's Republic of China
| | - Qi Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, People's Republic of China.
| | - Yang Sun
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, People's Republic of China.
| |
Collapse
|
17
|
Błochowiak K, Celichowski P, Kempisty B, Iwanik K, Nowicki M. Transcriptomic Profile of Genes Encoding Proteins Involved in Pathogenesis of Sjögren's Syndrome Related Xerostomia-Molecular and Clinical Trial. J Clin Med 2020; 9:jcm9103299. [PMID: 33066537 PMCID: PMC7602267 DOI: 10.3390/jcm9103299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/01/2020] [Accepted: 10/10/2020] [Indexed: 01/14/2023] Open
Abstract
Sjögren’s syndrome (SS) is characterized by xerostomia. We aimed to investigate and compare gene expressions in the labial salivary glands of SS patients with xerostomia SS (sicca) and without xerostomia SS (non-sicca) and of healthy subjects (HS) by means of microarray analysis, and to find genes involved in xerostomia. The study group comprised 11 SS patients (3 SS (sicca) and 8 SS (non-sicca)) and 9 HS. The relative gene expression changes were validated with RT-qPCR in the larger study group. Among the differently expressed genes belonging to the “secretion” ontology group with a fold change >2 and with a p value < 0.05, the Transmembrane P24 Trafficking Protein 10 (TMED10), Protein Disulfide Isomerase Family A Member 4 (PDIA4), Calnexin (CANX), Amyloid Beta Precursor Protein (APP), and Transmembrane BAX Inhibitor Motif Containing 6 (TMBIM6) gene expressions in both SS (sicca) and SS (non-sicca) groups were lower than in HS. Significant correlations were observed between TMED10, PDIA4, and CANX gene expression in SS (sicca) patients compared to the controls. There were no differences between the SS (sicca) and SS (non-sicca) study groups in the expression of the aforementioned genes. Results indicate their role in the endoplasmic reticulum system, their overlapping function and the loss of the APP neuroprotective function in xerostomia. It has a multifactorial origin and can be triggered by disturbances to the various signaling pathways in saliva secretion.
Collapse
Affiliation(s)
- Katarzyna Błochowiak
- Department of Oral Surgery and Periodontology, Poznan University of Medical Sciences, 61-812 Poznan, Poland
- Correspondence: ; Tel.: +48-608-836-850
| | - Piotr Celichowski
- Department of Histology and Embryology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (P.C.); (B.K.); (M.N.)
| | - Bartosz Kempisty
- Department of Histology and Embryology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (P.C.); (B.K.); (M.N.)
- Department of Anatomy, Poznan University of Medical Sciences, 61-701 Poznan, Poland
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Katarzyna Iwanik
- Department of Clinical Pathomorphology, Poznan University of Medical Sciences, 60-355 Poznan, Poland;
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (P.C.); (B.K.); (M.N.)
| |
Collapse
|
18
|
Shastri S, Shinde T, Perera AP, Gueven N, Eri R. Idebenone Protects against Spontaneous Chronic Murine Colitis by Alleviating Endoplasmic Reticulum Stress and Inflammatory Response. Biomedicines 2020; 8:biomedicines8100384. [PMID: 32998266 PMCID: PMC7601570 DOI: 10.3390/biomedicines8100384] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/18/2022] Open
Abstract
Endoplasmic reticulum (ER) stress in intestinal secretory goblet cells has been linked to the development of ulcerative colitis (UC). Emerging evidence suggests that the short chain quinone drug idebenone displays anti-inflammatory activity in addition to its potent antioxidant and mitochondrial electron donor properties. This study evaluated the impact of idebenone in Winnie mice, that are characterized by spontaneous chronic intestinal inflammation and ER stress caused by a missense mutation in the mucin MUC2 gene. Idebenone (200 mg/kg) was orally administered daily to 5-6 weeks old Winnie mice over a period of 21 days. Idebenone treatment substantially improved body weight gain, disease activity index (DAI), colon length and histopathology score. Immunohistochemistry revealed increased expression of MUC2 protein in goblet cells, consistent with increased MUC2 mRNA levels. Furthermore, idebenone significantly reduced the expression of the ER stress markers C/EBP homologous protein (CHOP), activating transcription factor 6 (ATF6) and X-box binding protein-1 (XBP-1) at both mRNA and protein levels. Idebenone also effectively reduced pro-inflammatory cytokine levels in colonic explants. Taken together, these results indicate that idebenone could represent a potential therapeutic approach against human UC by its strong anti-inflammatory activity and its ability to reduce markers of ER stress.
Collapse
Affiliation(s)
- Sonia Shastri
- Gut Health Laboratory, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston 7250, Tasmania, Australia; (T.S.); (A.P.P.)
- Correspondence: (S.S.); (R.E.); Tel.: +61-4-4992-4236 (S.S.); +61-3-6226-5017 (R.E.)
| | - Tanvi Shinde
- Gut Health Laboratory, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston 7250, Tasmania, Australia; (T.S.); (A.P.P.)
- Centre for Food Innovation, Tasmanian Institute of Agriculture, University of Tasmania, Launceston 7250, Tasmania, Australia
| | - Agampodi Promoda Perera
- Gut Health Laboratory, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston 7250, Tasmania, Australia; (T.S.); (A.P.P.)
| | - Nuri Gueven
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart 7005, Tasmania, Australia;
| | - Rajaraman Eri
- Gut Health Laboratory, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston 7250, Tasmania, Australia; (T.S.); (A.P.P.)
- Correspondence: (S.S.); (R.E.); Tel.: +61-4-4992-4236 (S.S.); +61-3-6226-5017 (R.E.)
| |
Collapse
|
19
|
Gao F, Fan H. Heat shock protein 5 and inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2020; 28:802-806. [DOI: 10.11569/wcjd.v28.i16.802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease is a kind of chronic recurrent intestinal inflammatory disease whose occurrence and development are affected by the integrity of the mucosal barrier. As the main component of the mucosal barrier, intestinal epithelial cells mainly include Paneth cells, goblet cells, etc. Heat shock protein 5 is a key factor for endoplasmic reticulum stress, and it affects the survival and apoptosis of intestinal epithelial cells mainly through endoplasmic reticulum stress pathways, and then participates in the process of inflammatory bowel disease.
Collapse
Affiliation(s)
- Fei Gao
- Department of Integrated Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Heng Fan
- Department of Integrated Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
20
|
Intestinal Immune Homeostasis and Inflammatory Bowel Disease: A Perspective on Intracellular Response Mechanisms. GASTROINTESTINAL DISORDERS 2020. [DOI: 10.3390/gidisord2030024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of inflammatory bowel disease (IBD) involves perturbation of intestinal immune homeostasis in genetically susceptible individuals. A mutual interplay between intestinal epithelial cells (IECs) and gut resident microbes maintains a homeostatic environment across the gut. An idiopathic gastrointestinal (GI) complication triggers aberrant physiological stress in the epithelium and peripheral myeloid cells, leading to a chronic inflammatory condition. Indeed, events in the endoplasmic reticulum (ER) and mitochondria contribute to orchestrating intracellular mechanisms such as the unfolded protein response (UPR) and oxidative stress, respectively, to resolve aberrant cellular stress. This review highlights the signaling cascades encrypted within ER and mitochondria in IECs and/or myeloid cells to dissipate chronic stress in maintaining intestinal homeostasis.
