1
|
Yin Z, Zhang J, Zhao M, Liu J, Xu Y, Peng S, Pan W, Wei C, Zheng Z, Liu S, Qin JJ, Wan J, Wang M. EDIL3/Del-1 prevents aortic dissection through enhancing internalization and degradation of apoptotic vascular smooth muscle cells. Autophagy 2024; 20:2405-2425. [PMID: 38873925 DOI: 10.1080/15548627.2024.2367191] [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: 12/04/2023] [Revised: 05/28/2024] [Accepted: 06/08/2024] [Indexed: 06/15/2024] Open
Abstract
Thoracic aortic dissection (TAD) is a severe disease, characterized by numerous apoptotic vascular smooth muscle cells (VSMCs). EDIL3/Del-1 is a secreted protein involved in macrophage efferocytosis in acute inflammation. Here, we aimed to investigate whether EDIL3 promoted the internalization and degradation of apoptotic VSMCs during TAD. The levels of EDIL3 were decreased in the serum and aortic tissue from TAD mice. Global edil3 knockout (edil3-/-) mice and edil3-/- bone marrow chimeric mice exhibited a considerable exacerbation in β-aminopropionitrile monofumarate (BAPN)-induced TAD, accompanied with increased apoptotic VSMCs accumulating in the damaged aortic tissue. Two types of phagocytes, RAW264.7 cells and bone marrow-derived macrophages (BMDMs) were used for in vitro efferocytosis assay. edil3-deficient phagocytes exhibited inefficient internalization and degradation of apoptotic VSMCs. Instead, EDIL3 promoted the internalization phase through interacting with phosphatidylserine (PtdSer) on apoptotic VSMCs and binding to the macrophage ITGAV/αv-ITGB3/β3 integrin. In addition, EDIL3 accelerated the degradation phase through activating LC3-associated phagocytosis (LAP). Mechanically, following the engulfment, EDIL3 enhanced the activity of SMPD1/acid sphingomyelinase in the phagosome through blocking ITGAV-ITGB3 integrin, which facilitates phagosomal reactive oxygen species (ROS) production by NAPDH oxidase CYBB/NOX2. Furthermore, exogenous EDIL3 supplementation alleviated BAPN-induced TAD and promoted apoptotic cell clearance. EDIL3 may be a novel factor for the prevention and treatment of TAD.Abbreviations: BAPN: β-aminopropionitrile monofumarate; BMDM: bone marrow-derived macrophage; C12FDG: 5-dodecanoylaminofluorescein-di-β-D-galactopyranoside; CTRL: control; CYBB/NOX2: cytochrome b-245, beta polypeptide; DCFH-DA: 2',7'-dichlorofluorescin diacetate; EDIL3/Del-1: EGF-like repeats and discoidin I-like domains 3; EdU: 5-ethynyl-2'-deoxyuridine; EVG: elastic van Gieson; H&E: hematoxylin and eosin; IL: interleukin; LAP: LC3-associated phagocytosis; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; NAC: N-acetylcysteine; PtdSer: phosphatidylserine; rEDIL3: recombinant EDIL3; ROS: reactive oxygen species; SMPD1: sphingomyelin phosphodiesterase 1; TAD: thoracic aortic dissection; TEM: transmission electron microscopy; VSMC: vascular smooth muscle cell; WT: wild-type.
Collapse
Affiliation(s)
- Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Siqi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
2
|
Jiang D, Yue H, Liang WT, Wu Z. Developmental endothelial locus 1: the present and future of an endogenous factor in vessels. Front Physiol 2024; 15:1347888. [PMID: 39206385 PMCID: PMC11350114 DOI: 10.3389/fphys.2024.1347888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Developmental Endothelial Locus-1 (DEL-1), also known as EGF-like repeat and discoidin I-like domain-3 (EDIL3), is increasingly recognized for its multifaceted roles in immunoregulation and vascular biology. DEL-1 is a protein that is mainly produced by endothelial cells. It interacts with various integrins to regulate the behavior of immune cells, such as preventing unnecessary recruitment and inflammation. DEL-1 also helps in resolving inflammation by promoting efferocytosis, which is the process of clearing apoptotic cells. Its potential as a therapeutic target in immune-mediated blood disorders, cardiovascular diseases, and cancer metastasis has been spotlighted due to its wide-ranging implications in vascular integrity and pathology. However, there are still unanswered questions about DEL-1's precise functions and mechanisms. This review provides a comprehensive examination of DEL-1's activity across different vascular contexts and explores its potential clinical applications. It underscores the need for further research to resolve existing controversies and establish the therapeutic viability of DEL-1 modulation.
Collapse
Affiliation(s)
| | | | - Wei-Tao Liang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhong Wu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Lim JH, Neuwirth A, Chung KJ, Grossklaus S, Soehnlein O, Hajishengallis G, Chavakis T. Formyl peptide receptor 2 regulates dendritic cell metabolism and Th17 cell differentiation during neuroinflammation. Front Immunol 2024; 15:1354074. [PMID: 39148732 PMCID: PMC11324504 DOI: 10.3389/fimmu.2024.1354074] [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: 12/11/2023] [Accepted: 07/18/2024] [Indexed: 08/17/2024] Open
Abstract
Formyl peptide receptor 2 (FPR2) is a receptor for formylated peptides and specific pro-resolving mediators, and is involved in various inflammatory processes. Here, we aimed to elucidate the role of FPR2 in dendritic cell (DC) function and autoimmunity-related central nervous system (CNS) inflammation by using the experimental autoimmune encephalomyelitis (EAE) model. EAE induction was accompanied by increased Fpr2 mRNA expression in the spinal cord. FPR2-deficient (Fpr2 KO) mice displayed delayed onset of EAE compared to wild-type (WT) mice, associated with reduced frequencies of Th17 cells in the inflamed spinal cord at the early stage of the disease. However, FPR2 deficiency did not affect EAE severity after the disease reached its peak. FPR2 deficiency in mature DCs resulted in decreased expression of Th17 polarizing cytokines IL6, IL23p19, IL1β, and thereby diminished the DC-mediated activation of Th17 cell differentiation. LPS-activated FPR2-deficient DCs showed upregulated Nos2 expression and nitric oxide (NO) production, as well as reduced oxygen consumption rate and impaired mitochondrial function, including decreased mitochondrial superoxide levels, lower mitochondrial membrane potential and diminished expression of genes related to the tricarboxylic acid cycle and genes related to the electron transport chain, as compared to WT DCs. Treatment with a NO inhibitor reversed the reduced Th17 cell differentiation in the presence of FPR2-deficient DCs. Together, by regulating DC metabolism, FPR2 enhances the production of DC-derived Th17-polarizing cytokines and hence Th17 cell differentiation in the context of neuroinflammation.
Collapse
Affiliation(s)
- Jong-Hyung Lim
- Laboratory of Innate Immunity and Inflammation, Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ales Neuwirth
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sylvia Grossklaus
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Oliver Soehnlein
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - George Hajishengallis
- Laboratory of Innate Immunity and Inflammation, Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
4
|
Jia M, Fu H, Jiang X, Wang L, Xu J, Barnes PJ, Adcock IM, Liu Y, He S, Zhang F, Yao L, Sun P, Yao X. DEL-1, as an anti-neutrophil transepithelial migration molecule, inhibits airway neutrophilic inflammation in asthma. Allergy 2024; 79:1180-1194. [PMID: 37681299 DOI: 10.1111/all.15882] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 08/05/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Neutrophil migration into the airways is a key process in neutrophilic asthma. Developmental endothelial locus-1 (DEL-1), an extracellular matrix protein, is a neutrophil adhesion inhibitor that attenuates neutrophilic inflammation. METHODS Levels of DEL-1 were measured in exhaled breath condensate (EBC) and serum in asthma patients by ELISA. DEL-1 modulation of neutrophil adhesion and transepithelial migration was examined in a co-culture model in vitro. The effects of DEL-1-adenoviral vector-mediated overexpression on ovalbumin/lipopolysaccharide (OVA/LPS)-induced neutrophilic asthma were studied in mice in vivo. RESULTS DEL-1 was primarily expressed in human bronchial epithelial cells and was decreased in asthma patients. Serum DEL-1 concentrations were reduced in patients with severe asthma compared with normal subjects (567.1 ± 75.3 vs. 276.8 ± 29.36 pg/mL, p < .001) and were negatively correlated to blood neutrophils (r = -0.2881, p = .0384) and neutrophil-to-lymphocyte ratio (NLR) (r = -0.5469, p < .0001). DEL-1 concentrations in the EBC of severe asthmatic patients (113.2 ± 8.09 pg/mL) were also lower than normal subjects (193.0 ± 7.61 pg/mL, p < .001) and were positively correlated with the asthma control test (ACT) score (r = 0.3678, p = .0035) and negatively related to EBC IL-17 (r = -0.3756, p = .0131), myeloperoxidase (MPO) (r = -0.5967, p = .0055), and neutrophil elastase (NE) (r = -0.5488, p = .0009) expression in asthma patients. Neutrophil adhesion and transepithelial migration in asthma patients were associated with LFA-1 binding to ICAM-1 and inhibited by DEL-1. DEL-1 mRNA and protein expression in human bronchial epithelial cells were regulated by IL-17. Exogenous DEL-1 inhibited IL-17-enhanced neutrophil adhesion and migration. DEL-1 expression was decreased while neutrophil infiltration was increased in the airway of a murine model of neutrophilic asthma. This was prevented by DEL-1 overexpression. CONCLUSIONS DEL-1 down-regulation leads to increased neutrophil migration across bronchial epithelial cells and is associated with neutrophilic airway inflammation in asthma.
Collapse
Affiliation(s)
- Man Jia
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Heng Fu
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyu Jiang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lina Wang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiayan Xu
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Peter J Barnes
- Airway Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Yi Liu
- Department of Allergy, Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shujuan He
- Department of Respiratory Medicine, Nanjing Red Cross Hospital, Nanjing, China
| | - Fan Zhang
- Department of Respiratory Medicine, Nanjing Red Cross Hospital, Nanjing, China
| | - Lei Yao
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Peng Sun
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Yao
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Sirisereephap K, Tamura H, Lim JH, Surboyo MDC, Isono T, Hiyoshi T, Rosenkranz AL, Sato-Yamada Y, Domon H, Ikeda A, Hirose T, Sunazuka T, Yoshiba N, Okada H, Terao Y, Maeda T, Tabeta K, Chavakis T, Hajishengallis G, Maekawa T. A novel macrolide-Del-1 axis to regenerate bone in old age. iScience 2024; 27:108798. [PMID: 38261928 PMCID: PMC10797555 DOI: 10.1016/j.isci.2024.108798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/24/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
Aging is associated with increased susceptibility to chronic inflammatory bone loss disorders, such as periodontitis, in large part due to the impaired regenerative potential of aging tissues. DEL-1 exerts osteogenic activity and promotes bone regeneration. However, DEL-1 expression declines with age. Here we show that systemically administered macrolide antibiotics and a non-antibiotic erythromycin derivative, EM-523, restore DEL-1 expression in 18-month-old ("aged") mice while promoting regeneration of bone lost due to naturally occurring age-related periodontitis. These compounds failed to induce bone regeneration in age-matched DEL-1-deficient mice. Consequently, these drugs promoted DEL-1-dependent functions, including alkaline phosphatase activity and osteogenic gene expression in the periodontal tissue while inhibiting osteoclastogenesis, leading to net bone growth. Macrolide-treated aged mice exhibited increased skeletal bone mass, suggesting that this treatment may be pertinent to systemic bone loss disorders. In conclusion, we identified a macrolide-DEL-1 axis that can regenerate bone lost due to aging-related disease.
Collapse
Affiliation(s)
- Kridtapat Sirisereephap
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
- Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Hikaru Tamura
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Jong-Hyung Lim
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Meircurius Dwi Condro Surboyo
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
- Faculty of Dentistry, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Toshihito Isono
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Takumi Hiyoshi
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Andrea L. Rosenkranz
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Yurie Sato-Yamada
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Hisanori Domon
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Akari Ikeda
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tomoyasu Hirose
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Toshiaki Sunazuka
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Nagako Yoshiba
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Hiroyuki Okada
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Takeyasu Maeda
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Koichi Tabeta
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tomoki Maekawa
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| |
Collapse
|
6
|
Vergadi E, Kolliniati O, Lapi I, Ieronymaki E, Lyroni K, Alexaki VI, Diamantaki E, Vaporidi K, Hatzidaki E, Papadaki HA, Galanakis E, Hajishengallis G, Chavakis T, Tsatsanis C. An IL-10/DEL-1 axis supports granulopoiesis and survival from sepsis in early life. Nat Commun 2024; 15:680. [PMID: 38263289 PMCID: PMC10805706 DOI: 10.1038/s41467-023-44178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/03/2023] [Indexed: 01/25/2024] Open
Abstract
The limited reserves of neutrophils are implicated in the susceptibility to infection in neonates, however the regulation of neutrophil kinetics in infections in early life remains poorly understood. Here we show that the developmental endothelial locus (DEL-1) is elevated in neonates and is critical for survival from neonatal polymicrobial sepsis, by supporting emergency granulopoiesis. Septic DEL-1 deficient neonate mice display low numbers of myeloid-biased multipotent and granulocyte-macrophage progenitors in the bone marrow, resulting in neutropenia, exaggerated bacteremia, and increased mortality; defects that are rescued by DEL-1 administration. A high IL-10/IL-17A ratio, observed in newborn sepsis, sustains tissue DEL-1 expression, as IL-10 upregulates while IL-17 downregulates DEL-1. Consistently, serum DEL-1 and blood neutrophils are elevated in septic adult and neonate patients with high serum IL-10/IL-17A ratio, and mortality is lower in septic patients with high serum DEL-1. Therefore, IL-10/DEL-1 axis supports emergency granulopoiesis, prevents neutropenia and promotes sepsis survival in early life.
