1
|
Zhang M, Liu T, Luo L, Zhang Y, Chen Q, Wang F, Xie Y. Common diagnostic biomarkers and molecular mechanisms of Helicobacter pylori infection and inflammatory bowel disease. Front Immunol 2024; 15:1492810. [PMID: 39712025 PMCID: PMC11659760 DOI: 10.3389/fimmu.2024.1492810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
Background Helicobacter pylori (H. pylori) may be present in the intestinal mucosa of patients with inflammatory bowel disease (IBD), which is a chronic inflammation of the gastrointestinal tract. The role of H. pylori in the pathogenesis of IBD remains unclear. In this study, bioinformatics techniques were used to investigate the correlation and co-pathogenic pathways between H. pylori and IBD. Methods The following matrix data were downloaded from the GEO database: H. pylori-associated gastritis, GSE233973 and GSE27411; and IBD, GSE3365 and GSE179285. Differential gene analysis was performed via the limma software package in the R environment. A protein-protein interaction (PPI) network of DEGs was constructed via the STRING database. Cytoscape software, through the CytoHubba plugin, filters the PPI subnetwork and identifies Hub genes. Validation of the Hub genes was performed in the validation set. Immune analysis was conducted via the CIBERSORT algorithm. Transcription factor interaction and small molecule drug analyses of the Hub genes were also performed. Results Using the GSE233973 and GSE3365 datasets, 151 differentially expressed genes (DEGs) were identified. GO enrichment analysis revealed involvement in leukocyte migration and chemotaxis, response to lipopolysaccharides, response to biostimulatory stimuli, and regulation of interleukin-8 (IL-8) production. Ten Hub genes (TLR4, IL10, CXCL8, IL1B, TLR2, CXCR2, CCL2, IL6, CCR1 and MMP-9) were identified via the PPI network and Cytoscape software. Enrichment analysis of the Hub genes focused on the lipopolysaccharide response, bacterial molecular response, biostimulatory response and leukocyte movement. Validation using the GSE27411 and GSE179285 datasets revealed that MMP-9 was significantly upregulated in both the H. pylori and IBD groups. The CIBERSORT algorithm revealed immune infiltration differences between the control and disease groups of IBD patients. Additionally, the CMap database identified the top 11 small molecule compounds across 10 cell types, including TPCA-1, AS-703026 and memantine, etc. Conclusion Our study revealed the co-pathogenic mechanism between H. pylori and IBD and identified 10 Hub genes related to cellular immune regulation and signal transduction. The expression of MMP-9 is significantly upregulated in both H. pylori infection and IBD. This study provides a new perspective for exploring the prevention and treatment of H. pylori infection and IBD.
Collapse
Affiliation(s)
- Minglin Zhang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tong Liu
- Department of General Surgery, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Lijun Luo
- School of Medical Laboratory Science, Hebei North University, Zhangjiakou, Hebei, China
| | - Yi Zhang
- Department of General Surgery, The First People's Hospital of Qingzhen City, Guiyang, Guizhou, China
| | - Qijiao Chen
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Fen Wang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuxin Xie
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
2
|
Wu Y, Shen J. Unraveling the intricacies of neutrophil extracellular traps in inflammatory bowel disease: Pathways, biomarkers, and promising therapies. Cytokine Growth Factor Rev 2024; 80:156-167. [PMID: 39438227 DOI: 10.1016/j.cytogfr.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024]
Abstract
The development of inflammatory bowel disease (IBD), including ulcerative colitis and Crohn's disease, involves various factors and is characterized by persistent inflammation of the mucosal lining. However, the role of neutrophils in this process remains controversial. Neutrophil extracellular traps (NETs), which consist of chromatin, antimicrobial proteins, and oxidative enzymes, are released by neutrophils to trap pathogens. They are also involved in various immune-mediated and vascular diseases. NETs act as a vital defense mechanisms at the gut-mucosal interface and are frequently exposed to bacterial, viral, and fungal threats. However, they can also contribute to inflammation and worsen imbalances in the gut bacteria. Recent studies have suggested that NETs have a significant impact on IBD development. Previous studies have shown increased levels of NETs in tissue and blood samples from patients with IBD, as well as in experimental colitis mouse models. Therefore, this review discusses how NETs are formed and their role in the pathophysiology of IBD. It discusses how NETs may lead to tissue damage and contribute to IBD-associated complications. Moreover, non-invasive biomarkers are needed to replace invasive procedures such as endoscopy to better evaluate the disease status. Given the crucial role of NETs in IBD progression, this review focuses on potential NET biomarkers that can help predict the evolution of IBD. Furthermore, this review identifies potential therapeutic targets for regulating NET production, which could expand the range of available treatment options for IBD.
Collapse
Affiliation(s)
- Yilin Wu
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai 200127, China; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China; Shanghai Institute of Digestive Disease, No.160 PuJian Road, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai 200127, China; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China; Shanghai Institute of Digestive Disease, No.160 PuJian Road, China.
| |
Collapse
|
3
|
Yatsenko T, Rios R, Nogueira T, Salama Y, Takahashi S, Adachi E, Tabe Y, Hattori N, Osada T, Naito T, Takahashi K, Hattori K, Heissig B. The influence of 4G/5G polymorphism in the plasminogen-activator-inhibitor-1 promoter on COVID-19 severity and endothelial dysfunction. Front Immunol 2024; 15:1445294. [PMID: 39281671 PMCID: PMC11392769 DOI: 10.3389/fimmu.2024.1445294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/08/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction Plasminogen activator inhibitor-1 (PAI-1) is linked to thrombosis and endothelial dysfunction in severe COVID-19. The +43 G>A PAI-1 and 4G/5G promoter polymorphism can influence PAI-1 expression. The 4G5G PAI-1 promoter gene polymorphism constitutes the 4G4G, 4G5G, and 5G5G genotypes. However, the impact of PAI-1 polymorphisms on disease severity or endothelial dysfunction remains unclear. Methods Clinical data, sera, and peripheral blood mononuclear cells (PBMCs) of COVID-19 patients were studied. Results Comorbidities and clinical biomarkers did not correlate with genotypes in either polymorphism. However, differences between fibrinolytic factors and interleukin-1β (IL-1β) were identified in genotypes of the 4G/5G but not the 43 G>A PAI polymorphism. Patients with the 4G4G genotype of the 4G/5G polymorphism showed high circulating PAI-1, mainly complexed with plasminogen activators, and low IL-1β and plasmin levels, indicating suppressed fibrinolysis. NFκB was upregulated in PBMCs of COVID-19 patients with the 4G4G genotype. Discussion Mechanistically, IL-1β enhanced PAI-1 expression in 4G4G endothelial cells, preventing the generation of plasmin and cleavage products like angiostatin, soluble uPAR, and VCAM1. We identified inflammation-induced endothelial dysfunction coupled with fibrinolytic system overactivation as a risk factor for patients with the 5G5G genotype.
Collapse
Affiliation(s)
- Tetiana Yatsenko
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Enzymes Chemistry and Biochemistry, Palladin Institute of Biochemistry of National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Ricardo Rios
- Institute of Computing, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Tatiane Nogueira
- Institute of Computing, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Yousef Salama
- An-Najah Center for Cancer and Stem Cell Research, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Satoshi Takahashi
- Division of Clinical Precision Research Platform, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Eisuke Adachi
- Department of Infectious Diseases and Applied Immunology, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Yoko Tabe
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Nobutaka Hattori
- Center for Genome and Regenerative Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Taro Osada
- Department of Gastroenterology, Juntendo University, Urayasu Hospital, Urayasu-shi, Japan
| | - Toshio Naito
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Koichi Hattori
- Center for Genome and Regenerative Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Beate Heissig
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
4
|
Gui H, Chen X, Nie Y, Zhang X. Enhancing the revelation of key genes and interaction networks in non-small cell lung cancer with major depressive disorder: A bioinformatics analysis. Health Sci Rep 2024; 7:e2167. [PMID: 38933422 PMCID: PMC11199184 DOI: 10.1002/hsr2.2167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/01/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Background and Aims Lung cancer is ranked as the second most prevalent form of cancer worldwide. Nonsmall cell lung cancer (NSCLC) represents the predominant histological subtype. Research suggests that one-third of lung cancer patients also experiencing depression. Antidepressants play an indispensable role in the management of NSCLC. Despite significant advancements in treatment, lung cancer patients still face a high mortality rate. Major depressive disorder (MDD) and related antidepressants involved in treatment efficacy and prognosis of NSCLC. However, there has been a lack of screening and analysis regarding genes and networks associated with both NSCLC and MDD. Methods To investigate the correlation between MDD and NSCLC, our discovery and validation analysis included four datasets from the Gene Expression Omnibus database from NSCLC or MDD. Differential gene expression (DEGs) analysis, GO and KEGG Pathway, and protein-protein interaction network analyzes to identify hub genes, networks, and associated observations link between MDD and NSCLC. Results The analysis of two datasets yielded a total of 84 downregulated and 52 upregulated DEGs. Pathway enrichment analyzes indicated that co-upregulated genes were enriched in the regulation of positive regulation of cellular development, collagen-containing extracellular matrix (ECM), cytokine binding, and axon guidance. We identified 20 key genes, which were further analyzed using the MCODE plugin to identify two core subnetworks. The integration of functionally similar genes provided valuable insights into the potential involvement of these hub genes in diverse biological processes including angiogenesis humoral immune response regulation inflammatory response organization ECM network. Conclusion We have identified a total of 136 DEGs that participate in multiple biological signaling pathways. A total of 20 hub genes have demonstrated robust associations, potentially indicating novel diagnostic and therapeutic targets for both diseases.
