1
|
Giambra V, Caldarelli M, Franza L, Rio P, Bruno G, di Iasio S, Mastrogiovanni A, Gasbarrini A, Gambassi G, Cianci R. The Role of Notch Signaling and Gut Microbiota in Autoinflammatory Diseases: Mechanisms and Future Views. Biomedicines 2025; 13:768. [PMID: 40299348 PMCID: PMC12024679 DOI: 10.3390/biomedicines13040768] [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: 01/30/2025] [Revised: 03/06/2025] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
Notch signaling is an evolutionarily conserved, multifunctional pathway involved in cell fate determination and immune modulation and contributes to the pathogenesis of autoinflammatory diseases. Emerging evidence reveals a bidirectional interaction between Notch and the gut microbiota (GM), whereby GM composition is capable of modulating Notch signaling through the binding of microbial elements to Notch receptors, leading to immune modulation. Furthermore, Notch regulates the GM by promoting SCFA-producing bacteria while suppressing proinflammatory strains. Beneficial microbes, such as Lactobacillus and Akkermansia muciniphila, modulate Notch and reduce proinflammatory cytokine production (such as IL-6 and TNF-α). The interaction between GM and Notch can either amplify or attenuate inflammatory pathways in inflammatory bowel diseases (IBDs), Behçet's disease, and PAPA syndrome. Together, these findings provide novel therapeutic perspectives for autoinflammatory diseases by targeting the GM via probiotics or inhibiting Notch signaling. This review focuses on Notch-GM crosstalk and how GM-based and/or Notch-targeted approaches may modulate immune responses and promote better clinical outcomes.
Collapse
Affiliation(s)
- Vincenzo Giambra
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (V.G.)
| | - Mario Caldarelli
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Laura Franza
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
- Department of Emergency Medicine, AOU Modena, 41125 Modena, Italy
| | - Pierluigi Rio
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Gaja Bruno
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (V.G.)
| | - Serena di Iasio
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (V.G.)
| | - Andrea Mastrogiovanni
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Giovanni Gambassi
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| |
Collapse
|
2
|
Xu L, Liu W, Huang X, Sun T, Mei L, Liu M, Ren Z, Wang M, Zheng H, Wang Q, Li D, Wang Q, Ke X. Sinomenine hydrochloride improves DSS-induced colitis in mice through inhibition of the Notch signaling pathway. BMC Gastroenterol 2024; 24:451. [PMID: 39695403 DOI: 10.1186/s12876-024-03546-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE To study the therapeutic effect of sinomenine hydrochloride (SH) on dextran sodium sulfate (DSS)-induced colitis in mice as an animal model and the changes of Notch signaling pathway in colon tissue of mice after treatment. METHODS Twenty-four mice were randomly divided into control group, model group, SH low-dose group (20 mg/kg) and SH high-dose group (60 mg/kg), with 6 mice in each group. Disease activity index (DAI), colonic mucosal injury index and colonic histopathological score were calculated. The expression levels of related genes, proteins in Notch signaling pathway and inflammatory factors were quantified. RESULTS SH can significantly reduce the symptoms of colitis mice, and can significantly reduce the DAI score (Model: 3.44 ± 0.27; SH-20: 2.50 ± 0.18; SH-60: 1.89 ± 0.17; P < 0.001) and histopathological injury degree (Model: 7.67 ± 0.52; SH-20: 5.17 ± 0.75, P < 0.01; SH-60: 3.33 ± 0.52, P < 0.001). SH can down-regulate the expression levels of Notch1, NICD1, Jagged1 and Hes1 proteins in colon tissue of colitis mice (Model: 1.92 ± 0.16, 1.83 ± 0.21, 2.23 ± 0.22, 1.91 ± 0.17; SH-20: 1.56 ± 0.12, 1.39 ± 0.13, 1.58 ± 0.12, 1.38 ± 0.11; SH-60: 1.24 ± 0.09, 1.23 ± 0.10, 1.23 ± 0.11, 1.22 ± 0.09; P < 0.01), and reduce the contents of serum pro-inflammatory cytokines TNF-α, IL-1β and IL-6 (Model: 718.53 ± 81.81, 51.62 ± 2.80, 444.07 ± 67.77; SH-20: 544.72 ± 90.03, 34.10 ± 2.90, 345.43 ± 43.40; SH-60: 434.11 ± 71.75, 29.44 ± 3.70, 236.11 ± 29.35; P < 0.001). CONCLUSION The therapeutic effect of SH on DSS-induced colitis in mice may be related to inhibiting the overactivation of Notch signaling pathway.