Collapse
|
21
|
Rees WD, Stahl M, Jacobson K, Bressler B, Sly LM, Vallance BA, Steiner TS. Enteroids Derived From Inflammatory Bowel Disease Patients Display Dysregulated Endoplasmic Reticulum Stress Pathways, Leading to Differential Inflammatory Responses and Dendritic Cell Maturation. J Crohns Colitis 2020; 14:948-961. [PMID: 31796949 DOI: 10.1093/ecco-jcc/jjz194] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Endoplasmic reticulum [ER] stress in intestinal epithelial cells [IECs] contributes to the pathogenesis of inflammatory bowel disease [IBD]. We hypothesized that ER stress changes innate signalling in human IECs, augmenting toll-like receptor [TLR] responses and inducing pro-inflammatory changes in underlying dendritic cells [DCs]. METHODS Caco-2 cells and primary human colon-derived enteroid monolayers were exposed to ATP [control stressor] or thapsigargin [Tg] [ER stress inducer], and were stimulated with the TLR5 agonist flagellin. Cytokine release was measured by an enzyme immunoassay. ER stress markers CHOP, GRP78 and XBP1s/u were measured via quantitative PCR and Western blot. Monocyte-derived DCs [moDCs] were cultured with the IEC supernatants and their activation state was measured. Responses from enteroids derived from IBD patients and healthy control participants were compared. RESULTS ER stress enhanced flagellin-induced IL-8 release from Caco-2 cells and enteroids. Moreover, conditioned media activated DCs to become pro-inflammatory, with increased expression of CD80, CD86, MHCII, IL-6, IL-15 and IL-12p70 and decreased expression of CD103 and IL-10. Flagellin-induced IL-8 production correlated with DC activation, suggesting a common stress pathway. Moreover, there were distinct differences in cytokine expression and basal ER stress between IBD and healthy subject-derived enteroid monolayers, suggesting a dysregulated ER stress pathway in IBD-derived enteroids. CONCLUSIONS Cellular stress enhances TLR5 responses in IECs, leading to increased DC activation, indicating a previously unknown mechanistic link between epithelial ER stress and immune activation in IBD. Furthermore, dysregulated ER stress may be propagated from the intestinal epithelial stem cell niche in IBD patients.
Collapse
Affiliation(s)
- William D Rees
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Martin Stahl
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Kevan Jacobson
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Brian Bressler
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Providence Health, Vancouver, BC, Canada
| | - Laura M Sly
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Bruce A Vallance
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Theodore S Steiner
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada
| |
Collapse
|
22
|
Dextran Sodium Sulfate-Induced Impairment of Protein Trafficking and Alterations in Membrane Composition in Intestinal Caco-2 Cell Line. Int J Mol Sci 2020; 21:ijms21082726. [PMID: 32326391 PMCID: PMC7215722 DOI: 10.3390/ijms21082726] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/09/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022] Open
Abstract
A key morphological feature of inflammatory bowel disease (IBD) is the loss of the barrier function of intestinal epithelial cells. The present study investigates endoplasmic reticulum (ER) stress in addition to alterations in protein and membrane trafficking in a dextran sulfate sodium (DSS)-induced IBD-like phenotype of intestinal Caco-2 cells in culture. DSS treatment significantly reduced the transepithelial electric resistance (TEER) and increased the epithelial permeability of Caco-2 cells, without affecting their viability. This was associated with an alteration in the expression levels of inflammatory factors in addition to an increase in the expression of the ER stress protein markers, namely immunoglobulin-binding protein (BiP), C/EBP homologous protein (CHOP), activation transcription factor 4 (ATF4), and X-box binding protein (XBP1). The DSS-induced ER-stress resulted in impaired intracellular trafficking and polarized sorting of sucrase-isomaltase (SI) and dipeptidyl peptidase-4 (DPPIV), which are normally sorted to the apical membrane via association with lipid rafts. The observed impaired sorting was caused by reduced cholesterol levels and subsequent distortion of the lipid rafts. The data presented confirm perturbation of ER homeostasis in DSS-treated Caco-2 cells, accompanied by impairment of membrane and protein trafficking resulting in altered membrane integrity, cellular polarity, and hence disrupted barrier function.
Collapse
|
23
|
ER stress activation in the intestinal mucosa but not in mesenteric adipose tissue is associated with inflammation in Crohn's disease patients. PLoS One 2019; 14:e0223105. [PMID: 31557250 PMCID: PMC6762147 DOI: 10.1371/journal.pone.0223105] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/12/2019] [Indexed: 12/21/2022] Open
Abstract
Chronic/abnormal activation of endoplasmic reticulum (ER) stress is linked to the exacerbation of the inflammatory process and has been recently linked to Crohn’s disease (CD) pathophysiology. We investigated the intestinal mucosa and the mesenteric adipose tissue (MAT) collected from CD patients with active disease (CD group) and from non-IBD patients (CTR group) to study ER stress activation and to address tissue-specific modulation in CD. The intestinal mucosa of CD patients showed an upregulation in the expression of ER stress related genes, including ATF3, DNAJC3, STC2, DDIT3, CALR, HSPA5 and HSP90B1. Results showed that EIF2AK3 gene was upregulated, along with increased protein expression of p-eIF2α and p-eIF2α/eIF2α ratio. Additionally, ERN1 gene expression was upregulated, along with an increased spliced/activated form sXBP1 protein. Despite the upregulation of ATF6 gene expression in the intestinal mucosa of CD patients, no differences were found in ATF6 protein expression. Lastly, the analysis of MAT revealed unchanged levels of ER stress markers along with no differences in the activation of UPR. However, chaperone gene expression was modulated in the MAT of CD patients. To conclude, our results address tissue-specific differences in UPR activation in CD and point the ER stress as an important pro-inflammatory mechanism in CD, specifically in the intestinal mucosa.
Collapse
|
24
|
Markota A, Metzger R, Heiseke AF, Jandl L, Dursun E, Eisenächer K, Reindl W, Haller D, Krug AB. Comparison of iron-reduced and iron-supplemented semisynthetic diets in T cell transfer colitis. PLoS One 2019; 14:e0218332. [PMID: 31276514 PMCID: PMC6611680 DOI: 10.1371/journal.pone.0218332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/30/2019] [Indexed: 12/18/2022] Open
Abstract
Clinical observations in inflammatory bowel disease patients and experimental studies in rodents suggest that iron in the intestinal lumen derived from iron-rich food or oral iron supplementation could exacerbate inflammation and that iron depletion from the diet could be protective. To test the hypothesis that dietary iron reduction is protective against colitis development, the impact of iron reduction in the diet below 10 mg/kg on the course of CD4+ CD62L+ T cell transfer colitis was investigated in adult C57BL/6 mice. Weight loss as well as clinical and histological signs of inflammation were comparable between mice pretreated with semisynthetic diets with either < 10mg/kg iron content or supplemented with 180 mg/kg iron in the form of ferrous sulfate or hemin. Accumulation and activation of Ly6Chigh monocytes, changes in dendritic cell subset composition and induction of proinflammatory Th1/Th17 cells in the inflamed colon were not affected by the iron content of the diets. Thus, dietary iron reduction did not protect adult mice against severe intestinal inflammation in T cell transfer induced colitis.
Collapse
Affiliation(s)
- Anamarija Markota
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Rebecca Metzger
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Alexander F. Heiseke
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Lisa Jandl
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Ezgi Dursun
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Katharina Eisenächer
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Wolfgang Reindl
- Klinikum Mannheim, II. Medizinische Klinik, Mannheim, Germany
| | - Dirk Haller
- Chair for Nutrition and Immunology, Technical University Munich, Freising, Germany
| | - Anne B. Krug
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
- * E-mail:
| |
Collapse
|
25
|
Grootjans J, Krupka N, Hosomi S, Matute JD, Hanley T, Saveljeva S, Gensollen T, Heijmans J, Li H, Limenitakis JP, Ganal-Vonarburg SC, Suo S, Luoma AM, Shimodaira Y, Duan J, Shih DQ, Conner ME, Glickman JN, Fuhler GM, Palm NW, de Zoete MR, van der Woude CJ, Yuan GC, Wucherpfennig KW, Targan SR, Rosenstiel P, Flavell RA, McCoy KD, Macpherson AJ, Kaser A, Blumberg RS. Epithelial endoplasmic reticulum stress orchestrates a protective IgA response. Science 2019; 363:993-998. [PMID: 30819965 PMCID: PMC6637967 DOI: 10.1126/science.aat7186] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 09/16/2018] [Accepted: 02/08/2019] [Indexed: 12/25/2022]
Abstract
Immunoglobulin A (IgA) is the major secretory immunoglobulin isotype found at mucosal surfaces, where it regulates microbial commensalism and excludes luminal factors from contacting intestinal epithelial cells (IECs). IgA is induced by both T cell-dependent and -independent (TI) pathways. However, little is known about TI regulation. We report that IEC endoplasmic reticulum (ER) stress induces a polyreactive IgA response, which is protective against enteric inflammation. IEC ER stress causes TI and microbiota-independent expansion and activation of peritoneal B1b cells, which culminates in increased lamina propria and luminal IgA. Increased numbers of IgA-producing plasma cells were observed in healthy humans with defective autophagy, who are known to exhibit IEC ER stress. Upon ER stress, IECs communicate signals to the peritoneum that induce a barrier-protective TI IgA response.