Collapse
Affiliation(s)
- Eleni Vergadi
- Department of Paediatrics, School of Medicine, University of Crete, Heraklion, Greece.
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece.
| | - Ourania Kolliniati
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Ioanna Lapi
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Eleftheria Ieronymaki
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Konstantina Lyroni
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Eleni Diamantaki
- Department of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Katerina Vaporidi
- Department of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Eleftheria Hatzidaki
- Department of Neonatology/Neonatal Intensive Care Unit, School of Medicine, University of Crete, Heraklion, Greece
| | - Helen A Papadaki
- Department of Hematology, School of Medicine, University of Crete, Heraklion, Greece
| | - Emmanouil Galanakis
- Department of Paediatrics, School of Medicine, University of Crete, Heraklion, Greece
| | - George Hajishengallis
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christos Tsatsanis
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
7
|
Gauthier T, Martin-Rodriguez O, Chagué C, Daoui A, Ceroi A, Varin A, Bonnefoy F, Valmary-Degano S, Couturier M, Behlke S, Saas P, Cartron PF, Perruche S. Amelioration of experimental autoimmune encephalomyelitis by in vivo reprogramming of macrophages using pro-resolving factors. J Neuroinflammation 2023; 20:307. [PMID: 38124095 PMCID: PMC10734130 DOI: 10.1186/s12974-023-02994-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Reinstating inflammation resolution represents an innovative concept to regain inflammation control in diseases marked by chronic inflammation. While most therapeutics target inflammatory molecules and inflammatory effector cells and mediators, targeting macrophages to initiate inflammation resolution to control neuroinflammation has not yet been attempted. Resolution-phase macrophages are critical in the resolution process to regain tissue homeostasis, and are programmed through the presence and elimination of apoptotic leukocytes. Hence, inducing resolution-phase macrophages might represent an innovative therapeutic approach to control and terminate dysregulated neuroinflammation. METHODS Here, we investigated if the factors released by in vitro induced resolution-phase macrophages (their secretome) are able to therapeutically reprogram macrophages to control neuroinflammation in the model of experimental autoimmune encephalomyelitis (EAE). RESULTS We found that injection of the pro-resolutive secretome reduced demyelination and decreased inflammatory cell infiltration in the CNS, notably through the in vivo reprogramming of macrophages at the epigenetic level. Adoptive transfer experiments with in vivo or in vitro reprogrammed macrophages using such pro-resolutive secretome confirmed the stability and transferability of this acquired therapeutic activity. CONCLUSIONS Overall, our data confirm the therapeutic activity of a pro-resolution secretome in the treatment of ongoing CNS inflammation, via the epigenetic reprogramming of macrophages and open with that a new therapeutic avenue for diseases marked by neuroinflammation.
Collapse
Affiliation(s)
- Thierry Gauthier
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000, Besançon, France
| | | | - Cécile Chagué
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000, Besançon, France
| | - Anna Daoui
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000, Besançon, France
| | - Adam Ceroi
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000, Besançon, France
| | - Alexis Varin
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000, Besançon, France
| | - Francis Bonnefoy
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000, Besançon, France
- MED'INN'Pharma, 25000, Besancon, France
| | | | | | | | - Philippe Saas
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000, Besançon, France
| | - Pierre-François Cartron
- Team "Apoptosis and Tumor Progression" CRCINA-INSERM U1232, Université de Nantes Nantes, LaBEX IGO, REpiCGO, EpiSAVMEN, LaBCT, Institut de Cancérologie de L'Ouest (ICO), 44000, Nantes, France
| | - Sylvain Perruche
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000, Besançon, France.
- MED'INN'Pharma, 25000, Besancon, France.
| |
Collapse
|
8
|
Zhao M, Zheng Z, Yin Z, Zhang J, Peng S, Liu J, Pan W, Wei C, Xu Y, Qin JJ, Wan J, Wang M. DEL-1 deficiency aggravates pressure overload-induced heart failure by promoting neutrophil infiltration and neutrophil extracellular traps formation. Biochem Pharmacol 2023; 218:115912. [PMID: 37956894 DOI: 10.1016/j.bcp.2023.115912] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023]
Abstract
Recent studies have shown that neutrophils play an important role in the development and progression of heart failure. Developmental endothelial locus-1 (DEL-1) is an anti-inflammatory glycoprotein that has been found to have protective effects in various cardiovascular diseases. However, the role of DEL-1 in chronic heart failure is not well understood. In a mouse model of pressure overload-induced non-ischemic cardiac failure, we found that neutrophil infiltration in the heart increased and DEL-1 levels decreased in the early stages of heart failure. DEL-1 deficiency worsened pressure overload-induced cardiac dysfunction and remodeling in mice. Mechanistically, DEL-1 deficiency promotes neutrophil infiltration and the formation of neutrophil extracellular traps (NETs) through the regulation of P38 signaling. In vitro experiments showed that DEL-1 can inhibit P38 signaling and NETs formation in mouse neutrophils in a MAC-1-dependent manner. Depleting neutrophils, inhibiting NETs formation, and inhibiting P38 signaling all reduced the exacerbation of heart failure caused by DEL-1 deletion. Overall, our findings suggest that DEL-1 deficiency worsens pressure overload-induced heart failure by promoting neutrophil infiltration and NETs formation.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China; Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| |
Collapse
|
9
|
Sun Y, Boyko T, Marecic O, Struck D, Mann RK, Andrew TW, Lopez M, Tong X, Goodman SB, Yang F, Longaker MT, Chan CKF, Yang GP. Del1 Is a Growth Factor for Skeletal Progenitor Cells in the Fracture Callus. Biomolecules 2023; 13:1214. [PMID: 37627279 PMCID: PMC10452420 DOI: 10.3390/biom13081214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Failure to properly form bone or integrate surgical implants can lead to morbidity and additional surgical interventions in a significant proportion of orthopedic surgeries. While the role of skeletal stem cells (SSCs) in bone formation and repair is well-established, very little is known about the factors that regulate the downstream Bone, Cartilage, Stromal, Progenitors (BCSPs). BCSPs, as transit amplifying progenitor cells, undergo multiple mitotic divisions to expand the pool of lineage committed progenitors allowing stem cells to preserve their self-renewal and stemness. Del1 is a protein widely expressed in the skeletal system, but its deletion led to minimal phenotype changes in the uninjured mouse. In this paper, we demonstrate that Del1 is a key regulator of BCSP expansion following injury. In Del1 knockout mice, there is a significant reduction in the number of BCSPs which leads to a smaller callus and decreased bone formation compared with wildtype (WT) littermates. Del1 serves to promote BCSP proliferation and prevent apoptosis in vivo and in vitro. Moreover, exogenous Del1 promotes proliferation of aged human BCSPs. Our results highlight the potential of Del1 as a therapeutic target for improving bone formation and implant success. Del1 injections may improve the success of orthopedic surgeries and fracture healing by enhancing the proliferation and survival of BCSPs, which are crucial for generating new bone tissue during the process of bone formation and repair.
Collapse
Affiliation(s)
- Yuxi Sun
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Tatiana Boyko
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (R.K.M.); (T.W.A.); (M.T.L.)
| | - Owen Marecic
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (R.K.M.); (T.W.A.); (M.T.L.)
| | - Danielle Struck
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (R.K.M.); (T.W.A.); (M.T.L.)
| | - Randall K. Mann
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (R.K.M.); (T.W.A.); (M.T.L.)
| | - Tom W. Andrew
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (R.K.M.); (T.W.A.); (M.T.L.)
| | - Michael Lopez
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (R.K.M.); (T.W.A.); (M.T.L.)
| | - Xinming Tong
- Department of Orthopedic Surgery, Stanford University, Stanford, CA 94305, USA; (X.T.); (S.B.G.); (F.Y.)
| | - Stuart B. Goodman
- Department of Orthopedic Surgery, Stanford University, Stanford, CA 94305, USA; (X.T.); (S.B.G.); (F.Y.)
| | - Fan Yang
- Department of Orthopedic Surgery, Stanford University, Stanford, CA 94305, USA; (X.T.); (S.B.G.); (F.Y.)
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (R.K.M.); (T.W.A.); (M.T.L.)
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Charles K. F. Chan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (R.K.M.); (T.W.A.); (M.T.L.)
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - George P. Yang
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Birmingham VA Medical Center, Birmingham, AL 35233, USA
| |
Collapse
|
10
|
Prakash AV, Welliver RR, Mirmire S, Baron S, Hicar MD. Presence of coronary aneurysms during Kawasaki Disease (KD) correlates with lower levels of autoantibodies to both full form and spliced variant of immune regulator Del-1. Immunol Lett 2023; 256-257:34-41. [PMID: 37019289 DOI: 10.1016/j.imlet.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023]
Abstract
Kawasaki disease (KD), a rare multisystem inflammatory condition that predominantly affects children under six years of age, is the leading cause of childhood-acquired heart disease in developed countries. The pathogenesis is unknown, but studies support that an infectious stimulus triggers an autoimmune reaction in a genetically susceptible child. Recent studies demonstrated an association with autoantibody response to Del-1 (also known as EDIL3) in children with KD. Del-1 is an extracellular matrix protein that is expressed both in macrophages and vascular endothelium. Del-1 has an anti-inflammatory role by preventing leucocyte migration to inflammatory sites. Del-1 has two expression variants and genetic variants of Del-1 have been associated with the risk of intracranial aneurysms. Due to the physiologic plausibility for a role during KD, we chose to assess if autoantibodies against DEL-1 are seen in a larger cohort of children with KD and to assess if responses correlated to aneurysm formation. Contrary to prior findings, in comparison to febrile controls, autoantibodies were not overall higher in children with KD. Elevation in Post-IVIG samples in comparison to pre-IVIG and convalescent samples supports the commonality of anti-Del-1 antibodies. Autoantibodies were notably lower in children with KD who had coronary Z score elevations in comparison to those who did not.
Collapse
Affiliation(s)
- Aviraag Vijaya Prakash
- Jacobs School of Medicine and Public Health, Department of Pediatrics, University at Buffalo, Buffalo, New York.
| | - R Ross Welliver
- Jacobs School of Medicine and Public Health, Department of Pediatrics, University at Buffalo, Buffalo, New York.
| | - Sanjiti Mirmire
- Houston Methodist Hospital, Department of Neurology, Houston, Texas.
| | - Sarah Baron
- Jacobs School of Medicine and Public Health, Department of Pediatrics, University at Buffalo, Buffalo, New York.
| | - Mark D Hicar
- Jacobs School of Medicine and Public Health, Department of Pediatrics, University at Buffalo, Buffalo, New York.
| |
Collapse
|
11
|
Hajishengallis G, Chavakis T. Mechanisms and Therapeutic Modulation of Neutrophil-Mediated Inflammation. J Dent Res 2022; 101:1563-1571. [PMID: 35786033 PMCID: PMC9703529 DOI: 10.1177/00220345221107602] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Neutrophils are abundant, short-lived myeloid cells that are readily recruitable to sites of inflammation, where they serve as first-line defense against infection and other types of insult to the host. In recent years, there has been increased understanding on the involvement of neutrophils in chronic inflammatory diseases, where they may act as direct effectors of destructive inflammation. However, destructive tissue inflammation is also instigated in settings of neutrophil paucity, suggesting that neutrophils also mediate critical homeostatic functions. The activity of neutrophils is regulated by a variety of local tissue factors. In addition, systemic metabolic conditions, such as hypercholesterolemia and hyperglycemia, affect the production and mobilization of neutrophils from the bone marrow. Moreover, according to the recently emerged concept of innate immune memory, the functions of neutrophils can be enhanced through the process of trained granulopoiesis. This process may have both beneficial and potentially destructive effects, depending on context, that is, protective against infections and tumors, while destructive in the context of chronic inflammatory conditions. Although we are far from a complete understanding of the mechanisms underlying the regulation and function of neutrophils, current insights enable the development of targeted therapeutic interventions that can restrain neutrophil-mediated inflammation in chronic inflammatory diseases, such as periodontitis.