Collapse
Affiliation(s)
- Huan Gui
- School of MedicineGuizhou UniversityGuiyangChina
- Department of Hyperbaric OxygenPeople's Hospital of Qianxinan Buyi and Miao Minority Autonomous PrefectureXingyiChina
| | - Xulong Chen
- School of MedicineGuizhou UniversityGuiyangChina
- Department of UrologyAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Yingjie Nie
- School of MedicineGuizhou UniversityGuiyangChina
- Department of Science and ResearchHong Kong University Shenzhen HospitalShenzhenChina
| | - Xiangyan Zhang
- School of MedicineGuizhou UniversityGuiyangChina
- NHC Key Laboratory of Pulmonary Immunological DiseasesGuizhou Provincial People's HospitalGuiyangChina
| |
Collapse
|
5
|
Li X, Yao Z, Qian J, Li H, Li H. Lactate Protects Intestinal Epithelial Barrier Function from Dextran Sulfate Sodium-Induced Damage by GPR81 Signaling. Nutrients 2024; 16:582. [PMID: 38474712 DOI: 10.3390/nu16050582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
The dysregulation of the intestinal epithelial barrier significantly contributes to the inflammatory progression of ulcerative colitis. Recent studies have indicated that lactate, produced by gut bacteria or derived from fermented foods, plays a key role in modulating inflammation via G-protein-coupled receptor 81 (GPR81). In this study, we aimed to investigate the potential role of GPR81 in the progression of colitis and to assess the impact of lactate/GPR81 signaling on intestinal epithelial barrier function. Our findings demonstrated a downregulation of GPR81 protein expression in patients with colitis. Functional verification experiments showed that Gpr81-deficient mice exhibited more severe damage to the intestinal epithelial barrier and increased susceptibility to DSS-induced colitis, characterized by exacerbated oxidative stress, elevated inflammatory cytokine secretion, and impaired expression of tight-junction proteins. Mechanistically, we found that lactate could suppress TNF-α-induced MMP-9 expression and prevent the disruption of tight-junction proteins by inhibiting NF-κB activation through GPR81 in vitro. Furthermore, our study showed that dietary lactate could preserve intestinal epithelial barrier function against DSS-induced damage in a GPR81-dependent manner in vivo. Collectively, these results underscore the crucial involvement of the lactate/GPR81 signaling pathway in maintaining intestinal epithelial barrier function, providing a potential therapeutic strategy for ulcerative colitis.
Collapse
Affiliation(s)
- Xiaojing Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhijie Yao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jin Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hongling Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Haitao Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
6
|
Urokinase-type plasminogen activator blockade ameliorates experimental colitis in mice. Sci Rep 2023; 13:2899. [PMID: 36806262 PMCID: PMC9938860 DOI: 10.1038/s41598-023-29824-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 02/10/2023] [Indexed: 02/20/2023] Open
Abstract
Although several angiogenesis-related factors are reportedly involved in the pathogenesis of ulcerative colitis (UC), the mechanisms by which they contribute to disease are unclear. We first examined the expression of angiogenesis-related factors in inflamed colorectal tissue of UC patients using antibody array, and identified the 5 factors with highest expression, which included matrix metalloproteinase-8, urokinase-type plasminogen activator (uPA), angiostatin/plasminogen, hepatocyte growth factor and endoglin. Subsequent real-time PCR experiments using additional colorectal tissues revealed that uPA mRNA levels were significantly higher in inflamed tissues than in non-inflamed tissues, and significantly correlated with the severity of UC. Mirror section immunohistochemistry revealed that uPA was expressed in the neutrophils of inflamed colorectal tissues. We administered dextran sulfate sodium (DSS) in drinking water to uPA knockout (uPA-/-) mice, and found that the disease activity index in uPA-/- mice was marginally lower and the histological score in uPA-/- mice was significantly lower than those in wild-type mice, suggesting the importance of uPA in colitis. When an uPA-selective inhibitor, UK122, was administered to DSS-treated C57BL6J mice, the disease activity index and histological score in those mice were significantly lower compared with control mice. Multiple cytokine/chemokine assay using colorectal tissues from uPA-/- and UK122-treated mice revealed significantly lowered level of RANTES. In conclusion, uPA was highly expressed in neutrophils of the inflamed mucosa of UC patients, and the expression level correlated with the severity of UC. Genetic uPA deletion or pharmacological uPA blockade significantly ameliorated colitis in mice, concomitant with downregulation of RANTES.
Collapse
|
7
|
The serine protease plasmin plays detrimental roles in epithelial sodium channel activation and podocyte injury in Dahl salt-sensitive rats. Hypertens Res 2023; 46:50-62. [PMID: 36241707 DOI: 10.1038/s41440-022-01064-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/06/2022] [Accepted: 09/26/2022] [Indexed: 02/03/2023]
Abstract
Salt-sensitive hypertension is associated with poor clinical outcomes. The epithelial sodium channel (ENaC) in the kidney plays pivotal roles in sodium reabsorption and blood pressure regulation, in which its γ subunit is activated by extracellular serine proteases. In proteinuric nephropathies, plasmin filtered through injured glomeruli reportedly activates γENaC in the distal nephron and causes podocyte injury. We previously reported that Dahl salt-sensitive (DS) rats fed a high-salt (HS) diet developed hypertension and proteinuria along with γENaC activation and that a synthetic serine protease inhibitor, camostat mesilate, mitigated these changes. However, the role of plasmin in DS rats remained unclear. In this study, we evaluated the relationship between plasmin and hypertension as well as podocyte injury and the effects of plasmin inhibitors in DS rats. Five-week-old DS rats were divided into normal-salt diet, HS diet, and HS+plasmin inhibitor (either tranexamic acid [TA] or synthetic plasmin inhibitor YO-2) groups. After blood pressure measurement and 24 h urine collection over 5 weeks, rats were sacrificed for biochemical analyses. The HS group displayed severe hypertension and proteinuria together with activation of plasmin in urine and γENaC in the kidney, which was significantly attenuated by YO-2 but not TA. YO-2 inhibited the attachment of plasmin(ogen) to podocytes and alleviated podocyte injury by inhibiting apoptosis and inflammatory/profibrotic cytokines. YO-2 also suppressed upregulation of protease-activated receptor-1 and phosphorylated ERK1/2. These results indicate an important role of plasmin in the development of salt-sensitive hypertension and related podocyte injury, suggesting plasmin inhibition as a potential therapeutic strategy.
Collapse
|
8
|
Prudovsky I, Kacer D, Zucco VV, Palmeri M, Falank C, Kramer R, Carter D, Rappold J. Tranexamic acid: Beyond antifibrinolysis. Transfusion 2022; 62 Suppl 1:S301-S312. [PMID: 35834488 DOI: 10.1111/trf.16976] [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: 11/30/2021] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022]
Abstract
Tranexamic acid (TXA) is a popular antifibrinolytic drug widely used in hemorrhagic trauma patients and cardiovascular, orthopedic, and gynecological surgical patients. TXA binds plasminogen and prevents its maturation to the fibrinolytic enzyme plasmin. A number of studies have demonstrated the broad life-saving effects of TXA in trauma, superior to those of other antifibrinolytic agents. Besides preventing fibrinolysis and blood loss, TXA has been reported to suppress posttraumatic inflammation and edema. Although the efficiency of TXA transcends simple inhibition of fibrinolysis, little is known about its mechanisms of action besides the suppression of plasmin maturation. Understanding the broader effects of TXA at the cell, organ, and organism levels are required to elucidate its potential mechanisms of action transcending antifibrinolytic activity. In this article, we provide a brief review of the current clinical use of TXA and then focus on the effects of TXA beyond antifibrinolytics such as its anti-inflammatory activity, protection of the endothelial and epithelial monolayers, stimulation of mitochondrial respiration, and suppression of melanogenesis.
