1
|
Li F, Bai Y, Guan Z, Ji X, Zhan X, Gao Y, Zhong W, Rao Z. Dexmedetomidine attenuates sepsis-associated acute lung injury by regulating macrophage efferocytosis through the ROS/ADAM10/AXL pathway. Int Immunopharmacol 2024; 142:112832. [PMID: 39362816 DOI: 10.1016/j.intimp.2024.112832] [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: 01/21/2024] [Revised: 03/20/2024] [Accepted: 07/26/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND The lungs are highly susceptible to damage during sepsis, with severe lung injury potentially progressing to acute respiratory distress syndrome and even fatal sepsis. Effective efferocytosis of apoptotic cells is crucial in alleviating inflammation and tissue injury. METHODS We established a septic lung injury mouse model via intraperitoneal injection of lipopolysaccharide. Lung injury was assessed by histology, immunofluorescence, neutrophil immunohistochemistry staining, and cytokine detection. We extracted alveolar macrophages by bronchoalveolar lavage and primary macrophages from mouse bone marrow to investigate the regulatory effects of Dexmedetomidine (DEX) on efferocytosis. We further validated the molecular mechanisms underlying the regulation of macrophage efferocytosis by DEX through knockdown of AXL expression. Additionally, we examined the efferocytic ability of monocytes isolated from patients. RESULTS We discovered that DEX treatment effectively alleviated pulmonary injury and inflammation. Lipopolysaccharide reduced macrophage efferocytosis and AXL expression which were reversed by DEX. We also found DEX inhibited the increased activation of A Disintegrin And Metalloproteinase 10 (ADAM10) and the production of soluble AXL. Moreover, our findings demonstrated that DEX decreased the elevated ROS production linked to higher ADAM10 activation. Blocking AXL negated DEX's benefits on efferocytosis and lung protection. Efferocytosis in monocytes from septic lung injury patients was notably lower than in healthy individuals. CONCLUSION Our findings demonstrated that DEX treatment effectively reduces septic lung injury by promoting macrophage efferocytosis through ROS/ADAM10/AXL signaling pathwway.
Collapse
Affiliation(s)
- Fei Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, China; Department of Anesthesiology, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Yan Bai
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, China
| | - Zhu Guan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, China
| | - Xingyue Ji
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, China
| | - Xinyu Zhan
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), 210029 Nanjing, China
| | - Yiyun Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), 210029 Nanjing, China
| | - Weizhe Zhong
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), 210029 Nanjing, China.
| | - Zhuqing Rao
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, China.
| |
Collapse
|
2
|
Gharzia FG, Aljohmani A, Beck A, Philipp SE, Yildiz D. Regulation of ADAM10 activity through microdomain-dependent intracellular calcium changes. Cell Commun Signal 2024; 22:531. [PMID: 39497138 PMCID: PMC11533308 DOI: 10.1186/s12964-024-01891-5] [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: 06/11/2024] [Accepted: 10/12/2024] [Indexed: 11/06/2024] Open
Abstract
A disintegrin and metalloproteinases (ADAMs) are transmembrane proteases that cleave other proteins close to the surface in a process called shedding. The prominent member ADAM10 has been linked to several pathologies such as Alzheimer's disease, bacterial infection, cancer development and metastasis. Although the regulation of the ADAM10 activity by calcium influx and calmodulin inhibition has been reported, the spatiotemporal regulation of Ca2+-dependent ADAM10 activation and the required source of Ca2+ ions have not been thoroughly studied. In the present study, we observed the rapid Ca2+-dependent activation of ADAM10 in A549 lung carcinoma cells upon stimulation with ionomycin. The calmodulin-inhibitors trifluoperazine and ophiobolin A mediated delayed activation of ADAM10, which apparently did not depend on intracellular Ca2+ in the case of trifluoperazine. Furthermore, the surface translocation and release of ADAM10 in extracellular vesicles exhibited different kinetics and were only partially linked to catalytic activation. Finally, ADAM10 activation was observed after the entry of Ca2+ through certain channels, such as canonical members of transient receptor potential (TRP) channels. Therefore, the opening of particular channels for Ca2+ entry points and subsequent Ca2+ flux as well as the temporal aspects of the consequent increase in Ca2+ levels, must be considered for future therapeutic options involving the increasing or decreasing ADAM10 activity.
Collapse
Affiliation(s)
| | - Ahmad Aljohmani
- Molecular Pharmacology, PZMS, Saarland University, Campus Homburg Building 46, 66421, Homburg, Germany
| | - Andreas Beck
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, Homburg, Germany
| | - Stephan E Philipp
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, Homburg, Germany
| | - Daniela Yildiz
- Molecular Pharmacology, PZMS, Saarland University, Campus Homburg Building 46, 66421, Homburg, Germany.
| |
Collapse
|
3
|
Abdalla AM, Miao Y, Ming N, Ouyang C. ADAM10 modulates the efficacy of T-cell-mediated therapy in solid tumors. Immunol Cell Biol 2024; 102:907-923. [PMID: 39417304 DOI: 10.1111/imcb.12826] [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: 04/23/2024] [Revised: 08/15/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
T-cell-mediated therapeutic strategies are the most potent effectors of cancer immunotherapy. However, an essential barrier to this therapy in solid tumors is disrupting the anti-cancer immune response, cancer-immunity cycle, T-cell priming, trafficking and T-cell cytotoxic capacity. Thus, reinforcing the anti-cancer immune response is needed to improve the effectiveness of T-cell-mediated therapy. Tumor-associated protease ADAM10, endothelial cells (ECs) and cytotoxic CD8+ T cells engage in complex communication via adhesion, transmigration and chemotactic mechanisms to facilitate an anti-cancer immune response. The precise impact of ADAM10 on the intricate mechanisms underlying these interactions remains unclear. This paper broadly explores how ADAM10, through different routes, influences the efficacy of T-cell-mediated therapy. ADAM10 cleaves CD8+ T-cell-targeting genes and impacts their expression and specificity. In addition, ADAM10 mediates the interactions of adhesion molecules with T cells and influences CD8+ T-cell activity and trafficking. Thus, understanding the role of ADAM10 in these events may lead to innovative strategies for advancing T-cell-mediated therapies.
Collapse
Affiliation(s)
- Ahmed Me Abdalla
- School of Biological Sciences and Technology, University of Jinan, Jinan, China
- Department of Biochemistry, College of Applied Science, University of Bahri, Khartoum, Sudan
| | - Yu Miao
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, Gansu, China
- Department of Phase 1 Clinical and Research Ward, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Ning Ming
- School of Biological Sciences and Technology, University of Jinan, Jinan, China
| | - Chenxi Ouyang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Noera G, Bertolini A, Calzà L, Gori M, Pitino A, D'Arrigo G, Egan CG, Tripepi G. Effect of early administration of tetracosactide on mortality and host response in critically ill patients requiring rescue surgery: a sensitivity analysis of the STOPSHOCK phase 3 randomized controlled trial. Mil Med Res 2024; 11:56. [PMID: 39160574 PMCID: PMC11331742 DOI: 10.1186/s40779-024-00555-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 07/12/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Undifferentiated shock is recognized as a criticality state that is transitional in immune-mediated topology for casual risk of lethal microcirculatory dysfunction. This was a sensitivity analysis of a drug (tetracosactide; TCS10) targeting melanocortin receptors (MCRs) in a phase 3 randomized controlled trial to improve cardiovascular surgical rescue outcome by reversing mortality and hemostatic disorders. METHODS Sensitivity analysis was based on a randomized, two-arm, multicenter, double-blind, controlled trial. The Naïve Bayes classifier was performed by density-based sensitivity index for principal strata as proportional hazard model of 30-day surgical risk mortality according to European System for Cardiac Operative Risk Evaluation inputs-outputs in 100 consecutive cases (from August to September 2013 from Emilia Romagna region, Italy). Patients included an agent-based TCS10 group (10 mg, single intravenous bolus before surgery; n = 56) and control group (n = 44) and the association with cytokines, lactate, and bleeding-blood transfusion episodes with the prior-risk log-odds for mortality rate in time-to-event was analyzed. RESULTS Thirty-day mortality was significantly improved in the TCS10 group vs. control group (0 vs. 8 deaths, P < 0.0001). Baseline levels of interleukin (IL)-6, IL-10, and lactate were associated with bleeding episodes, independent of TCS10 treatment [odds ratio (OR) = 1.90, 95% confidence interval (CI) 1.39-2.79; OR = 1.53, 95%CI 1.17-2.12; and OR = 2.92, 95%CI 1.40-6.66, respectively], while baseline level of Fms-like tyrosine kinase 3 ligand (Flt3L) was associated with lower bleeding rates in TCS10-treated patients (OR = 0.31, 95%CI 0.11-0.90, P = 0.03). For every 8 TCS10-treated patients, 1 bleeding case was avoided. Blood transfusion episodes were significantly reduced in the TCS10 group compared to the control group (OR = 0.32, 95%CI 0.14-0.73, P = 0.01). For every 4 TCS10-treated patients, 1 transfusion case was avoided. CONCLUSIONS Sensitivity index underlines the quality target product profile of TCS10 in the runway of emergency casualty care. To introduce the technology readiness level in real-life critically ill patients, further large-scale studies are required. TRIAL REGISTRATION European Union Drug Regulating Authorities Clinical Trials Database (EudraCT Number: 2007-006445-41 ).