Collapse
Affiliation(s)
- Linxia Xu
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Wei Liu
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Xixiang Huang
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Tong Sun
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Letian Mei
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Man Liu
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Zhi Ren
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Meng Wang
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Hailun Zheng
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Qiangwu Wang
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Dapeng Li
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Qizhi Wang
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China
| | - Xiquan Ke
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Bengbu, Anhui, 233000, China.
| |
Collapse
|
3
|
Abdel-Naby DH, El-Sheikh MM, Abd El-Rahman SS, El-Hamoly T. GSK-3β/Notch-1 Activation Promotes Radiation-Induced Renal Damage: The Role of Gallic Acid in Mitigation of Nephrotoxicity. ENVIRONMENTAL TOXICOLOGY 2024; 39:4871-4883. [PMID: 38894622 DOI: 10.1002/tox.24361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 04/20/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
Despite the therapeutic advances in treating malignancies, the efficient radiotherapeutic approaches with deprived adverse reactions still represent a potential clinical inquiry. The current study aims to elucidate the role of gallic acid (GA) in modifying the hazardous renal cytotoxicity induced by acute exposure to radiation. The MTT test was used to evaluate the viability of Vero cells exposed to 2 Gy gamma radiation with or without incubation of GA. In an in vivo model, male Wistar rats were divided into four experimental groups (n = 6): Control, Irradiated (IRR, 5 Gy), GA (100 mg/kg, i.p.) + IRR, and Glycogen synthase kinase inhibitor (GSKI, 3 mg/kg, i.p.) + IRR. Based on the MTT toxicity assay, from 0 and up to 5 μM dosages of GA did not demonstrate any cytotoxicity to Vero cells. The optimal GA dose that could protect the cells from radiation was 5 μM. Furthermore, GA exerted a protective effect from gamma radiation on renal tissue as indicated by corrected renal functions, decreased LDH level in serum, and balanced oxidative status, which is indicated by decreased tissue contents of NOx and TBARS with a significant increase of reduced GSH. These outcomes were inferred by the upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) expression. The overall molecular impact of radiation in damaging the renal tissue may be explained by modifying the upstream AKT activity and its downstream targets GSK-3β/Notch-1. Here, we concluded that the anticipated adverse reaction in the course of radiation exposure could be protected by daily administration of GA.
Collapse
Affiliation(s)
- Doaa H Abdel-Naby
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Marwa M El-Sheikh
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Sahar S Abd El-Rahman
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Tarek El-Hamoly
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
4
|
Liu F, Schrack JA, Walston J, Mathias RA, Windham BG, Grams ME, Coresh J, Walker KA. Mid-life plasma proteins associated with late-life prefrailty and frailty: a proteomic analysis. GeroScience 2024; 46:5247-5265. [PMID: 38856871 PMCID: PMC11336072 DOI: 10.1007/s11357-024-01219-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/21/2024] [Indexed: 06/11/2024] Open
Abstract
Physical frailty is a syndrome that typically manifests in later life, although the pathogenic process causing physical frailty likely begins decades earlier. To date, few studies have examined the biological signatures in mid-life associated with physical frailty later in life. Among 4,189 middle-aged participants (57.8 ± 5.0 years, 55.8% women) from the Atherosclerosis Risk in Community (ARIC) study, we evaluated the associations of 4,955 plasma proteins (log 2-transformed and standardized) measured using the SomaScan platform with their frailty status approximately 20 years later. Using multinomial logistic regression models adjusting for demographics, health behaviors, kidney function, total cholesterol, and comorbidities, 12 and 221 proteins were associated with prefrailty and frailty in later life, respectively (FDR p < 0.05). Top frailty-associated proteins included neurocan core protein (NCAN, OR = 0.66), fatty acid-binding protein heart (FABP3, OR = 1.62) and adipocyte (FABP4, OR = 1.65), as well proteins involved in the contactin-1 (CNTN1), toll-like receptor 5 (TLR5), and neurogenic locus notch homolog protein 1 (NOTCH1) signaling pathway relevant to skeletal muscle regeneration, myelination, and inflammation. Pathway analyses suggest midlife dysregulation of inflammation, metabolism, extracellular matrix, angiogenesis, and lysosomal autophagy among those at risk for late-life frailty. After further adjusting for midlife body mass index (BMI) - an established frailty risk factor - only CNTN1 (OR = 0.75) remained significantly associated with frailty. Post-hoc analyses demonstrated that the top 41 midlife frailty-associated proteins mediate 32% of the association between mid-life BMI and late-life frailty. Our findings provide new insights into frailty etiology earlier in the life course, enhancing the potential for prevention.