Collapse
Affiliation(s)
- Igor Prudovsky
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, Maine, USA
| | - Doreen Kacer
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, Maine, USA
| | - Victoria Vieira Zucco
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, Maine, USA
| | - Monica Palmeri
- Maine Medical Center Cardiovascular Institute, Maine Medical Center, Portland, Maine, USA
| | - Carolyne Falank
- Department of Trauma, Maine Medical Center, Maine Medical Center, Portland, Maine, USA
| | - Robert Kramer
- Maine Medical Center Cardiovascular Institute, Maine Medical Center, Portland, Maine, USA
| | - Damien Carter
- Department of Trauma, Maine Medical Center, Maine Medical Center, Portland, Maine, USA
| | - Joseph Rappold
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, Maine, USA.,Department of Trauma, Maine Medical Center, Maine Medical Center, Portland, Maine, USA
| |
Collapse
|
9
|
Cao Q, Mertens RT, Sivanathan KN, Cai X, Xiao P. Macrophage orchestration of epithelial and stromal cell homeostasis in the intestine. J Leukoc Biol 2022; 112:313-331. [PMID: 35593111 PMCID: PMC9543232 DOI: 10.1002/jlb.3ru0322-176r] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 11/06/2022] Open
Abstract
The intestinal tract is a complex ecosystem where numerous cell types of epithelial, immune, neuronal, and endothelial origin coexist in an intertwined, highly organized manner. The functional equilibrium of the intestine relies heavily on the proper crosstalk and cooperation among each cell population. Furthermore, macrophages are versatile, innate immune cells that participate widely in the modulation of inflammation and tissue remodeling. Emerging evidence suggest that macrophages are central in orchestrating tissue homeostasis. Herein, we describe how macrophages interact with epithelial cells, neurons, and other types of mesenchymal cells under the context of intestinal inflammation, followed by the therapeutic implications of cellular crosstalk pertaining to the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Qian Cao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Randall Tyler Mertens
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Kisha Nandini Sivanathan
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Xuechun Cai
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China.,Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
10
|
Ben Moftah M, Eswayah A. Intricate relationship between SARS-CoV-2-induced shedding and cytokine storm generation: A signaling inflammatory pathway augmenting COVID-19. HEALTH SCIENCES REVIEW (OXFORD, ENGLAND) 2022; 2:100011. [PMID: 35013738 PMCID: PMC8734057 DOI: 10.1016/j.hsr.2021.100011] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 12/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), through its ability to induce cytokine release syndrome, can set up a generalized inflammatory response together with activating multiple inflammatory pathways, which contributes to a dramatic increase in the number of mortalities and morbidities worldwide. Reportedly, the manipulative nature of coronavirus disease 2019 (COVID-19), which targets the immune system, often focuses on specific inflammation-related pathways, usually confined to interleukins and tumor necrosis factor-α (TNF-α), with a great emphasis on therapeutic approaches targeting the inhibition of these inflammatory mediators. The involvement of a disintegrin and metalloprotease 17 (ADAM-17) and matrix metalloproteinase-9 (MMP-9) in the pathogenesis of COVID-19, through their ability to potentiate the cytokine storm during an episode of SARS-CoV-2 infection, often goes unnoticed. In this review, the intricate relationship between ADAM-17 and MMP-9 together with angiotensin-converting enzyme 2 (ACE-2) as the main target for SARS-CoV-2 is highlighted in detail through a compilation of evidence-based literature; thus, we shed light on a proposed inflammatory pathway that COVID-19 may exploit to provoke an inflammatory response of a complex nature. Conclusively, our proposed mechanism acts as a means to developing a therapeutic approach aimed at modulating the intricate communication between ADAM-17 and MMP-9, where a great emphasis on the role of ACE-2 shedding and subsequent elevation in angiotensin II (Ang-II) levels is crucial to understanding the awry inflammatory response in patients with COVID-19. From this concept, designing a therapeutic strategy targeting multiple inflammatory mediators and enzymes simultaneously is another approach to unravel this global pandemic.
Collapse
Affiliation(s)
- Moayed Ben Moftah
- Department of Medicinal and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tripoli, Tripoli, Libya
| | - Asma Eswayah
- Department of Medicinal and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tripoli, Tripoli, Libya
| |
Collapse
|
11
|
Hayashi Y, Nakase H. The Molecular Mechanisms of Intestinal Inflammation and Fibrosis in Crohn’s Disease. Front Physiol 2022; 13:845078. [PMID: 35222098 PMCID: PMC8874128 DOI: 10.3389/fphys.2022.845078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/26/2022] [Indexed: 12/20/2022] Open
Abstract
Crohn’s disease (CD) is an inflammatory bowel disease (IBD) with repeated remissions and relapses. As the disease progresses, fibrosis and narrowing of the intestine occur, leading to severe complications such as intestinal obstruction. Endoscopic balloon dilatation, surgical stricture plasty, and bowel resection have been performed to treat intestinal stenosis. The clinical issue is that some patients with CD have a recurrence of intestinal stenosis even after the medical treatments. On the other hand, there exist no established medical therapies to prevent stenosis. With the progressive intestinal inflammation, cytokines and growth factors, including transforming growth factor (TGF-β), stimulate intestinal myofibroblasts, contributing to fibrosis of the intestine, smooth muscle hypertrophy, and mesenteric fat hypertrophy. Therefore, chronically sustained inflammation has long been considered a cause of intestinal fibrosis and stenosis. Still, even after the advent of biologics and tighter control of inflammation, intestinal fibrosis’s surgical rate has not necessarily decreased. It is essential to elucidate the mechanisms involved in intestinal fibrosis in CD from a molecular biological level to overcome clinical issues. Recently, much attention has been paid to several key molecules of intestinal fibrosis: peroxisome proliferator-activating receptor gamma (PPARγ), toll-like receptor 4 (TLR4), adherent-invasive Escherichia coli (AIEC), Th17 immune response, and plasminogen activator inhibitor 1 (PAI-1). As a major problem in the treatment of CD, the pathophysiology of patients with CD is not the same and varies depending on each patient. It is necessary to integrate these key molecules for a better understanding of the mechanism of intestinal inflammation and fibrosis.
Collapse
|
12
|
Das L, Banki MA, Azmoon P, Pizzo D, Gonias SL. Enzymatically Inactive Tissue-Type Plasminogen Activator Reverses Disease Progression in the Dextran Sulfate Sodium Mouse Model of Inflammatory Bowel Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:590-601. [PMID: 33465348 PMCID: PMC8101053 DOI: 10.1016/j.ajpath.2021.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
Enzymatically inactive tissue-type plasminogen activator (EI-tPA) does not activate fibrinolysis, but interacts with the N-methyl-d-aspartate receptor (NMDA-R) and low-density lipoprotein receptor-related protein-1 (LRP1) in macrophages to block innate immune system responses mediated by toll-like receptors. Herein, we examined the ability of EI-tPA to treat colitis in mice, induced by dextran sulfate sodium. In two separate studies, designed to generate colitis of differing severity, a single dose of EI-tPA administered after inflammation established significantly improved disease parameters. EI-tPA-treated mice demonstrated improved weight gain. Stools improved in character and became hemoccult negative. Abdominal tenderness decreased. Colon shortening significantly decreased in EI-tPA-treated mice, suggesting attenuation of irreversible tissue damage and remodeling. Furthermore, histopathologic evidence of disease decreased in the distal 25% of the colon in EI-tPA-treated mice. EI-tPA did not decrease the number of CD45-positive leukocytes or F4/80-positive macrophage-like cells detected in extracts of colons from dextran sulfate sodium-treated mice as assessed by flow cytometry. However, multiple colon cell types expressed the NMDA-R, suggesting the ability of diverse cells, including CD3-positive cells, CD103-positive cells, Ly6G-positive cells, and epithelial cell adhesion molecule-positive epithelial cells to respond to EI-tPA. Mesenchymal cells that line intestinal crypts and provide barrier function expressed LRP1, thereby representing another potential target for EI-tPA. These results demonstrate that the NMDA-R/LRP1 receptor system may be a target for drug development in diseases characterized by tissue damage and chronic inflammation.