Collapse
Affiliation(s)
- Giorgio Noera
- Health Ricerca e Sviluppo, Global Contractor for STOPSHOCK National Plan of Military Research Ministry of Defence, Rome, 00187, Italy.
| | - Alfio Bertolini
- Department of Medicine and Division of Clinical Pharmacology, School of Medicine, UNIMORE, Policlinico, Modena, 41124, Italy
| | - Laura Calzà
- IRET Foundation, Ozzano Dell' Emilia, Bologna, 40064, Italy
| | - Mercedes Gori
- Institute of Clinical Physiology (IFC-CNR), Section of Rome, Rome, 00185, Italy
| | - Annalisa Pitino
- Institute of Clinical Physiology (IFC-CNR), Section of Rome, Rome, 00185, Italy
| | - Graziella D'Arrigo
- National Research Council-Institute of Clinical Physiology, Reggio Calabria, 89124, Italy
| | | | - Giovanni Tripepi
- National Research Council-Institute of Clinical Physiology, Reggio Calabria, 89124, Italy
| |
Collapse
|
5
|
Babendreyer A, Kieselhorst J, Rinkens C, Lyashenko AM, Düsterhöft S, Jahr H, Craveiro RB, Wolf M, Ludwig A. Downregulation of the metalloproteinases ADAM10 or ADAM17 promotes osteoclast differentiation. Cell Commun Signal 2024; 22:322. [PMID: 38863060 PMCID: PMC11167776 DOI: 10.1186/s12964-024-01690-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: 03/27/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Bone resorption is driven through osteoclast differentiation by macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-Β ligand (RANKL). We noted that a disintegrin and metalloproteinase (ADAM) 10 and ADAM17 are downregulated at the expression level during osteoclast differentiation of the murine monocytic cell line RAW264.7 in response to RANKL. Both proteinases are well known to shed a variety of single-pass transmembrane molecules from the cell surface. We further showed that inhibitors of ADAM10 or ADAM17 promote osteoclastic differentiation and furthermore enhance the surface expression of receptors for RANKL and M-CSF on RAW264.7 cells. Using murine bone marrow-derived monocytic cells (BMDMCs), we demonstrated that a genetic deficiency of ADAM17 or its required regulator iRhom2 leads to increased osteoclast development in response to M-CSF and RANKL stimulation. Moreover, ADAM17-deficient osteoclast precursor cells express increased levels of the receptors for RANKL and M-CSF. Thus, ADAM17 negatively regulates osteoclast differentiation, most likely through shedding of these receptors. To assess the time-dependent contribution of ADAM10, we blocked this proteinase by adding a specific inhibitor on day 0 of BMDMC stimulation with M-CSF or on day 7 of subsequent stimulation with RANKL. Only ADAM10 inhibition beginning on day 7 increased the size of developing osteoclasts indicating that ADAM10 suppresses osteoclast differentiation at a later stage. Finally, we could confirm our findings in human peripheral blood mononuclear cells (PBMCs). Thus, downregulation of either ADAM10 or ADAM17 during osteoclast differentiation may represent a novel regulatory mechanism to enhance their differentiation process. Enhanced bone resorption is a critical issue in osteoporosis and is driven through osteoclast differentiation by specific osteogenic mediators. The present study demonstrated that the metalloproteinases ADAM17 and ADAM10 critically suppress osteoclast development. This was observed for a murine cell line, for isolated murine bone marrow cells and for human blood cells by either preferential inhibition of the proteinases or by gene knockout. As a possible mechanism, we studied the surface expression of critical receptors for osteogenic mediators on developing osteoclasts. Our findings revealed that the suppressive effects of ADAM17 and ADAM10 on osteoclastogenesis can be explained in part by the proteolytic cleavage of surface receptors by ADAM10 and ADAM17, which reduces the sensitivity of these cells to osteogenic mediators. We also observed that osteoclast differentiation was associated with the downregulation of ADAM10 and ADAM17, which reduced their suppressive effects. We therefore propose that this downregulation serves as a feedback loop for enhancing osteoclast development.
Collapse
Affiliation(s)
- Aaron Babendreyer
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Julia Kieselhorst
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Cindy Rinkens
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Anastasia M Lyashenko
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Holger Jahr
- Institute of Anatomy and Cell Biology, University Hospital RWTH Aachen, Aachen, Germany
- Institute of Structural Mechanics and Lightweight Design, RWTH Aachen University, Aachen, Germany
| | - Rogerio B Craveiro
- Department of Orthodontics, University Hospital RWTH Aachen, Aachen, Germany
| | - Michael Wolf
- Department of Orthodontics, University Hospital RWTH Aachen, Aachen, Germany
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany.
| |
Collapse
|
6
|
Cheetham CJ, McKelvey MC, McAuley DF, Taggart CC. Neutrophil-Derived Proteases in Lung Inflammation: Old Players and New Prospects. Int J Mol Sci 2024; 25:5492. [PMID: 38791530 PMCID: PMC11122108 DOI: 10.3390/ijms25105492] [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: 04/23/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Neutrophil-derived proteases are critical to the pathology of many inflammatory lung diseases, both chronic and acute. These abundant enzymes play roles in key neutrophil functions, such as neutrophil extracellular trap formation and reactive oxygen species release. They may also be released, inducing tissue damage and loss of tissue function. Historically, the neutrophil serine proteases (NSPs) have been the main subject of neutrophil protease research. Despite highly promising cell-based and animal model work, clinical trials involving the inhibition of NSPs have shown mixed results in lung disease patients. As such, the cutting edge of neutrophil-derived protease research has shifted to proteases that have had little-to-no research in neutrophils to date. These include the cysteine and serine cathepsins, the metzincins and the calpains, among others. This review aims to outline the previous work carried out on NSPs, including the shortcomings of some of the inhibitor-orientated clinical trials. Our growing understanding of other proteases involved in neutrophil function and neutrophilic lung inflammation will then be discussed. Additionally, the potential of targeting these more obscure neutrophil proteases will be highlighted, as they may represent new targets for inhibitor-based treatments of neutrophil-mediated lung inflammation.
Collapse
Affiliation(s)
- Coby J. Cheetham
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK;
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| |
Collapse
|
7
|
Maas SL, Donners MMPC, van der Vorst EPC. ADAM10 and ADAM17, Major Regulators of Chronic Kidney Disease Induced Atherosclerosis? Int J Mol Sci 2023; 24:ijms24087309. [PMID: 37108478 PMCID: PMC10139114 DOI: 10.3390/ijms24087309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Chronic kidney disease (CKD) is a major health problem, affecting millions of people worldwide, in particular hypertensive and diabetic patients. CKD patients suffer from significantly increased cardiovascular disease (CVD) morbidity and mortality, mainly due to accelerated atherosclerosis development. Indeed, CKD not only affects the kidneys, in which injury and maladaptive repair processes lead to local inflammation and fibrosis, but also causes systemic inflammation and altered mineral bone metabolism leading to vascular dysfunction, calcification, and thus, accelerated atherosclerosis. Although CKD and CVD individually have been extensively studied, relatively little research has studied the link between both diseases. This narrative review focuses on the role of a disintegrin and metalloproteases (ADAM) 10 and ADAM17 in CKD and CVD and will for the first time shed light on their role in CKD-induced CVD. By cleaving cell surface molecules, these enzymes regulate not only cellular sensitivity to their micro-environment (in case of receptor cleavage), but also release soluble ectodomains that can exert agonistic or antagonistic functions, both locally and systemically. Although the cell-specific roles of ADAM10 and ADAM17 in CVD, and to a lesser extent in CKD, have been explored, their impact on CKD-induced CVD is likely, yet remains to be elucidated.
Collapse
Affiliation(s)
- Sanne L Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Marjo M P C Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), 80336 Munich, Germany
| |
Collapse
|
8
|
Liao S, Lin Y, Liu L, Yang S, Lin Y, He J, Shao Y. ADAM10-a "multitasker" in sepsis: focus on its posttranslational target. Inflamm Res 2023; 72:395-423. [PMID: 36565333 PMCID: PMC9789377 DOI: 10.1007/s00011-022-01673-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 07/25/2022] [Accepted: 11/30/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Sepsis has a complex pathogenesis in which the uncontrolled systemic inflammatory response triggered by infection leads to vascular barrier disruption, microcirculation dysfunction and multiple organ dysfunction syndrome. Numerous recent studies reveal that a disintegrin and metalloproteinase 10 (ADAM10) acts as a "molecular scissor" playing a pivotal role in the inflammatory response during sepsis by regulating proteolysis by cleaving various membrane protein substrates, including proinflammatory cytokines, cadherins and Notch, which are involved in intercellular communication. ADAM10 can also act as the cellular receptor for Staphylococcus aureus α-toxin, leading to lethal sepsis. However, its substrate-specific modulation and precise targets in sepsis have not yet to be elucidated. METHODS We performed a computer-based online search using PubMed and Google Scholar for published articles concerning ADAM10 and sepsis. CONCLUSIONS In this review, we focus on the functions of ADAM10 in sepsis-related complex endothelium-immune cell interactions and microcirculation dysfunction through the diversity of its substrates and its enzymatic activity. In addition, we highlight the posttranslational mechanisms of ADAM10 at specific subcellular sites, or in multimolecular complexes, which will provide the insight to intervene in the pathophysiological process of sepsis caused by ADAM10 dysregulation.