Collapse
Affiliation(s)
- Joep Grootjans
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
- Amsterdam University Medical Center, University of Amsterdam, Department of Gastroenterology and Hepatology and Tygat Institute for Liver and Intestinal Research, Meibergdreef 9, Amsterdam, Netherlands
| | - Niklas Krupka
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
- Maurice Müller Laboratories (DBMR), Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, Murtenstrasse 35, University of Bern, 3010 Bern, Switzerland
| | - Shuhei Hosomi
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3, Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Juan D Matute
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
- Division of Neonatology, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Thomas Hanley
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Svetlana Saveljeva
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Thomas Gensollen
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Jarom Heijmans
- Amsterdam University Medical Center, University of Amsterdam, Department of Internal Medicine, Tygat Institute for Liver and Intestinal Research, Meibergdreef 9, Amsterdam, Netherlands
| | - Hai Li
- Maurice Müller Laboratories (DBMR), Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, Murtenstrasse 35, University of Bern, 3010 Bern, Switzerland
| | - Julien P Limenitakis
- Maurice Müller Laboratories (DBMR), Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, Murtenstrasse 35, University of Bern, 3010 Bern, Switzerland
| | - Stephanie C Ganal-Vonarburg
- Maurice Müller Laboratories (DBMR), Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, Murtenstrasse 35, University of Bern, 3010 Bern, Switzerland
| | - Shengbao Suo
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Adrienne M Luoma
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Yosuke Shimodaira
- F. Widjaja Foundation, Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jinzhi Duan
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - David Q Shih
- F. Widjaja Foundation, Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Margaret E Conner
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jonathan N Glickman
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Marcel R de Zoete
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - C Janneke van der Woude
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Stephan R Targan
- F. Widjaja Foundation, Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, Rosalind-Franklin-Str. 12, 24105 Kiel, Germany
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Kathy D McCoy
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Andrew J Macpherson
- Maurice Müller Laboratories (DBMR), Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, Murtenstrasse 35, University of Bern, 3010 Bern, Switzerland
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Junjappa RP, Patil P, Bhattarai KR, Kim HR, Chae HJ. IRE1α Implications in Endoplasmic Reticulum Stress-Mediated Development and Pathogenesis of Autoimmune Diseases. Front Immunol 2018; 9:1289. [PMID: 29928282 PMCID: PMC5997832 DOI: 10.3389/fimmu.2018.01289] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 05/22/2018] [Indexed: 12/15/2022] Open
Abstract
Inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) is the most prominent and evolutionarily conserved endoplasmic reticulum (ER) membrane protein. This transduces the signal of misfolded protein accumulation in the ER, named as ER stress, to the nucleus as “unfolded protein response (UPR).” The ER stress-mediated IRE1α signaling pathway arbitrates the yin and yang of cell life. IRE1α has been implicated in several physiological as well as pathological conditions, including immune disorders. Autoimmune diseases are caused by abnormal immune responses that develop due to genetic mutations and several environmental factors, including infections and chemicals. These factors dysregulate the cell immune reactions, such as cytokine secretion, antigen presentation, and autoantigen generation. However, the mechanisms involved, in which these factors induce the onset of autoimmune diseases, are remaining unknown. Considering that these environmental factors also induce the UPR, which is expected to have significant role in secretory cells and immune cells. The role of the major UPR molecule, IRE1α, in causing immune responses is well identified, but its role in inducing autoimmunity and the pathogenesis of autoimmune diseases has not been clearly elucidated. Hence, a better understanding of the role of IRE1α and its regulatory mechanisms in causing autoimmune diseases could help to identify and develop the appropriate therapeutic strategies. In this review, we mainly center the discussion on the molecular mechanisms of IRE1α in the pathophysiology of autoimmune diseases.
Collapse
Affiliation(s)
- Raghu Patil Junjappa
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| | - Prakash Patil
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| | - Kashi Raj Bhattarai
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| | - Hyung-Ryong Kim
- Graduate School, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | - Han-Jung Chae
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| |
Collapse
|
27
|
Choi S, Snider JM, Olakkengil N, Lambert JM, Anderson AK, Ross-Evans JS, Cowart LA, Snider AJ. Myristate-induced endoplasmic reticulum stress requires ceramide synthases 5/6 and generation of C14-ceramide in intestinal epithelial cells. FASEB J 2018; 32:5724-5736. [PMID: 29768040 DOI: 10.1096/fj.201800141r] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Saturated fatty acids (SFAs) have been shown to induce endoplasmic reticulum (ER) stress and chronic inflammatory responses, as well as alter sphingolipid metabolism. Disruptions in ER stress and sphingolipid metabolism have also been implicated in intestinal inflammation. Therefore, to elucidate the roles of SFAs in ER stress and inflammation in intestinal epithelial cells, we examined myristate (C14:0) and palmitate (C16:0). Myristate, but not palmitate, induced ER stress signaling, including activation of inositol-requiring enzyme 1 (IRE1) and X-box binding protein 1 (XBP1) signaling. Myristate significantly increased C14-ceramide levels, whereas palmitate increased several long-chain ceramides. To define the role of ceramide synthases (CerSs) in myristate-induced ER stress, we used the pharmacologic inhibitor, fumonisin B1 (FB1), and small interfering RNA (siRNA) for CerS5 and 6, the primary isoforms that are involved in C14-ceramide generation. FB1 and siRNA for CerS5 or 6 suppressed myristate-induced C14-ceramide generation and XBP1 splicing (XBP1s). Moreover, increased XBP1s induced the downstream expression of IL-6 in a CerS5/6-dependent manner. In addition, a myristate-enriched milk fat-based diet, but not a lard-based diet, increased C14-ceramide, XBP1s, and IL-6 expression in vivo. Taken together, our data suggest that myristate modulates ER stress and cytokine production in the intestinal epithelium via CerS5/6 and C14-ceramide generation.-Choi, S., Snider, J. M., Olakkengil, N., Lambert, J. M., Anderson, A. K., Ross-Evans, J. S., Cowart, L. A., Snider, A. J. Myristate-induced endoplasmic reticulum stress requires ceramide synthases 5/6 and generation of C14-ceramide in intestinal epithelial cells.