Collapse
Affiliation(s)
- G. Hajishengallis
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - T. Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Sachsen, Germany
| |
Collapse
|
12
|
Zhao M, Zheng Z, Li C, Wan J, Wang M. Developmental endothelial locus-1 in cardiovascular and metabolic diseases: A promising biomarker and therapeutic target. Front Immunol 2022; 13:1053175. [PMID: 36518760 PMCID: PMC9742254 DOI: 10.3389/fimmu.2022.1053175] [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: 09/25/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiovascular and metabolic diseases (CVMDs) are a leading cause of death worldwide and impose a major socioeconomic burden on individuals and healthcare systems, underscoring the urgent need to develop new drug therapies. Developmental endothelial locus-1 (DEL-1) is a secreted multifunctional domain protein that can bind to integrins and play an important role in the occurrence and development of various diseases. Recently, DEL-1 has attracted increased interest for its pharmacological role in the treatment and/or management of CVMDs. In this review, we present the current knowledge on the predictive and therapeutic role of DEL-1 in a variety of CVMDs, such as atherosclerosis, hypertension, cardiac remodeling, ischemic heart disease, obesity, and insulin resistance. Collectively, DEL-1 is a promising biomarker and therapeutic target for CVMDs.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Chenfei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China,*Correspondence: Menglong Wang, ; Jun Wan,
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China,*Correspondence: Menglong Wang, ; Jun Wan,
| |
Collapse
|
13
|
Christoforakis Z, Dermitzaki E, Paflioti E, Katrinaki M, Deiktakis M, H Tosounidis T, Ballalis K, Tsatsanis C, Venihaki M, Kontakis G. Correlation of systemic metabolic inflammation with knee osteoarthritis. Hormones (Athens) 2022; 21:457-466. [PMID: 35764781 DOI: 10.1007/s42000-022-00381-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 06/14/2022] [Indexed: 01/15/2023]
Abstract
PURPOSE The aim of this study was to analyze local and systematic inflammatory status in knee osteoarthritis (KOA), focusing on intra-articular and remote adipose tissue depots, and to explore its potential association with metabolic syndrome (MetS). METHODS Patients (n = 27) with end-stage KOA were enrolled in the study and samples from infrapatellar fat pad (IFP), synovium, subcutaneous adipose tissue (SAT), synovial fluid (SF), and serum were collected. In homogenates from the tissues, mRNA expression of developmental endothelial locus-1 (DEL-1) was determined. Interleukin 6 (IL-6) and interleukin 8 (IL-8) were measured in tissues and SF and serum samples by enzyme-linked immunosorbent assay. RESULTS Fifteen patients fulfilled MetS criteria (w-MetS group) and 12 did not (non-MetS). In the entire population, IL-6 levels were significantly higher in IFP compared to synovium (median (interquartile range), 26.05 (26.16) vs. 15.75 (14.8) pg/mg of total protein, p = 0.043), but not to SAT (17.89 (17.9) pg/mg); IL-8 levels were significantly higher in IFP (17.3 (19.3) pg/mg) and SAT (24.2 (26) pg/mg) when compared to synovium (8.45 (6.17) pg/mg) (p = 0.029 and < 0.001, respectively). Significantly higher IL-6 concentrations in SF were detected in w-MetS patients compared to non-MetS (194.8 (299) vs. 64.1 (86.9) pg/ml, p = 0.027). Finally, DEL-1 mRNA expression was higher in IFP compared to synovium (eightfold, p = 0.019). CONCLUSIONS Our findings support the critical role of IFP in knee joint homeostasis and progression of KOA. Furthermore, in KOA patients w-MetS, SAT is thought to play an important role in intra-knee inflammation via secretion of soluble inflammatory mediators, such as IL-6.
Collapse
Affiliation(s)
- Zacharias Christoforakis
- Department of Orthopaedics and Traumatology, University Hospital of Heraklion, Crete, Greece
- Department of Orthopaedics, General Hospital of Agios Nikolaos, Crete, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Eirini Dermitzaki
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Eleni Paflioti
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Marianna Katrinaki
- Department of Clinical Chemistry-Biochemistry, University Hospital of Heraklion, Crete, Greece
| | - Michail Deiktakis
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Theodoros H Tosounidis
- Department of Orthopaedics and Traumatology, University Hospital of Heraklion, Crete, Greece
| | - Konstantine Ballalis
- Department of Orthopaedics and Traumatology, University Hospital of Heraklion, Crete, Greece
| | - Christos Tsatsanis
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Maria Venihaki
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Crete, Greece.
| | - George Kontakis
- Department of Orthopaedics and Traumatology, University Hospital of Heraklion, Crete, Greece
| |
Collapse
|
14
|
Li R, Zeng J, Ren T. Expression of DEL-1 in alveolar epithelial cells prevents lipopolysaccharide-induced inflammation, oxidative stress, and eosinophil recruitment in acute lung injury. Int Immunopharmacol 2022; 110:108961. [PMID: 35764019 DOI: 10.1016/j.intimp.2022.108961] [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: 03/24/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 11/05/2022]
Abstract
Bacterial infection is a major cause of acute lung injury (ALI). Developmental endothelial locus-1 (DEL-1) is an immunomodulatory mediator secreted by the endothelial cells. This study aimed to investigate the role of DEL-1 in lipopolysaccharide (LPS)-induced ALI in mouse models and its ability to regulate on eosinophil recruitment. Male C57BL/6 mice were administered an adeno-associated virus (AAV)-mediated DEL-1 overexpression vector via intratracheal injection. Twenty-one days after vector instillation, mice were challenged with LPS (5 mg/kg body weight). Lung injury was evaluated using haematoxylin-eosin staining, flow cytometry, enzyme-linked immunosorbnent assay, quantitative real-time polymerase chain reaction, western blotting, immunohistochemistry and immunofluorescence analyses. DEL-1 was expressed in alveolar epithelial cells of mice. Compared with that in the control group, DEL-1 was expressed at low levels in the lungs of LPS-challenged mice. LPS injured the lungs in mice, as evidenced by an increase in alveolar wall thickness, inflammatory cell infiltration in the stroma, and alveolar collapse. AAV-mediated DEL-1 overexpression attenuated LPS-induced lung injury and inhibited the release of TNF-α, IL-6, and IL-1β. DEL-1 overexpression also attenuated LPS-induced oxidative stress by decreasing lactic dehydrogenase (LDH), myeloperoxidase (MPO), malondialdehyde (MDA), and reactive oxygen species (ROS) activities and increasing superoxide dismutase (SOD) activity. In addition, DEL-1 prevented eosinophil recruitment into lung tissues and inhibited eotaxin production. This study revealed the beneficial role of DEL-1 in preventing LPS-induced ALI in mice. Therefore, DEL-1 can protect lung tissues against LPS-induced inflammation, oxidative stress, and eosinophil recruitment.
Collapse
Affiliation(s)
- Ruirui Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, Xinjiang Uygur Autonomous Region, 832008, P.R. China.
| | - Jianqiong Zeng
- Cardiovascular Surgery CCU, Foshan First People's Hospital, Foshan, Guangdong, 528000, P.R. China
| | - Tao Ren
- Three departments of cardiology, The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, Xinjiang Uygur Autonomous Region, 832008, P.R. China
| |
Collapse
|
15
|
Cheng M, Wang H, Yang G, Cheng Y, Yang Z, Chen X, Liu Y, Sun Z. Sustained developmental endothelial locus-1 overexpression promotes spinal cord injury recovery in mice through the SIRT1/SERCA2 signaling pathway. Brain Res Bull 2022; 181:65-76. [DOI: 10.1016/j.brainresbull.2022.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/04/2022] [Accepted: 01/22/2022] [Indexed: 11/16/2022]
|
16
|
Becker GM, Burke JM, Lewis RM, Miller JE, Morgan JLM, Rosen BD, Van Tassell CP, Notter DR, Murdoch BM. Variants Within Genes EDIL3 and ADGRB3 are Associated With Divergent Fecal Egg Counts in Katahdin Sheep at Weaning. Front Genet 2022; 13:817319. [PMID: 35360858 PMCID: PMC8960952 DOI: 10.3389/fgene.2022.817319] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/02/2022] [Indexed: 12/20/2022] Open
Abstract
Gastrointestinal nematodes (GIN) pose a severe threat to sheep production worldwide. Anthelmintic drug resistance coupled with growing concern regarding potential environmental effects of drug use have demonstrated the necessity of implementing other methods of GIN control. The aim of this study was to test for genetic variants associated with resistance or susceptibility to GIN in Katahdin sheep to improve the current understanding of the genetic mechanisms responsible for host response to GIN. Linear regression and case-control genome-wide association studies were conducted with high-density genotype data and cube-root transformed weaning fecal egg counts (tFEC) of 583 Katahdin sheep. The case-control GWAS identified two significant SNPs (P-values 1.49e-08 to 1.01e-08) within introns of the gene adhesion G protein-coupled receptor B3 (ADGRB3) associated with lower fecal egg counts. With linear regression, four significant SNPs (P-values 7.82e-08 to 3.34e-08) were identified within the first intron of the gene EGF-like repeats and discoidin domains 3 (EDIL3). These identified SNPs were in very high linkage disequilibrium (r2 of 0.996–1), and animals with alternate homozygous genotypes had significantly higher median weaning tFEC phenotypes compared to all other genotypes. Significant SNPs were queried through public databases to identify putative transcription factor binding site (TFBS) and potential lncRNA differences between reference and alternate alleles. Changes in TFBS were predicted at two SNPs, and one significant SNP was found to be within a predicted lncRNA sequence with greater than 90% similarity to a known lncRNA in the bovine genome. The gene EDIL3 has been described in other species for its roles in the inhibition and resolution of inflammation. Potential changes of EDIL3 expression mediated through lncRNA expression and/or transcription factor binding may impact the overall immune response and reduce the ability of Katahdin sheep to control GIN infection. This study lays the foundation for further research of EDIL3 and ADGRB3 towards understanding genetic mechanisms of susceptibility to GIN, and suggests these SNPs may contribute to genetic strategies for improving parasite resistance traits in sheep.
Collapse
Affiliation(s)
- Gabrielle M. Becker
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Joan M. Burke
- USDA, ARS, Dale Bumpers Small Farms Research Center, Booneville, AR, United States
| | - Ronald M. Lewis
- Department of Animal Science, University of Nebraska–Lincoln, Lincoln, NE, United States
| | - James E. Miller
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | | | - Benjamin D. Rosen
- USDA, ARS, Animal Genomics and Improvement Laboratory, Beltsville, MD, United States
| | - Curtis P. Van Tassell
- USDA, ARS, Animal Genomics and Improvement Laboratory, Beltsville, MD, United States
| | - David R. Notter
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Brenda M. Murdoch
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, United States
- *Correspondence: Brenda M. Murdoch,
| |
Collapse
|
17
|
Failer T, Amponsah-Offeh M, Neuwirth A, Kourtzelis I, Subramanian P, Mirtschink P, Peitzsch M, Matschke K, Tugtekin SM, Kajikawa T, Li X, Steglich A, Gembardt F, Wegner AC, Hugo C, Hajishengallis G, Chavakis T, Deussen A, Todorov V, Kopaliani I. Developmental endothelial locus-1 protects from hypertension-induced cardiovascular remodeling via immunomodulation. J Clin Invest 2022; 132:126155. [PMID: 35133978 PMCID: PMC8920341 DOI: 10.1172/jci126155] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/02/2022] [Indexed: 11/25/2022] Open
Abstract
The causative role of inflammation in hypertension-related cardiovascular diseases is evident and calls for development of specific immunomodulatory therapies. We tested the therapeutic efficacy and mechanisms of action of developmental endothelial locus-1 (DEL-1), an endogenous antiinflammatory factor, in angiotensin II– (ANGII–) and deoxycorticosterone acetate–salt–induced (DOCA-salt–induced) cardiovascular organ damage and hypertension. By using mice with endothelial overexpression of DEL-1 (EC-Del1 mice) and performing preventive and interventional studies by injecting recombinant DEL-1 in mice, we showed that DEL-1 improved endothelial function and abrogated aortic adventitial fibrosis, medial thickening, and loss of elastin. DEL-1 also protected the mice from cardiac concentric hypertrophy and interstitial and perivascular coronary fibrosis and improved left ventricular function and myocardial coronary perfusion. DEL-1 prevented aortic stiffness and abolished the progression of hypertension. Mechanistically, DEL-1 acted by inhibiting αvβ3 integrin–dependent activation of pro-MMP2 in mice and in human isolated aorta. Moreover, DEL-1 stabilized αvβ3 integrin–dependent CD25+FoxP3+ Treg numbers and IL-10 levels, which were associated with decreased recruitment of inflammatory cells and reduced production of proinflammatory cytokines in cardiovascular organs. The demonstrated effects and immune-modulating mechanisms of DEL-1 in abrogation of cardiovascular remodeling and progression of hypertension identify DEL-1 as a potential therapeutic factor.