Collapse
Affiliation(s)
- Shuanglin Liao
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
| | - Yao Lin
- The Key Laboratory of Organ Dysfunction and Protection Translational Medicine, Jieyang Medical Research Center, Jieyang People’s Hospital, Tianfu Road 107, Rongcheng District, Jieyang, 522000 Guangdong China
| | - Lizhen Liu
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
| | - Shuai Yang
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
| | - YingYing Lin
- The Key Laboratory of Organ Dysfunction and Protection Translational Medicine, Jieyang Medical Research Center, Jieyang People’s Hospital, Tianfu Road 107, Rongcheng District, Jieyang, 522000 Guangdong China
| | - Junbing He
- The Key Laboratory of Organ Dysfunction and Protection Translational Medicine, Jieyang Medical Research Center, Jieyang People’s Hospital, Tianfu Road 107, Rongcheng District, Jieyang, 522000 Guangdong China
| | - Yiming Shao
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
- grid.410560.60000 0004 1760 3078The Key Laboratory of Sepsis Translational Medicine, Guangdong Medical University, Zhanjiang, Guangdong China
| |
Collapse
|
9
|
Sharma D, Singh NK. The Biochemistry and Physiology of A Disintegrin and Metalloproteinases (ADAMs and ADAM-TSs) in Human Pathologies. Rev Physiol Biochem Pharmacol 2023; 184:69-120. [PMID: 35061104 DOI: 10.1007/112_2021_67] [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] [Indexed: 02/07/2023]
Abstract
Metalloproteinases are a group of proteinases that plays a substantial role in extracellular matrix remodeling and its molecular signaling. Among these metalloproteinases, ADAMs (a disintegrin and metalloproteinases) and ADAM-TSs (ADAMs with thrombospondin domains) have emerged as highly efficient contributors mediating proteolytic processing of various signaling molecules. ADAMs are transmembrane metalloenzymes that facilitate the extracellular domain shedding of membrane-anchored proteins, cytokines, growth factors, ligands, and their receptors and therefore modulate their biological functions. ADAM-TSs are secretory, and soluble extracellular proteinases that mediate the cleavage of non-fibrillar extracellular matrix proteins. ADAMs and ADAM-TSs possess pro-domain, metalloproteinase, disintegrin, and cysteine-rich domains in common, but ADAM-TSs have characteristic thrombospondin motifs instead of the transmembrane domain. Most ADAMs and ADAM-TSs are activated by cleavage of pro-domain via pro-protein convertases at their N-terminus, hence directing them to various signaling pathways. In this article, we are discussing not only the structure and regulation of ADAMs and ADAM-TSs, but also the importance of these metalloproteinases in various human pathophysiological conditions like cardiovascular diseases, colorectal cancer, autoinflammatory diseases (sepsis/rheumatoid arthritis), Alzheimer's disease, proliferative retinopathies, and infectious diseases. Therefore, based on the emerging role of ADAMs and ADAM-TSs in various human pathologies, as summarized in this review, these metalloproteases can be considered as critical therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Deepti Sharma
- Department of Ophthalmology, Visual and Anatomical Sciences, Integrative Biosciences Center (IBio), Wayne State University School of Medicine, Detroit, MI, USA
| | - Nikhlesh K Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, Integrative Biosciences Center (IBio), Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
10
|
Klapproth E, Witt A, Klose P, Wiedemann J, Vavilthota N, Künzel SR, Kämmerer S, Günscht M, Sprott D, Lesche M, Rost F, Dahl A, Rauch E, Kattner L, Weber S, Mirtschink P, Kopaliani I, Guan K, Lorenz K, Saftig P, Wagner M, El-Armouche A. Targeting cardiomyocyte ADAM10 ectodomain shedding promotes survival early after myocardial infarction. Nat Commun 2022; 13:7648. [PMID: 36496449 PMCID: PMC9741599 DOI: 10.1038/s41467-022-35331-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
After myocardial infarction the innate immune response is pivotal in clearing of tissue debris as well as scar formation, but exaggerated cytokine and chemokine secretion with subsequent leukocyte infiltration also leads to further tissue damage. Here, we address the value of targeting a previously unknown a disintegrin and metalloprotease 10 (ADAM10)/CX3CL1 axis in the regulation of neutrophil recruitment early after MI. We show that myocardial ADAM10 is distinctly upregulated in myocardial biopsies from patients with ischemia-driven cardiomyopathy. Intriguingly, upon MI in mice, pharmacological ADAM10 inhibition as well as genetic cardiomycyte-specific ADAM10 deletion improves survival with markedly enhanced heart function and reduced scar size. Mechanistically, abolished ADAM10-mediated CX3CL1 ectodomain shedding leads to diminished IL-1β-dependent inflammation, reduced neutrophil bone marrow egress as well as myocardial tissue infiltration. Thus, our data shows a conceptual insight into how acute MI induces chemotactic signaling via ectodomain shedding in cardiomyocytes.
Collapse
Affiliation(s)
- Erik Klapproth
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anke Witt
- grid.4488.00000 0001 2111 7257Department of Physiology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Pauline Klose
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Johanna Wiedemann
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nikitha Vavilthota
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stephan R. Künzel
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Susanne Kämmerer
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mario Günscht
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - David Sprott
- grid.4488.00000 0001 2111 7257Department of Physiology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mathias Lesche
- grid.4488.00000 0001 2111 7257DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Fabian Rost
- grid.4488.00000 0001 2111 7257DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Andreas Dahl
- grid.4488.00000 0001 2111 7257DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | | | | | - Silvio Weber
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Peter Mirtschink
- grid.4488.00000 0001 2111 7257Institute of Clinical Chemistry and Laboratory Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Irakli Kopaliani
- grid.4488.00000 0001 2111 7257Department of Physiology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kaomei Guan
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kristina Lorenz
- grid.8379.50000 0001 1958 8658Institute of Pharmacology and Toxicology, Julius-Maximilians-University of Würzburg, Würzburg, Germany ,grid.419243.90000 0004 0492 9407Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany
| | - Paul Saftig
- grid.9764.c0000 0001 2153 9986Biochemical Institute, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Michael Wagner
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany ,grid.4488.00000 0001 2111 7257Rhythmology, Clinic of Internal Medicine and Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Ali El-Armouche
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
11
|
The Autism Spectrum Disorder-Associated Bacterial Metabolite p-Cresol Derails the Neuroimmune Response of Microglial Cells Partially via Reduction of ADAM17 and ADAM10. Int J Mol Sci 2022; 23:ijms231911013. [PMID: 36232346 PMCID: PMC9570133 DOI: 10.3390/ijms231911013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
The bacterial metabolite 4-methylphenol (para-cresol or p-cresol) and its derivative p-cresyl sulfate (pCS) are elevated in the urine and feces of children with autism spectrum disorder (ASD). It has been shown that p-cresol administration induces social behavior deficits and repetitive behavior in mice. However, the mechanisms of p-cresol, specifically its metabolite pCS that can reach the brain, in ASD remain to be investigated. The pCS has been shown to inhibit LPS-stimulated inflammatory response. A Disintegrin And Metalloprotease 10 (ADAM10) and A Disintegrin And Metalloprotease 17 (ADAM17) are thought to regulate microglial immune response by cleaving membrane-bound proteins. In the present study, a neuroinflammation model of LPS-activated BV2 microglia has been used to unveil the potential molecular mechanism of pCS in ASD pathogenesis. In microglial cells pCS treatment decreases the expression or maturation of ADAM10 and ADAM17. In addition, pCS treatment attenuates TNF-α and IL-6 releases as well as phagocytosis activity of microglia. In in vitro ADAM10/17 inhibition experiments, either ADAM10 or ADAM17 inhibition reduces constitutive and LPS-activated release of TNF-α, TNFR-1 and IL-6R by microglial cells, while it increases constitutive and LPS-activated microglial phagocytotic activity. The in vivo results further confirm the involvement of ADAM10 and ADAM17 in ASD pathogenesis. In in utero VPA-exposed male mice, elevated concentration in serum of p-cresol-associated metabolites pCS and p-cresyl glucuronide (pCG) is associated with a VPA-induced increased ADAM10 maturation, and a decreased ADAM17 maturation that is related with attenuated levels of soluble TNF-α and TGF-β1 in the mice brain. Overall, the present study demonstrates a partial role of ADAM10 and ADAM17 in the derailed innate immune response of microglial cells associated with pCS-induced ASD pathogenesis.
Collapse
|
12
|
Bienstein M, Minond D, Schwaneberg U, Davari MD, Yildiz D. In Silico and Experimental ADAM17 Kinetic Modeling as Basis for Future Screening System for Modulators. Int J Mol Sci 2022; 23:1368. [PMID: 35163294 PMCID: PMC8835787 DOI: 10.3390/ijms23031368] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/12/2022] [Accepted: 01/23/2022] [Indexed: 11/21/2022] Open
Abstract
Understanding the mechanisms of modulators' action on enzymes is crucial for optimizing and designing pharmaceutical substances. The acute inflammatory response, in particular, is regulated mainly by a disintegrin and metalloproteinase (ADAM) 17. ADAM17 processes several disease mediators such as TNFα and APP, releasing their soluble ectodomains (shedding). A malfunction of this process leads to a disturbed inflammatory response. Chemical protease inhibitors such as TAPI-1 were used in the past to inhibit ADAM17 proteolytic activity. However, due to ADAM17's broad expression and activity profile, the development of active-site-directed ADAM17 inhibitor was discontinued. New 'exosite' (secondary substrate binding site) inhibitors with substrate selectivity raised the hope of a substrate-selective modulation as a promising approach for inflammatory disease therapy. This work aimed to develop a high-throughput screen for potential ADAM17 modulators as therapeutic drugs. By combining experimental and in silico methods (structural modeling and docking), we modeled the kinetics of ADAM17 inhibitor. The results explain ADAM17 inhibition mechanisms and give a methodology for studying selective inhibition towards the design of pharmaceutical substances with higher selectivity.
Collapse
Affiliation(s)
- Marian Bienstein
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany; (M.B.); (U.S.)
| | - Dmitriy Minond
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Ulrich Schwaneberg
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany; (M.B.); (U.S.)
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstraße 50, 52056 Aachen, Germany
| | - Mehdi D. Davari
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle, Germany
| | - Daniela Yildiz
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Center for Human and Molecular Biology (ZHMB), University of Saarland, Kirrbergerstr., 66421 Homburg, Germany
| |
Collapse
|
13
|
Pseudomonas aeruginosa Triggered Exosomal Release of ADAM10 Mediates Proteolytic Cleavage in Trans. Int J Mol Sci 2022; 23:ijms23031259. [PMID: 35163191 PMCID: PMC8835980 DOI: 10.3390/ijms23031259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
Pneumonia is a life-threatening disease often caused by infection with Streptococcus pneumoniae and Pseudomonas aeruginosa. Many of the mediators (e.g., TNF, IL-6R) and junction molecules (e.g., E-cadherin) orchestrating inflammatory cell recruitment and loss of barrier integrity are proteolytically cleaved through a disintegrin and metalloproteinases (ADAMs). We could show by Western blot, surface expression analysis and measurement of proteolytic activity in cell-based assays, that ADAM10 in epithelial cells is upregulated and activated upon infection with Pseudomonas aeruginosa and Exotoxin A (ExoA), but not upon infection with Streptococcus pneumoniae. Targeting ADAM10 by pharmacological inhibition or gene silencing, we demonstrated that this activation was critical for cleavage of E-cadherin and modulated permeability and epithelial integrity. Stimulation with heat-inactivated bacteria revealed that the activation was based on the toxin repertoire rather than the interaction with the bacterial particle itself. Furthermore, calcium imaging experiments showed that the ExoA action was based on the induction of calcium influx. Investigating the extracellular vesicles and their proteolytic activity, we could show that Pseudomonas aeruginosa triggered exosomal release of ADAM10 and proteolytic cleavage in trans. This newly described mechanism could constitute an essential mechanism causing systemic inflammation in patients suffering from Pseudomonas aeruginosa-induced pneumonia stimulating future translational studies.