Collapse
Affiliation(s)
- Lipsa Das
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Michael A Banki
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Pardis Azmoon
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Donald Pizzo
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Steven L Gonias
- Department of Pathology, University of California San Diego, La Jolla, California.
| |
Collapse
|
13
|
Jiang Y, Zhao X, Yu J, Wang Q, Wen C, Huang L. Deciphering potential pharmacological mechanism of Sha-Shen-Mai-Dong decoction on primary Sjogren's syndrome. BMC Complement Med Ther 2021; 21:79. [PMID: 33648502 PMCID: PMC7923330 DOI: 10.1186/s12906-021-03257-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 02/22/2021] [Indexed: 12/29/2022] Open
Abstract
Background Sha-Shen-Mai-Dong decoction (SSMD) is a classical prescription widely used in primary Sjogren’s Syndrome (pSS) therapy. This study aims to explore the potential pharmacological mechanism of SSMD on pSS. Methods Active components of SSMD were obtained from Traditional Chinese Medicine Integrative Database and Traditional Chinese Medicine Systems Pharmacology databases and targets of SSMD were predicted by Pharmmapper and STITCH database. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were carried out to explore the function characteristics of SSMD. The expression matrix of microarray of pSS was obtained from Gene Expression Omnibus and we obtained 162 differentially expressed genes (DEGs). Protein-protein interaction (PPI) networks were constructed to identify the hub targets. Principal component analysis (PCA) and molecular docking were conducted to further elucidate the possibility of SSMD for pSS. Results SSMD contained a total of 1056 active components, corresponding to 88 targets, among which peripheral myelin protein 2(PMP2), androgen receptor (AR) and glutamic acid decarboxylase 1(GAD1) are associated with multiple active components in SSMD and may be the core targets. Moreover, these targets were closely related to tissue pathological injury in SS, such as lacrimal gland, salivary gland and nervous system injury. GO and KEGG analysis showed that 88 targets enriched in REDOX process, transcriptional regulation and negative regulation of apoptosis process. Besides, SSMD may influence the cell proliferation, gene transcription through regulating Ras and cAMP-related signaling pathways. In addition, SSMD may show effects on immune regulation, such as macrophage differentiation, Toll-like receptor 4 signaling pathway and T-helper 1 in SS. Moreover, PPI network suggested that FN1, MMP-9 may be the hub targets in SSMD. Result of PCA and molecular docking analysis further determined the feasibility of SSMD in treating pSS. Conclusion SSMD can regulate multiple biological processes by virtue of its multiple active components, thus showing prominent advantage in the treatment of pSS. The discovery of active ingredients and targets in SSMD provides valuable resources for drug research and development for pSS. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-021-03257-7.
Collapse
Affiliation(s)
- Yuepeng Jiang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310058, China
| | - Xiaoxuan Zhao
- Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Jie Yu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310058, China
| | - Qiao Wang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310058, China
| | - Chengping Wen
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310058, China.
| | - Lin Huang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310058, China.
| |
Collapse
|
14
|
Exploration of Active Site-Directed Plasmin Inhibitors: Beyond Tranexamic Acid. Processes (Basel) 2021. [DOI: 10.3390/pr9020329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Plasmin (Plm), a trypsin-like serine protease, is responsible for fibrinolysis pathway and pathologic events, such as angiogenesis, tumor invasion, and metastasis, and alters the expression of cytokines. A growing body of data indicates that a Plm inhibitor is a potential candidate as an anti-inflammatory and anti-cancer agent. A class of active site-directed plasmin inhibitors containing tranexamic acid residue has been designed. As evidenced by docking studies, the inhibitor binds to the active site not to the lysine binding site (LBS) in plasmin, thus preventing plasmin from digesting the substrate. Further optimization of the series, concerning both activity and selectivity, led to the second generation of inhibitors. This review focuses on the Plm inhibitory activity-structure relationship of Plm inhibitors with the goal of realizing their design and clinical application.
Collapse
|
15
|
Heissig B, Salama Y, Takahashi S, Osada T, Hattori K. The multifaceted role of plasminogen in inflammation. Cell Signal 2020; 75:109761. [PMID: 32861744 PMCID: PMC7452830 DOI: 10.1016/j.cellsig.2020.109761] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 01/01/2023]
Abstract
A fine-tuned activation and deactivation of proteases and their inhibitors are involved in the execution of the inflammatory response. The zymogen/proenzyme plasminogen is converted to the serine protease plasmin, a key fibrinolytic factor by plasminogen activators including tissue-type plasminogen activator (tPA). Plasmin is part of an intricate protease network controlling proteins of initial hemostasis/coagulation, fibrinolytic and complement system. Activation of these protease cascades is required to mount a proper inflammatory response. Although best known for its ability to dissolve clots and cleave fibrin, recent studies point to the importance of fibrin-independent functions of plasmin during acute inflammation and inflammation resolution. In this review, we provide an up-to-date overview of the current knowledge of the enzymatic and cytokine-like effects of tPA and describe the role of tPA and plasminogen receptors in the regulation of the inflammatory response with emphasis on the cytokine storm syndrome such as observed during coronavirus disease 2019 or macrophage activation syndrome. We discuss tPA as a modulator of Toll like receptor signaling, plasmin as an activator of NFkB signaling, and summarize recent studies on the role of plasminogen receptors as controllers of the macrophage conversion into the M2 type and as mediators of efferocytosis during inflammation resolution.
Collapse
Affiliation(s)
- Beate Heissig
- Department of Immunological Diagnosis, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan.
| | - Yousef Salama
- An-Najah Center for Cancer and Stem Cell Research, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine.
| | - Satoshi Takahashi
- Department of Hematology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| | - Taro Osada
- Department of Gastroenterology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi, 279-0021 Chiba, Japan.
| | - Koichi Hattori
- Center for Genomic & Regenerative Medicine, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan.
| |
Collapse
|
16
|
Xiao L, Jin H, Duan W, Hou Y. Roles of N-terminal Annexin A2 phosphorylation sites and miR-206 in colonic adenocarcinoma. Life Sci 2020; 253:117740. [PMID: 32376265 DOI: 10.1016/j.lfs.2020.117740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/15/2022]
Abstract
AIMS Annexin A2 (ANXA2) is closely associated with tumor malignancy and its N-terminus includes a vital domain for its function. The aims are to explore the correlation between the sites (Tyr23, Ser1, Ser11 and Ser25) in the domain and its roles. MAIN METHODS We re-expressed ANXA2 with mutated sites in ANXA2-deleted human colonic adenocarcinoma cell line caco2 (ANXA2-/-caco2). A series of analyses were used to determine the correlation of each site with ANXA2 activation, cell malignancy enhancement and motility-associated microstructural development. Bioinformatics and luciferase reporter assays were employed to validate ANXA2-targeted miRNAs. KEY FINDINGS The in vitro results showed that all single and multiple mutations of the ANXA2 N-terminal sites inhibited ANXA2 phosphorylation at different levels and subsequently inhibited the proliferation, motility, and polymerization of F-actin and β-tubulin in caco2 cells. Motility-associated microstructures were significantly remodeled when these sites were mutated. The forced expression of miR-206 significantly suppressed the proliferation, motility and epithelial-mesenchymal transition (EMT) of caco2 cells. The in vivo results showed that all the ANXA2 N-terminal site mutations and forced expression of miR-206 significantly inhibited tumor growth. Overall, this study demonstrated that the sites of the ANXA2 N-terminus, especially Tyr23, play crucial roles in maintaining the high malignancy of colonic adenocarcinoma. Furthermore, miR-206 targets ANXA2 and plays a role as a cancer suppressor in colonic adenocarcinoma. SIGNIFICANCE Our study provided evidence that further elucidates the molecular mechanism of ANXA2 and its roles in colonic adenocarcinoma and suggested potential targets of ANXA2 for colonic adenocarcinoma therapy by using miR-206 as a novel strategy.