Collapse
Affiliation(s)
- Fangyu Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Jennifer A Schrack
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Center On Aging and Health, Johns Hopkins University, Baltimore, MD, USA
| | - Jeremy Walston
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Rasika A Mathias
- Genomics and Precision Health Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infection Disease, Bethesda, MD, USA
| | - B Gwen Windham
- Department of Medicine, MIND Center, University of Mississippi Medical Center, Jackson, MS, USA
| | - Morgan E Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Division of Precision Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Population Health and Medicine, Optimal Aging Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute On Aging, Baltimore, MD, USA
| |
Collapse
|
5
|
Erkert L, Gamez-Belmonte R, Kabisch M, Schödel L, Patankar JV, Gonzalez-Acera M, Mahapatro M, Bao LL, Plattner C, Kühl AA, Shen J, Serneels L, De Strooper B, Neurath MF, Wirtz S, Becker C. Alzheimer's disease-related presenilins are key to intestinal epithelial cell function and gut immune homoeostasis. Gut 2024; 73:1618-1631. [PMID: 38684238 DOI: 10.1136/gutjnl-2023-331622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
OBJECTIVE Mutations in presenilin genes are the major cause of Alzheimer's disease. However, little is known about their expression and function in the gut. In this study, we identify the presenilins Psen1 and Psen2 as key molecules that maintain intestinal homoeostasis. DESIGN Human inflammatory bowel disease (IBD) and control samples were analysed for Psen1 expression. Newly generated intestinal epithelium-specific Psen1-deficient, Psen2-deficient and inducible Psen1/Psen2 double-deficient mice were used to dissect the functional role of presenilins in intestinal homoeostasis. RESULTS Psen1 expression was regulated in experimental gut inflammation and in patients with IBD. Induced deletion of Psen1 and Psen2 in mice caused rapid weight loss and spontaneous development of intestinal inflammation. Mice exhibited epithelial barrier disruption with bacterial translocation and deregulation of key pathways for nutrient uptake. Wasting disease was independent of gut inflammation and dysbiosis, as depletion of microbiota rescued Psen-deficient animals from spontaneous colitis development but not from weight loss. On a molecular level, intestinal epithelial cells lacking Psen showed impaired Notch signalling and dysregulated epithelial differentiation. CONCLUSION Overall, our study provides evidence that Psen1 and Psen2 are important guardians of intestinal homoeostasis and future targets for barrier-promoting therapeutic strategies in IBD.
Collapse
Affiliation(s)
- Lena Erkert
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Reyes Gamez-Belmonte
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Melanie Kabisch
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Lena Schödel
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Jay V Patankar
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Miguel Gonzalez-Acera
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Mousumi Mahapatro
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Li-Li Bao
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
| | - Christina Plattner
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Anja A Kühl
- iPATH.Berlin, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jie Shen
- Department of Neurology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lutgarde Serneels
- VIB Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
- UK Dementia Research Institute@UCL, University College London, London, UK
| | - Markus F Neurath
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen-Nürnberg, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
6
|
Ning H, Liu J, Tan J, Yi M, Lin X. The role of the Notch signalling pathway in the pathogenesis of ulcerative colitis: from the perspective of intestinal mucosal barrier. Front Med (Lausanne) 2024; 10:1333531. [PMID: 38249980 PMCID: PMC10796567 DOI: 10.3389/fmed.2023.1333531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024] Open
Abstract
Ulcerative colitis is a common digestive disorder worldwide, with increasing incidence in recent years. It is an urgent problem to be solved, as it seriously affects and threatens the health and life of the global population. Studies have shown that dysfunction of the intestinal mucosal barrier is a critical pathogenic factor and molecular basis of ulcerative colitis, and some scholars have described it as a "barrier organ disease." While the Notch signalling pathway affects a series of cellular processes, including proliferation, differentiation, development, migration, and apoptosis. Therefore, it can regulate intestinal stem cells, CD4+ T cells, innate lymphoid cells, macrophages, and intestinal microbiota and intervene in the chemical, physical, immune, and biological mucosal barriers in cases of ulcerative colitis. The Notch signalling pathway associated with the pathogenesis of ulcerative colitis has distinct characteristics, with good regulatory effects on the mucosal barrier. However, research on ulcerative colitis has mainly focused on immune regulation, anti-inflammatory activity, and antioxidant stress; therefore, the study of the Notch signalling pathway suggests the possibility of understanding the pathogenesis of ulcerative colitis from another perspective. In this article we explore the role and mechanism of the Notch signalling pathway in the pathogenesis of ulcerative colitis from the perspective of the intestinal mucosal barrier to provide new targets and theoretical support for further research on the pathogenesis and clinical treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Hang Ning
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jiemin Liu
- Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jiaqian Tan
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Mengni Yi
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaoyuan Lin
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
7
|
Yoon HJ, Lee S, Kim TY, Yu SE, Kim HS, Chung YS, Chung S, Park S, Shin YC, Wang EK, Noh J, Kim HJ, Ku CR, Koh H, Kim CS, Park JS, Shin YM, Sung HJ. Sprayable nanomicelle hydrogels and inflammatory bowel disease patient cell chips for development of intestinal lesion-specific therapy. Bioact Mater 2022; 18:433-445. [PMID: 35415304 PMCID: PMC8971598 DOI: 10.1016/j.bioactmat.2022.03.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/07/2022] [Accepted: 03/20/2022] [Indexed: 12/16/2022] Open
Abstract
All-in-one treatments represent a paradigm shift in future medicine. For example, inflammatory bowel disease (IBD) is mainly diagnosed by endoscopy, which could be applied for not only on-site monitoring but also the intestinal lesion-targeted spray of injectable hydrogels. Furthermore, molecular conjugation to the hydrogels would program both lesion-specific adhesion and drug-free therapy. This study validated this concept of all-in-one treatment by first utilizing a well-known injectable hydrogel that underwent efficient solution-to-gel transition and nanomicelle formation as a translatable component. These properties enabled spraying of the hydrogel onto the intestinal walls during endoscopy. Next, peptide conjugation to the hydrogel guided endoscopic monitoring of IBD progress upon adhesive gelation with subsequent moisturization of inflammatory lesions, specifically by nanomicelles. The peptide was designed to mimic the major component that mediates intestinal interaction with Bacillus subtilis flagellin during IBD initiation. Hence, the peptide-guided efficient adhesion of the hydrogel nanomicelles onto Toll-like receptor 5 (TLR5) as the main target of flagellin binding and Notch-1. The peptide binding potently suppressed inflammatory signaling without drug loading, where TLR5 and Notch-1 operated collaboratively through downstream actions of tumor necrosis factor-alpha. The results were produced using a human colorectal cell line, clinical IBD patient cells, gut-on-a-chip, a mouse IBD model, and pig experiments to validate the translational utility. Injectable nanomicelle hydrogel for all-in-one treatment of intestinal inflammation. Spraying of the hydrogel onto the intestinal walls during endoscopy. Peptide-guided detection and moisturization of inflammatory lesions.