Collapse
Affiliation(s)
- Songhwa Choi
- Department of Biochemistry, Stony Brook University, Stony Brook, New York, USA.,Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Justin M Snider
- Department of Biochemistry, Stony Brook University, Stony Brook, New York, USA.,Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Nicole Olakkengil
- Department of Biochemistry, Stony Brook University, Stony Brook, New York, USA
| | - Johana M Lambert
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Andrea K Anderson
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jessica S Ross-Evans
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA.,Hunter Holmes McGuire Veterans' Affairs Medical Center, Richmond, Virginia, USA
| | - Ashley J Snider
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA.,Cancer Center, Stony Brook University, Stony Brook, New York, USA.,Northport Veterans Affairs Medical Center, Northport, New York, USA
| |
Collapse
|
28
|
Perinatal supplementation of 4-phenylbutyrate and glutamine attenuates endoplasmic reticulum stress and improves colonic epithelial barrier function in rats born with intrauterine growth restriction. J Nutr Biochem 2018; 55:104-112. [DOI: 10.1016/j.jnutbio.2017.12.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/30/2017] [Accepted: 12/21/2017] [Indexed: 02/07/2023]
|
29
|
Lopes F, Keita ÅV, Saxena A, Reyes JL, Mancini NL, Al Rajabi A, Wang A, Baggio CH, Dicay M, van Dalen R, Ahn Y, Carneiro MBH, Peters NC, Rho JM, MacNaughton WK, Girardin SE, Jijon H, Philpott DJ, Söderholm JD, McKay DM. ER-stress mobilization of death-associated protein kinase-1-dependent xenophagy counteracts mitochondria stress-induced epithelial barrier dysfunction. J Biol Chem 2018; 293:3073-3087. [PMID: 29317503 DOI: 10.1074/jbc.ra117.000809] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/21/2017] [Indexed: 12/14/2022] Open
Abstract
The gut microbiome contributes to inflammatory bowel disease (IBD), in which bacteria can be present within the epithelium. Epithelial barrier function is decreased in IBD, and dysfunctional epithelial mitochondria and endoplasmic reticulum (ER) stress have been individually associated with IBD. We therefore hypothesized that the combination of ER and mitochondrial stresses significantly disrupt epithelial barrier function. Here, we treated human colonic biopsies, epithelial colonoids, and epithelial cells with an uncoupler of oxidative phosphorylation, dinitrophenol (DNP), with or without the ER stressor tunicamycin and assessed epithelial barrier function by monitoring internalization and translocation of commensal bacteria. We also examined barrier function and colitis in mice exposed to dextran sodium sulfate (DSS) or DNP and co-treated with DAPK6, an inhibitor of death-associated protein kinase 1 (DAPK1). Contrary to our hypothesis, induction of ER stress (i.e. the unfolded protein response) protected against decreased barrier function caused by the disruption of mitochondrial function. ER stress did not prevent DNP-driven uptake of bacteria; rather, specific mobilization of the ATF6 arm of ER stress and recruitment of DAPK1 resulted in enhanced autophagic killing (xenophagy) of bacteria. Of note, epithelia with a Crohn's disease-susceptibility mutation in the autophagy gene ATG16L1 exhibited less xenophagy. Systemic delivery of the DAPK1 inhibitor DAPK6 increased bacterial translocation in DSS- or DNP-treated mice. We conclude that promoting ER stress-ATF6-DAPK1 signaling in transporting enterocytes counters the transcellular passage of bacteria evoked by dysfunctional mitochondria, thereby reducing the potential for metabolic stress to reactivate or perpetuate inflammation.
Collapse
Affiliation(s)
- Fernando Lopes
- From the Gastrointestinal Research Group, Departments of Physiology and Pharmacology and
| | - Åsa V Keita
- the Department of Clinical and Experimental Medicine, Division of Surgery, Linköping University, Linköping 581 83, Sweden, and
| | - Alpana Saxena
- From the Gastrointestinal Research Group, Departments of Physiology and Pharmacology and
| | - Jose Luis Reyes
- From the Gastrointestinal Research Group, Departments of Physiology and Pharmacology and
| | - Nicole L Mancini
- From the Gastrointestinal Research Group, Departments of Physiology and Pharmacology and
| | - Ala Al Rajabi
- From the Gastrointestinal Research Group, Departments of Physiology and Pharmacology and
| | - Arthur Wang
- From the Gastrointestinal Research Group, Departments of Physiology and Pharmacology and
| | - Cristiane H Baggio
- From the Gastrointestinal Research Group, Departments of Physiology and Pharmacology and
| | - Michael Dicay
- From the Gastrointestinal Research Group, Departments of Physiology and Pharmacology and
| | - Rob van Dalen
- the Departments of Laboratory Medicine and Pathobiology and
| | - Younghee Ahn
- the Departments of Pediatrics, Clinical Neurosciences, and Physiology and Pharmacology and
| | - Matheus B H Carneiro
- the Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Nathan C Peters
- the Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Jong M Rho
- the Departments of Pediatrics, Clinical Neurosciences, and Physiology and Pharmacology and
| | - Wallace K MacNaughton
- From the Gastrointestinal Research Group, Departments of Physiology and Pharmacology and
| | | | - Humberto Jijon
- Medicine, Calvin, Joan, and Phoebe Snyder Institute for Chronic Diseases, and
| | - Dana J Philpott
- Immunology, University of Toronto, Toronto, Ontario M5S1A1, Canada
| | - Johan D Söderholm
- the Department of Clinical and Experimental Medicine, Division of Surgery, Linköping University, Linköping 581 83, Sweden, and
| | - Derek M McKay
- From the Gastrointestinal Research Group, Departments of Physiology and Pharmacology and
| |
Collapse
|
30
|
Pineda E, Perdomo D. Entamoeba histolytica under Oxidative Stress: What Countermeasure Mechanisms Are in Place? Cells 2017; 6:cells6040044. [PMID: 29160807 PMCID: PMC5755502 DOI: 10.3390/cells6040044] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/17/2017] [Accepted: 11/17/2017] [Indexed: 02/06/2023] Open
Abstract
Entamoeba histolytica is the causative agent of human amoebiasis; it affects 50 million people worldwide and causes approximately 100,000 deaths per year. Entamoeba histolytica is an anaerobic parasite that is primarily found in the colon; however, for unknown reasons, it can become invasive, breaching the gut barrier and migrating toward the liver causing amoebic liver abscesses. During the invasive process, it must maintain intracellular hypoxia within the oxygenated human tissues and cellular homeostasis during the host immune defense attack when it is confronted with nitric oxide and reactive oxygen species. But how? This review will address the described and potential mechanisms available to counter the oxidative stress generated during invasion and the possible role that E. histolytica’s continuous endoplasmic reticulum (Eh-ER) plays during these events.
Collapse
Affiliation(s)
- Erika Pineda
- Laboratory of Fundamental Microbiology and Pathogenicity (MFP), University of Bordeaux, CNRS UMR-5234, 33000 Bordeaux, France.
| | - Doranda Perdomo
- Laboratory of Fundamental Microbiology and Pathogenicity (MFP), University of Bordeaux, CNRS UMR-5234, 33000 Bordeaux, France.
| |
Collapse
|
31
|
Hosomi S, Grootjans J, Tschurtschenthaler M, Krupka N, Matute JD, Flak MB, Martinez-Naves E, Gomez Del Moral M, Glickman JN, Ohira M, Lanier LL, Kaser A, Blumberg R. Intestinal epithelial cell endoplasmic reticulum stress promotes MULT1 up-regulation and NKG2D-mediated inflammation. J Exp Med 2017; 214:2985-2997. [PMID: 28747426 PMCID: PMC5626394 DOI: 10.1084/jem.20162041] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 05/25/2017] [Accepted: 07/10/2017] [Indexed: 12/25/2022] Open
Abstract
Hosomi et al. show that intestinal epithelial cell–specific deletion of X-box–binding protein 1, an unfolded protein response–related transcription factor, results in CHOP-dependent increased expression of specific natural killer group 2 member D (NKG2D) ligands. This activates NKG2D-expressing intraepithelial group 1 ILCs and promotes small intestinal inflammation. Endoplasmic reticulum (ER) stress is commonly observed in intestinal epithelial cells (IECs) and can, if excessive, cause spontaneous intestinal inflammation as shown by mice with IEC-specific deletion of X-box–binding protein 1 (Xbp1), an unfolded protein response–related transcription factor. In this study, Xbp1 deletion in the epithelium (Xbp1ΔIEC) is shown to cause increased expression of natural killer group 2 member D (NKG2D) ligand (NKG2DL) mouse UL16-binding protein (ULBP)–like transcript 1 and its human orthologue cytomegalovirus ULBP via ER stress–related transcription factor C/EBP homology protein. Increased NKG2DL expression on mouse IECs is associated with increased numbers of intraepithelial NKG2D-expressing group 1 innate lymphoid cells (ILCs; NK cells or ILC1). Blockade of NKG2D suppresses cytolysis against ER-stressed epithelial cells in vitro and spontaneous enteritis in vivo. Pharmacological depletion of NK1.1+ cells also significantly improved enteritis, whereas enteritis was not ameliorated in Recombinase activating gene 1−/−;Xbp1ΔIEC mice. These experiments reveal innate immune sensing of ER stress in IECs as an important mechanism of intestinal inflammation.