Collapse
Affiliation(s)
- Li Xiao
- College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Xi'an, Shaanxi 710119, China
| | - Huijuan Jin
- College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Xi'an, Shaanxi 710119, China
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Yingchun Hou
- College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
17
|
Aota K, Ono S, Yamanoi T, Kani K, Momota Y, Azuma M. MMP-9 Inhibition Suppresses Interferon-γ-Induced CXCL10 Production in Human Salivary Gland Ductal Cells. Inflammation 2020; 42:2148-2158. [PMID: 31440939 DOI: 10.1007/s10753-019-01079-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gene expression profiling of lip salivary gland (LSG) has shown that C-X-C motif chemokine 10 (CXCL10) and matrix metalloproteinase 9 (MMP9) expression is upregulated in primary Sjögren's syndrome (pSS) patients. Although CXCL10 and MMP-9 are both associated with pSS pathogenesis, the potential relationship between these two factors has not been investigated. In this study, we used LSG sections from pSS patients and human salivary gland cell lines to investigate the relationship between CXCL10 and MMP-9. Immunofluorescence analyses revealed that CXCL10 and MMP-9 were co-expressed in the LSG of pSS patients, particularly in expanded ductal cells. Furthermore, RT-qPCR analyses on human salivary gland ductal NS-SV-DC cells confirmed that CXCL10 expression was induced by interferon (IFN)-γ, whereas that of MMP9 was stimulated by IFN-α, tumor necrosis factor-α, and interleukin-1β. Remarkably, MMP-9 inhibition in IFN-γ-stimulated NS-SV-DC cells significantly decreased CXCL10 mRNA and secreted protein levels. Further analyses established that MMP-9 inhibition in IFN-γ-stimulated NS-SV-DC cells decreased STAT1 phosphorylation and hence suppressed IFN-γ signaling. Collectively, these results suggest that in addition to its reported role in the destruction of acinar structures, MMP-9 is involved in the IFN-γ-induced production of CXCL10 in pSS lesions. We believe that our findings open the door to the development of novel treatments for pSS, based on the modulation of MMP-9 activity.
Collapse
Affiliation(s)
- Keiko Aota
- Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8504, Japan.
| | - Shinji Ono
- Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8504, Japan
| | - Tomoko Yamanoi
- Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8504, Japan
| | - Koichi Kani
- Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8504, Japan
| | - Yukihiro Momota
- Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8504, Japan
| | - Masayuki Azuma
- Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8504, Japan
| |
Collapse
|
18
|
Imai J, Yahata T, Ichikawa H, Ibrahim AA, Yazawa M, Sumiyoshi H, Inagaki Y, Matsushima M, Suzuki T, Mine T, Ando K, Miyata T, Hozumi K. Inhibition of plasminogen activator inhibitor-1 attenuates against intestinal fibrosis in mice. Intest Res 2020; 18:219-228. [PMID: 32050315 PMCID: PMC7206341 DOI: 10.5217/ir.2019.00037] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 10/07/2019] [Indexed: 12/16/2022] Open
Abstract
Background/Aims Intestinal fibrosis is a major complication of Crohn’s disease (CD). The profibrotic protein transforming growth factor-β (TGF-β) has been considered to be critical for the induction of the fibrotic program. TGF-β has the ability to induce not only the expression of extracellular matrix (ECM) including collagen, but also the production of plasminogen activator inhibitor-1 (PAI-1) that prevents enzymatic degradation of the ECM during the onset of fibrotic diseases. However, the significance of PAI-1 in the developing intestinal fibrosis has not been fully understood. In the present study, we examined the actual expression of PAI-1 in fibrotic legion of intestinal inflammation and its correlation with the abnormal ECM deposition. Methods Chronic intestinal inflammation was induced in BALB/c mice using 8 repeated intrarectal injections of 2,4,6-trinitrobenzene sulfonic acid (TNBS). TM5275, a PAI-1 inhibitor, was orally administered as a carboxymethyl cellulose suspension each day for 2 weeks after the sixth TNBS injection. Results Using a publicly available dataset (accession number, GSE75214) and TNBS-treated mice, we observed increases in PAI-1 transcripts at active fibrotic lesions in both patients with CD and mice with chronic intestinal inflammation. Oral administration of TM5275 immediately after the onset of intestinal fibrosis upregulated MMP-9 (matrix metalloproteinase 9) and decreased collagen accumulation, resulting in attenuation of the fibrogenesis in TNBS-treated mice. Conclusions PAI-1-mediated fibrinolytic system facilitates collagen degradation suppression. Hence, PAI-1 inhibitor could be applied as an anti-fibrotic drug in CD treatment.
Collapse
Affiliation(s)
- Jin Imai
- Center for Matrix Biology and Medicine, Tokai University School of Medicine, Kanagawa, Japan.,Department of Gastroenterology, Tokai University School of Medicine, Kanagawa, Japan
| | - Takashi Yahata
- Research Center for Regenerative Medicine, Tokai University School of Medicine, Kanagawa, Japan.,Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Hitoshi Ichikawa
- Department of Gastroenterology, Tokai University School of Medicine, Kanagawa, Japan
| | - Abd Aziz Ibrahim
- Research Center for Regenerative Medicine, Tokai University School of Medicine, Kanagawa, Japan.,Department of Hematology and Oncology, Tokai University School of Medicine, Kanagawa, Japan
| | - Masaki Yazawa
- Department of Immunology, Tokai University School of Medicine, Kanagawa, Japan
| | - Hideaki Sumiyoshi
- Center for Matrix Biology and Medicine, Tokai University School of Medicine, Kanagawa, Japan.,Department of Regenerative Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Tokai University School of Medicine, Kanagawa, Japan.,Department of Regenerative Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Masashi Matsushima
- Department of Gastroenterology, Tokai University School of Medicine, Kanagawa, Japan
| | - Takayoshi Suzuki
- Department of Gastroenterology, Tokai University School of Medicine, Kanagawa, Japan
| | - Tetsuya Mine
- Department of Gastroenterology, Tokai University School of Medicine, Kanagawa, Japan
| | - Kiyoshi Ando
- Research Center for Regenerative Medicine, Tokai University School of Medicine, Kanagawa, Japan.,Department of Hematology and Oncology, Tokai University School of Medicine, Kanagawa, Japan
| | - Toshio Miyata
- Division of Molecular Medicine and Therapy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Katsuto Hozumi
- Center for Matrix Biology and Medicine, Tokai University School of Medicine, Kanagawa, Japan.,Research Center for Regenerative Medicine, Tokai University School of Medicine, Kanagawa, Japan.,Department of Immunology, Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
19
|
Li T, Wang C, Liu Y, Li B, Zhang W, Wang L, Yu M, Zhao X, Du J, Zhang J, Dong Z, Jiang T, Xie R, Ma R, Fang S, Zhou J, Shi J. Neutrophil Extracellular Traps Induce Intestinal Damage and Thrombotic Tendency in Inflammatory Bowel Disease. J Crohns Colitis 2020; 14:240-253. [PMID: 31325355 DOI: 10.1093/ecco-jcc/jjz132] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Despite the presence of neutrophil extracellular traps [NETs] in inflamed colon having been confirmed, the role of NETs, especially the circulating NETs, in the progression and thrombotic tendency of inflammatory bowel disease [IBD] remains elusive. We extended our previous study to prove that NETs constitute a central component in the progression and prothrombotic state of IBD. METHODS In all 48 consecutive patients with IBD were studied. Acute colitis was induced by the treatment of C57BL/6 mice with 3.5% dextran sulphate sodium [DSS] in drinking water for 6 days. Peripheral blood neutrophils and sera were collected from IBD patients and murine colitis models. Exposed phosphatidylserine [PS] was analysed with flow cytometry and confocal microscopy. Procoagulant activity was evaluated using clotting time, purified coagulation complex, and fibrin formation assays. RESULTS We observed higher plasma NET levels and presence of NETs in colon tissue in patients with active IBD. More importantly, NETs were induced in mice with DSS colitis, and inhibition of NET release attenuated colitis as well as colitis-associated tumorigenesis. NET degradation through DNase administration decreased cytokine levels during DSS-induced colitis. In addition, DNase treatment also significantly attenuated the accelerated thrombus formation and platelet activation observed in DSS-induced colitis. NETs triggered PS-positive microparticle release and PS exposure on platelets and endothelial cells partially through TLR2 and TLR4, converting them to a procoagulant phenotype. CONCLUSIONS NETs exacerbate colon tissue damage and drive thrombotic tendency during active IBD. Strategies directed against NET formation may offer a potential therapeutic approach for the treatment of IBD.