Collapse
|
14
|
Jordan PM, Gerstmeier J, Pace S, Bilancia R, Rao Z, Börner F, Miek L, Gutiérrez-Gutiérrez Ó, Arakandy V, Rossi A, Ialenti A, González-Estévez C, Löffler B, Tuchscherr L, Serhan CN, Werz O. Staphylococcus aureus-Derived α-Hemolysin Evokes Generation of Specialized Pro-resolving Mediators Promoting Inflammation Resolution. Cell Rep 2021; 33:108247. [PMID: 33053344 PMCID: PMC7729929 DOI: 10.1016/j.celrep.2020.108247] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/02/2020] [Accepted: 09/17/2020] [Indexed: 12/19/2022] Open
Abstract
Underlying mechanisms of how infectious inflammation is resolved by the host are incompletely understood. One hallmark of inflammation resolution is the activation of specialized pro-resolving mediators (SPMs) that enhance bacterial clearance and promote tissue repair. Here, we reveal α-hemolysin (Hla) from Staphylococcus aureus as a potent elicitor of SPM biosynthesis in human M2-like macrophages and in the mouse peritoneum through selective activation of host 15-lipoxygenase-1 (15-LOX-1). S. aureus-induced SPM formation in M2 is abolished upon Hla depletion or 15-LOX-1 knockdown. Isolated Hla elicits SPM formation in M2 that is reverted by inhibition of the Hla receptor ADAM10. Lipid mediators derived from Hla-treated M2 accelerate planarian tissue regeneration. Hla but not zymosan provokes substantial SPM formation in the mouse peritoneum, devoid of leukocyte infiltration and pro-inflammatory cytokine secretion. Besides harming the host, Hla may also exert beneficial functions by stimulating SPM production to promote the resolution of infectious inflammation. Jordan et al. reveal that α-hemolysin from Staphylococcus aureus stimulates specialized pro-resolving mediator (SPM) formation through activation of 15-lipoxygenase-1 in human macrophages involving ADAM10. The host may exploit α-hemolysin as an SPM inducer to better cope with S. aureus infections and to promote inflammation resolution and tissue regeneration.
Collapse
Affiliation(s)
- Paul M Jordan
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany
| | - Jana Gerstmeier
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany.
| | - Simona Pace
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany
| | - Rossella Bilancia
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany; Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Zhigang Rao
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany
| | - Friedemann Börner
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany
| | - Laura Miek
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany
| | | | - Vandana Arakandy
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Antonietta Rossi
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Armando Ialenti
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | | | - Bettina Löffler
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Lorena Tuchscherr
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Oliver Werz
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany.
| |
Collapse
|
15
|
Urban J, Suchankova M, Ganovska M, Leksa V, Sandor F, Tedlova E, Konig B, Bucova M. The Role of CX3CL1 and ADAM17 in Pathogenesis of Diffuse Parenchymal Lung Diseases. Diagnostics (Basel) 2021; 11:diagnostics11061074. [PMID: 34208027 PMCID: PMC8230701 DOI: 10.3390/diagnostics11061074] [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: 05/12/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
Fractalkine (CX3CL1) is a unique chemokine that functions as a chemoattractant for effector cytotoxic lymphocytes and macrophages expressing fractalkine receptor CX3CR1. CX3CL1 exists in two forms—a soluble and a membrane-bound form. The soluble CX3CL1 is released from cell membranes by proteolysis by the TNF-α-converting enzyme/disintegrin-like metalloproteinase 17 (TACE/ADAM17) and ADAM10. In this study, we evaluated the diagnostic relevance and potential roles of CX3CL1 and ADAM17 in the pathogenesis of diffuse parenchymal lung diseases (DPLDs) in the human population. The concentration of CX3CL1 and ADAM17 was measured by the enzyme-linked immunosorbent assay (ELISA) test in bronchoalveolar lavage fluids of patients suffering from different DPLDs. The concentration of CX3CL1 was significantly higher in patients suffering from idiopathic pulmonary fibrosis (IPF) and hypersensitivity pneumonitis patients compared to the control group. A significantly higher concentration of CX3CL1 was measured in fibrotic DPLDs compared to non-fibrotic DLPD patients. We found a positive correlation of CX3CL1 levels with the number of CD8+ T cells, and a negative correlation with CD4+ T cells in BALF and diffusion capacity for carbon monoxide. The concentration of ADAM17 was significantly lower in the IPF group compared to the other DPLD groups. We noticed a significantly higher CX3CL1/ADAM17 ratio in the IPF group compared to the other DPLD groups. We suggest that CX3CL1 has a distinctive role in the pathogenesis of DPLDs. The level of CX3CL1 strongly correlates with the severity of lung parenchyma impairment. The results suggest that high values of CX3CL1/ADAM17 could be diagnostic markers for IPF.
Collapse
Affiliation(s)
- Jan Urban
- 4th Department of Pneumology and Phthisiology, National Institute for Tuberculosis, Respiratory Diseases and Thoracic Surgery, 059 84 Vysne Hagy, Slovakia
- Institute of Immunology, Faculty of Medicine Comenius University, 811 08 Bratislava, Slovakia; (M.S.); (M.B.)
- Correspondence: ; Tel.: +421-524-414-252
| | - Magda Suchankova
- Institute of Immunology, Faculty of Medicine Comenius University, 811 08 Bratislava, Slovakia; (M.S.); (M.B.)
| | - Martina Ganovska
- Department of Clinical Laboratories, National Institute for Tuberculosis, Respiratory Diseases and Thoracic Surgery, 059 84 Vysne Hagy, Slovakia;
| | - Vladimir Leksa
- Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia;
- Centre for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University of Vienna, A-9010 Vienna, Austria
| | - Frantisek Sandor
- Department of Pneumology and Phthisiology, Faculty of Medicine Comenius University and University Hospital, 821 01 Bratislava, Slovakia; (F.S.); (E.T.)
| | - Eva Tedlova
- Department of Pneumology and Phthisiology, Faculty of Medicine Comenius University and University Hospital, 821 01 Bratislava, Slovakia; (F.S.); (E.T.)
| | - Brian Konig
- Department of Operations Research and Econometrics, Faculty of Economic Informatics, University of Economics in Bratislava, 852 35 Bratislava, Slovakia;
- Institute of Economic Research of Slovak Academy of Sciences, 811 05 Bratislava, Slovakia
| | - Maria Bucova
- Institute of Immunology, Faculty of Medicine Comenius University, 811 08 Bratislava, Slovakia; (M.S.); (M.B.)
| |
Collapse
|
16
|
Sluiter TJ, van Buul JD, Huveneers S, Quax PHA, de Vries MR. Endothelial Barrier Function and Leukocyte Transmigration in Atherosclerosis. Biomedicines 2021; 9:328. [PMID: 33804952 PMCID: PMC8063931 DOI: 10.3390/biomedicines9040328] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/24/2022] Open
Abstract
The vascular endothelium is a highly specialized barrier that controls passage of fluids and migration of cells from the lumen into the vessel wall. Endothelial cells assist leukocytes to extravasate and despite the variety in the specific mechanisms utilized by different leukocytes to cross different vascular beds, there is a general principle of capture, rolling, slow rolling, arrest, crawling, and ultimately diapedesis via a paracellular or transcellular route. In atherosclerosis, the barrier function of the endothelium is impaired leading to uncontrolled leukocyte extravasation and vascular leakage. This is also observed in the neovessels that grow into the atherosclerotic plaque leading to intraplaque hemorrhage and plaque destabilization. This review focuses on the vascular endothelial barrier function and the interaction between endothelial cells and leukocytes during transmigration. We will discuss the role of endothelial dysfunction, transendothelial migration of leukocytes and plaque angiogenesis in atherosclerosis.