Collapse
Affiliation(s)
- Hyo-Jin Yoon
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Songhyun Lee
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Tae Young Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung Eun Yu
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hye-Seon Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Young Shin Chung
- Department of Obstetrics and Gynecology, Institution of Women's Life Medical Science, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Seyong Chung
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Suji Park
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yong Cheol Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Eun Kyung Wang
- Department of Internal Medicine, Endocrinology, Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jihye Noh
- Department of Pediatrics, Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyun Jung Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Cheol Ryong Ku
- Department of Internal Medicine, Endocrinology, Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hong Koh
- Department of Pediatrics, Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Chang-Soo Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Numais Co., Ltd., Korea Seoul 04799, Republic of Korea
| | - Joon-Sang Park
- Department of Computer Engineering, Hongik University, Seoul, 04066, Republic of Korea
- Corresponding author.
| | - Young Min Shin
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Corresponding author.
| | - Hak-Joon Sung
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Corresponding author.
| |
Collapse
|
8
|
Badr AM, Alkharashi LA, Sherif IO, Alanteet AA, Alotaibi HN, Mahran YF. IL-17/Notch1/STAT3 Pathway Contributes to 5-Fluorouracil-Induced Intestinal Mucositis in Rats: Amelioration by Thymol Treatment. Pharmaceuticals (Basel) 2022; 15:1412. [PMID: 36422542 PMCID: PMC9693479 DOI: 10.3390/ph15111412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
5-Fluorouracil (5-FU) is an anticancer drug with intestinal mucositis (IM) as a deleterious side effect. Thymol is a monoterpene phenol which has been reported to possess an antioxidant and anti-inflammatory activity versus 5-FU-induced IM. The Notch pathway affects multiple cellular activities, such as cellular proliferation, in addition to inflammatory responses modulation. Accordingly, this work was carried out in order to elucidate the role of the Notch pathway in 5-FU-induced IM and to further elucidate the immunomodulatory protective mechanisms of thymol. Experimental rats were divided randomly into four groups: Control, 5-FU, 5-FU+thymol (60 mg/kg/day), and 5-FU+thymol (120 mg/kg/day). 5-FU was injected intraperitoneally at a dose of 150 mg/kg on days 6 and 7, while thymol was orally administered daily for 11 days. By the end of the study, intestinal tissues were collected for the determination of IL-17, CD4, CD8, Notch1, Hes-1, pSTAT3, and STAT-3 protein expressions. The effect of thymol on 5-FU cytotoxicity was also examined using WST1 assay. 5-FU induced a marked increase in IL-17 levels, along with a marked downregulation of CD4 and the upregulation of CD8, Notch1, Hes-1 protein expressions, and activation of STAT3 in the intestinal tissue when compared with the control group. Thymol ameliorated the changes that occurred in these parameters. Additionally, cytotoxicity testing revealed that thymol augmented the antiproliferative action of 5-FU against breast and colorectal human cancer cell lines. This study was the first to show that the IL-17/Notch1/STAT3 pathway is involved in the molecular mechanism of 5-FU-induced IM, as well as the immunomodulatory activity of thymol.