Collapse
Affiliation(s)
- Tao Li
- Department of Hematology, the First Hospital, Harbin Medical University, Nangang District, Harbin, PR, China.,Key Laboratory of Myocardial Ischemia, Ministry of Education, Heilongjiang, China
| | - Chunxu Wang
- Department of Hematology, the First Hospital, Harbin Medical University, Nangang District, Harbin, PR, China
| | - Yingmiao Liu
- Department of Stomatology of the First Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Baorong Li
- Department of Stomatology of the First Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Wujian Zhang
- Department of General Surgery of the First Hospital, Harbin Medical University, Heilongjiang, China
| | - Lixiu Wang
- Department of Cardiology of the First Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Muxin Yu
- Department of Hematology, the First Hospital, Harbin Medical University, Nangang District, Harbin, PR, China
| | - Xinyi Zhao
- Department of Cardiology of the Second Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Jingwen Du
- Department of Hematology, the First Hospital, Harbin Medical University, Nangang District, Harbin, PR, China
| | - Jinming Zhang
- Department of Gastroenterology of the Fourth Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Zengxiang Dong
- Department of Cardiology of the First Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Tao Jiang
- Department of General Surgery of the First Hospital, Harbin Medical University, Heilongjiang, China
| | - Rui Xie
- Department of Oncology of The Third Hospital, Harbin Medical University, Heilongjiang, China
| | - Ruishuang Ma
- Department of Oncology of The Third Hospital, Harbin Medical University, Heilongjiang, China
| | - Shaohong Fang
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Heilongjiang, China
| | - Jin Zhou
- Department of Hematology, the First Hospital, Harbin Medical University, Nangang District, Harbin, PR, China
| | - Jialan Shi
- Department of Hematology, the First Hospital, Harbin Medical University, Nangang District, Harbin, PR, China.,Medicine Departments of Surgery, Brigham and Women's Hospital, VA Boston Healthcare System and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Schroder WA, Hirata TD, Le TT, Gardner J, Boyle GM, Ellis J, Nakayama E, Pathirana D, Nakaya HI, Suhrbier A. SerpinB2 inhibits migration and promotes a resolution phase signature in large peritoneal macrophages. Sci Rep 2019; 9:12421. [PMID: 31455834 PMCID: PMC6712035 DOI: 10.1038/s41598-019-48741-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 08/12/2019] [Indexed: 12/17/2022] Open
Abstract
SerpinB2 (plasminogen activator inhibitor type 2) has been called the "undecided serpin" with no clear consensus on its physiological role, although it is well described as an inhibitor of urokinase plasminogen activator (uPA). In macrophages, pro-inflammatory stimuli usually induce SerpinB2; however, expression is constitutive in Gata6+ large peritoneal macrophages (LPM). Interrogation of expression data from human macrophages treated with a range of stimuli using a new bioinformatics tool, CEMiTool, suggested that SerpinB2 is most tightly co- and counter-regulated with genes associated with cell movement. Using LPM from SerpinB2-/- and SerpinB2R380A (active site mutant) mice, we show that migration on Matrigel was faster than for their wild-type controls. Confocal microscopy illustrated that SerpinB2 and F-actin staining overlapped in focal adhesions and lamellipodia. Genes associated with migration and extracellular matrix interactions were also identified by RNA-Seq analysis of migrating RPM from wild-type and SerpinB2R380A mice. Subsequent gene set enrichment analyses (GSEA) suggested SerpinB2 counter-regulates many Gata6-regulated genes associated with migration. These data argue that the role of SerpinB2 in macrophages is inhibition of uPA-mediated plasmin generation during cell migration. GSEA also suggested that SerpinB2 expression (likely via ensuing modulation of uPA-receptor/integrin signaling) promotes the adoption of a resolution phase signature.
Collapse
Affiliation(s)
- Wayne A Schroder
- QIMR Berghofer Medical Research Institute, Brisbane, Qld, 4029, Australia
| | - Thiago D Hirata
- School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Thuy T Le
- QIMR Berghofer Medical Research Institute, Brisbane, Qld, 4029, Australia
| | - Joy Gardner
- QIMR Berghofer Medical Research Institute, Brisbane, Qld, 4029, Australia
| | - Glen M Boyle
- QIMR Berghofer Medical Research Institute, Brisbane, Qld, 4029, Australia
| | - Jonathan Ellis
- QIMR Berghofer Medical Research Institute, Brisbane, Qld, 4029, Australia
| | - Eri Nakayama
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Dilan Pathirana
- QIMR Berghofer Medical Research Institute, Brisbane, Qld, 4029, Australia
| | - Helder I Nakaya
- School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, Brisbane, Qld, 4029, Australia.
| |
Collapse
|
21
|
Hiramoto K, Yamate Y, Sugiyama D, Matsuda K, Iizuka Y, Yamaguchi T. Ameliorative effect of tranexamic acid on physiological skin aging and its sex difference in mice. Arch Dermatol Res 2019; 311:545-553. [PMID: 31147768 DOI: 10.1007/s00403-019-01938-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/17/2019] [Accepted: 05/24/2019] [Indexed: 02/01/2023]
Abstract
An effective method to protect the skin from natural aging is unknown. Therefore, in this study, we examined the ameliorative effects of tranexamic acid on natural skin aging. In addition, we examined the sex difference in the effect exhibited by tranexamic acid. We bred hairless mice without ultraviolet ray irradiation and physical stress for 2 years. During the study period, mice were orally administered tranexamic acid (12 mg/kg/day) three times per week. Development of signs of skin aging was found to be ameliorated by tranexamic acid. Furthermore, synthetic inhibition of plasmin was observed following tranexamic acid treatment. The synthetic reinforcement of hyaluronic acid by an increase in the number of epidermal cells and the degradative inhibition of extracellular matrix (ECM) by matrix metalloproteinase (MMP) suppression were observed. These results indicate that natural skin aging was ameliorated by tranexamic acid via the regulation of the plasmin/TGF-β/epidermal cells/hyaluronic acid and plasmin/MMPs/ECM signal transmission pathways. Taken together, sex difference was observed for the ameliorative effect of tranexamic acid on skin aging, with a stronger effect observed in females than in males. More importantly, we found that the synthesis of hyaluronic acid was stronger in female mice than in male mice.
Collapse
Affiliation(s)
- Keiichi Hiramoto
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagakicho, Suzuka, Mie, 513-8670, Japan.
| | - Yurika Yamate
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagakicho, Suzuka, Mie, 513-8670, Japan
| | - Daijiro Sugiyama
- R&D Department, Daiichi Sankyo Healthcare Co., LTD., 3-14-10 Nihonbashi, Chuo-ku, Tokyo, 103-8234, Japan
| | - Kazunari Matsuda
- R&D Department, Daiichi Sankyo Healthcare Co., LTD., 3-14-10 Nihonbashi, Chuo-ku, Tokyo, 103-8234, Japan
| | - Yasutaka Iizuka
- R&D Department, Daiichi Sankyo Healthcare Co., LTD., 3-14-10 Nihonbashi, Chuo-ku, Tokyo, 103-8234, Japan
| | - Tomohiko Yamaguchi
- R&D Department, Daiichi Sankyo Healthcare Co., LTD., 3-14-10 Nihonbashi, Chuo-ku, Tokyo, 103-8234, Japan
| |
Collapse
|
22
|
Structural studies of plasmin inhibition. Biochem Soc Trans 2019; 47:541-557. [DOI: 10.1042/bst20180211] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/28/2019] [Accepted: 01/31/2019] [Indexed: 12/24/2022]
Abstract
Abstract
Plasminogen (Plg) is the zymogen form of the serine protease plasmin (Plm), and it plays a crucial role in fibrinolysis as well as wound healing, immunity, tissue remodeling and inflammation. Binding to the targets via the lysine-binding sites allows for Plg activation by plasminogen activators (PAs) present on the same target. Cellular uptake of fibrin degradation products leads to apoptosis, which represents one of the pathways for cross-talk between fibrinolysis and tissue remodeling. Therapeutic manipulation of Plm activity plays a vital role in the treatments of a range of diseases, whereas Plm inhibitors are used in trauma and surgeries as antifibrinolytic agents. Plm inhibitors are also used in conditions such as angioedema, menorrhagia and melasma. Here, we review the rationale for the further development of new Plm inhibitors, with a particular focus on the structural studies of the active site inhibitors of Plm. We compare the binding mode of different classes of inhibitors and comment on how it relates to their efficacy, as well as possible future developments.
Collapse
|
23
|
How Adhesion Molecule Patterns Change While Neutrophils Traffic through the Lung during Inflammation. Mediators Inflamm 2019; 2019:1208086. [PMID: 30944544 PMCID: PMC6421765 DOI: 10.1155/2019/1208086] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/13/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023] Open
Abstract
In acute pulmonary inflammation, polymorphonuclear cells (PMNs) pass a transendothelial barrier from the circulation into the lung interstitium followed by a transepithelial migration into the alveolar space. These migration steps are regulated differentially by a concept of adhesion molecules and remain-despite decades of research-incompletely understood. Current knowledge of changes in the expression pattern of adhesion molecules mainly derives from in vitro studies or from studies in extrapulmonary organ systems, where regulation of adhesion molecules differs significantly. In a murine model of lung inflammation, we determined the expression pattern of nine relevant neutrophilic adhesion molecules on their way through the different compartments of the lung. We used a flow cytometry-based technique that allowed describing spatial distribution of the adhesion molecules expressed on PMNs during their migration through the lung in detail. For example, the highest expression of CD29 was found in the intravascular compartment, highlighting its impact on the initial adhesion to the endothelium. CD47 showed its peak of expression on the later phase of transendothelial migration, whereas CD11b and CD54 expression peaked interstitial. A pivotal role for transepithelial migration was found for the adhesion molecule CD172a. Thereby, expression may correlate with functional impact for specific migration steps. In vitro studies further confirmed our in vivo findings. In conclusion, we are the first to determine the changes in expression patterns of relevant adhesion molecules on their migration through the different compartments of the lung. These findings may help to further understand the regulation of neutrophil trafficking in the lung.