Collapse
Affiliation(s)
- Thijs J. Sluiter
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jaap D. van Buul
- Sanquin Research and Landsteiner Laboratory, Leeuwenhoek Centre for Advanced Microscopy, Swammerdam Institute for Life Sciences, University of Amsterdam, 1066 CX Amsterdam, The Netherlands;
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Paul H. A. Quax
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Margreet R. de Vries
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
17
|
Appel D, Hummel R, Weidemeier M, Endres K, Gölz C, Schäfer MKE. Pharmacologic Inhibition of ADAM10 Attenuates Brain Tissue Loss, Axonal Injury and Pro-inflammatory Gene Expression Following Traumatic Brain Injury in Mice. Front Cell Dev Biol 2021; 9:661462. [PMID: 33791311 PMCID: PMC8005610 DOI: 10.3389/fcell.2021.661462] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/23/2021] [Indexed: 12/30/2022] Open
Abstract
The α-secretase A disintegrin and metalloprotease 10 (ADAM10) regulates various physiological and pathophysiological processes. Despite its broad functional implications during development, plasticity, and disease, no pharmacological approaches to inhibit ADAM10 in acute brain injury have been reported. Here, we examined the effects of the ADAM10 inhibitor GI254023X on the neurological and histopathological outcome after experimental traumatic brain injury (TBI). C57BL/6N mice were subjected to the controlled cortical impact (CCI) model of TBI or sham procedure and received GI254023X or vehicle during the acute phase of injury (n = 40, 100 mg/kg, 25% DMSO, 0.1 M Na2CO3, intraperitoneal, 30 min and 24 h after TBI). GI254023X treatment did not improve neurological deficits from 1 to 7 days post-injury (dpi) but animals treated with GI254023X exhibited smaller brain lesions compared to vehicle treatment. Determination of brain mRNA expression by quantitative PCR showed that TBI-induced up-regulation of Adam10 and Adam17 was not influenced by GI254023X but the up-regulation of the matrix metalloproteinase genes Mmp2 and Mmp9 was attenuated. GI254023X treatment further increased the T cell marker Cd247 but did not affect blood brain barrier integrity, as assessed by Occludin mRNA expression and IgG brain extravasation. However, in agreement with neuroprotective effects of ADAM10 inhibition, GI254023X treatment attenuated axonal injury, as indicated by decreased generation of spectrin breakdown products (SBDPs) and decreased immunostaining using anti-non-phosphorylated neurofilament (SMI-32). Interestingly, reduced axonal injury in GI254023X-treated animals coincided with subtle mRNA dysregulation in the glutamate receptor subunit genes Gria1 and Grin2b. Quantitative PCR also revealed that GI254023X mitigated up-regulation of the pro-inflammatory markers Il6, Tnfa, and Lcn2 but not the up-regulation of the pan-microglia marker Aif1, the M2 microglia marker Arg1 and the reactive astrocyte marker Gfap. Taken together, the ADAM10 inhibitor GI254023X attenuates brain tissue loss, axonal injury and pro-inflammatory gene expression in the CCI model of TBI. These results suggest that ADAM10 may represent a therapeutic target in the acute phase of TBI.
Collapse
Affiliation(s)
- Dominik Appel
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Regina Hummel
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Martin Weidemeier
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Kristina Endres
- Focus Program Translational Neurosciences (FTN) of the Johannes Gutenberg-University, Mainz, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Christina Gölz
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Michael K. E. Schäfer
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
- Focus Program Translational Neurosciences (FTN) of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immunotherapy (FZI), Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
18
|
Pereira Vatanabe I, Peron R, Mantellatto Grigoli M, Pelucchi S, De Cesare G, Magalhães T, Manzine PR, Figueredo Balthazar ML, Di Luca M, Marcello E, Cominetti MR. ADAM10 Plasma and CSF Levels Are Increased in Mild Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22052416. [PMID: 33670873 PMCID: PMC7957802 DOI: 10.3390/ijms22052416] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 01/03/2023] Open
Abstract
ADAM10 is the main α-secretase that participates in the non-amyloidogenic cleavage of amyloid precursor protein (APP) in neurons, inhibiting the production of β-amyloid peptide (Aβ) in Alzheimer’s disease (AD). Strong recent evidence indicates the importance of the localization of ADAM10 for its activity as a protease. In this study, we investigated ADAM10 activity in plasma and CSF samples of patients with amnestic mild cognitive impairment (aMCI) and mild AD compared with cognitively healthy controls. Our results indicated that plasma levels of soluble ADAM10 were significantly increased in the mild AD group, and that in these samples the protease was inactive, as determined by activity assays. The same results were observed in CSF samples, indicating that the increased plasma ADAM10 levels reflect the levels found in the central nervous system. In SH-SY5Y neuroblastoma cells, ADAM10 achieves its major protease activity in the fraction obtained from plasma membrane lysis, where the mature form of the enzyme is detected, confirming the importance of ADAM10 localization for its activity. Taken together, our results demonstrate the potential of plasma ADAM10 to act as a biomarker for AD, highlighting its advantages as a less invasive, easier, faster, and lower-cost processing procedure, compared to existing biomarkers.
Collapse
Affiliation(s)
- Izabela Pereira Vatanabe
- Department of Gerontology, Federal University of São Carlos, São Carlos 13565-905, Brazil; (I.P.V.); (R.P.); (M.M.G.); (P.R.M.)
| | - Rafaela Peron
- Department of Gerontology, Federal University of São Carlos, São Carlos 13565-905, Brazil; (I.P.V.); (R.P.); (M.M.G.); (P.R.M.)
| | - Marina Mantellatto Grigoli
- Department of Gerontology, Federal University of São Carlos, São Carlos 13565-905, Brazil; (I.P.V.); (R.P.); (M.M.G.); (P.R.M.)
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, Universitá Degli Studi di Milano, 20133 Milan, Italy; (S.P.); (G.D.C.); (M.D.L.)
| | - Giulia De Cesare
- Department of Pharmacological and Biomolecular Sciences, Universitá Degli Studi di Milano, 20133 Milan, Italy; (S.P.); (G.D.C.); (M.D.L.)
| | - Thamires Magalhães
- Department of Neurology, University of Campinas, Campinas 13083-887, Brazil; (T.M.); (M.L.F.B.)
| | - Patricia Regina Manzine
- Department of Gerontology, Federal University of São Carlos, São Carlos 13565-905, Brazil; (I.P.V.); (R.P.); (M.M.G.); (P.R.M.)
| | | | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, Universitá Degli Studi di Milano, 20133 Milan, Italy; (S.P.); (G.D.C.); (M.D.L.)
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Universitá Degli Studi di Milano, 20133 Milan, Italy; (S.P.); (G.D.C.); (M.D.L.)
- Correspondence: (E.M.); (M.R.C.); Tel.: +39-02-5031-8314 (E.M.); +55-16-3306-6663 (M.R.C.)
| | - Marcia Regina Cominetti
- Department of Gerontology, Federal University of São Carlos, São Carlos 13565-905, Brazil; (I.P.V.); (R.P.); (M.M.G.); (P.R.M.)
- Correspondence: (E.M.); (M.R.C.); Tel.: +39-02-5031-8314 (E.M.); +55-16-3306-6663 (M.R.C.)
| |
Collapse
|
19
|
Zhou M, Lin Y, Lu L, Zhang Z, Guo W, Peng G, Zhang W, Zhu Z, Wu Z, Mo M, Yang X, Zhu X, Chen C, Chen X, Xu P. Association of ADAM10 gene variants with sporadic Parkinson's disease in Chinese Han population. J Gene Med 2021; 23:e3319. [PMID: 33527480 DOI: 10.1002/jgm.3319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 01/07/2021] [Accepted: 01/24/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide. Genetic factors play important roles in PD risk. rs653765 and rs514049 of ADAM10 were reported to be associated with Alzheimer's disease (AD) in Caucasian population; however, the association of the two variants with PD in Chinese Han population remains unknown. The present investigation aimed to explore the possible association of ADAM10 variants with PD in Chinese Han population. METHODS We enrolled 565 PD patients and 518 healthy controls to conduct a case-control study. DNA samples were extracted from peripheral blood leukocytes, and the genotypes were determined by utilization of MassARRAY platform. Plasma levels were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS We found CC genotype of rs514049 was associated with an increased risk of PD (OR (95% CI) = 3.776 (1.127-11.217), p = 0.018). The C allele frequency of rs514049 was significantly higher in PD group (OR (95% CI) = 1.328 (1.031-1.709), p = 0.028), especially in male subgroup (OR (95% CI) = 1.484 (1.053-2.092), p = 0.024). However, there was no significant difference in the genotype or allele frequencies for rs653765 within the groups. Plasma levels were significantly decreased in PD patients compared with controls (p < 0.001). CONCLUSIONS Our data suggested that C allele of rs514049 in ADAM10 may increase the risk of PD in Chinese Han population, especially in males. The decreased plasma levels are probably involved in PD development.
Collapse
Affiliation(s)
- Miaomiao Zhou
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuwan Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lin Lu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiling Zhang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenyuan Guo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guoyou Peng
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenlong Zhang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ziting Zhu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhuohua Wu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingshu Mo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinling Yang
- Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaoqin Zhu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Chaojun Chen
- Department of Neurology, Guangzhou Chinese Medical Integrated Hospital (Huadu), Guangzhou, China
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
20
|
Morsing SKH, Rademakers T, Brouns SLN, van Stalborch AMD, Donners MMPC, van Buul JD. ADAM10-Mediated Cleavage of ICAM-1 Is Involved in Neutrophil Transendothelial Migration. Cells 2021; 10:cells10020232. [PMID: 33504031 PMCID: PMC7911467 DOI: 10.3390/cells10020232] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 01/24/2023] Open
Abstract
To efficiently cross the endothelial barrier during inflammation, neutrophils first firmly adhere to the endothelial surface using the endothelial adhesion molecule ICAM-1. Upon actual transmigration, the release from ICAM-1 is required. While Integrin LFA1/Mac1 de-activation is one described mechanism that leads to this, direct cleavage of ICAM-1 from the endothelium represents a second option. We found that a disintegrin and metalloprotease 10 (ADAM10) cleaves the extracellular domain of ICAM-1 from the endothelial surface. Silencing or inhibiting endothelial ADAM10 impaired the efficiency of neutrophils to cross the endothelium, suggesting that neutrophils use endothelial ADAM10 to dissociate from ICAM-1. Indeed, when measuring transmigration kinetics, neutrophils took almost twice as much time to finish the diapedesis step when ADAM10 was silenced. Importantly, we found increased levels of ICAM-1 on the transmigrating neutrophils when crossing an endothelial monolayer where such increased levels were not detected when neutrophils crossed bare filters. Using ICAM-1-GFP-expressing endothelial cells, we show that ICAM-1 presence on the neutrophils can also occur by membrane transfer from the endothelium to the neutrophil. Based on these findings, we conclude that endothelial ADAM10 contributes in part to neutrophil transendothelial migration by cleaving ICAM-1, thereby supporting the release of neutrophils from the endothelium during the final diapedesis step.