Collapse
Affiliation(s)
- Amira M. Badr
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11211, Saudi Arabia
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Layla A. Alkharashi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11211, Saudi Arabia
| | - Iman O. Sherif
- Emergency Hospital, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Alaa A. Alanteet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11211, Saudi Arabia
| | - Hind N. Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11211, Saudi Arabia
| | - Yasmen F. Mahran
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| |
Collapse
|
9
|
Singh SB, Coffman CN, Carroll-Portillo A, Varga MG, Lin HC. Notch Signaling Pathway Is Activated by Sulfate Reducing Bacteria. Front Cell Infect Microbiol 2021; 11:695299. [PMID: 34336718 PMCID: PMC8319767 DOI: 10.3389/fcimb.2021.695299] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/28/2021] [Indexed: 12/05/2022] Open
Abstract
Sulfate Reducing Bacteria (SRB), usually rare residents of the gut, are often found in increased numbers (called a SRB bloom) in inflammatory conditions such as Inflammatory Bowel Disease (IBD), pouchitis, and periodontitis. However, the underlying mechanisms of this association remain largely unknown. Notch signaling, a conserved cell-cell communication pathway, is usually involved in tissue development and differentiation. Dysregulated Notch signaling is observed in inflammatory conditions such as IBD. Lipolysaccharide and pathogens also activate Notch pathway in macrophages. In this study, we tested whether Desulfovibrio, the most dominant SRB genus in the gut, may activate Notch signaling. RAW 264.7 macrophages were infected with Desulfovibrio vulgaris (DSV) and analyzed for the expression of Notch signaling pathway-related proteins. We found that DSV induced protein expression of Notch1 receptor, Notch intracellular domain (NICD) and p21, a downstream Notch target, in a dose-and time-dependent manner. DSV also induced the expression of pro-IL1β, a precursor of IL-1β, and SOCS3, a regulator of cytokine signaling. The gamma secretase inhibitor DAPT or Notch siRNA dampened DSV-induced Notch-related protein expression as well the expression of pro-IL1β and SOCS3. Induction of Notch-related proteins by DSV was not affected by TLR4 -IN -C34(C34), a TLR4 receptor antagonist. Additionally, cell-free supernatant of DSV-infected macrophages induced NICD expression in uninfected macrophages. DSV also activated Notch pathway in the human epithelial cell line HCT116 and in mouse small intestine. Thus, our study uncovers a novel mechanism by which SRB interact with host cells by activating Notch signaling pathway. Our study lays a framework for examining whether the Notch pathway induced by SRB contributes to inflammation in conditions associated with SRB bloom and whether it can be targeted as a therapeutic approach to treat these conditions.
Collapse
Affiliation(s)
- Sudha B Singh
- Biomedical Research Institute of New Mexico, New Mexico VA Health Care System, Albuquerque, NM, United States
| | - Cristina N Coffman
- Biomedical Research Institute of New Mexico, New Mexico VA Health Care System, Albuquerque, NM, United States
| | - Amanda Carroll-Portillo
- Division of Gastroenterology and Hepatology, Department of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Matthew G Varga
- Biomedical Research Institute of New Mexico, New Mexico VA Health Care System, Albuquerque, NM, United States
| | - Henry C Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, University of New Mexico, Albuquerque, NM, United States.,Medicine Service, New Mexico VA Health Care System, Albuquerque, NM, United States
| |
Collapse
|
10
|
Keewan E, Beg S, Naser SA. Anti-TNF-α agents Modulate SARS-CoV-2 Receptors and Increase the Risk of Infection Through Notch-1 Signaling. Front Immunol 2021; 12:641295. [PMID: 34025650 PMCID: PMC8134694 DOI: 10.3389/fimmu.2021.641295] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/22/2021] [Indexed: 12/16/2022] Open
Abstract
Although millions of patients with underlining conditions are treated primarily with anti-TNF-α agents, little is known about the safety of this standard therapy during the coronavirus disease-2019 (COVID-19) pandemic. In this study, we investigated the effect of anti-TNF-α monoclonal antibodies on the cellular entry mechanism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and increasing the risk of COVID-19 development. We focused on the expression of angiotensin-converting enzyme II (ACE2), type II transmembrane serine proteases (TMPRSS2)/TNF-α converting enzyme (TACE) ratio. We also investigated the involvement of Notch-1 signaling and its downstream influence on IL-6, myeloid cell leukemia sequence-1(MCL-1) in the anti-TNF-α mode of action and increased the susceptibility to Mycobacterium avium subspecies paratuberculosis (MAP) infection. Surprisingly, anti-TNF-α downregulated ACE2 expression by 0.46-fold and increased TMPRSS2/TACE ratio by 44% in THP-1 macrophages. Treatment of macrophages with rIL-6 also downregulated ACE2 and increased TMPRSS2/TACE ratio by 54%. Interestingly, anti-TNF-α treatment upregulated Notch-1, IL-6, and MCL-1 by 1.3, 1.2, and 1.9-fold, respectively, and increased viability and burden of MAP infection in macrophages. Blocking Notch signaling doubled ACE2 expression, decreased TMPRSS2/TACE ratio by 38%, and reduced MAP viability by 56%. In a small group of patients, ACE2 level was significantly lower in the plasma from rheumatoid arthritis (RA) patients on anti-TNF-α treatment compared to healthy control. The data in this critical study demonstrated that through Notch-1/IL-6 signaling, anti-TNF-α agents decreased ACE2 expression and shedding through TMPRSS2/TACE modulation and increased the susceptibility to infection. Overall, this study warns against anti-TNF-α therapy in some patients with underlining inflammatory conditions during the COVID-19 pandemic. The findings should impact current guidelines regarding treatment decisions of patients on anti-TNF-α during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Esra'a Keewan
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Shazia Beg
- UCF Health, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Saleh A Naser
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
11
|
Christopoulos PF, Gjølberg TT, Krüger S, Haraldsen G, Andersen JT, Sundlisæter E. Targeting the Notch Signaling Pathway in Chronic Inflammatory Diseases. Front Immunol 2021; 12:668207. [PMID: 33912195 PMCID: PMC8071949 DOI: 10.3389/fimmu.2021.668207] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
The Notch signaling pathway regulates developmental cell-fate decisions and has recently also been linked to inflammatory diseases. Although therapies targeting Notch signaling in inflammation in theory are attractive, their design and implementation have proven difficult, at least partly due to the broad involvement of Notch signaling in regenerative and homeostatic processes. In this review, we summarize the supporting role of Notch signaling in various inflammation-driven diseases, and highlight efforts to intervene with this pathway by targeting Notch ligands and/or receptors with distinct therapeutic strategies, including antibody designs. We discuss this in light of lessons learned from Notch targeting in cancer treatment. Finally, we elaborate on the impact of individual Notch members in inflammation, which may lay the foundation for development of therapeutic strategies in chronic inflammatory diseases.