Collapse
|
24
|
Hiramoto K, Yamate Y, Sugiyama D, Matsuda K, Iizuka Y, Yamaguchi T. Tranexamic Acid Ameliorates Nonmelanoma Skin Cancer Induced by Long‐term Ultraviolet A Irradiation. Photochem Photobiol 2018; 95:612-617. [DOI: 10.1111/php.13025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/06/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Keiichi Hiramoto
- Department of Pharmaceutical Sciences Suzuka University of Medical Science Suzuka Mie Japan
| | - Yurika Yamate
- Department of Pharmaceutical Sciences Suzuka University of Medical Science Suzuka Mie Japan
| | - Daijiro Sugiyama
- Department of Pharmaceutical Sciences Suzuka University of Medical Science Suzuka Mie Japan
| | - Kazunari Matsuda
- R&D Department Daiichi Sankyo Healthcare Co., LTD. Chuo‐ku Tokyo Japan
| | - Yasutaka Iizuka
- R&D Department Daiichi Sankyo Healthcare Co., LTD. Chuo‐ku Tokyo Japan
| | | |
Collapse
|
25
|
Eissa N, Hussein H, Hendy GN, Bernstein CN, Ghia JE. Chromogranin-A and its derived peptides and their pharmacological effects during intestinal inflammation. Biochem Pharmacol 2018; 152:315-326. [PMID: 29656116 DOI: 10.1016/j.bcp.2018.04.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 04/10/2018] [Indexed: 02/07/2023]
Abstract
The gastrointestinal tract is the largest endocrine organ that produces a broad range of active peptides. Mucosal changes during inflammation alter the distribution and products of enteroendocrine cells (EECs) that play a role in immune activation and regulation of gut homeostasis by mediating communication between the nervous, endocrine and immune systems. Patients with inflammatory bowel disease (IBD) typically have altered expression of chromogranin (CHG)-A (CHGA), a major soluble protein secreted by EECs that functions as a pro-hormone. CHGA gives rise to several bioactive peptides that have direct or indirect effects on intestinal inflammation. In IBD, CHGA and its derived peptides are correlated with the disease activity. In this review we describe the potential immunomodulatory roles of CHGA and its derived peptides and their clinical relevance during the progression of intestinal inflammation. Targeting CHGA and its derived peptides could be of benefit for the diagnosis and clinical management of IBD patients.
Collapse
Affiliation(s)
- Nour Eissa
- Department of Immunology, College of Medicine, University of Manitoba, Winnipeg, MB, Canada; Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada; IBD Clinical and Research Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Hayam Hussein
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Ohio State University, Columbus, OH, USA
| | - Geoffrey N Hendy
- Metabolic Disorders and Complications, McGill University Health Centre-Research Institute, Departments of Medicine, Physiology, and Human Genetics, McGill University, Montréal, QC, Canada
| | - Charles N Bernstein
- IBD Clinical and Research Centre, University of Manitoba, Winnipeg, MB, Canada; Section of Gastroenterology, Department of Internal Medicine, College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Jean-Eric Ghia
- Department of Immunology, College of Medicine, University of Manitoba, Winnipeg, MB, Canada; Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada; IBD Clinical and Research Centre, University of Manitoba, Winnipeg, MB, Canada; Section of Gastroenterology, Department of Internal Medicine, College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
26
|
Liu J, Luan W, Zhang Y, Gu J, Shi Y, Yang Y, Feng Z, Qi F. HDAC6 interacts with PTPN1 to enhance melanoma cells progression. Biochem Biophys Res Commun 2018; 495:2630-2636. [DOI: 10.1016/j.bbrc.2017.12.145] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 12/23/2017] [Indexed: 01/05/2023]
|
27
|
Matrix metalloproteinases as regulators of inflammatory processes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2036-2042. [DOI: 10.1016/j.bbamcr.2017.05.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/06/2017] [Accepted: 05/09/2017] [Indexed: 12/26/2022]
|
28
|
Ge F, Zhu S, Liu L, Yan J, Ji Y, Sun Z. Anti-inflammatory effects of Fagopyrum cymosum administered as a potential drug for ulcerative colitis. Exp Ther Med 2017; 14:4745-4754. [PMID: 29201175 PMCID: PMC5704331 DOI: 10.3892/etm.2017.5153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 06/29/2017] [Indexed: 12/21/2022] Open
Abstract
Fagopyrum cymosum (Trev.) Meisn (Fag), which belongs to the Polygonaceae family, has been widely used to treat inflammatory diseases. Previous studies have revealed that Fag components exhibit anti-inflammatory activities; however, their potential use in treating inflammatory bowel disease (IBD) has not been explored. In the present study, mice with 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis were used as a model of IBD. Fag extract was orally administered for 3 days following the induction of colitis and the conventional drug, salicylazosulfapyridine (SASP), was used as a control. The results revealed that Fag significantly ameliorated TNBS-induced body weight loss and colonic shortening in mice (P<0.05). Furthermore, Fag suppressed levels of proinflammatory cytokines and reduced macrophage infiltration into colonic tissues (P<0.05). To further verify the anti-inflammatory effects of Fag at the molecular level, a murine macrophage cell line, Raw264.7, was used. Nuclear translocation of nuclear factor (NF)-κB p65 and the phosphorylation of inhibitor of NF-κB (IκB) were assessed using western blotting. The results demonstrated that Fag inhibited the production of proinflammatory cytokines via inhibiting NF-κB p65 nuclear translocation and IκB phosphorylation (P<0.05). Furthermore, the clinical study results revealed that Fag had significantly fewer side effects (P<0.05) and served as a better anti-inflammatory drug for ulcerative colitis compared with SASP.
Collapse
Affiliation(s)
- Fei Ge
- Department of Gastroenterology, Haian Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Haian, Jiangsu 226600, P.R. China.,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Shilin Zhu
- Department of Gastroenterology, Haian Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Haian, Jiangsu 226600, P.R. China
| | - Lina Liu
- Department of Liver Disease, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Jing Yan
- First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Yu Ji
- Department of Gastroenterology, Haian Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Haian, Jiangsu 226600, P.R. China
| | - Zhiguang Sun
- First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
29
|
Eiamboonsert S, Salama Y, Watarai H, Dhahri D, Tsuda Y, Okada Y, Hattori K, Heissig B. The role of plasmin in the pathogenesis of murine multiple myeloma. Biochem Biophys Res Commun 2017; 488:387-392. [PMID: 28501622 DOI: 10.1016/j.bbrc.2017.05.062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/10/2017] [Indexed: 12/28/2022]
Abstract
Aside from a role in clot dissolution, the fibrinolytic factor, plasmin is implicated in tumorigenesis. Although abnormalities of coagulation and fibrinolysis have been reported in multiple myeloma patients, the biological roles of fibrinolytic factors in multiple myeloma (MM) using in vivo models have not been elucidated. In this study, we established a murine model of fulminant MM with bone marrow and extramedullar engraftment after intravenous injection of B53 cells. We found that the fibrinolytic factor expression pattern in murine B53 MM cells is similar to the expression pattern reported in primary human MM cells. Pharmacological targeting of plasmin using the plasmin inhibitors YO-2 did not change disease progression in MM cell bearing mice although systemic plasmin levels was suppressed. Our findings suggest that although plasmin has been suggested to be a driver for disease progression using clinical patient samples in MM using mostly in vitro studies, here we demonstrate that suppression of plasmin generation or inhibition of plasmin cannot alter MM progression in vivo.