Collapse
Affiliation(s)
- Shuhei Hosomi
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Joep Grootjans
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Department of Gastroenterology and Hepatology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Markus Tschurtschenthaler
- Department of Medicine, Division of Gastroenterology, University of Cambridge, Cambridge, England, UK
| | - Niklas Krupka
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Juan D Matute
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Magdalena B Flak
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Eduardo Martinez-Naves
- Department of Microbiology and Immunology, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Gomez Del Moral
- Department of Cell Biology, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Mizuki Ohira
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA.,Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA
| | - Arthur Kaser
- Department of Gastroenterology and Hepatology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Richard Blumberg
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
32
|
Chong WC, Shastri MD, Eri R. Endoplasmic Reticulum Stress and Oxidative Stress: A Vicious Nexus Implicated in Bowel Disease Pathophysiology. Int J Mol Sci 2017; 18:E771. [PMID: 28379196 PMCID: PMC5412355 DOI: 10.3390/ijms18040771] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023] Open
Abstract
The endoplasmic reticulum (ER) is a complex protein folding and trafficking organelle. Alteration and discrepancy in the endoplasmic reticulum environment can affect the protein folding process and hence, can result in the production of misfolded proteins. The accumulation of misfolded proteins causes cellular damage and elicits endoplasmic reticulum stress. Under such stress conditions, cells exhibit reduced functional synthesis, and will undergo apoptosis if the stress is prolonged. To resolve the ER stress, cells trigger an intrinsic mechanism called an unfolded protein response (UPR). UPR is an adaptive signaling process that triggers multiple pathways through the endoplasmic reticulum transmembrane transducers, to reduce and remove misfolded proteins and improve the protein folding mechanism, in order to improve and maintain endoplasmic reticulum homeostasis. An increasing number of studies support the view that oxidative stress has a strong connection with ER stress. During the protein folding process, reactive oxygen species are produced as by-products, leading to impaired reduction-oxidation (redox) balance conferring oxidative stress. As the protein folding process is dependent on redox homeostasis, the oxidative stress can disrupt the protein folding mechanism and enhance the production of misfolded proteins, causing further ER stress. It is proposed that endoplasmic reticulum stress and oxidative stress together play significant roles in the pathophysiology of bowel diseases.
Collapse
Affiliation(s)
- Wai Chin Chong
- School of Health Science, University of Tasmania, Newnham TAS 7248, Australia.
| | - Madhur D Shastri
- School of Health Science, University of Tasmania, Newnham TAS 7248, Australia.
| | - Rajaraman Eri
- School of Health Science, University of Tasmania, Newnham TAS 7248, Australia.
| |
Collapse
|
33
|
Kunde DA, Chong WC, Nerurkar PV, Ahuja KD, Just J, Smith JA, Guven N, Eri RD. Bitter melon protects against ER stress in LS174T colonic epithelial cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:2. [PMID: 28049460 PMCID: PMC5210302 DOI: 10.1186/s12906-016-1522-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 12/16/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Bitter Melon (BM) has been used as a functional food in traditional Chinese and Indian medicine for many generations and has gained a great deal of attention due to its apparent benefits in moderating some of the pathogenic processes in a variety of inflammatory conditions. BM extract (BME) has been shown to possess strong anti-oxidant properties. In addition, it can ameliorate oxidative stress and potentially ER stress. There is increasing evidence that oxidative and ER stress are major contributors for intestinal secretory cell dysfunction which leads to local inflammation and disease pathogenesis that are hallmarks of inflammatory bowel diseases (IBD). Hence, the search for potential therapeutics against ER stress and oxidative stress in intestinal epithelial secretory cells may provide valuable resources for the management of IBD. The aim of the present study was to investigate the effects of BME in ameliorating ER stress in colonic epithelial cells. METHODS Human colonic adenocarcinoma LS174T cells were used for the assessment of BME effects on colonic epithelial cells in vitro. Cell viability was assessed using trypan blue exclusion and the effect of BME in ameliorating tunicamycin (TM)-induced ER stress was determined by analysing the mRNA expression of the common ER stress markers; ATF6, XBP1, GRP78, CHOP and PERK by quantitative RT-PCR and GRP78 and CHOP by western blot. RESULTS In the absence of ER stress, BME exhibited no cell toxicity up to 2.0% w/v and no significant effect on the basal mRNA expression of ER stress markers in LS174T cells. In contrast, pre-treatment of LS174T cells with BME followed by induction of ER stress resulted in a significant decrease in mRNA expression of ATF6, XBP1, GRP78, CHOP and PERK and protein expression of GRP78 and CHOP. Co-treatment during induction of ER stress and post- treatment following induction of ER Stress in LS174T cells resulted in a lower but still significant reduction in mRNA expression levels of most ER stress markers. CONCLUSIONS This is one of the first studies demonstrating the efficacy of BME in reducing expression of ER stress markers in colonic epithelial cells suggesting the potential of BME as a dietary intervention in ameliorating ER stress and oxidation in IBD. Interestingly, while the most significant effect was seen with pre-treatment of cells with BME there was a reduced but still significant effect when co-treated or even post-treated. This suggests that BME may even be effective in modulating ER stress in the face of an existing cell stress environment.
Collapse
|
34
|
C/EBP homologous protein promotes NSAID-activated gene 1-linked pro-inflammatory signals and enterocyte invasion by enteropathogenic Escherichia coli. Microbes Infect 2016; 19:110-121. [PMID: 27771295 DOI: 10.1016/j.micinf.2016.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 08/24/2016] [Accepted: 10/12/2016] [Indexed: 01/18/2023]
Abstract
NSAID-activated Gene 1 (NAG-1) is a prognostic indicator of chronic inflammatory diseases and aggressive tumors. Among the stress sentinels in response to infection by enteropathogenic Escherichia coli (EPEC) or other pathogenic E. coli, C/EBP homologous protein (CHOP), a representative stress-regulated transcription factor, was prominently increased and assessed for its involvement in NAG-1-mediated pathogenic cellular responses. NAG-1 expression was transcriptionally upregulated by CHOP, which promoted chemokine production through sustained NF-κB activation. Mechanistically, NF-κB activation by NAG-1 was due to TGFβ-activated kinase 1 (TAK-1)-mediated pathway rather than SMAD-associated signals. Moreover, CHOP and subsequent TAK-1-linked signals were also involved in bacterial invasion into human cells. Therefore, CHOP as an infection-induced sentinel played crucial roles in induction of NAG-1 and subsequent prolonged activation of pro-inflammatory responses to EPEC infection or related chronic pathogenic states.
Collapse
|
35
|
Heindryckx F, Binet F, Ponticos M, Rombouts K, Lau J, Kreuger J, Gerwins P. Endoplasmic reticulum stress enhances fibrosis through IRE1α-mediated degradation of miR-150 and XBP-1 splicing. EMBO Mol Med 2016; 8:729-44. [PMID: 27226027 PMCID: PMC4931288 DOI: 10.15252/emmm.201505925] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 04/16/2016] [Accepted: 04/20/2016] [Indexed: 01/08/2023] Open
Abstract
ER stress results in activation of the unfolded protein response and has been implicated in the development of fibrotic diseases. In this study, we show that inhibition of the ER stress-induced IRE1α signaling pathway, using the inhibitor 4μ8C, blocks TGFβ-induced activation of myofibroblasts in vitro, reduces liver and skin fibrosis in vivo, and reverts the fibrotic phenotype of activated myofibroblasts isolated from patients with systemic sclerosis. By using IRE1α(-/-) fibroblasts and expression of IRE1α-mutant proteins lacking endoribonuclease activity, we confirmed that IRE1α plays an important role during myofibroblast activation. IRE1α was shown to cleave miR-150 and thereby to release the suppressive effect that miR-150 exerted on αSMA expression through c-Myb. Inhibition of IRE1α was also demonstrated to block ER expansion through an XBP-1-dependent pathway. Taken together, our results suggest that ER stress could be an important and conserved mechanism in the pathogenesis of fibrosis and that components of the ER stress pathway may be therapeutically relevant for treating patients with fibrotic diseases.