Collapse
Affiliation(s)
| | - Torleif T. Gjølberg
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Centre for Eye Research and Department of Ophthalmology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Stig Krüger
- Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Guttorm Haraldsen
- Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jan Terje Andersen
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Eirik Sundlisæter
- Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
12
|
Lin QN, Liu YD, Guo SE, Zhou R, Huang Q, Zhang ZM, Qin X. Schisandrin B ameliorates high-glucose-induced vascular endothelial cells injury by regulating the Noxa/Hsp27/NF-κB signaling pathway. Biochem Cell Biol 2019; 97:681-692. [PMID: 30817212 DOI: 10.1139/bcb-2018-0321] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: To address the molecular mechanism of the anti-inflammation effects of schisandrin B (Sch B) in atherosclerosis, we examined injured HMEC-1, HBMEC, and HUVEC-12 cells induced by high glucose (HG). Methods: Western blot was performed to detect the levels of the proteins Hsp27, Noxa, TLR5, p-IκBα, and p-p65 in HG-induced cells, while ELISA was used to analyze the inflammatory cytokines TNF-α, IL-6, MCP-1, and IL-1β in cells with Hsp27 or Noxa stable expression. Results: Overexpression of Hsp27 upregulated the inflammatory cytokines and the release of IκBα, promoted transportation of p65 into the nucleus, and lastly, affected the inflammation process, while Sch B counteracted the upregulation. In addition, the effect of Noxa overexpression, which is different from Hsp27 overexpression, was consistent with that of Sch B treatment. Conclusions: Sch B may inhibit the inflammatory cascade and alleviate the injury to HMEC-1, HBMEC, and HUEVC-12 cells caused by HG by regulating the Noxa/Hsp27/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Qiu-Ning Lin
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Yong-Dong Liu
- Department of Vascular Surgery, Affiliated Liutie Central Hospital & Clinical Medical College of Guangxi Medical University, Liuzhou 545007, P.R. China
| | - Si-En Guo
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Rui Zhou
- Department of Hepatobiliary Surgery, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang 537120, P.R. China
| | - Qun Huang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Zhan-Man Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Xiao Qin
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| |
Collapse
|
13
|
Kaur H, Moreau R. Role of mTORC1 in intestinal epithelial repair and tumorigenesis. Cell Mol Life Sci 2019; 76:2525-2546. [PMID: 30944973 PMCID: PMC11105546 DOI: 10.1007/s00018-019-03085-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/08/2019] [Accepted: 03/25/2019] [Indexed: 12/15/2022]
Abstract
mTORC1 signaling is the prototypical pathway regulating protein synthesis and cell proliferation. mTORC1 is active in stem cells located at the base of intestinal crypts but silenced as transit-amplifying cells differentiate into enterocytes or secretory cells along the epithelium. After an insult or injury, self-limiting and controlled activation of mTORC1 is critical for the renewal and repair of intestinal epithelium. mTORC1 promotes epithelial cell renewal by driving cryptic stem cell division, and epithelial cell repair by supporting the dedifferentiation and proliferation of enterocytes or secretory cells. Under repeated insult or injury, mTORC1 becomes constitutively active, triggering an irreversible return to stemness, cell division, proliferation, and inflammation among dedifferentiated epithelial cells. Epithelium-derived cytokines promulgate inflammation within the lamina propria, which in turn releases inflammatory factors that act back on the epithelium where undamaged intestinal epithelial cells participate in the pervading state of inflammation and become susceptible to tumorigenesis.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Régis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
14
|
Fu D, Xiao C, Xie Y, Gao J, Ye S. MiR-3926 inhibits synovial fibroblasts proliferation and inflammatory cytokines secretion through targeting toll like receptor 5. Gene 2018; 687:200-206. [PMID: 30412746 DOI: 10.1016/j.gene.2018.11.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 09/17/2018] [Accepted: 11/04/2018] [Indexed: 01/07/2023]
Abstract
Rheumatoid arthritis synovial fibroblasts (RASFs) play a key role in the pathogenesis of rheumatoid arthritis (RA). This study was aimed to investigate the effects of miR-3926 on the biological activities of RASFs. The results showed that miR-3926 was significantly down-regulated in RASFs and RA synovial tissue. Overexpression of miR-3926 significantly inhibited RASFs proliferation and decreased the secretion of inflammatory cytokines including TNF-α, IL-1β and IL-6 in RASFs. TLR5 was identified to be a direct target of miR-3926. TLR5 showed an opposite expression trends with miR-3926 in RASFs and RA synovial tissue. Overexpression of miR-3926 led to a reduction of endogenous TLR5 in RASFs, whereas down-regulation of miR-3926 increased TLR5 expression. Knocking down of TLR5 significantly inhibited RASFs proliferation and inflammatory cytokines secretion. Rescue experiments with a miR-3926-resistant variant of TLR5 showed that overexpression of TLR5 restored RASFs proliferation and inflammatory cytokines secretion in miR-3926-overexpressing RASFs. In conclusion, miR-3926 is downregulated in RA synovial tissues and its overexpression caused the inhibitory effects on RASF proliferation and inflammatory cytokines secretion by targeting TLR5. The miR-3926/TLR5 pathway may represent a novel target for prevention and treatment of RA.