Collapse
Affiliation(s)
- Theresa Failer
- Department of Physiology, Technische Universität Dresden, Dresden, Germany
| | | | - Aleš Neuwirth
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ioannis Kourtzelis
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Pallavi Subramanian
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Klaus Matschke
- Department of Cardiac Surgery, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sems M Tugtekin
- Department of Cardiac Surgery, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Tetsuhiro Kajikawa
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, United States of America
| | - Xiaofei Li
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, United States of America
| | - Anne Steglich
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Florian Gembardt
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Annika C Wegner
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Christian Hugo
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - George Hajishengallis
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, United States of America
| | | | - Andreas Deussen
- Department of Physiology, Technische Universität Dresden, Dresden, Germany
| | - Vladimir Todorov
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Irakli Kopaliani
- Department of Physiology, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
18
|
The Efficacy of Fecal Microbiota Transplantation in Experimental Autoimmune Encephalomyelitis: Transcriptome and Gut Microbiota Profiling. J Immunol Res 2021; 2021:4400428. [PMID: 34938813 PMCID: PMC8687821 DOI: 10.1155/2021/4400428] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/19/2021] [Accepted: 11/25/2021] [Indexed: 12/14/2022] Open
Abstract
Objective To study the protective effect of fecal microbiota transplantation (FMT) on experimental autoimmune encephalomyelitis (EAE) and reveal its potential intestinal microflora-dependent mechanism through analyses of the intestinal microbiota and spinal cord transcriptome in mice. Method We measured the severity of disease by clinical EAE scores and H&E staining. Gut microbiota alteration in the gut and differentially expressed genes (DEGs) in the spinal cord were analyzed through 16S rRNA and transcriptome sequencing. Finally, we analyzed associations between the relative abundance of intestinal microbiota constituents and DEGs. Results We observed that clinical EAE scores were lower in the EAE+FMT group than in the EAE group. Meanwhile, mice in the EAE+FMT group also had a lower number of infiltrating cells. The results of 16S rRNA sequence analysis showed that FMT increased the relative abundance of Firmicutes and Proteobacteria and reduced the abundance of Bacteroides and Actinobacteria. Meanwhile, FMT could modulate gut microbiota balance, especially via increasing the relative abundance of g_Adlercreutzia, g_Sutterella, g_Prevotella_9, and g_Tyzzerella_3 and decreasing the relative abundance of g_Turicibacter. Next, we analyzed the transcriptome of mouse spinal cord tissue and found that 1476 genes were differentially expressed between the EAE and FMT groups. The analysis of these genes showed that FMT mainly participated in the inflammatory response. Correlation analysis between gut microbes and transcriptome revealed that the relative abundance of Adlercreutzia was correlated with the expression of inflammation-related genes negatively, including Casp6, IL1RL2 (IL-36R), IL-17RA, TNF, CCL3, CCR5, and CCL8, and correlated with the expression of neuroprotection-related genes positively, including Snap25, Edil3, Nrn1, Cpeb3, and Gpr37. Conclusion Altogether, FMT may selectively regulate gene expression to improve inflammation and maintain the stability of the intestinal environment in a gut microbiota-dependent manner.
Collapse
|
19
|
Wang H, Li X, Kajikawa T, Shin J, Lim JH, Kourtzelis I, Nagai K, Korostoff JM, Grossklaus S, Naumann R, Chavakis T, Hajishengallis G. Stromal cell-derived DEL-1 inhibits Tfh cell activation and inflammatory arthritis. J Clin Invest 2021; 131:e150578. [PMID: 34403362 PMCID: PMC8483759 DOI: 10.1172/jci150578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 08/12/2021] [Indexed: 11/17/2022] Open
Abstract
The secreted protein developmental endothelial locus 1 (DEL-1) regulates inflammatory cell recruitment and protects against inflammatory pathologies in animal models. Here, we investigated DEL-1 in inflammatory arthritis using collagen-induced arthritis (CIA) and collagen Ab-induced arthritis (CAIA) models. In both models, mice with endothelium-specific overexpression of DEL-1 were protected from arthritis relative to WT controls, whereas arthritis was exacerbated in DEL-1-deficient mice. Compared with WT controls, mice with collagen VI promoter-driven overexpression of DEL-1 in mesenchymal cells were protected against CIA but not CAIA, suggesting a role for DEL-1 in the induction of the arthritogenic Ab response. Indeed, DEL-1 was expressed in perivascular stromal cells of the lymph nodes and inhibited Tfh and germinal center B cell responses. Mechanistically, DEL-1 inhibited DC-dependent induction of Tfh cells by targeting the LFA-1 integrin on T cells. Overall, DEL-1 restrained arthritis through a dual mechanism, one acting locally in the joints and associated with the anti-recruitment function of endothelial cell-derived DEL-1; the other mechanism acting systemically in the lymph nodes and associated with the ability of stromal cell-derived DEL-1 to restrain Tfh responses. DEL-1 may therefore be a promising therapeutic for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Hui Wang
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xiaofei Li
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tetsuhiro Kajikawa
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jieun Shin
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jong-Hyung Lim
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ioannis Kourtzelis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Kosuke Nagai
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jonathan M. Korostoff
- Department of Periodontics, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sylvia Grossklaus
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ronald Naumann
- Transgenic Core Facility, Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
20
|
Li M, Zhong D, Li G. Regulatory role of local tissue signal Del-1 in cancer and inflammation: a review. Cell Mol Biol Lett 2021; 26:31. [PMID: 34217213 PMCID: PMC8254313 DOI: 10.1186/s11658-021-00274-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/28/2021] [Indexed: 12/29/2022] Open
Abstract
Developmental endothelial locus-1 (Del-1) is a secretory, multifunctional domain protein. It can bind to integrins and phosphatidylserine. As a local tissue signal, it plays a regulatory role in the cancer microenvironment and inflammation. Del-1 has destructive effects in most cancers and is associated with the progression and invasion of some cancers. In contrast, Del-1 also plays a protective role in inflammation. Del-1 regulates inflammation by regulating the generation of neutrophils in bone marrow, inhibiting the recruitment and migration of neutrophils and accelerating the clearance of neutrophils by macrophages. Del-1 and IL-17 are reciprocally regulated, and their balance maintains immune system homeostasis. Del-1 is expected to become a new therapeutic target for inflammatory disorders such as multiple sclerosis.
Collapse
Affiliation(s)
- Meng Li
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Road, Harbin, 150001, Heilongjiang, China
| | - Di Zhong
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Road, Harbin, 150001, Heilongjiang, China.
| | - Guozhong Li
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Road, Harbin, 150001, Heilongjiang, China
| |
Collapse
|
21
|
Chung JW, Kim HT, Ha YS, Lee EH, Chun SY, Lee CH, Byeon KH, Choi SH, Lee JN, Kim BS, Kim TH, Yoo ES, Yoon GS, Baek MC, Kwon TG. Identification of a novel non-invasive biological marker to overcome the shortcomings of PSA in diagnosis and risk stratification for prostate cancer: Initial prospective study of developmental endothelial locus-1 protein. PLoS One 2021; 16:e0250254. [PMID: 33901217 PMCID: PMC8075267 DOI: 10.1371/journal.pone.0250254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/01/2021] [Indexed: 12/27/2022] Open
Abstract
Objective This prospective study sought to clarify the developmental endothelial locus-1 (Del-1) protein as values of diagnosis and risk stratification of prostate cancer (PCa). Design From February 2017 to December 2019, a total 458 patients who underwent transrectal ultrasound guided prostate biopsy or surgery of benign prostatic hyperplasia agreed to research of Del-1 protein. We prospectively compared and analyzed the Del-1 protein and prostate specific antigen (PSA) in relation to the patients’ demographic and clinicopathological characteristics. Results Mean age was 68.86±8.55 years. Mean PSA and Del-1 protein was 21.72±89.37, 0.099±0.145, respectively. Two hundred seventy-six (60.3%) patients were diagnosed as PCa. Among them, 181 patients underwent radical prostatectomy (RP). There were significant differences in Del-1 protein between benign and PCa group (0.066±0.131 vs 0.121±0.149, respectively, p<0.001). When we set the cut-off value of del-1 protein as 0.120, in patients with 3≤PSA≤8, positive predictive value and specificity of Del-1 protein (≥0.120) for predicting PCa were 88.9% (56/63) and 93.5% (101/108), respectively. Among 181 patients who underwent RP, there were significant differences in Del-1 protein according to stage (pT2 vs pT3a vs ≥pT3b) (0.113±0.078, 0.171±0.121, 0.227±0.161, respectively, p<0.001) and to Gleason score (6 (3+3) or 7 (3+4) vs 7 (4+3) or 8 (4+4) vs 9 or 10) (0.134±0.103, 0.150±0.109, 0.212±0.178, respectively, P = 0.044). Multivariate analysis showed that PSA, Del-1 protein and high Gleason score (≥9) were the independent prognostic factors for predicting higher pT stage (≥3b). Furthermore, age, PSA and Del-1 protein were independent prognostic factors for predicting significant PCa. Conclusion Patients with PCa showed higher expression of Del-1 protein than benign patients. Del-1 protein increased with the stage and Gleason score of PCa. Collaboration with PSA, Del-1 protein can be a non-invasive useful marker for diagnosis and risk stratification of PCa.
Collapse
Affiliation(s)
- Jae-Wook Chung
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyun Tae Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Yun-Sok Ha
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Hye Lee
- Biomedical Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - So Young Chun
- Biomedical Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Chan-Hyeong Lee
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Kyeong Hyeon Byeon
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seock Hwan Choi
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jun Nyung Lee
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Bum Soo Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Tae-Hwan Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Sang Yoo
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ghil Suk Yoon
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Korea
- * E-mail: (MCB); (TGK)
| | - Tae Gyun Kwon
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Joint Institute for Regenerative Medicine, Kyungpook National University, Daegu, Republic of Korea
- * E-mail: (MCB); (TGK)
| |
Collapse
|
22
|
Hajishengallis G, Chavakis T. DEL-1: a potential therapeutic target in inflammatory and autoimmune disease? Expert Rev Clin Immunol 2021; 17:549-552. [PMID: 33870840 DOI: 10.1080/1744666x.2021.1915771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- George Hajishengallis
- Penn Dental Medicine, Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Triantafyllos Chavakis
- Faculty of Medicine, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
23
|
Lee SJ, Jeong JH, Lee J, Park HY, Jung JH, Kang J, Kim EA, Park NJY, Park JY, Lee IH, Chae YS. MicroRNA-496 inhibits triple negative breast cancer cell proliferation by targeting Del-1. Medicine (Baltimore) 2021; 100:e25270. [PMID: 33832090 PMCID: PMC8036068 DOI: 10.1097/md.0000000000025270] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 03/05/2021] [Indexed: 01/05/2023] Open
Abstract
Del-1 has been linked to the pathogenesis of various cancers, including breast cancer. However, the regulation of Del-1 expression remains unclear. We previously reported the interaction between microRNA-137 (miR-137) and the Del-1 gene. In this study, we investigated miR-496 and miR-137 as regulators of Del-1 expression in triple negative breast cancer (TNBC). Del-1 mRNA and miR-496 were measured by quantitative PCR in breast cancer cells (MDA-MB-231, MCF7, SK-BR3, and T-47D) and tissues from 30 patients with TNBC. The effects of miR-496 on cell proliferation, migration, and invasion were determined with MTT, wound healing, and Matrigel transwell assays, respectively. In MDA-MB-231 cells, miR-496 levels were remarkably low and Del-1 mRNA levels were higher than in other breast cancer cell lines. Luciferase reporter assays revealed that miR-496 binds the 3'-UTR of Del-1 and Del-1 expression is downregulated by miR-496 mimics. Furthermore, miR-496 inhibited the proliferation, migration, and invasion of MDA-MB-231 cells. The effects of miR-496 on cell proliferation were additive with those of miR-137, another miRNA that regulates Del-1 expression. Moreover, in the 30 TNBC specimens, miR-496 was downregulated (P < .005) and the levels of Del-1 in the plasma were significantly elevated as compared with in normal controls (P = .0142). The Cancer Genome Atlas (TCGA) data showed the correlation of miR-496 expression with better overall survival in patients with early TNBC. In in silico and in vitro analyses, we showed that Del-1 is a target of miR-496 in TNBC and thereby affects cancer progression. Our findings suggest that miR-496 and miR-137 additively target Del-1 and act as modulating factors in TNBC. They are potentially new biomarkers for patients with TNBC.