Collapse
Affiliation(s)
- Femke Heindryckx
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - François Binet
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Markella Ponticos
- Centre for Rheumatology and Connective Tissue Diseases, University College London, London, UK
| | - Krista Rombouts
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Johan Kreuger
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Pär Gerwins
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden Department of Radiology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
36
|
Grootjans J, Kaser A, Kaufman RJ, Blumberg RS. The unfolded protein response in immunity and inflammation. Nat Rev Immunol 2016; 16:469-84. [PMID: 27346803 DOI: 10.1038/nri.2016.62] [Citation(s) in RCA: 549] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The unfolded protein response (UPR) is a highly conserved pathway that allows the cell to manage endoplasmic reticulum (ER) stress that is imposed by the secretory demands associated with environmental forces. In this role, the UPR has increasingly been shown to have crucial functions in immunity and inflammation. In this Review, we discuss the importance of the UPR in the development, differentiation, function and survival of immune cells in meeting the needs of an immune response. In addition, we review current insights into how the UPR is involved in complex chronic inflammatory diseases and, through its role in immune regulation, antitumour responses.
Collapse
Affiliation(s)
- Joep Grootjans
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts 02115, USA
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts 02115, USA
| |
Collapse
|
37
|
Melhem H, Hansmannel F, Bressenot A, Battaglia-Hsu SF, Billioud V, Alberto JM, Gueant JL, Peyrin-Biroulet L. Methyl-deficient diet promotes colitis and SIRT1-mediated endoplasmic reticulum stress. Gut 2016; 65:595-606. [PMID: 25608526 DOI: 10.1136/gutjnl-2014-307030] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 01/04/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Methyl donor deficiency (MDD) aggravates experimental colitis in rats and increases endoplasmic reticulum (ER) stress through decreased sirtuin 1 (SIRT1) in neuronal cells and myocardium. ER stress plays a key role in IBD pathogenesis. AIM We investigated whether the influence of MDD on colitis resulted from an ER stress response triggered by decreased SIRT1 expression. DESIGN The unfolded protein response (UPR), chaperones proteins, heat shock factor protein 1 (HSF1) and SIRT1 were examined in rats with MDD and dextran sulfate sodium (DSS)-induced colitis in a Caco-2 cell model with stable expression of transcobalamin-oleosin (TO) chimera, which impairs cellular availability of vitamin B12, and in IBD. The effects of SIRT1 activation were studied both in vitro and in vivo. RESULTS MDD aggravated DSS-induced colitis clinically, endoscopically and histologically. MDD activated ER stress pathways, with increased phosphorylate-PKR-like ER kinase, P-eiF-2α, P-IRE-1α, activating transcription factor (ATF)6, XBP1-S protein and ATF4 mRNA expression levels in rats. This was accompanied by reduced SIRT1 expression level and greater acetylation of HSF1, in relation with a dramatic decrease of chaperones (binding immunoglobulin protein (BIP), heat shock protein (HSP)27 and HSP90). Adding either vitamin B12, S-adenosylmethionine or an SIRT1 activator (SRT1720) reduced the UPR in vitro. In rats, SIRT1 activation by SRT1720 prevented colitis by reducing HSF1 acetylation and increasing expression of BIP, HSP27 and HSP90. Immunohistochemistry showed impaired expression of SIRT1 in the colonic epithelium of patients with IBD. CONCLUSIONS SIRT1 is a master regulator of ER stress and severity of experimental colitis in case of MDD. It could deserve further interest as a therapeutic target of IBD.
Collapse
Affiliation(s)
- Hassan Melhem
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Franck Hansmannel
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Aude Bressenot
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Syue-Fang Battaglia-Hsu
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Vincent Billioud
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Jean Marc Alberto
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Jean Louis Gueant
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Laurent Peyrin-Biroulet
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| |
Collapse
|
38
|
Curcumin as a therapeutic agent in the chemoprevention of inflammatory bowel disease. Drug Discov Today 2016; 21:843-9. [PMID: 26995272 DOI: 10.1016/j.drudis.2016.03.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 02/17/2016] [Accepted: 03/08/2016] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel diseases (IBD), mainly Crohn's disease (CD) and ulcerative colitis (UC) are chronic ailments of the gastrointestinal tract, characterized by recurrent inflammation. Current therapeutic strategies are based on the mitigation of symptoms, including inflammatory remission and healing of mucosal manifestations. Extensive studies have suggested that continuous oxidative damage can lead to the inflammatory signaling cascade in IBD. Curcumin, a potent modulator of cell signaling, is popular for its antioxidant and anti-inflammatory activities, and has already been shown remarkable therapeutic results in IBD. Here, we review and discuss the effects of curcumin as a therapeutic agent in the chemoprevention of IBD.
Collapse
|
39
|
Morales Fénero CI, Colombo Flores AA, Câmara NOS. Inflammatory diseases modelling in zebrafish. World J Exp Med 2016; 6:9-20. [PMID: 26929916 PMCID: PMC4759353 DOI: 10.5493/wjem.v6.i1.9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 10/20/2015] [Accepted: 12/18/2015] [Indexed: 02/06/2023] Open
Abstract
The ingest of diets with high content of fats and carbohydrates, low or no physical exercise and a stressful routine are part of the everyday lifestyle of most people in the western world. These conditions are triggers for different diseases with complex interactions between the host genetics, the metabolism, the immune system and the microbiota, including inflammatory bowel diseases (IBD), obesity and diabetes. The incidence of these disorders is growing worldwide; therefore, new strategies for its study are needed. Nowadays, the majority of researches are in use of murine models for understand the genetics, physiopathology and interaction between cells and signaling pathways to find therapeutic solutions to these diseases. The zebrafish, a little tropical water fish, shares 70% of our genes and conserves anatomic and physiological characteristics, as well as metabolical pathways, with mammals, and is rising as a new complementary model for the study of metabolic and inflammatory diseases. Its high fecundity, fast development, transparency, versatility and low cost of maintenance makes the zebrafish an interesting option for new researches. In this review, we offer a discussion of the existing genetic and induced zebrafish models of two important Western diseases that have a strong inflammatory component, the IBD and the obesity.
Collapse
|
40
|
Zheng L, Dai YC, Zhang YL, Chen X, Fang CY, Tang ZP. Role of endoplasmic reticulum stress signaling molecule PERK in bowel mucosal injury in ulcerative colitis. Shijie Huaren Xiaohua Zazhi 2015; 23:5493-5498. [DOI: 10.11569/wcjd.v23.i34.5493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Endoplasmic reticulum stress (ERS) is the main focus in the study of the pathogenesis of ulcerative colitis, and it protects and repairs the intestinal epithelial cell (IEC) injury through unfolded protein response (UPR). Protein kinase R-like ER kinase (PERK) is an endoplasmic reticulum-localized type I transmembrane protein, with serine/threonine protein kinase activity. IECs are one of cell populations with the most vigorous metabolism and have abundant endoplasmic reticulum. Early ERS can activate PERK-eIF2 alpha channel and inhibit the synthesis of proteins to protect cells. However, sustained severe ERS promotes cell damage and death, activates nuclear factor-kappa B in IECs, causes the secretion of a variety of inflammatory cytokines, and promotes the occurrence of inflammatory lesions.
Collapse
|
41
|
Sreedhar R, Arumugam S, Thandavarayan RA, Giridharan VV, Karuppagounder V, Pitchaimani V, Afrin R, Harima M, Nakamura T, Ueno K, Nakamura M, Suzuki K, Watanabe K. Toki-shakuyaku-san, a Japanese kampo medicine, reduces colon inflammation in a mouse model of acute colitis. Int Immunopharmacol 2015; 29:869-875. [DOI: 10.1016/j.intimp.2015.08.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 08/20/2015] [Accepted: 08/24/2015] [Indexed: 02/07/2023]
|
42
|
Sreedhar R, Arumugam S, Karuppagounder V, Thandavarayan RA, Giridharan VV, Pitchaimani V, Afrin MR, Harima M, Nakamura T, Nakamura M, Suzuki K, Watanabe K. Jumihaidokuto effectively inhibits colon inflammation and apoptosis in mice with acute colitis. Int Immunopharmacol 2015; 29:957-963. [DOI: 10.1016/j.intimp.2015.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/05/2015] [Accepted: 10/06/2015] [Indexed: 02/07/2023]
|
43
|
Hosomi S, Kaser A, Blumberg RS. Role of endoplasmic reticulum stress and autophagy as interlinking pathways in the pathogenesis of inflammatory bowel disease. Curr Opin Gastroenterol 2015; 31:81-8. [PMID: 25426970 PMCID: PMC4592163 DOI: 10.1097/mog.0000000000000144] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW The purpose of this study is to provide an overview of the role of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) in inflammatory bowel disease (IBD). RECENT FINDINGS Human genetic studies have identified several UPR-related genes and autophagy-related genes as IBD risk loci. Impairment of each branch of the UPR causes spontaneous enteritis or creates higher susceptibility for intestinal inflammation in model systems. Deficiency of either UPR or autophagy in small intestinal epithelial cells promotes each other's compensatory engagement, which is especially prominent in Paneth cells such that, in the absence of both, severe spontaneous enteritis emerges. SUMMARY Interactions between the UPR and autophagy exhibit critical synergistic interactions within the intestinal epithelium and especially Paneth cells that are of considerable importance to the maintenance of homeostasis. When dysfunctional in the Paneth cell, spontaneous inflammation can emerge that may extend beyond the epithelium providing direct experimental evidence that subsets of Crohn's disease may emanate from primary Paneth cell disturbances.