Collapse
Affiliation(s)
- Sofia K. H. Morsing
- Molecular Cell Biology Lab, Department Molecular and Cellular Homeostasis, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (S.K.H.M.); (T.R.); (S.L.N.B.); (A.-M.D.v.S.)
| | - Timo Rademakers
- Molecular Cell Biology Lab, Department Molecular and Cellular Homeostasis, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (S.K.H.M.); (T.R.); (S.L.N.B.); (A.-M.D.v.S.)
| | - Sanne L. N. Brouns
- Molecular Cell Biology Lab, Department Molecular and Cellular Homeostasis, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (S.K.H.M.); (T.R.); (S.L.N.B.); (A.-M.D.v.S.)
| | - Anne-Marieke D. van Stalborch
- Molecular Cell Biology Lab, Department Molecular and Cellular Homeostasis, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (S.K.H.M.); (T.R.); (S.L.N.B.); (A.-M.D.v.S.)
| | - Marjo M. P. C. Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
- Correspondence: (M.M.P.C.D.); (J.D.v.B.); Tel.: +31-43-3877167 (M.M.P.C.D.); +31-20-5121219 (J.D.v.B.); Fax: +31-20-5123310 (J.D.v.B.)
| | - Jaap D. van Buul
- Molecular Cell Biology Lab, Department Molecular and Cellular Homeostasis, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (S.K.H.M.); (T.R.); (S.L.N.B.); (A.-M.D.v.S.)
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), Section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, 1066 CX Amsterdam, The Netherlands
- Correspondence: (M.M.P.C.D.); (J.D.v.B.); Tel.: +31-43-3877167 (M.M.P.C.D.); +31-20-5121219 (J.D.v.B.); Fax: +31-20-5123310 (J.D.v.B.)
| |
Collapse
|
21
|
Seifert A, Düsterhöft S, Wozniak J, Koo CZ, Tomlinson MG, Nuti E, Rossello A, Cuffaro D, Yildiz D, Ludwig A. The metalloproteinase ADAM10 requires its activity to sustain surface expression. Cell Mol Life Sci 2021; 78:715-732. [PMID: 32372373 PMCID: PMC7873107 DOI: 10.1007/s00018-020-03507-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/05/2020] [Accepted: 03/17/2020] [Indexed: 12/25/2022]
Abstract
The metalloproteinase ADAM10 critically contributes to development, inflammation, and cancer and can be controlled by endogenous or synthetic inhibitors. Here, we demonstrate for the first time that loss of proteolytic activity of ADAM10 by either inhibition or loss of function mutations induces removal of the protease from the cell surface and the whole cell. This process is temperature dependent, restricted to mature ADAM10, and associated with an increased internalization, lysosomal degradation, and release of mature ADAM10 in extracellular vesicles. Recovery from this depletion requires de novo synthesis. Functionally, this is reflected by loss and recovery of ADAM10 substrate shedding. Finally, ADAM10 inhibition in mice reduces systemic ADAM10 levels in different tissues. Thus, ADAM10 activity is critically required for its surface expression in vitro and in vivo. These findings are crucial for development of therapeutic ADAM10 inhibition strategies and may showcase a novel, physiologically relevant mechanism of protease removal due to activity loss.
Collapse
Affiliation(s)
- Anke Seifert
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Justyna Wozniak
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Chek Z Koo
- School of Biosciences, University of Birmingham, Birmingham, UK
| | | | - Elisa Nuti
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | | | - Daniela Yildiz
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Aachen, Germany.
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
| |
Collapse
|
22
|
Aljohmani A, Yildiz D. A Disintegrin and Metalloproteinase-Control Elements in Infectious Diseases. Front Cardiovasc Med 2020; 7:608281. [PMID: 33392273 PMCID: PMC7772189 DOI: 10.3389/fcvm.2020.608281] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Despite recent advances in treatment strategies, infectious diseases are still under the leading causes of death worldwide. Although the activation of the inflammatory cascade is one prerequisite of defense, persistent and exuberant immune response, however, may lead to chronicity of inflammation predisposing to a temporal or permanent tissue damage not only of the site of infection but also among different body organs. The initial response to invading pathogens is mediated by the recognition through various pattern-recognition receptors along with cellular engulfment resulting in a coordinated release of soluble effector molecules and cytokines aiming to terminate the external stimuli. Members of the ‘a disintegrin and metalloproteinase’ (ADAM) family have the capability to proteolytically cleave transmembrane molecules close to the plasma membrane, a process called ectodomain shedding. In fact, in infectious diseases dysregulation of numerous ADAM substrates such as junction molecules (e.g., E-cadherin, VE-cadherin, JAM-A), adhesion molecules (e.g., ICAM-1, VCAM-1, L-selectin), and chemokines and cytokines (e.g., CXCL16, TNF-α) has been observed. The alpha-cleavage by ADAM proteases represents a rate limiting step for downstream regulated intramembrane proteolysis (RIPing) of several substrates, which influence cellular differentiation, cell signaling pathways and immune modulation. Both the substrates mentioned above and RIPing crucially contribute to a systematic damage in cardiovascular, endocrine, and/or gastrointestinal systems. This review will summarize the current knowledge of ADAM function and the subsequent RIPing in infectious diseases (e.g., pathogen recognition and clearance) and discuss the potential long-term effect on pathophysiological changes such as cardiovascular diseases.
Collapse
Affiliation(s)
- Ahmad Aljohmani
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - Daniela Yildiz
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| |
Collapse
|
23
|
Seifert A, Wozniak J, Düsterhöft S, Kasparek P, Sedlacek R, Dreschers S, Orlikowsky TW, Yildiz D, Ludwig A. The iRhom2/ADAM17 Axis Attenuates Bacterial Uptake by Phagocytes in a Cell Autonomous Manner. Int J Mol Sci 2020; 21:ijms21175978. [PMID: 32825187 PMCID: PMC7503280 DOI: 10.3390/ijms21175978] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/11/2020] [Accepted: 08/18/2020] [Indexed: 12/31/2022] Open
Abstract
Uptake of bacteria by phagocytes is a crucial step in innate immune defence. Members of the disintegrin and metalloproteinase (ADAM) family critically control the immune response by limited proteolysis of surface expressed mediator molecules. Here, we investigated the significance of ADAM17 and its regulatory adapter molecule iRhom2 for bacterial uptake by phagocytes. Inhibition of metalloproteinase activity led to increased phagocytosis of pHrodo labelled Gram-negative and -positive bacteria (E. coli and S. aureus, respectively) by human and murine monocytic cell lines or primary phagocytes. Bone marrow-derived macrophages showed enhanced uptake of heat-inactivated and living E. coli when they lacked either ADAM17 or iRhom2 but not upon ADAM10-deficiency. In monocytic THP-1 cells, corresponding short hairpin RNA (shRNA)-mediated knockdown confirmed that ADAM17, but not ADAM10, promoted phagocytosis of E. coli. The augmented bacterial uptake occurred in a cell autonomous manner and was accompanied by increased release of the chemokine CXCL8, less TNFα release and only minimal changes in the surface expression of the receptors TNFR1, TLR6 and CD36. Inhibition experiments indicated that the enhanced bacterial phagocytosis after ADAM17 knockdown was partially dependent on TNFα-activity but not on CXCL8. This novel role of ADAM17 in bacterial uptake needs to be considered in the development of ADAM17 inhibitors as therapeutics.
Collapse
Affiliation(s)
- Anke Seifert
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany; (A.S.); (J.W.); (S.D.)
| | - Justyna Wozniak
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany; (A.S.); (J.W.); (S.D.)
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany; (A.S.); (J.W.); (S.D.)
| | - Petr Kasparek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 25250 Vestec, Czech Republic; (P.K.); (R.S.)
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 25250 Vestec, Czech Republic; (P.K.); (R.S.)
| | - Stephan Dreschers
- Department of Neonatology, University Children’s Hospital, 52074 Aachen, Germany; (S.D.); (T.W.O.)
| | - Thorsten W. Orlikowsky
- Department of Neonatology, University Children’s Hospital, 52074 Aachen, Germany; (S.D.); (T.W.O.)
| | - Daniela Yildiz
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany; (A.S.); (J.W.); (S.D.)
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, 66424 Homburg, Germany
- Correspondence: (D.Y.); (A.L.); Tel.: +49-241-8035771 (A.L.); Fax: +49-241-8082433 (A.L.)
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany; (A.S.); (J.W.); (S.D.)
- Correspondence: (D.Y.); (A.L.); Tel.: +49-241-8035771 (A.L.); Fax: +49-241-8082433 (A.L.)
| |
Collapse
|
24
|
Yang J, LeBlanc ME, Cano I, Saez-Torres KL, Saint-Geniez M, Ng YS, D'Amore PA. ADAM10 and ADAM17 proteases mediate proinflammatory cytokine-induced and constitutive cleavage of endomucin from the endothelial surface. J Biol Chem 2020; 295:6641-6651. [PMID: 32193206 DOI: 10.1074/jbc.ra119.011192] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/17/2020] [Indexed: 12/30/2022] Open
Abstract
Contact between inflammatory cells and endothelial cells (ECs) is a crucial step in vascular inflammation. Recently, we demonstrated that the cell-surface level of endomucin (EMCN), a heavily O-glycosylated single-transmembrane sialomucin, interferes with the interactions between inflammatory cells and ECs. We have also shown that, in response to an inflammatory stimulus, EMCN is cleared from the cell surface by an unknown mechanism. In this study, using adenovirus-mediated overexpression of a tagged EMCN in human umbilical vein ECs, we found that treatment with tumor necrosis factor α (TNF-α) or the strong oxidant pervanadate leads to loss of cell-surface EMCN and increases the levels of the C-terminal fragment of EMCN 3- to 4-fold. Furthermore, treatment with the broad-spectrum matrix metalloproteinase inhibitor batimastat (BB94) or inhibition of ADAM metallopeptidase domain 10 (ADAM10) and ADAM17 with two small-molecule inhibitors, GW280264X and GI254023X, or with siRNA significantly reduced basal and TNFα-induced cell-surface EMCN cleavage. Release of the C-terminal fragment of EMCN by TNF-α treatment was blocked by chemical inhibition of ADAM10 alone or in combination with ADAM17. These results indicate that cell-surface EMCN undergoes constitutive cleavage and that TNF-α treatment dramatically increases this cleavage, which is mediated predominantly by ADAM10 and ADAM17. As endothelial cell-surface EMCN attenuates leukocyte-EC interactions during inflammation, we propose that EMCN is a potential therapeutic target to manage vascular inflammation.