Collapse
Affiliation(s)
- Di Fu
- Department of Rheumatology, First Affiliated Hospital of Guangzhou Medical University, No.151 Yanjiangxi Road, Guangzhou, Guangdong 510120, PR China
| | - Chuyin Xiao
- Department of Rheumatology, First Affiliated Hospital of Guangzhou Medical University, No.151 Yanjiangxi Road, Guangzhou, Guangdong 510120, PR China
| | - Yingying Xie
- Department of Rheumatology, First Affiliated Hospital of Guangzhou Medical University, No.151 Yanjiangxi Road, Guangzhou, Guangdong 510120, PR China
| | - Jianquan Gao
- Department of Rheumatology, First Affiliated Hospital of Guangzhou Medical University, No.151 Yanjiangxi Road, Guangzhou, Guangdong 510120, PR China
| | - Shanhui Ye
- Department of Rheumatology, First Affiliated Hospital of Guangzhou Medical University, No.151 Yanjiangxi Road, Guangzhou, Guangdong 510120, PR China.
| |
Collapse
|
15
|
Liu Y, Dong Y, Zhu X, Fan H, Xu M, Chen Q, Nan Z, Wu H, Deng S, Liu X, Zuo D, Yang J. MiR-155 inhibition ameliorates 2, 4, 6-Trinitrobenzenesulfonic acid (TNBS)-induced experimental colitis in rat via influencing the differentiation of Th17 cells by Jarid2. Int Immunopharmacol 2018; 64:401-410. [PMID: 30253332 DOI: 10.1016/j.intimp.2018.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 08/23/2018] [Accepted: 09/08/2018] [Indexed: 01/19/2023]
Abstract
Th17 cells play an important role in the immune imbalance and inflammatory state in colonic mucosa of Inflammatory Bowel Disease (IBD) and to clarify the mechanism that affect the differentiation of Th17 cells will help us find a new target for the treatment of IBD. MiR-155 which is reported to have an important role in regulating immune system function is also detected to be significantly up-regulated in colonic tissues of IBD patients. However, whether and how miR-155 affects the differentiation of Th17 cells in the colon of IBD patients is still worth studying. Here, we investigated the role of miR-155 in TNBS-induced rat colitis. Firstly, we found that the disease activity index (DAI) and Colon pathological changes were significantly reduced (P < 0.05) by using miR-155 inhibition sequences delivered by lentiviral vector, which revealed that miR-155 inhibition ameliorated TNBS-Induced experimental colitis. Then, we carried out flow cytometry, ELISA, qRT-PCR, and found that in TNBS+miR-155 inhibition group, the proportion of Th17 cells in spleens and mesenteric lymph nodes (MLNs) and the level of the Th17 cell-associated cytokines IL-6, IL-17A, IL-17F and IL-21 in colon tissues were significantly reduced (P < 0.05), which revealed that miR-155 inhibition regulated the differentiation and function of Th17 cells. Finally, we discovered that Jarid2 was significantly elevated (P < 0.05) by miR-155 inhibition and notch1 expression was inversely correlated with Jarid2 by using Immunohistochemistry and western blot. This study suggests that miR-155 inhibition ameliorates TNBS-induced colitis by regulating the Th17 cells differentiation and function and Jarid2/notch1 is closely related with the process.