Collapse
Affiliation(s)
- Shuhei Hosomi
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
| | - Richard S. Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Negroni A, Prete E, Vitali R, Cesi V, Aloi M, Civitelli F, Cucchiara S, Stronati L. Endoplasmic reticulum stress and unfolded protein response are involved in paediatric inflammatory bowel disease. Dig Liver Dis 2014; 46:788-94. [PMID: 24953208 DOI: 10.1016/j.dld.2014.05.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/09/2014] [Accepted: 05/18/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Endoplasmic reticulum stress and unfolded protein response have been recently associated with the development of inflammatory bowel diseases in adults. We aimed at assessing the involvement of these pathways also in paediatric inflammatory bowel disease by analysing the expression of the main genes involved in endoplasmic reticulum stress and correlating them with the degree of intestinal inflammation. METHODS Real-time PCR and Western blot analysis of the expression of the endoplasmic reticulum stress marker HSPA5 and of selected genes representing the three pathways of unfolded protein response (IRE-XBP1, PERK-ATF4, ATF6p90-p50) in inflamed and uninflamed biopsies from 28 inflammatory bowel disease paediatric patients and 10 controls. RESULTS HSPA5, PDIA4, as well as unspliced and spliced XBP1 mRNAs were significantly increased in patients' inflamed colonic mucosa compared to uninflamed mucosa and controls. HSPA5, PDIA4, ATF6, and phospho-IRE proteins were also upregulated, indicating the activation of the IRE-XBP1 and ATF6p90-p50 branches of unfolded protein response. A positive significant correlation between interleukin-8 levels, as a marker of inflammation, and upregulated genes was found in the inflamed colonic mucosa. CONCLUSION A deregulation of the genes involved in the endoplasmic reticulum stress and unfolded protein response pathways may be a key component of the inflammatory response in paediatric patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Anna Negroni
- Department of Radiobiology and Human Health, ENEA, Rome, Italy.
| | - Enrica Prete
- Department of Radiobiology and Human Health, ENEA, Rome, Italy
| | - Roberta Vitali
- Department of Radiobiology and Human Health, ENEA, Rome, Italy
| | - Vincenzo Cesi
- Department of Radiobiology and Human Health, ENEA, Rome, Italy
| | - Marina Aloi
- Department of Pediatrics and Infantile Neuropsychiatry, Paediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Rome, Italy
| | - Fortunata Civitelli
- Department of Pediatrics and Infantile Neuropsychiatry, Paediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Rome, Italy
| | - Salvatore Cucchiara
- Department of Pediatrics and Infantile Neuropsychiatry, Paediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Rome, Italy
| | - Laura Stronati
- Department of Radiobiology and Human Health, ENEA, Rome, Italy
| |
Collapse
|
45
|
Cao SS, Kaufman RJ. Endoplasmic reticulum stress and oxidative stress in cell fate decision and human disease. Antioxid Redox Signal 2014; 21:396-413. [PMID: 24702237 PMCID: PMC4076992 DOI: 10.1089/ars.2014.5851] [Citation(s) in RCA: 936] [Impact Index Per Article: 85.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE The endoplasmic reticulum (ER) is a specialized organelle for the folding and trafficking of proteins, which is highly sensitive to changes in intracellular homeostasis and extracellular stimuli. Alterations in the protein-folding environment cause accumulation of misfolded proteins in the ER that profoundly affect a variety of cellular signaling processes, including reduction-oxidation (redox) homeostasis, energy production, inflammation, differentiation, and apoptosis. The unfolded protein response (UPR) is a collection of adaptive signaling pathways that evolved to resolve protein misfolding and restore an efficient protein-folding environment. RECENT ADVANCES Production of reactive oxygen species (ROS) has been linked to ER stress and the UPR. ROS play a critical role in many cellular processes and can be produced in the cytosol and several organelles, including the ER and mitochondria. Studies suggest that altered redox homeostasis in the ER is sufficient to cause ER stress, which could, in turn, induce the production of ROS in the ER and mitochondria. CRITICAL ISSUES Although ER stress and oxidative stress coexist in many pathologic states, whether and how these stresses interact is unknown. It is also unclear how changes in the protein-folding environment in the ER cause oxidative stress. In addition, how ROS production and protein misfolding commit the cell to an apoptotic death and contribute to various degenerative diseases is unknown. FUTURE DIRECTIONS A greater fundamental understanding of the mechanisms that preserve protein folding homeostasis and redox status will provide new information toward the development of novel therapeutics for many human diseases.
Collapse
Affiliation(s)
- Stewart Siyan Cao
- 1 Degenerative Diseases Program, Sanford Burnham Medical Research Institute , La Jolla, California
| | | |
Collapse
|
46
|
Vrecenak JD, Mattei P. Fast-track management is safe and effective after bowel resection in children with Crohn's disease. J Pediatr Surg 2014; 49:99-102; discussion 102-3. [PMID: 24439590 DOI: 10.1016/j.jpedsurg.2013.09.038] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 09/30/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND "Fast-track" management (FT) challenges traditional postoperative tenets in order to minimize discomfort and optimize inpatient care. We examined the outcomes of consecutively performed laparoscopic-assisted ileocecectomy for Crohn's disease (CD), with particular focus on FT's effects in patients with underlying bowel inflammation. METHODS We retrospectively reviewed all patients undergoing isolated laparoscopic-assisted ileocecectomy for CD at our institution between 12/2000 and 12/2010, excluding patients with multiple areas of surgical CD, bladder involvement, or age >18years. RESULTS Seventy-one patients aged 8-18years underwent isolated laparoscopic-assisted ileocecectomy for CD, of which 45 met FT criteria. Individual practice patterns primarily determined which patients were FT-managed. FT management led to decreased length of stay (LOS), time to first stool, time to full diet, and intravenous narcotic use. No significant difference in complications or disease progression was observed between the two groups during 2-year follow up. CONCLUSIONS Our results suggest that FT is safe and effective in patients with CD. In a chronically ill population, counseling patients and families to expect early discharge is critical to the success of this strategy. Despite CD-related GI pathology, FT patients realized benefits in terms of LOS, time to bowel function, and narcotic use without any increase in complications.
Collapse
Affiliation(s)
| | - Peter Mattei
- The Children's Hospital of Philadelphia, Philadelphia, PA.
| |
Collapse
|
47
|
Mei Y, Thompson MD, Cohen RA, Tong X. Endoplasmic Reticulum Stress and Related Pathological Processes. JOURNAL OF PHARMACOLOGICAL & BIOMEDICAL ANALYSIS 2013; 1:1000107. [PMID: 24611136 PMCID: PMC3942890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The endoplasmic reticulum (ER) plays a pivotal role in lipid and protein biosynthesis as well as calcium store regulation, which determines its essential role in cell function. Hypoxia, nutrient deprivation, perturbation of redox status and aberrant calcium regulation can all trigger the ER stress response, which is mediated through three main sensors, namely inositol requiring element-1 (IRE-1), protein kinase-like ER kinase (PERK) and activating transcription factor 6 (ATF6). This review explores the interaction of ER stress and ER stress-associated pathological processes, including inflammation, apoptosis, aberrant autophagy, mitochondrial dysfunction and hypoxic responses. In addition, the correlation of ER stress with lipid and calcium homeostasis and dysregulation, and its role in disease development is also presented. Improved understanding of ER stress and its cofactors in pathological processes may provide new perspective on disease development and control.