Collapse
Affiliation(s)
- Soo Jung Lee
- Department of Oncology/Hematology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital
| | | | - Jeeyeon Lee
- Breast & Thyroid Surgery, Kyungpook National University, Kyungpook National University Chilgok Hospital
| | - Ho Yong Park
- Breast & Thyroid Surgery, Kyungpook National University, Kyungpook National University Chilgok Hospital
| | - Jin Hyang Jung
- Breast & Thyroid Surgery, Kyungpook National University, Kyungpook National University Chilgok Hospital
| | | | - Eun Ae Kim
- Exosome Convergence Research Center, School of Medicine, Kyungpook National University
| | - Nora Jee-Young Park
- Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Ji-Young Park
- Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - In Hee Lee
- Department of Oncology/Hematology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital
| | - Yee Soo Chae
- Department of Oncology/Hematology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital
| |
Collapse
|
24
|
Tamura H, Maekawa T, Domon H, Hiyoshi T, Hirayama S, Isono T, Sasagawa K, Yonezawa D, Takahashi N, Oda M, Maeda T, Tabeta K, Terao Y. Effects of Erythromycin on Osteoclasts and Bone Resorption via DEL-1 Induction in Mice. Antibiotics (Basel) 2021; 10:antibiotics10030312. [PMID: 33803007 PMCID: PMC8002756 DOI: 10.3390/antibiotics10030312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 01/14/2023] Open
Abstract
Macrolides are used to treat various infectious diseases, including periodontitis. Furthermore, macrolides are known to have immunomodulatory effects; however, the underlying mechanism of their action remains unclear. DEL-1 has emerged as an important factor in homeostatic immunity and osteoclastogenesis. Specifically, DEL-1 is downregulated in periodontitis tissues. Therefore, in the present study, we investigated whether the osteoclastogenesis inhibitory effects of erythromycin (ERM) are mediated through upregulation of DEL-1 expression. We used a ligature-induced periodontitis model in C57BL/6Ncrl wild-type or DEL-1-deficient mice and in vitro cell-based mechanistic studies to investigate how ERM inhibits alveolar bone resorption. As a result of measuring alveolar bone resorption and gene expression in the tooth ligation model, ERM treatment reduced bone loss by increasing DEL-1 expression and decreasing the expression of osteoclast-related factors in wild-type mice. In DEL-1-deficient mice, ERM failed to suppress bone loss and gene expression of osteoclast-related factors. In addition, ERM treatment downregulated osteoclast differentiation and calcium resorption in in vitro experiments with mouse bone marrow-derived macrophages. In conclusion, ERM promotes the induction of DEL-1 in periodontal tissue, which may regulate osteoclastogenesis and decrease inflammatory bone resorption. These findings suggest that ERM may exert immunomodulatory effects in a DEL-1-dependent manner.
Collapse
Affiliation(s)
- Hikaru Tamura
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (H.T.); (T.H.); (S.H.); (T.I.); (K.S.); (Y.T.)
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan;
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (N.T.); (K.T.)
| | - Tomoki Maekawa
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (H.T.); (T.H.); (S.H.); (T.I.); (K.S.); (Y.T.)
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan;
- Correspondence: (T.M.); (H.D.); Tel.: +81-25-227-2828 (T.M.); +81-227-2840 (H.D.)
| | - Hisanori Domon
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (H.T.); (T.H.); (S.H.); (T.I.); (K.S.); (Y.T.)
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan;
- Correspondence: (T.M.); (H.D.); Tel.: +81-25-227-2828 (T.M.); +81-227-2840 (H.D.)
| | - Takumi Hiyoshi
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (H.T.); (T.H.); (S.H.); (T.I.); (K.S.); (Y.T.)
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (N.T.); (K.T.)
| | - Satoru Hirayama
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (H.T.); (T.H.); (S.H.); (T.I.); (K.S.); (Y.T.)
| | - Toshihito Isono
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (H.T.); (T.H.); (S.H.); (T.I.); (K.S.); (Y.T.)
| | - Karin Sasagawa
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (H.T.); (T.H.); (S.H.); (T.I.); (K.S.); (Y.T.)
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (N.T.); (K.T.)
| | - Daisuke Yonezawa
- Division of Oral Science for Health Promotion, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan;
| | - Naoki Takahashi
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (N.T.); (K.T.)
| | - Masataka Oda
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Yamashita 607-8414, Japan;
| | - Takeyasu Maeda
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan;
| | - Koichi Tabeta
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (N.T.); (K.T.)
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (H.T.); (T.H.); (S.H.); (T.I.); (K.S.); (Y.T.)
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan;
| |
Collapse
|
25
|
Lee SJ, Lee J, Jung JH, Park HY, Moon PG, Chae YS, Baek MC. Exosomal Del-1 as a Potent Diagnostic Marker for Breast Cancer: Prospective Cohort Study. Clin Breast Cancer 2021; 21:e748-e756. [PMID: 33722523 DOI: 10.1016/j.clbc.2021.02.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/26/2020] [Accepted: 02/08/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND The differential diagnostic role of plasma developmental endothelial locus-1 (Del-1) was proposed in our previous study. Therefore the current study aimed to confirm the diagnostic role and explore the prognostic role of exosomal Del-1 in a prospective cohort of female patients with breast cancer. PATIENTS AND METHODS To determine the optimal sampling time for the postoperative Del-1 measurements, blood was serially collected on days 1, 3, 5, and 7 after surgery in 22 patients (cohort 1). Thereafter, 111 female patients with breast cancer were prospectively enrolled (cohort 2) to compare exosomal Del-1 levels before and after surgery. RESULTS Among the subsequent prospective cohort, 107 patients (96.4%) showed a high exosomal Del-1 level (optical density [OD] value > 0.5) at the time of diagnosis. Of these patients, 101 (94.6%) in this high-level group showed normalized Del-1 levels postoperatively, representing a significant difference (mean OD value, 1.232 vs. 0.196; P < .00001). High postoperative Del-1 level was significantly associated with a worse disease-free survival adjusted to the clinicopathological characteristics (hazard ratio, 24.0; P = .0011). CONCLUSION This study confirmed the normalization of exosomal Del-1 after surgery, indicating exosomal Del-1 as a potent diagnostic biomarker for breast cancer. In addition, because a high Del-1 level after surgery was associated with early relapse, this suggests exosomal Del-1 as a potential prognostic marker by identifying the existence of residual cancer.
Collapse
Affiliation(s)
- Soo Jung Lee
- School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Jeeyeon Lee
- School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Breast & Thyroid Surgery, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Jin Hyang Jung
- School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Breast & Thyroid Surgery, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Ho Yong Park
- School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Breast & Thyroid Surgery, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Pyong-Gon Moon
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Yee Soo Chae
- School of Medicine, Kyungpook National University, Daegu, South Korea; Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, Daegu, South Korea.
| | - Moon-Chang Baek
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea; Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
26
|
Li X, Colamatteo A, Kalafati L, Kajikawa T, Wang H, Lim JH, Bdeir K, Chung KJ, Yu X, Fusco C, Porcellini A, De Simone S, Matarese G, Chavakis T, De Rosa V, Hajishengallis G. The DEL-1/β3 integrin axis promotes regulatory T cell responses during inflammation resolution. J Clin Invest 2021; 130:6261-6277. [PMID: 32817592 DOI: 10.1172/jci137530] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023] Open
Abstract
FOXP3+CD4+ regulatory T cells (Tregs) are critical for immune homeostasis and respond to local tissue cues, which control their stability and function. We explored here whether developmental endothelial locus-1 (DEL-1), which, like Tregs, increases during resolution of inflammation, promotes Treg responses. DEL-1 enhanced Treg numbers and function at barrier sites (oral and lung mucosa). The underlying mechanism was dissected using mice lacking DEL-1 or expressing a point mutant thereof, or mice with T cell-specific deletion of the transcription factor RUNX1, identified by RNA sequencing analysis of the DEL-1-induced Treg transcriptome. Specifically, through interaction with αvβ3 integrin, DEL-1 promoted induction of RUNX1-dependent FOXP3 expression and conferred stability of FOXP3 expression upon Treg restimulation in the absence of exogenous TGF-β1. Consistently, DEL-1 enhanced the demethylation of the Treg-specific demethylated region (TSDR) in the mouse Foxp3 gene and the suppressive function of sorted induced Tregs. Similarly, DEL-1 increased RUNX1 and FOXP3 expression in human conventional T cells, promoting their conversion into induced Tregs with increased TSDR demethylation, enhanced stability, and suppressive activity. We thus uncovered a DEL-1/αvβ3/RUNX1 axis that promotes Treg responses at barrier sites and offers therapeutic options for modulating inflammatory/autoimmune disorders.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alessandra Colamatteo
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II," Naples, Italy
| | - Lydia Kalafati
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases, Dresden, Germany, and German Cancer Research Center, Heidelberg, Germany
| | - Tetsuhiro Kajikawa
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hui Wang
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jong-Hyung Lim
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Khalil Bdeir
- Department of Pathology and Laboratory Medicine and
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Xiang Yu
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Clorinda Fusco
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II," Naples, Italy
| | - Antonio Porcellini
- Dipartimento di Biologia, Università degli Studi di Napoli "Federico II," Complesso Universitario di Monte Santangelo, Naples, Italy
| | - Salvatore De Simone
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Naples, Italy
| | - Giuseppe Matarese
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II," Naples, Italy.,Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Naples, Italy
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Veronica De Rosa
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Naples, Italy.,Unità di Neuroimmunologia, Fondazione Santa Lucia, Rome, Italy
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
27
|
Kwon CH, Sun JL, Kim MJ, Abd El-Aty AM, Jeong JH, Jung TW. Clinically confirmed DEL-1 as a myokine attenuates lipid-induced inflammation and insulin resistance in 3T3-L1 adipocytes via AMPK/HO-1- pathway. Adipocyte 2020; 9:576-586. [PMID: 32954935 PMCID: PMC7714434 DOI: 10.1080/21623945.2020.1823140] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Regular exercise is the first line of therapy for treating obesity-mediated metabolic disorders, including insulin resistance. It has been reported that developmental endothelial locus-1 (DEL-1) enhances macrophage efferocytosis, resulting in inflammation clearance as well as improves insulin resistance in skeletal muscle. However, the relationship between exercise and DEL-1, and the effects of DEL-1 on insulin signalling in adipocytes have not been fully elucidated to date. Protein expression levels were determined by Western blot analysis. Cells were transfected with small interfering (si) RNA to suppress gene expression. Lipid accumulation levels were detected using Oil red-O staining. Proinflammatory cytokine secretion levels were measured using ELISA. DEL-1 expression levels were induced in the skeletal muscle of people who exercised using microarray analysis. Recombinant DEL-1 augmented AMP-activated protein kinase (AMPK) phosphorylation and haem oxygenase (HO)-1 expression to alleviating inflammation and impairment of insulin signalling in 3T3-L1 adipocytes treated with palmitate. siRNA of AMPK or HO-1 also mitigated the effects of DEL-1 on inflammation and insulin resistance. DEL-1 ameliorates inflammation and insulin resistance in differentiated 3T3-L1 cells via AMPK/HO-1 signalling, suggesting that DEL-1 may be the exercise-mediated therapeutic target for treating insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Chang Hyuk Kwon
- Center for Bioinformatics, EONE Laboratories, Incheon, Republic of Korea
| | - Jaw Long Sun
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Myeong Jun Kim
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - A. M. Abd El-Aty
- State Key Laboratory of Biobased Material and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
28
|
Hyun YM, Seo SU, Choi WS, Kwon HJ, Kim DY, Jeong S, Kang GY, Yi E, Kim M, Ryu HJ, Looney MR, Choi EY, Kim HS. Endogenous DEL-1 restrains melanoma lung metastasis by limiting myeloid cell-associated lung inflammation. SCIENCE ADVANCES 2020; 6:6/45/eabc4882. [PMID: 33158867 PMCID: PMC7673713 DOI: 10.1126/sciadv.abc4882] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/18/2020] [Indexed: 05/07/2023]
Abstract
Distant metastasis represents the primary cause of cancer-associated death. Pulmonary metastasis is most frequently seen in many cancers, largely driven by lung inflammation. Components from primary tumor or recruited leukocytes are known to facilitate metastasis formation. However, contribution of target site-specific host factor to metastasis is poorly understood. Here, we show that developmental endothelial locus-1 (DEL-1), an anti-inflammatory factor abundant in the lung and down-regulated by inflammatory insults, protects from melanoma lung metastasis independently of primary tumor development and systemic immunosurveillance. DEL-1 deficiency is associated with gene profiles that favor metastatic progression with inflammation and defective immunosurveillance. Mechanistically, DEL-1 deficiency primarily influences Ly6G+ neutrophil accumulation in lung metastatic niche, leading to IL-17A up-regulation from γδ T cells and reduced antimetastatic NK cells. In support, neutrophil depletion or recombinant DEL-1 treatment profoundly reverses these effects. Thus, our results identify DEL-1 as a previously unrecognized link between tumor-induced inflammation and pulmonary metastasis.