Collapse
Affiliation(s)
- Jinling Yang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02115.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Michelle E LeBlanc
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02115.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Issahy Cano
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02115.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Kahira L Saez-Torres
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02115.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Magali Saint-Geniez
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02115.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Yin-Shan Ng
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02115.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02115 .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115.,Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
25
|
Abstract
Proteolysis has emerged as a key post-translational regulator of the function of molecules on the cell surface and in the extracellular milieu. In principle, proteolysis can activate or inactivate a substrate, or can change its functional properties. ADAMs (a disintegrin and metalloprotease) and ADAMTS (a disintegrin-like and metalloprotease domain with thrombospondin type 1 repeats) proteases are related members of a superfamily of metallo-endopeptidases that also includes MMPs and astacins. ADAMs are integral membrane proteins that typically cleave other membrane anchored proteins, whereas ADAMTS proteases lack a membrane anchor, and process both cell-surface and secreted molecules, the latter mostly extracellular matrix (ECM) components. ADAMs are implicated in fertilization, neurogenesis, in regulating the function of ligands for the EGF-receptor, and in the release of proteins such as the pro-inflammatory cytokine TNFα from the plasma membrane. ADAMTS proteases have key roles in embryonic development, including lung development, the molecular maturation of von Willebrand factor and procollagen as well as organization of fibrillin microfibrils in ECM, and are implicated in the pathogenesis of diverse lung and airway disorders. Here, we provide a general overview of the biochemical properties and physiological functions of ADAMs and ADAMTS proteases and describe their relevance to lung and airway disorders.
Collapse
|
26
|
Zhou Y, Lei J, Xie Q, Wu L, Jin S, Guo B, Wang X, Yan G, Zhang Q, Zhao H, Zhang J, Zhang X, Wang J, Gu J, Liu X, Ye D, Miao H, Serhan CN, Li Y. Fibrinogen-like protein 2 controls sepsis catabasis by interacting with resolvin Dp5. SCIENCE ADVANCES 2019; 5:eaax0629. [PMID: 31763448 PMCID: PMC6853772 DOI: 10.1126/sciadv.aax0629] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 09/17/2019] [Indexed: 06/10/2023]
Abstract
The mechanisms that drive programmed resolution of inflammation remain elusive. Here, we report the temporal regulation of soluble (s) and transmembrane (m) fibrinogen-like protein 2 (Fgl2) during inflammation and show that both sFgl2 and mFgl2 correlate with the outcome. The expression and ectodomain shedding of Fgl2 are respectively promoted by miR-466l and metalloproteinases (ADAM10 and ADAM17) during inflammation resolution. Deficiency of Fgl2 enhances polymorphonuclear neutrophil (PMN) infiltration but impairs macrophage (MΦ) maturation and phagocytosis and inhibits the production of n-3 docosapentaenoic acid-derived resolvin 5 (RvDp5). In contrast, administration of sFgl2 blunts PMN infiltration as well as promotes PMN apoptosis and RvDp5 biosynthesis. By activating ALX/FPR2, RvDp5 enhances sFgl2 secretion via ADAM17 and synergistically accelerates resolution of inflammation. These results uncover a previously unknown endogenous programmed mechanism by which Fgl2 regulates resolution of inflammation and shed new light on clinical sepsis treatments.
Collapse
Affiliation(s)
- Yu Zhou
- Clinical Medicine Research Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Juan Lei
- Clinical Medicine Research Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Qichao Xie
- Department of Oncology, Third Affiliated Hospital, Chongqing Medical University, Chongqing 401120, China
| | - Lei Wu
- Clinical Medicine Research Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Shengwei Jin
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan Road, Wenzhou, Zhejiang Province 325027, China
| | - Bo Guo
- Maternal and Child Health Research Institute, Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
| | - Xiang Wang
- Clinical Medicine Research Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Guifang Yan
- Clinical Medicine Research Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Qi Zhang
- Clinical Medicine Research Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Huakan Zhao
- Clinical Medicine Research Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Jiangang Zhang
- Clinical Medicine Research Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xiao Zhang
- Clinical Medicine Research Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Jingchun Wang
- Clinical Medicine Research Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Jiaqi Gu
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan Road, Wenzhou, Zhejiang Province 325027, China
| | - Xiaoli Liu
- Family Planning Department, Chongqing Health Center for Women and Children, Chongqing 401147, China
| | - Duyun Ye
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongming Miao
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing 400038, China
| | - Charles N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Hale Transformative Medicine Building, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Yongsheng Li
- Clinical Medicine Research Center, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| |
Collapse
|
27
|
Park JH, Choi JY, Jo C, Koh YH. Involvement of ADAM10 in acrolein-induced astrocytic inflammation. Toxicol Lett 2019; 318:44-49. [PMID: 31639409 DOI: 10.1016/j.toxlet.2019.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/05/2019] [Accepted: 10/10/2019] [Indexed: 12/29/2022]
Abstract
Acrolein is a neurotoxin produced through lipid peroxidation in the brain affected by ischemic stroke, which results in neuronal cell injury and inflammation. However the mechanism underlying acrolein-induced brain inflammation remains unclear. Therefore we examined how acrolein leads to astrocytic inflammation. It was found that acrolein increased the levels of NLRP3 and cleaved caspase-1, which led to the maturation of interleukin-1β (IL-1β). ELISA assay results, which showed that acrolein increased the secreted IL-1β, further supported acrolein-induced astrocytic inflammation. Acrolein increased ADAM10 protein levels and the cleavage of N-cadherin. The ADAM10 inhibitor, GI 254023X blocked N-cadherin cleavage by acrolein, suggesting that ADAM10 is an upstream of N-cadherin. Furthermore, we found that acrolein activated p38 MAPK and NF-κB p65, while pretreatment with p38 MAPK inhibitor, SB203580 and GI 254023X inhibited NF-κB p65 activation and NLRP3 inflammasome. This suggests that p38 MAPK mediates the activation of NF-κB p65, which is associated with NLRP3 expression. Finally, we showed that acrolein induced cell toxicity and decrease of EAAT1 expression, suggesting that acrolein may induce a loss of glutamate uptake function. In conclusion, we demonstrate that acrolein induces astrocytic inflammation through NLRP3 inflammasome, which is regulated by ADAM10 and attributed to p38 MAPK-activated NF-κB p65 activity.
Collapse
Affiliation(s)
- Jung Hyun Park
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2(i)-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| | - Ji-Young Choi
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2(i)-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| | - Chulman Jo
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2(i)-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| | - Young Ho Koh
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2(i)-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea.
| |
Collapse
|
28
|
Status update on iRhom and ADAM17: It's still complicated. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1567-1583. [PMID: 31330158 DOI: 10.1016/j.bbamcr.2019.06.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023]
Abstract
Several membrane-bound proteins with a single transmembrane domain are subjected to limited proteolysis at the cell surface. This cleavage leads to the release of their biologically active ectodomains, which can trigger different signalling pathways. In many cases, this ectodomain shedding is mediated by members of the family of a disintegrins and metalloproteinases (ADAMs). ADAM17 in particular is responsible for the cleavage of several proinflammatory mediators, growth factors, receptors and adhesion molecules. Due to its direct involvement in the release of these signalling molecules, ADAM17 can be positively and negatively involved in various physiological processes as well as in inflammatory, fibrotic and malignant pathologies. This central role of ADAM17 in a variety of processes requires strict multi-level regulation, including phosphorylation, various conformational changes and endogenous inhibitors. Recent research has shown that an early, crucial control mechanism is interaction with certain adapter proteins identified as iRhom1 and iRhom2, which are pseudoproteases of the rhomboid superfamily. Thus, iRhoms have also a decisive influence on physiological and pathophysiological signalling processes regulated by ADAM17. Their characteristic gene expression profiles, the specific consequences of gene knockouts and finally the occurrence of disease-associated mutations suggest that iRhom1 and iRhom2 undergo different gene regulation in order to fulfil their function in different cell types and are therefore only partially redundant. Therefore, there is not only interest in ADAM17, but also in iRhoms as therapeutic targets. However, to exploit the therapeutic potential, the regulation of ADAM17 activity and in particular its interaction with iRhoms must be well understood.
Collapse
|
29
|
Li H, Han S, Sun Q, Yao Y, Li S, Yuan C, Zhang B, Jing B, Wu J, Song Y, Wang H. Long non-coding RNA CDKN2B-AS1 reduces inflammatory response and promotes cholesterol efflux in atherosclerosis by inhibiting ADAM10 expression. Aging (Albany NY) 2019; 11:1695-1715. [PMID: 30926762 PMCID: PMC6461186 DOI: 10.18632/aging.101863] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/06/2019] [Indexed: 06/09/2023]
Abstract
INTRODUCTION Long non-coding RNAs (lncRNAs) play key roles in the development of atherosclerosis through the inflammatory pathway. This study aimed to investigate the role of lncRNA cyclin-dependent kinase inhibitor 2B antisense RNA 1 (CDKN2B-AS1) in atherosclerosis via its function in A disintegrin and metalloprotease 10 (ADAM10). METHODS Initially, the expression of CDKN2B-AS1 and ADAM10 in atherosclerotic plaque tissues and THP-1 macrophage-derived foam cells was determined, after which the cholesterol efflux rate of macrophages was calculated. Interaction between CDKN2B-AS1 and ADAM10 was analyzed, after which, expression of CDKN2B-AS1 and ADAM10 were altered to explore their effects on inflammatory response and cholesterol efflux. The aforementioned findings were further intended to be validated by the atherosclerosis mouse model experiments. RESULTS Atherosclerotic plaque tissue and THP-1 macrophage-derived foam cells exhibited downregulated CDKN2B-AS1 and upregulated ADAM10. Upon overexpressing CDKN2B-AS1 or silencing ADAM10, lipid accumulation was reduced and cholesterol efflux was increased. CDKN2B-AS1 located in the nucleus could bind to DNA methyltransferase 1 (DNMT1) to enhance methylation of ADAM10 promoter, leading to suppressed atherosclerotic inflammatory response and promoted cholesterol efflux. CONCLUSION Altogether, lncRNA CDKN2B-AS1 can inhibit the transcription of ADAM10 via DNMT1-mediated ADAM10 DNA methylation, consequently preventing inflammatory response of atherosclerosis and promoting cholesterol efflux.