Collapse
Affiliation(s)
- Yujin Liu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yalan Dong
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiwen Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Meng Xu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qianyun Chen
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhen Nan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Wu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuangjiao Deng
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xingxing Liu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Dongmei Zuo
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jia Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
16
|
Poulsen LLC, Edelmann RJ, Krüger S, Diéguez-Hurtado R, Shah A, Stav-Noraas TE, Renzi A, Szymanska M, Wang J, Ehling M, Benedito R, Kasprzycka M, Bækkevold E, Sundnes O, Midwood KS, Scott H, Collas P, Siebel CW, Adams RH, Haraldsen G, Sundlisæter E, Hol J. Inhibition of Endothelial NOTCH1 Signaling Attenuates Inflammation by Reducing Cytokine-Mediated Histone Acetylation at Inflammatory Enhancers. Arterioscler Thromb Vasc Biol 2018; 38:854-869. [PMID: 29449332 DOI: 10.1161/atvbaha.117.310388] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/23/2018] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Endothelial upregulation of adhesion molecules serves to recruit leukocytes to inflammatory sites and appears to be promoted by NOTCH1; however, current models based on interactions between active NOTCH1 and NF-κB components cannot explain the transcriptional selectivity exerted by NOTCH1 in this context. APPROACH AND RESULTS Observing that Cre/Lox-induced conditional mutations of endothelial Notch modulated inflammation in murine contact hypersensitivity, we found that IL (interleukin)-1β stimulation induced rapid recruitment of RELA (v-rel avian reticuloendotheliosis viral oncogene homolog A) to genomic sites occupied by NOTCH1-RBPJ (recombination signal-binding protein for immunoglobulin kappa J region) and that NOTCH1 knockdown reduced histone H3K27 acetylation at a subset of NF-κB-directed inflammatory enhancers. CONCLUSIONS Our findings reveal that NOTCH1 signaling supports the expression of a subset of inflammatory genes at the enhancer level and demonstrate how key signaling pathways converge on chromatin to coordinate the transition to an infla mmatory endothelial phenotype.
Collapse
Affiliation(s)
- Lars la Cour Poulsen
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Reidunn Jetne Edelmann
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Stig Krüger
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Rodrigo Diéguez-Hurtado
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Akshay Shah
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Tor Espen Stav-Noraas
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Anastasia Renzi
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Monika Szymanska
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Junbai Wang
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Manuel Ehling
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Rui Benedito
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Monika Kasprzycka
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Espen Bækkevold
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Olav Sundnes
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Kim S Midwood
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Helge Scott
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Philippe Collas
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Christian W Siebel
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Ralf H Adams
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Guttorm Haraldsen
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.).
| | - Eirik Sundlisæter
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| | - Johanna Hol
- From the Department of Pathology, Oslo University Hospital Rikshospitalet (L.l.C.P., R.J.E., S.K., T.E.S.-N., A.R., M.S., J.W., M.K., E.B., O.S., H.S., G.H., E.S., J.H.), Department of Pathology, Institute for Clinical Medical Sciences (H.S., G.H.) and Department of Molecular Medicine, Institute for Basal Medical Sciences (A.S., P.C.), University of Oslo, Norway; Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, University of Münster, Germany (R.D.-H., M.E., R.B., R.H.A.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, United Kingdom (K.S.M.); and Department of Discovery Oncology, Genentech, Inc, South San Francisco, CA (C.W.S.)
| |
Collapse
|
17
|
N-3 vs. n-6 fatty acids differentially influence calcium signalling and adhesion of inflammatory activated monocytes: impact of lipid rafts. Inflamm Res 2016; 65:881-894. [DOI: 10.1007/s00011-016-0971-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 04/01/2016] [Accepted: 06/28/2016] [Indexed: 11/30/2022] Open
|
18
|
Cheng YL, Choi Y, Sobey CG, Arumugam TV, Jo DG. Emerging roles of the γ-secretase-notch axis in inflammation. Pharmacol Ther 2014; 147:80-90. [PMID: 25448038 DOI: 10.1016/j.pharmthera.2014.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/03/2014] [Indexed: 12/14/2022]
Abstract
γ-Secretase is a distinct proteolytic complex required for the activation of many transmembrane proteins. The cleavage of substrates by γ-secretase plays diverse biological roles in producing essential products for the organism. More than 90 transmembrane proteins have been reported to be substrates of γ-secretase. Two of the most widely known and studied of these substrates are the amyloid precursor protein (APP) and the Notch receptor, which are precursors for the generation of amyloid-β (Aβ) and the Notch intracellular domain (NICD), respectively. The wide spectrum of γ-secretase substrates has made analyses of the pathology of γ-secretase-related diseases and underlying mechanisms challenging. Inflammation is an important aspect of disease pathology that requires an in-depth analysis. γ-Secretase may contribute to disease development or progression by directly increasing and regulating production of pro-inflammatory cytokines. This review summarizes recent evidence for a role of γ-secretase in inflammatory diseases, and discusses the potential use of γ-secretase inhibitors as an effective future treatment option.
Collapse
Affiliation(s)
- Yi-Lin Cheng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Yuri Choi
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | | | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
19
|
Taniguchi K, Karin M. IL-6 and related cytokines as the critical lynchpins between inflammation and cancer. Semin Immunol 2014; 26:54-74. [PMID: 24552665 DOI: 10.1016/j.smim.2014.01.001] [Citation(s) in RCA: 519] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/06/2014] [Indexed: 11/17/2022]
Abstract
Inflammatory responses play pivotal roles in cancer development, including tumor initiation, promotion, progression, and metastasis. Cytokines are now recognized as important mediators linking inflammation and cancer, and are therefore potential therapeutic and preventive targets as well as prognostic factors. The interleukin (IL)-6 family of cytokines, especially IL-6 and IL-11, is highly up-regulated in many cancers and considered as one of the most important cytokine families during tumorigenesis and metastasis. This review discusses molecular mechanisms linking the IL-6 cytokine family to solid malignancies and their treatment.
Collapse
Affiliation(s)
- Koji Taniguchi
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; UC San Diego Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|