Collapse
Affiliation(s)
- Young-Min Hyun
- Department of Anatomy and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang-Uk Seo
- Mucosal Immunology Laboratory, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Woo Seon Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyung-Joon Kwon
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Young Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Soi Jeong
- Department of Anatomy and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gyeong-Yi Kang
- Department of Anatomy and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eunbi Yi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Minjung Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyun Jin Ryu
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mark R Looney
- Departments of Medicine and Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Eun Young Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Hun Sik Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
29
|
Maekawa T, Tamura H, Domon H, Hiyoshi T, Isono T, Yonezawa D, Hayashi N, Takahashi N, Tabeta K, Maeda T, Oda M, Ziogas A, Alexaki VI, Chavakis T, Terao Y, Hajishengallis G. Erythromycin inhibits neutrophilic inflammation and mucosal disease by upregulating DEL-1. JCI Insight 2020; 5:136706. [PMID: 32603314 DOI: 10.1172/jci.insight.136706] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/24/2020] [Indexed: 02/06/2023] Open
Abstract
Macrolide antibiotics exert antiinflammatory effects; however, little is known regarding their immunomodulatory mechanisms. In this study, using 2 distinct mouse models of mucosal inflammatory disease (LPS-induced acute lung injury and ligature-induced periodontitis), we demonstrated that the antiinflammatory action of erythromycin (ERM) is mediated through upregulation of the secreted homeostatic protein developmental endothelial locus-1 (DEL-1). Consistent with the anti-neutrophil recruitment action of endothelial cell-derived DEL-1, ERM inhibited neutrophil infiltration in the lungs and the periodontium in a DEL-1-dependent manner. Whereas ERM (but not other antibiotics, such as josamycin and penicillin) protected against lethal pulmonary inflammation and inflammatory periodontal bone loss, these protective effects of ERM were abolished in Del1-deficient mice. By interacting with the growth hormone secretagogue receptor and activating JAK2 in human lung microvascular endothelial cells, ERM induced DEL-1 transcription that was mediated by MAPK p38 and was CCAAT/enhancer binding protein-β dependent. Moreover, ERM reversed IL-17-induced inhibition of DEL-1 transcription, in a manner that was dependent not only on JAK2 but also on PI3K/AKT signaling. Because DEL-1 levels are severely reduced in inflammatory conditions and with aging, the ability of ERM to upregulate DEL-1 may lead to a novel approach for the treatment of inflammatory and aging-related diseases.
Collapse
Affiliation(s)
- Tomoki Maekawa
- Center for Advanced Oral Science.,Division of Microbiology and Infectious Diseases.,Division of Periodontology, and
| | - Hikaru Tamura
- Center for Advanced Oral Science.,Division of Microbiology and Infectious Diseases.,Division of Periodontology, and
| | - Hisanori Domon
- Center for Advanced Oral Science.,Division of Microbiology and Infectious Diseases
| | - Takumi Hiyoshi
- Center for Advanced Oral Science.,Division of Microbiology and Infectious Diseases
| | | | - Daisuke Yonezawa
- Center for Advanced Oral Science.,Division of Oral Science for Health Promotion, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Naoki Hayashi
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Yamashina, Japan
| | | | | | - Takeyasu Maeda
- Center for Advanced Oral Science.,Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Masataka Oda
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Yamashina, Japan
| | - Athanasios Ziogas
- Institute of Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany.,Centre for Cardiovascular Science, Queen's Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Yutaka Terao
- Center for Advanced Oral Science.,Division of Microbiology and Infectious Diseases
| | - George Hajishengallis
- Laboratory of Innate Immunity and Inflammation, Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
30
|
Yuh DY, Maekawa T, Li X, Kajikawa T, Bdeir K, Chavakis T, Hajishengallis G. The secreted protein DEL-1 activates a β3 integrin-FAK-ERK1/2-RUNX2 pathway and promotes osteogenic differentiation and bone regeneration. J Biol Chem 2020; 295:7261-7273. [PMID: 32280065 PMCID: PMC7247308 DOI: 10.1074/jbc.ra120.013024] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/08/2020] [Indexed: 12/25/2022] Open
Abstract
The integrin-binding secreted protein developmental endothelial locus-1 (DEL-1) is involved in the regulation of both the initiation and resolution of inflammation in different diseases, including periodontitis, an oral disorder characterized by inflammatory bone loss. Here, using a mouse model of bone regeneration and in vitro cell-based mechanistic studies, we investigated whether and how DEL-1 can promote alveolar bone regeneration during resolution of experimental periodontitis. Compared with WT mice, mice lacking DEL-1 or expressing a DEL-1 variant with an Asp-to-Glu substitution in the RGD motif ("RGE point mutant"), which does not interact with RGD-dependent integrins, exhibited defective bone regeneration. Local administration of DEL-1 or of its N-terminal segment containing the integrin-binding RGD motif, but not of the RGE point mutant, reversed the defective bone regeneration in the DEL-1-deficient mice. Moreover, DEL-1 (but not the RGE point mutant) promoted osteogenic differentiation of MC3T3-E1 osteoprogenitor cells or of primary calvarial osteoblastic cells in a β3 integrin-dependent manner. The ability of DEL-1 to promote in vitro osteogenesis, indicated by induction of osteogenic genes such as the master transcription factor Runt-related transcription factor-2 (Runx2) and by mineralized nodule formation, depended on its capacity to induce the phosphorylation of focal adhesion kinase (FAK) and of extracellular signal-regulated kinase 1/2 (ERK1/2). We conclude that DEL-1 can activate a β3 integrin-FAK-ERK1/2-RUNX2 pathway in osteoprogenitors and promote new bone formation in mice. These findings suggest that DEL-1 may be therapeutically exploited to restore bone lost due to periodontitis and perhaps other osteolytic conditions.
Collapse
Affiliation(s)
- Da-Yo Yuh
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Tomoki Maekawa
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104; Research Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Xiaofei Li
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Tetsuhiro Kajikawa
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Khalil Bdeir
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, 001069 Dresden, Germany
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104.
| |
Collapse
|
31
|
Inönü E, Kayis SA, Eskan MA, Hakki SS. Salivary Del‐1, IL‐17, and LFA‐1 levels in periodontal health and disease. J Periodontal Res 2020; 55:511-518. [DOI: 10.1111/jre.12738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 01/13/2020] [Accepted: 02/04/2020] [Indexed: 01/13/2023]
Affiliation(s)
- Elif Inönü
- Department of Periodontology Faculty of Dentistry Baskent University Ankara Turkey
| | - Seyit Ali Kayis
- Department of Biostatistics Faculty of Medicine Karabuk University Karabuk Turkey
| | - Mehmet Akif Eskan
- Department of Periodontics NOVA Southeastern University College of Dental Medicine Fort Lauderdale FL USA
| | - Sema S. Hakki
- Department of Periodontology Faculty of Dentistry Selcuk University Konya Turkey
| |
Collapse
|
32
|
Kim DY, Lee SH, Fu Y, Jing F, Kim WY, Hong SB, Song JA, Choe H, Ryu HJ, Kim M, Lim D, Kim MS, Yun CO, Lee T, Hyun H, Choi EY. Del-1, an Endogenous Inhibitor of TGF-β Activation, Attenuates Fibrosis. Front Immunol 2020; 11:68. [PMID: 32117240 PMCID: PMC7018852 DOI: 10.3389/fimmu.2020.00068] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/10/2020] [Indexed: 02/06/2023] Open
Abstract
Uncontrolled activation of transforming growth factor (TGF)-β results in a wide range of pathologic conditions. Therapeutic interventions to regulate TGF-β signaling during fibrosis have been developed but the effectiveness is still limited. Here, we show that developmental endothelial locus-1 (Del-1) ameliorates fibrosis in mice by inhibiting αv integrin-mediated activation of TGF-β. Del-1 bound to αvβ6 integrin, an important activator of TGF-β, and inhibited the binding of αvβ6 integrin to the latency-associated peptide (LAP), thereby suppressing αv integrin-mediated activation of TGF-β. Lack of Del-1 increased colocalization of αv integrin and LAP in the lungs, which was reversed by Del-1 supplementation. The crucial role of Del-1 in regulating TGF-β activity was recapitulated in a mouse model of fibrosis using an adenovirus expressing inactive TGF-β1. Del-1 supplementation improved the pathological characteristics of the mice and reduced mortality. Thus, we propose that Del-1 is a negative regulator of TGF-β activation and a potential anti-fibrotic factor.
Collapse
Affiliation(s)
- Dong-Young Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seung-Hwan Lee
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yan Fu
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Feifeng Jing
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Won-Young Kim
- Division of Critical Care Medicine, Department of Internal Medicine, Chung-Ang University Hospital, Seoul, South Korea
| | - Sang-Bum Hong
- Division of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jung-A Song
- Department of Physiology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Han Choe
- Department of Physiology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyun Jin Ryu
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Minjung Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dahae Lim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Min-Seon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea
| | - Taewon Lee
- Division of Applied Mathematical Sciences, College of Science and Technology, Korea University, Sejong, South Korea
| | - Hoon Hyun
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, South Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
33
|
Ziogas A, Maekawa T, Wiessner JR, Le TT, Sprott D, Troullinaki M, Neuwirth A, Anastasopoulou V, Grossklaus S, Chung KJ, Sperandio M, Chavakis T, Hajishengallis G, Alexaki VI. DHEA Inhibits Leukocyte Recruitment through Regulation of the Integrin Antagonist DEL-1. THE JOURNAL OF IMMUNOLOGY 2020; 204:1214-1224. [PMID: 31980574 DOI: 10.4049/jimmunol.1900746] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
Leukocytes are rapidly recruited to sites of inflammation via interactions with the vascular endothelium. The steroid hormone dehydroepiandrosterone (DHEA) exerts anti-inflammatory properties; however, the underlying mechanisms are poorly understood. In this study, we show that an anti-inflammatory mechanism of DHEA involves the regulation of developmental endothelial locus 1 (DEL-1) expression. DEL-1 is a secreted homeostatic factor that inhibits β2-integrin-dependent leukocyte adhesion, and the subsequent leukocyte recruitment and its expression is downregulated upon inflammation. Similarly, DHEA inhibited leukocyte adhesion to the endothelium in venules of the inflamed mouse cremaster muscle. Importantly, in a model of lung inflammation, DHEA limited neutrophil recruitment in a DEL-1-dependent manner. Mechanistically, DHEA counteracted the inhibitory effect of inflammation on DEL-1 expression. Indeed, whereas TNF reduced DEL-1 expression and secretion in endothelial cells by diminishing C/EBPβ binding to the DEL-1 gene promoter, DHEA counteracted the inhibitory effect of TNF via activation of tropomyosin receptor kinase A (TRKA) and downstream PI3K/AKT signaling that restored C/EBPβ binding to the DEL-1 promoter. In conclusion, DHEA restrains neutrophil recruitment by reversing inflammation-induced downregulation of DEL-1 expression. Therefore, the anti-inflammatory DHEA/DEL-1 axis could be harnessed therapeutically in the context of inflammatory diseases.
Collapse
Affiliation(s)
- Athanasios Ziogas
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany;
| | - Tomoki Maekawa
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104.,Research Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, 951-8514 Niigata, Japan
| | - Johannes R Wiessner
- Walter Brendel Centre of Experimental Medicine and Institute of Cardiovascular Physiology and Pathophysiology, BioMedical Centre, Ludwig Maximilians University of Munich, 81377 Planegg-Martinsried, Germany; and
| | - Thi Trang Le
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - David Sprott
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Maria Troullinaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ales Neuwirth
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Vasiliki Anastasopoulou
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Sylvia Grossklaus
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Kyoung-Jin Chung
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Markus Sperandio
- Walter Brendel Centre of Experimental Medicine and Institute of Cardiovascular Physiology and Pathophysiology, BioMedical Centre, Ludwig Maximilians University of Munich, 81377 Planegg-Martinsried, Germany; and
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, United Kingdom
| | - George Hajishengallis
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany;
| |
Collapse
|
34
|
Troullinaki M, Chen LS, Witt A, Pyrina I, Phieler J, Kourtzelis I, Chmelar J, Sprott D, Gercken B, Koutsilieris M, Chavakis T, Chatzigeorgiou A. Robo4-mediated pancreatic endothelial integrity decreases inflammation and islet destruction in autoimmune diabetes. FASEB J 2020; 34:3336-3346. [PMID: 31916652 DOI: 10.1096/fj.201900125rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/18/2022]
Abstract
In Type 1 Diabetes Mellitus (T1DM), leukocyte infiltration of the pancreatic islets and the resulting immune-mediated destruction of beta cells precede hyperglycemia and clinical disease symptoms. In this context, the role of the pancreatic endothelium as a barrier for autoimmunity- and inflammation-related destruction of the islets is not well studied. Here, we identified Robo4, expressed on endothelial cells, as a regulator of pancreatic vascular endothelial permeability during autoimmune diabetes. Circulating levels of Robo4 were upregulated in mice subjected to the Multiple Low-Dose Streptozotocin (MLDS) model of diabetes. Upon MLDS induction, Robo4-deficiency resulted in increased pancreatic vascular permeability, leukocyte infiltration to the islets and islet apoptosis, associated with reduced insulin levels and faster diabetes development. On the contrary, in vivo administration of Slit2 in mice modestly delayed the emergence of hyperglycaemia and ameliorated islet inflammation in MLDS-induced diabetes. Thus, Robo4-mediated endothelial barrier integrity reduces insulitis and islet destruction in autoimmune diabetes. Our findings highlight the importance of the endothelium as gatekeeper of pancreatic inflammation during T1DM development and may pave the way for novel Robo4-related therapeutic approaches for autoimmune diabetes.