Collapse
Affiliation(s)
- Salita Eiamboonsert
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yousef Salama
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Hiroshi Watarai
- Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Douaa Dhahri
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yuko Tsuda
- Faculty of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 850-8586, Japan
| | - Yoshio Okada
- Faculty of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 850-8586, Japan
| | - Koichi Hattori
- Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Center for Genome and Regenerative Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Beate Heissig
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Atopy Center, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| |
Collapse
|
30
|
Pharmacological targeting of plasmin prevents lethality in a murine model of macrophage activation syndrome. Blood 2017; 130:59-72. [PMID: 28325863 DOI: 10.1182/blood-2016-09-738096] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 03/10/2017] [Indexed: 12/12/2022] Open
Abstract
Macrophage activation syndrome (MAS) is a life-threatening disorder characterized by a cytokine storm and multiorgan dysfunction due to excessive immune activation. Although abnormalities of coagulation and fibrinolysis are major components of MAS, the role of the fibrinolytic system and its key player, plasmin, in the development of MAS remains to be solved. We established a murine model of fulminant MAS by repeated injections of Toll-like receptor-9 (TLR-9) agonist and d-galactosamine (DG) in immunocompetent mice. We found plasmin was excessively activated during the progression of fulminant MAS in mice. Genetic and pharmacological inhibition of plasmin counteracted MAS-associated lethality and other related symptoms. We show that plasmin regulates the influx of inflammatory cells and the production of inflammatory cytokines/chemokines. Collectively, our findings identify plasmin as a decisive checkpoint in the inflammatory response during MAS and a potential novel therapeutic target for MAS.
Collapse
|
31
|
Honjo K, Munakata S, Tashiro Y, Salama Y, Shimazu H, Eiamboonsert S, Dhahri D, Ichimura A, Dan T, Miyata T, Takeda K, Sakamoto K, Hattori K, Heissig B. Plasminogen activator inhibitor‐1 regulates macrophage‐dependent postoperative adhesion by enhancing EGF‐HER1 signaling in mice. FASEB J 2017; 31:2625-2637. [DOI: 10.1096/fj.201600871rr] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 02/21/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Kumpei Honjo
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Coloproctological Surgery Tokyo Japan
| | - Shinya Munakata
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Coloproctological Surgery Tokyo Japan
| | - Yoshihiko Tashiro
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Coloproctological Surgery Tokyo Japan
| | - Yousef Salama
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Hiroshi Shimazu
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Salita Eiamboonsert
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Douaa Dhahri
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Atsuhiko Ichimura
- United Centers for Advanced Research and Translational MedicineGraduate School of Medicine, Tohoku University Sendai Japan
| | - Takashi Dan
- United Centers for Advanced Research and Translational MedicineGraduate School of Medicine, Tohoku University Sendai Japan
| | - Toshio Miyata
- United Centers for Advanced Research and Translational MedicineGraduate School of Medicine, Tohoku University Sendai Japan
| | - Kazuyoshi Takeda
- Department of Immunology and Atopy CenterGraduate School of Medicine, Juntendo University Tokyo Japan
| | | | - Koichi Hattori
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Center for Genomic and Regenerative MedicineFaculty of Medicine Tokyo Japan
| | - Beate Heissig
- Division of Stem Cell Dynamics Center for Stem Cell Biology and Regenerative MedicineThe Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Immunology and Atopy CenterGraduate School of Medicine, Juntendo University Tokyo Japan
| |
Collapse
|
32
|
Afosah DK, Al-Horani RA, Sankaranarayanan NV, Desai UR. Potent, Selective, Allosteric Inhibition of Human Plasmin by Sulfated Non-Saccharide Glycosaminoglycan Mimetics. J Med Chem 2017; 60:641-657. [DOI: 10.1021/acs.jmedchem.6b01474] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Daniel K. Afosah
- Department of Medicinal Chemistry,
and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| | - Rami A. Al-Horani
- Department of Medicinal Chemistry,
and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| | - Nehru Viji Sankaranarayanan
- Department of Medicinal Chemistry,
and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| | - Umesh R. Desai
- Department of Medicinal Chemistry,
and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| |
Collapse
|
33
|
Hinkes S, Wuttke A, Saupe SM, Ivanova T, Wagner S, Knörlein A, Heine A, Klebe G, Steinmetzer T. Optimization of Cyclic Plasmin Inhibitors: From Benzamidines to Benzylamines. J Med Chem 2016; 59:6370-86. [DOI: 10.1021/acs.jmedchem.6b00606] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Stefan Hinkes
- Department of Pharmacy, Institute
of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| | - André Wuttke
- Department of Pharmacy, Institute
of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| | - Sebastian M. Saupe
- Department of Pharmacy, Institute
of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| | - Teodora Ivanova
- Department of Pharmacy, Institute
of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| | - Sebastian Wagner
- Department of Pharmacy, Institute
of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| | - Anna Knörlein
- Department of Pharmacy, Institute
of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| | - Andreas Heine
- Department of Pharmacy, Institute
of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| | - Gerhard Klebe
- Department of Pharmacy, Institute
of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| | - Torsten Steinmetzer
- Department of Pharmacy, Institute
of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| |
Collapse
|
34
|
Heissig B, Eiamboonsert S, Salama Y, Shimazu H, Dhahri D, Munakata S, Tashiro Y, Hattori K. Cancer therapy targeting the fibrinolytic system. Adv Drug Deliv Rev 2016; 99:172-179. [PMID: 26588878 DOI: 10.1016/j.addr.2015.11.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 10/27/2015] [Accepted: 11/11/2015] [Indexed: 12/17/2022]
Abstract
The tumor microenvironment is recognized as a key factor in the multiple stages of cancer progression, mediating local resistance, immune-escape and metastasis. Cancer growth and progression require remodeling of the tumor stromal microenvironment, such as the development of tumor-associated blood vessels, recruitment of bone marrow-derived cells and cytokine processing. Extracellular matrix breakdown achieved by proteases like the fibrinolytic factor plasmin and matrix metalloproteases is necessary for cell migration crucial for cancer invasion and metastasis. Key components of the fibrinolytic system are expressed in cells of the tumor microenvironment. Plasmin can control growth factor bioavailability, or the regulation of other proteases leading to angiogenesis, and inflammation. In this review, we will focus on the role of the fibrinolytic system in the tumor microenvironment summarizing our current understanding of the role of the fibrinolytic factors for the modulation of the local chemokine/cytokine milieu, resulting in myeloid cell recruitment, which can promote neoangiogenesis.
Collapse
|
35
|
Heissig B, Dhahri D, Eiamboonsert S, Salama Y, Shimazu H, Munakata S, Hattori K. Role of mesenchymal stem cell-derived fibrinolytic factor in tissue regeneration and cancer progression. Cell Mol Life Sci 2015; 72:4759-70. [PMID: 26350342 PMCID: PMC11113371 DOI: 10.1007/s00018-015-2035-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 08/03/2015] [Accepted: 08/31/2015] [Indexed: 12/21/2022]
Abstract
Tissue regeneration during wound healing or cancer growth and progression depends on the establishment of a cellular microenvironment. Mesenchymal stem cells (MSC) are part of this cellular microenvironment, where they functionally modulate cell homing, angiogenesis, and immune modulation. MSC recruitment involves detachment of these cells from their niche, and finally MSC migration into their preferred niches; the wounded area, the tumor bed, and the BM, just to name a few. During this recruitment phase, focal proteolysis disrupts the extracellular matrix (ECM) architecture, breaks cell-matrix interactions with receptors, and integrins, and causes the release of bioactive fragments from ECM molecules. MSC produce a broad array of proteases, promoting remodeling of the surrounding ECM through proteolytic mechanisms. The fibrinolytic system, with its main player plasmin, plays a crucial role in cell migration, growth factor bioavailability, and the regulation of other protease systems during inflammation, tissue regeneration, and cancer. Key components of the fibrinolytic cascade, including the urokinase plasminogen activator receptor (uPAR) and plasminogen activator inhibitor-1 (PAI-1), are expressed in MSC. This review will introduce general functional properties of the fibrinolytic system, which go beyond its known function of fibrin clot dissolution (fibrinolysis). We will focus on the role of the fibrinolytic system for MSC biology, summarizing our current understanding of the role of the fibrinolytic system for MSC recruitment and the functional consequences for tissue regeneration and cancer. Aspects of MSC origin, maintenance, and the mechanisms by which these cells contribute to altered protease activity in the microenvironment under normal and pathological conditions will also be discussed.
Collapse
Affiliation(s)
- Beate Heissig
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
- Atopy (Allergy) Center, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Douaa Dhahri
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Salita Eiamboonsert
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Yousef Salama
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Hiroshi Shimazu
- Division of Stem Cell Regulation, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Shinya Munakata
- Division of Stem Cell Regulation, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Koichi Hattori
- Division of Stem Cell Regulation, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Center for Genome and Regenerative Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|