Collapse
Affiliation(s)
- Yu Mei
- Vascular Biology Section, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Melissa D Thompson
- Vascular Biology Section, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Richard A Cohen
- Vascular Biology Section, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - XiaoYong Tong
- Vascular Biology Section, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| |
Collapse
|
48
|
Lu P, Struijs MC, Mei J, Witte-Bouma J, Korteland-van Male AM, de Bruijn ACJM, van Goudoever JB, Renes IB. Endoplasmic reticulum stress, unfolded protein response and altered T cell differentiation in necrotizing enterocolitis. PLoS One 2013; 8:e78491. [PMID: 24194940 PMCID: PMC3806824 DOI: 10.1371/journal.pone.0078491] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 09/12/2013] [Indexed: 12/15/2022] Open
Abstract
Background Endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR) play important roles in chronic intestinal inflammation. Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in preterm infants and is characterized by acute intestinal inflammation and necrosis. The objective of the study is to investigate the role of ER stress and the UPR in NEC patients. Methods Ileal tissues from NEC and control patients were obtained during surgical resection and/or at stoma closure. Splicing of XBP1 was detected using PCR, and gene expression was quantified using qPCR and Western blot. Results Splicing of XBP1 was only detected in a subset of acute NEC (A-NEC) patients, and not in NEC patients who had undergone reanastomosis (R-NEC). The other ER stress and the UPR pathways, PERK and ATF6, were not activated in NEC patients. A-NEC patients showing XBP1 splicing (A-NEC-XBP1s) had increased mucosal expression of GRP78, CHOP, IL6 and IL8. Similar results were obtained by inducing ER stress and the UPR invitro. A-NEC-XBP1s patients showed altered T cell differentiation indicated by decreased mucosal expression of RORC, IL17A and FOXP3. A-NEC-XBP1s patients additionally showed more severe morphological damage and a worse surgical outcome. Compared with A-NEC patients, R-NEC patients showed lower mucosal IL6 and IL8 expression and higher mucosal FOXP3 expression. Conclusions XBP1 splicing, ER stress and the UPR in NEC are associated with increased IL6 and IL8 expression levels, altered T cell differentiation and severe epithelial injury.
Collapse
Affiliation(s)
- Peng Lu
- Division of Neonatology, Department of Pediatrics, Erasmus MC-Sophia, Rotterdam, the Netherlands
- Department of Pediatrics, Emma Children’s Hospital - AMC, Amsterdam, The Netherlands
| | | | - Jiaping Mei
- Neonatal Intensive Care Unit, Shenzhen Maternal and Child Healthcare Hospital, Affiliated Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Janneke Witte-Bouma
- Division of Neonatology, Department of Pediatrics, Erasmus MC-Sophia, Rotterdam, the Netherlands
| | | | | | - Johannes B. van Goudoever
- Department of Pediatrics, Emma Children’s Hospital - AMC, Amsterdam, The Netherlands
- Department of Pediatrics, VU University Medical Center, Amsterdam, The Netherlands
| | - Ingrid B. Renes
- Division of Neonatology, Department of Pediatrics, Erasmus MC-Sophia, Rotterdam, the Netherlands
- Department of Pediatrics, Emma Children’s Hospital - AMC, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
49
|
Thakur PC, Davison JM, Stuckenholz C, Lu L, Bahary N. Dysregulated phosphatidylinositol signaling promotes endoplasmic-reticulum-stress-mediated intestinal mucosal injury and inflammation in zebrafish. Dis Model Mech 2013; 7:93-106. [PMID: 24135483 PMCID: PMC3882052 DOI: 10.1242/dmm.012864] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dysregulated phosphatidylinositol (PI) signaling has been implicated in human gastrointestinal (GI) malignancies and inflammatory states, underlining the need to study pathophysiological roles of PI in an in vivo genetic model. Here, we study the significance of PI in GI pathophysiology using the zebrafish mutant cdipthi559, which lacks PI synthesis, and unravel a crucial role of PI in intestinal mucosal integrity and inflammation. The cdipthi559 mutants exhibit abnormal villous architecture and disorganized proliferation of intestinal epithelial cells (IECs), with pathologies reminiscent of inflammatory bowel disease (IBD), including apoptosis of goblet cells, abnormal mucosecretion, bacterial overgrowth and leukocyte infiltration. The mutant IECs exhibit vacuolation, microvillus atrophy and impaired proliferation. The cdipthi559 gene expression profile shows enrichment of acute phase response signaling, and the endoplasmic reticulum (ER) stress factors hspa5 and xbp1 are robustly activated in the mutant GI tissue. Temporal electron micrographic analyses reveal that PI-deficient IECs undergo sequential ER-Golgi disruption, mitochondrial depletion, macroautophagy and cell death, consistent with chronic ER-stress-mediated cytopathology. Furthermore, pharmacological induction of ER stress by inhibiting protein glycosylation or PI synthase inhibition in leukocyte-specific reporter lines replicates the cdipthi559 inflammatory phenotype, suggesting a fundamental role of PI metabolism and ER stress in mucosal inflammation. Antibiotics and anti-inflammatory drugs resolved the inflammation, but not the autophagic necroapoptosis of IECs, suggesting that bacterial overgrowth can exacerbate ER stress pathology, whereas persistent ER stress is sufficient to trigger inflammation. Interestingly, the intestinal phenotype was partially alleviated by chemical chaperones, suggesting their therapeutic potential. Using zebrafish genetic and pharmacological models, this study demonstrates a newly identified link between intracellular PI signaling and ER-stress-mediated mucosal inflammation. The zebrafish cdipt mutants provide a powerful tool for dissecting the fundamental mechanisms of ER-stress-mediated human GI diseases and a platform to develop molecularly targeted therapies.
Collapse
Affiliation(s)
- Prakash C Thakur
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA
| | | | | | | | | |
Collapse
|
50
|
Bertrand J, Tennoune N, Marion-Letellier R, Goichon A, Chan P, Mbodji K, Vaudry D, Déchelotte P, Coëffier M. Evaluation of ubiquitinated proteins by proteomics reveals the role of the ubiquitin proteasome system in the regulation of Grp75 and Grp78 chaperone proteins during intestinal inflammation. Proteomics 2013; 13:3284-92. [PMID: 24030972 DOI: 10.1002/pmic.201300082] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 08/13/2013] [Accepted: 08/21/2013] [Indexed: 12/19/2022]
Abstract
The ubiquitin proteasome system (UPS) is the major pathway of intracellular protein degradation and may be involved in the pathophysiology of inflammatory bowel diseases or irritable bowel syndrome. UPS specifically degrades proteins tagged with an ubiquitin chain. We aimed to identify polyubiquitinated proteins during inflammatory response in intestinal epithelial HCT-8 cells by a proteomic approach. HCT-8 cells were incubated with interleukin 1β, tumor necrosis factor-α, and interferon-γ for 2 h. Total cellular protein extracts were separated by 2D gel electrophoresis and analyzed by an immunodetection using antiubiquitin antibody. Differential ubiquitinated proteins were then identified by LC-ESI MS/MS. Seven proteins were differentially ubiquitinated between control and inflammatory conditions. Three of them were chaperones: Grp75 and Hsc70 were more ubiquitinated (p < 0.05) and Grp78 was less ubiquitinated (p < 0.05) under inflammatory conditions. The results for Grp75 and Grp78 were then confirmed in HCT-8 cells and in 2-4-6-trinitrobenzen sulfonic acid induced colitis in rats mimicking inflammatory bowel disease by immunoprecipitation. No difference was observed in irritable bowel syndrome like model. In conclusion, we showed that a proteomic approach is suitable to identify ubiquitinated proteins and that UPS-regulated expression of Grp75 and Grp78 may be involved in inflammatory response. Further studies should lead to the identification of ubiquitin ligases responsible for Grp75 and Grp78 ubiquitination.
Collapse
Affiliation(s)
- Julien Bertrand
- INSERM Unit 1073, University of Rouen, Rouen, France; Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France
| | | | | | | | | | | | | | | | | |
Collapse
|