Collapse
Affiliation(s)
- Maria Troullinaki
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Lan-Sun Chen
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anke Witt
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Julia Phieler
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ioannis Kourtzelis
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jindrich Chmelar
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - David Sprott
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Antonios Chatzigeorgiou
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
35
|
DEL-1 ameliorates high-fat diet-induced insulin resistance in mouse skeletal muscle through SIRT1/SERCA2-mediated ER stress suppression. Biochem Pharmacol 2020; 171:113730. [DOI: 10.1016/j.bcp.2019.113730] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/21/2019] [Indexed: 12/21/2022]
|
36
|
Hajishengallis G. New developments in neutrophil biology and periodontitis. Periodontol 2000 2019; 82:78-92. [DOI: 10.1111/prd.12313] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- George Hajishengallis
- Department of Microbiology Penn Dental Medicine University of Pennsylvania Philadelphia Pennsylvania, USA
| |
Collapse
|
37
|
MicroRNA-137 Inhibits Cancer Progression by Targeting Del-1 in Triple-Negative Breast Cancer Cells. Int J Mol Sci 2019; 20:ijms20246162. [PMID: 31817673 PMCID: PMC6941134 DOI: 10.3390/ijms20246162] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) can be used to target a variety of human malignancy by targeting their oncogenes or tumor suppressor genes. The developmental endothelial locus-1 (Del-1) might be under miRNA regulation. This study investigated microRNA-137 (miR-137) function and Del-1 expression in triple-negative breast cancer (TNBC) cells and tissues. Del-1 mRNA and miRNA-137 levels were determined via qRT-PCR in breast cancer cells (MDA-MB-231, MCF7, SK-BR3, and T-47D) and tissues from 30 patients with TNBC. The effects of miR-137 on cell proliferation, migration, and invasion were determined using MTT assays, wound healing, and Matrigel transwell assays. The luciferase reporter assay revealed direct binding of miR-137 to the 3'-UTR of Del-1. miR-137 inhibited cell proliferation, migration, and invasion of MDA-MB-231 cells. Among the 30 TNBC specimens, miR-137 was downregulated and Del-1 level in plasma was significantly elevated relative to normal controls. It is concluded that miR-137 regulates Del-1 expression in TNBC by directly binding to the Del-1 gene and cancer progression. The results implicate miR-137 as a new therapeutic biomarker for patients with TNBC.
Collapse
|
38
|
Ruiz F, Vigne S, Pot C. Resolution of inflammation during multiple sclerosis. Semin Immunopathol 2019; 41:711-726. [PMID: 31732775 PMCID: PMC6881249 DOI: 10.1007/s00281-019-00765-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is a frequent autoimmune demyelinating disease of the central nervous system (CNS). There are three clinical forms described: relapsing-remitting multiple sclerosis (RRMS), the most common initial presentation (85%) among which, if not treated, about half will transform, into the secondary progressive multiple sclerosis (SPMS) and the primary progressive MS (PPMS) (15%) that is directly progressive without superimposed clinical relapses. Inflammation is present in all subsets of MS. The relapsing/remitting form could represent itself a particular interest for the study of inflammation resolution even though it remains incomplete in MS. Successful resolution of acute inflammation is a highly regulated process and dependent on mechanisms engaged early in the inflammatory response that are scarcely studied in MS. Moreover, recent classes of disease-modifying treatment (DMTs) that are effective against RRMS act by re-establishing the inflammatory imbalance, taking advantage of the pre-existing endogenous suppressor. In this review, we will discuss the active role of regulatory immune cells in inflammation resolution as well as the role of tissue and non-hematopoietic cells as contributors to inflammation resolution. Finally, we will explore how DMTs, more specifically induction therapies, impact the resolution of inflammation during MS.
Collapse
Affiliation(s)
- F Ruiz
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - S Vigne
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - C Pot
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland.
| |
Collapse
|
39
|
Yilmaz C, Karali K, Fodelianaki G, Gravanis A, Chavakis T, Charalampopoulos I, Alexaki VI. Neurosteroids as regulators of neuroinflammation. Front Neuroendocrinol 2019; 55:100788. [PMID: 31513776 DOI: 10.1016/j.yfrne.2019.100788] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/12/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023]
Abstract
Neuroinflammation is a physiological protective response in the context of infection and injury. However, neuroinflammation, especially if chronic, may also drive neurodegeneration. Neurodegenerative diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD) and traumatic brain injury (TBI), display inflammatory activation of microglia and astrocytes. Intriguingly, the central nervous system (CNS) is a highly steroidogenic environment synthesizing steroids de novo, as well as metabolizing steroids deriving from the circulation. Neurosteroid synthesis can be substantially affected by neuroinflammation, while, in turn, several steroids, such as 17β-estradiol, dehydroepiandrosterone (DHEA) and allopregnanolone, can regulate neuroinflammatory responses. Here, we review the role of neurosteroids in neuroinflammation in the context of MS, AD, PD and TBI and describe underlying molecular mechanisms. Moreover, we introduce the concept that synthetic neurosteroid analogues could be potentially utilized for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Canelif Yilmaz
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Kanelina Karali
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Georgia Fodelianaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
| |
Collapse
|
40
|
Kim WY, Lee SH, Kim DY, Ryu HJ, Chon GR, Park YY, Fu Y, Huh JW, Lim CM, Koh Y, Choi EY, Hong SB. Serum developmental endothelial locus-1 is associated with severity of sepsis in animals and humans. Sci Rep 2019; 9:13005. [PMID: 31506547 PMCID: PMC6737092 DOI: 10.1038/s41598-019-49564-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/27/2019] [Indexed: 12/16/2022] Open
Abstract
Disruption of the endothelial glycocalyx has a prominent role in the pathophysiology of sepsis. Developmental endothelial locus-1 (Del-1) is an endothelial-derived anti-inflammatory factor. We hypothesized that degradation of the endothelial glycocalyx during sepsis may increase serum Del-1. A mouse model of sepsis was created using cecal ligation and puncture. In septic mice, the endothelial glycocalyx was nearly completely degraded, with less formation of Del-1 in the endothelium and extracellular matrix than in control mice. Serum Del-1 levels were significantly increased in the septic mice with increasing severity of sepsis. Serum Del-1 levels were also measured in 84 patients with sepsis and septic shock and in 20 control subjects. The median serum Del-1 level in patients with sepsis was significantly higher than that in healthy controls. The high Del-1 group had higher illness severity scores and contained more patients with organ dysfunction than the low Del-1 group. The 90-day mortality rate was significantly higher in the high Del-1 group than in the low Del-1 group. Multivariate analysis indicated a tendency for a high serum Del-1 level to be associated with a higher mortality risk. Increased serum Del-1 may be a novel diagnostic biomarker of sepsis and an indicator of disease severity.
Collapse
Affiliation(s)
- Won-Young Kim
- Department of Internal Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Seung-Hwan Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Young Kim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyun Jin Ryu
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gyu Rak Chon
- Department of Internal Medicine, Cheongju St. Mary's Hospital, Cheongju, Republic of Korea
| | - Yun Young Park
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yan Fu
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Won Huh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chae-Man Lim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Younsuck Koh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Sang-Bum Hong
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
41
|
Hajishengallis G, Chavakis T. DEL-1-Regulated Immune Plasticity and Inflammatory Disorders. Trends Mol Med 2019; 25:444-459. [PMID: 30885428 DOI: 10.1016/j.molmed.2019.02.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/16/2022]
Abstract
In contrast to traditional immune cell-centered viewpoints, recent studies suggest that tissues are not passive recipients of immunity but have a 'regulatory say' over the host inflammatory response. Identification of tissue-derived homeostatic molecules regulating immune plasticity is seminal for understanding the inherent regulatory potential of different organs in the immune response. DEL-1 (developmental endothelial locus-1) is a secreted multidomain protein interacting with integrins and phospholipids and regulates, depending on its expression location, distinct stages of the host inflammatory response (from myelopoiesis over leukocyte recruitment to efferocytosis and resolution of inflammation). Here we synthesize recent evidence of DEL-1 as an exemplar local regulatory factor in the context of tissue immune plasticity and inflammatory disorders (such as periodontitis, multiple sclerosis, and pulmonary disorders), and discuss its potential as a therapeutic agent.
Collapse
Affiliation(s)
- George Hajishengallis
- Penn Dental Medicine, Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA.
| | - Triantafyllos Chavakis
- Faculty of Medicine, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
42
|
Zhu C, Li Z, Li B, Pfammatter M, Hornemann S, Aguzzi A. Unaltered prion disease in mice lacking developmental endothelial locus-1. Neurobiol Aging 2019; 76:208-213. [PMID: 30743056 DOI: 10.1016/j.neurobiolaging.2019.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 11/19/2022]
Abstract
Progression of prion diseases is driven by the accumulation of prions in the brain. Ablation of microglia or deletion of the eat-me-signal, milk-fat globule epidermal growth factor VIII (Mfge8), accelerates prion pathogenesis, suggesting that microglia defend the brain by phagocytosing prions. Similar to Mfge8, developmental endothelial locus-1 (Del-1) is a secreted protein that acts as an opsonin bridging phagocytes and apoptotic cells to facilitate phagocytosis. We therefore asked whether Del-1 might play a role in controlling prion pathogenesis. We assessed the anti-inflammatory and phagocytosis-promoting functions of Del-1 in prion disease and determined whether Del-1 complements Mfge8 in prion clearance in mice with a C57BL/6J genetic background. We found that Del-1 deficiency did not change prion disease progression or lesion patterns. In addition, prion clearance and scrapie prion protein deposition were unaltered in Del-1-deficient mice. In addition, prion-induced neuroinflammation was not affected by Del-1 deficiency. We conclude that Del-1 is not a major determinant of prion pathogenesis in this context.
Collapse
Affiliation(s)
- Caihong Zhu
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Zhihao Li
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Bei Li
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Manuela Pfammatter
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Simone Hornemann
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
43
|
DEL-1 promotes macrophage efferocytosis and clearance of inflammation. Nat Immunol 2018; 20:40-49. [PMID: 30455459 PMCID: PMC6291356 DOI: 10.1038/s41590-018-0249-1] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022]
Abstract
Resolution of inflammation is essential for tissue homeostasis and a promising approach to inflammatory disorders. Here we found that DEL-1, a secreted protein inhibiting leukocyte-endothelial adhesion and inflammation initiation, also functions as a non-redundant downstream effector in inflammation clearance. In human and murine periodontitis, waning of inflammation correlated with DEL-1 upregulation, whereas resolution of experimental periodontitis failed in DEL-1 deficiency. This concept was mechanistically substantiated in acute monosodium urate crystal-induced inflammation, where the pro-resolution function of DEL-1 was attributed to effective apoptotic neutrophil clearance (efferocytosis). DEL-1-mediated efferocytosis induced liver-X-receptor-dependent macrophage reprogramming to pro-resolving phenotype and was required for optimal production of at least certain specific pro-resolving mediators. Experiments in transgenic mice with cell-specific overexpression of DEL-1 linked its anti-leukocyte recruitment action to endothelial-derived DEL-1 and its efferocytic/pro-resolving action to macrophage-derived DEL-1. Thus, the compartmentalized expression of DEL-1 facilitates distinct homeostatic functions in an appropriate context that can be harnessed therapeutically.
Collapse
|
44
|
Hajishengallis G, Korostoff JM. Revisiting the Page & Schroeder model: the good, the bad and the unknowns in the periodontal host response 40 years later. Periodontol 2000 2018; 75:116-151. [PMID: 28758305 DOI: 10.1111/prd.12181] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In their classic 1976 paper, Page & Schroeder described the histopathologic events and the types of myeloid cells and lymphocytes involved in the initiation and progression of inflammatory periodontal disease. The staging of periodontal disease pathogenesis as 'initial', 'early', 'established' and 'advanced' lesions productively guided subsequent research in the field and remains fundamentally valid. However, major advances regarding the cellular and molecular mechanisms underlying the induction, regulation and effector functions of immune and inflammatory responses necessitate a reassessment of their work and its integration with emerging new concepts. We now know that each type of leukocyte is actually represented by functionally distinct subsets with different, or even conflicting, roles in immunity and inflammation. Unexpectedly, neutrophils, traditionally regarded as merely antimicrobial effectors in acute conditions and protagonists of the 'initial' lesion, are currently appreciated for their functional versatility and critical roles in chronic inflammation. Moreover, an entirely new field of study, osteoimmunology, has emerged and sheds light on the impact of immunoinflammatory events on the skeletal system. These developments and the molecular dissection of crosstalk interactions between innate and adaptive leukocytes, as well as between the immune system and local homeostatic mechanisms, offer a more nuanced understanding of the host response in periodontitis, with profound implications for treatment. At the same time, deeper insights have generated new questions, many of which remain unanswered. In this review, 40 years after Page & Schroeder proposed their model, we summarize enduring and emerging advances in periodontal disease pathogenesis.
Collapse
|
45
|
Fu Y, Tsauo J, Sun Y, Wang Z, Kim KY, Lee SH, Kim DY, Jing F, Lim D, Song HY, Hyun H, Choi EY. Developmental endothelial locus-1 prevents development of peritoneal adhesions in mice. Biochem Biophys Res Commun 2018; 500:783-789. [PMID: 29684347 DOI: 10.1016/j.bbrc.2018.04.158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/19/2018] [Indexed: 01/01/2023]
Abstract
Postoperative peritoneal adhesions, fibrous bands formed in the peritoneal cavity following surgery, represent a common, challenging and costly problem faced by surgeons and patients, for which effective therapeutic options are lacking. Since aberrant inflammation is one of the key mechanisms underlying peritoneal adhesion formation, here we set out to study the role of developmental endothelial locus-1 (Del-1), which has been recently identified as an endogenous inhibitor of inflammation, in the formation of postoperative peritoneal adhesions using a mouse model of peritoneal adhesions induced by ischemic buttons. Del-1-deficient mice had a higher incidence of adhesions, and their adhesions had higher quality and tenacity scores. Del-1 deficiency also led to enhanced inflammation mediators and collagen production. Finally, Del-1 supplementation decreased the incidence and severity of postoperative peritoneal adhesions. Taken together, these results indicate a protective role for Del-1 in postoperative peritoneal adhesion formation.
Collapse
Affiliation(s)
- Yan Fu
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jiaywei Tsauo
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Yucheng Sun
- Department of Orthopedic Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Zhe Wang
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Kun Yung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Seung-Hwan Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Young Kim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Feifeng Jing
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dahae Lim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ho-Young Song
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hoon Hyun
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea.
| | - Eun Young Choi
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
46
|
Yan S, Chen L, Zhao Q, Liu YN, Hou R, Yu J, Zhang H. Developmental endothelial locus-1 (Del-1) antagonizes Interleukin-17-mediated allergic asthma. Immunol Cell Biol 2018; 96:526-535. [PMID: 29437247 DOI: 10.1111/imcb.12023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 02/08/2018] [Accepted: 02/08/2018] [Indexed: 01/13/2023]
Abstract
Interleukin (IL)-17 is a major contributor to the pathogenesis of allergic asthma. Developmental endothelial locus-1 (Del-1) is an endothelial cell-secreted protein known to inhibit IL-17 expression. However, little is known about the association between Del-1 and IL-17 in the pathogenesis of allergic asthma. Using bronchoalveolar lavage fluid (BALF) and peripheral blood samples collected from allergic asthmatic patients and controls, we explored the role of Del-1 in relation to IL-17 in allergic asthma. We found that the negative correlation between Del-1 and IL-17 was significant in BALF of allergic asthmatics. Del-1 treatment inhibited the expression of IL-17, the differentiation of IL-17-secreting leukocytes and associated cytokines. Contrarily, IL-17 levels were increased after treatment with anti-Del-1 mAb. Consistent with this, Del-1 treatment led to downregulation of IL-5, CCL5 and IL-4, thus reducing secretion of eosinophil cationic protein. Furthermore, Del-1 significantly downregulated the expression of ICAM-1 and may have the potential to reduce leukocyte transendothelial migration. Our data demonstrate that Del-1 can negatively regulate IL-17 and its proinflammatory function, thereby limiting airway inflammation in allergic asthmatics, and suggest Del-1 as a potential candidate for prevention and treatment of allergic asthma.
Collapse
Affiliation(s)
- Shu Yan
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Li Chen
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Zhao
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ya-Nan Liu
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Hou
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Yu
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Zhang
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
47
|
Paetau S, Rolova T, Ning L, Gahmberg CG. Neuronal ICAM-5 Inhibits Microglia Adhesion and Phagocytosis and Promotes an Anti-inflammatory Response in LPS Stimulated Microglia. Front Mol Neurosci 2017; 10:431. [PMID: 29311819 PMCID: PMC5743933 DOI: 10.3389/fnmol.2017.00431] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/12/2017] [Indexed: 11/13/2022] Open
Abstract
The intercellular adhesion molecule-5 (ICAM-5) regulates neurite outgrowth and synaptic maturation. ICAM-5 overexpression in the hippocampal neurons induces filopodia formation in vitro. Since microglia are known to prune supernumerous synapses during development, we characterized the regulatory effect of ICAM-5 on microglia. ICAM-5 was released as a soluble protein from N-methyl-D-aspartic acid (NMDA)-treated neurons and bound by microglia. ICAM-5 promoted down-regulation of adhesion and phagocytosis in vitro. Microglia formed large cell clusters on ICAM-5-coated surfaces whereas they adhered and spread on the related molecule ICAM-1. ICAM-5 further reduced the secretion of the proinflammatory cytokines tumor necrosis factor α (TNF-α) and interleukin 1β (IL-1β), but on the contrary induced the secretion of the anti-inflammatory IL-10 from lipopolysaccharide (LPS) stimulated microglia. Thus, ICAM-5 might be involved in the regulation of microglia in both health and disease, playing an important neuroprotective role when the brain is under immune challenges and as a "don't-eat-me" signal when it is solubilized from active synapses.
Collapse
Affiliation(s)
- Sonja Paetau
- Laboratory of CG Gahmberg, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Taisia Rolova
- Laboratory of CG Gahmberg, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Lin Ning
- Laboratory of CG Gahmberg, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Carl G Gahmberg
- Laboratory of CG Gahmberg, Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
48
|
Mitroulis I, Chen LS, Singh RP, Kourtzelis I, Economopoulou M, Kajikawa T, Troullinaki M, Ziogas A, Ruppova K, Hosur K, Maekawa T, Wang B, Subramanian P, Tonn T, Verginis P, von Bonin M, Wobus M, Bornhäuser M, Grinenko T, Di Scala M, Hidalgo A, Wielockx B, Hajishengallis G, Chavakis T. Secreted protein Del-1 regulates myelopoiesis in the hematopoietic stem cell niche. J Clin Invest 2017; 127:3624-3639. [PMID: 28846069 DOI: 10.1172/jci92571] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 07/11/2017] [Indexed: 12/16/2022] Open
Abstract
Hematopoietic stem cells (HSCs) remain mostly quiescent under steady-state conditions but switch to a proliferative state following hematopoietic stress, e.g., bone marrow (BM) injury, transplantation, or systemic infection and inflammation. The homeostatic balance between quiescence, self-renewal, and differentiation of HSCs is strongly dependent on their interactions with cells that constitute a specialized microanatomical environment in the BM known as the HSC niche. Here, we identified the secreted extracellular matrix protein Del-1 as a component and regulator of the HSC niche. Specifically, we found that Del-1 was expressed by several cellular components of the HSC niche, including arteriolar endothelial cells, CXCL12-abundant reticular (CAR) cells, and cells of the osteoblastic lineage. Del-1 promoted critical functions of the HSC niche, as it regulated long-term HSC (LT-HSC) proliferation and differentiation toward the myeloid lineage. Del-1 deficiency in mice resulted in reduced LT-HSC proliferation and infringed preferentially upon myelopoiesis under both steady-state and stressful conditions, such as hematopoietic cell transplantation and G-CSF- or inflammation-induced stress myelopoiesis. Del-1-induced HSC proliferation and myeloid lineage commitment were mediated by β3 integrin on hematopoietic progenitors. This hitherto unknown Del-1 function in the HSC niche represents a juxtacrine homeostatic adaptation of the hematopoietic system in stress myelopoiesis.
Collapse
Affiliation(s)
- Ioannis Mitroulis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Lan-Sun Chen
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Rashim Pal Singh
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Ioannis Kourtzelis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Matina Economopoulou
- Department of Ophthalmology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Tetsuhiro Kajikawa
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maria Troullinaki
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Athanasios Ziogas
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Klara Ruppova
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Kavita Hosur
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tomoki Maekawa
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Baomei Wang
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Pallavi Subramanian
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Torsten Tonn
- Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Panayotis Verginis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and.,Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Malte von Bonin
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Manja Wobus
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Martin Bornhäuser
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Tatyana Grinenko
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Marianna Di Scala
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
| | - Andres Hidalgo
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain.,Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ben Wielockx
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and.,Center for Regenerative Therapies Dresden, Dresden, Germany
| | - George Hajishengallis
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and.,Center for Regenerative Therapies Dresden, Dresden, Germany
| |
Collapse
|
49
|
Folwaczny M, Karnesi E, Berger T, Paschos E. Clinical association between chronic periodontitis and the leukocyte extravasation inhibitors developmental endothelial locus-1 and pentraxin-3. Eur J Oral Sci 2017. [PMID: 28643381 DOI: 10.1111/eos.12357] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This clinical study aimed to determine whether periodontal disease is associated with expression of developmental endothelial locus-1 (Del-1) and pentraxin-3 (PTX-3), endogenous inhibitors of leukocyte extravasation in humans. Expression of DEL1, PTX3, interleukin-17A (IL17A), and lymphocyte function-associated antigen-1 (LFA1) was determined, using RT-PCR and melting curve analysis, in biopsies of gingival tissues from 95 patients: 42 with moderate periodontitis; 40 with severe periodontitis; and 13 healthy controls. Relative expression of DEL1 and PTX3 was statistically significantly weaker in patients with periodontitis than in the control subjects. On the contrary, both IL17A and LFA1 showed statistically significant stronger expression in patients with periodontitis than in healthy controls. Correlation analysis, performed using Spearman's test, showed that expression of DEL1 was statistically significantly linked to periodontitis (ρ = -0.103) and to age (ρ = -0.134), but not to the gender of the patient, and that expression of PTX3 was significantly correlated with periodontitis (ρ = -0.354). Expression of neutrophil extravasation inhibitors DEL1 and PTX3 show significant, but weak, association with the clinical manifestation of chronic periodontitis.
Collapse
Affiliation(s)
- Matthias Folwaczny
- Department of Operative Dentistry and Periodontology, Ludwig-Maximilians-University, Munich, Germany
| | - Evangelia Karnesi
- Department of Operative Dentistry and Periodontology, Ludwig-Maximilians-University, Munich, Germany
| | - Tamara Berger
- Department of Operative Dentistry and Periodontology, Ludwig-Maximilians-University, Munich, Germany
| | - Ekaterini Paschos
- Department of Orthodontics, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
50
|
Klotzsche-von Ameln A, Cremer S, Hoffmann J, Schuster P, Khedr S, Korovina I, Troullinaki M, Neuwirth A, Sprott D, Chatzigeorgiou A, Economopoulou M, Orlandi A, Hain A, Zeiher AM, Deussen A, Hajishengallis G, Dimmeler S, Chavakis T, Chavakis E. Endogenous developmental endothelial locus-1 limits ischaemia-related angiogenesis by blocking inflammation. Thromb Haemost 2017; 117:1150-1163. [PMID: 28447099 DOI: 10.1160/th16-05-0354] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 03/05/2017] [Indexed: 12/23/2022]
Abstract
We have recently identified endothelial cell-secreted developmental endothelial locus-1 (Del-1) as an endogenous inhibitor of β2-integrin-dependent leukocyte infiltration. Del-1 was previously also implicated in angiogenesis. Here, we addressed the role of endogenously produced Del-1 in ischaemia-related angiogenesis. Intriguingly, Del-1-deficient mice displayed increased neovascularisation in two independent ischaemic models (retinopathy of prematurity and hind-limb ischaemia), as compared to Del-1-proficient mice. On the contrary, angiogenic sprouting in vitro or ex vivo (aortic ring assay) and physiological developmental retina angiogenesis were not affected by Del-1 deficiency. Mechanistically, the enhanced ischaemic neovascularisation in Del-1-deficiency was linked to higher infiltration of the ischaemic tissue by CD45+ haematopoietic and immune cells. Moreover, Del-1-deficiency promoted β2-integrin-dependent adhesion of haematopoietic cells to endothelial cells in vitro, and the homing of hematopoietic progenitor cells and of immune cell populations to ischaemic muscles in vivo. Consistently, the increased hind limb ischaemia-related angiogenesis in Del-1 deficiency was completely reversed in mice lacking both Del-1 and the β2-integrin LFA-1. Additionally, enhanced retinopathy-associated neovascularisation in Del-1-deficient mice was reversed by LFA-1 blockade. Our data reveal a hitherto unrecognised function of endogenous Del-1 as a local inhibitor of ischaemia-induced angiogenesis by restraining LFA-1-dependent homing of pro-angiogenic haematopoietic cells to ischaemic tissues. Our findings are relevant for the optimisation of therapeutic approaches in the context of ischaemic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Emmanouil Chavakis
- Emmanouil Chavakis, MD, Dept. of Internal Medicine III, Goethe University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany, Tel.: +49 69 6301 4131, +49 69 6301 87965, Fax: +49 69 6301 83462, E-mail:
| |
Collapse
|