Collapse
Affiliation(s)
- Haocheng Li
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
- Equal contribution
| | - Song Han
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
- Equal contribution
| | - Qingfeng Sun
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Ye Yao
- Department of Cardiac Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Shiyong Li
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Chao Yuan
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Bo Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Bao Jing
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Jia Wu
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Ye Song
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Haiyang Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| |
Collapse
|
30
|
ADAM10 mediates malignant pleural mesothelioma invasiveness. Oncogene 2019; 38:3521-3534. [PMID: 30651596 PMCID: PMC6756017 DOI: 10.1038/s41388-018-0669-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/28/2018] [Accepted: 12/14/2018] [Indexed: 11/19/2022]
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer with limited therapeutic options and treatment efficiency. Even if the latency period between asbestos exposure, the main risk factor, and mesothelioma development is very long, the local invasion of mesothelioma is very rapid leading to a mean survival of one year after diagnosis. ADAM10 (A Disintegrin And Metalloprotease) sheddase targets membrane-bound substrates and its overexpression is associated with progression in several cancers. However, nothing is known about ADAM10 implication in MPM. In this study, we demonstrated higher ADAM10 expression levels in human MPM as compared to control pleural samples and in human MPM cell line. This ADAM10 overexpression was also observed in murine MPM samples. Two mouse mesothelioma cell lines were used in this study including one primary cell line obtained by repeated asbestos fibre injections. We show, in vitro, that ADAM10 targeting through shRNA and pharmacological (GI254023X) approaches reduced drastically mesothelioma cell migration and invasion, as well as for human mesothelioma cells treated with siRNA targeting ADAM10. Moreover, ADAM10 downregulation in murine mesothelioma cells significantly impairs MPM progression in vivo after intrapleural cell injection. We also demonstrate that ADAM10 sheddase downregulation decreases the production of a soluble N-cadherin fragment through membrane N-cadherin, which stimulated mesothelioma cell migration. Taken together, we demonstrate that ADAM10 is overexpressed in MPM and takes part to MPM progression through the generation of N-cadherin fragment that stimulates mesothelioma cell migration. ADAM10 inhibition is worth considering as a therapeutic perspective in mesothelioma context.
Collapse
|
31
|
Cai X, Dong J, Liu J, Zheng H, Kaweeteerawat C, Wang F, Ji Z, Li R. Multi-hierarchical profiling the structure-activity relationships of engineered nanomaterials at nano-bio interfaces. Nat Commun 2018; 9:4416. [PMID: 30356046 PMCID: PMC6200803 DOI: 10.1038/s41467-018-06869-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 10/02/2018] [Indexed: 12/13/2022] Open
Abstract
Increasing concerns over the possible risks of nanotechnology necessitates breakthroughs in structure-activity relationship (SAR) analyses of engineered nanomaterials (ENMs) at nano-bio interfaces. However, current nano-SARs are often based on univariate assessments and fail to provide tiered views on ENM-induced bio-effects. Here we report a multi-hierarchical nano-SAR assessment for a representative ENM, Fe2O3, by metabolomics and proteomics analyses. The established nano-SAR profile allows the visualizing of the contributions of seven basic properties of Fe2O3 to its diverse bio-effects. For instance, although surface reactivity is responsible for Fe2O3-induced cell migration, the inflammatory effects of Fe2O3 are determined by aspect ratio (nanorods) or surface reactivity (nanoplates). These nano-SARs are examined in THP-1 cells and animal lungs, which allow us to decipher the detailed mechanisms including NLRP3 inflammasome pathway and monocyte chemoattractant protein-1-dependent signaling. This study provides more insights for nano-SARs, and may facilitate the tailored design of ENMs to render them desired bio-effects.
Collapse
Affiliation(s)
- Xiaoming Cai
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, School of Public Health, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu 215123 China
| | - Jun Dong
- Wuhan Academy of Agricultural Science, Wuhan, Hubei 430000 China
| | - Jing Liu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, 116023 China
| | - Huizhen Zheng
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, School of Public Health, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu 215123 China
| | - Chitrada Kaweeteerawat
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Klong Nueng, 12120 Thailand
| | - Fangjun Wang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, 116023 China
| | - Zhaoxia Ji
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
- Living Proof, Inc., Cambridge, MA 02142 United States
| | - Ruibin Li
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, School of Public Health, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu 215123 China
| |
Collapse
|
32
|
|
33
|
Matthews AL, Koo CZ, Szyroka J, Harrison N, Kanhere A, Tomlinson MG. Regulation of Leukocytes by TspanC8 Tetraspanins and the "Molecular Scissor" ADAM10. Front Immunol 2018; 9:1451. [PMID: 30013551 PMCID: PMC6036176 DOI: 10.3389/fimmu.2018.01451] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/12/2018] [Indexed: 01/16/2023] Open
Abstract
A disintegrin and metalloproteinase 10 (ADAM10) is a ubiquitous transmembrane protein that functions as a "molecular scissor" to cleave the extracellular regions from its transmembrane target proteins. ADAM10 is well characterized as the ligand-dependent activator of Notch proteins, which control cell fate decisions. Indeed, conditional knockouts of ADAM10 in mice reveal impaired B-, T-, and myeloid cell development and/or function. ADAM10 cleaves many other leukocyte-expressed substrates. On B-cells, ADAM10 cleavage of the low-affinity IgE receptor CD23 promotes allergy and asthma, cleavage of ICOS ligand impairs antibody responses, and cleavage of the BAFF-APRIL receptor transmembrane activator and CAML interactor, and BAFF receptor, reduce B-cell survival. On microglia, increased ADAM10 cleavage of a rare variant of the scavenger receptor triggering receptor expressed on myeloid cells 2 may increase susceptibility to Alzheimer's disease. We and others recently showed that ADAM10 interacts with one of six different regulatory tetraspanin membrane proteins, which we termed the TspanC8 subgroup, comprising Tspan5, Tspan10, Tspan14, Tspan15, Tspan17, and Tspan33. The TspanC8s are required for ADAM10 exit from the endoplasmic reticulum, and emerging evidence suggests that they dictate ADAM10 subcellular localization and substrate specificity. Therefore, we propose that ADAM10 should not be regarded as a single scissor, but as six different scissors with distinct substrate specificities, depending on the associated TspanC8. In this review, we collate recent transcriptomic data to present the TspanC8 repertoires of leukocytes, and we discuss the potential role of the six TspanC8/ADAM10 scissors in leukocyte development and function.
Collapse
Affiliation(s)
- Alexandra L Matthews
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Chek Ziu Koo
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Justyna Szyroka
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Neale Harrison
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Aditi Kanhere
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Michael G Tomlinson
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
34
|
Akhter R, Shao Y, Shaw M, Formica S, Khrestian M, Leverenz JB, Bekris LM. Regulation of ADAM10 by miR-140-5p and potential relevance for Alzheimer's disease. Neurobiol Aging 2017; 63:110-119. [PMID: 29253717 DOI: 10.1016/j.neurobiolaging.2017.11.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 11/02/2017] [Accepted: 11/15/2017] [Indexed: 12/27/2022]
Abstract
Recent reports in Alzheimer's disease (AD) research suggest that alterations in microRNA (miRNA) expression are associated with disease pathology. Our previous studies suggest that A Disintegrin and Metalloproteinase 10 (ADAM10) expression is important in AD and could be modulated by an extended regulatory region that includes the 3' untranslated region. In this study, we have investigated the role of trans-acting factors in ADAM10 gene regulation. Our study shows that miRNA-140-5p has enhanced expression in the AD postmortem brain hippocampus using high-throughput miRNA arrays and quantitative real-time polymerase chain reaction. Interestingly, we have also seen that miRNA-140-5p seed sequence is present on 3' untranslated region of both ADAM10 and its transcription factor SOX2. The specific interaction of miRNA-140-5p with both ADAM10 and SOX2 signifies high regulatory importance of this miRNA in controlling ADAM10 expression. Thus, this investigation unravels mechanisms underlying ADAM10 downregulation by miR-140-5p and suggests that dysfunctional regulation of ADAM10 expression is exacerbated by AD-related neurotoxic effects. These findings underscore the importance of understanding the impact of trans-acting factors in the modulation of AD pathophysiology.
Collapse
Affiliation(s)
- Rumana Akhter
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Yvonne Shao
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - McKenzie Shaw
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Shane Formica
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Maria Khrestian
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lynn M Bekris
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
35
|
Amar S, Minond D, Fields GB. Clinical Implications of Compounds Designed to Inhibit ECM-Modifying Metalloproteinases. Proteomics 2017; 17. [PMID: 28613012 DOI: 10.1002/pmic.201600389] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/03/2017] [Indexed: 12/19/2022]
Abstract
Remodeling of the extracellular matrix (ECM) is crucial in development and homeostasis, but also has a significant role in disease progression. Two metalloproteinase families, the matrix metalloproteinases (MMPs) and a disintegrin and metalloproteases (ADAMs), participate in the remodeling of the ECM, either directly or through the liberation of growth factors and cell surface receptors. The correlation of MMP and ADAM activity to a variety of diseases has instigated numerous drug development programs. However, broad-based and Zn2+ -chelating MMP and ADAM inhibitors have fared poorly in the clinic. Selective MMP and ADAM inhibitors have been described recently based on (a) antibodies or antibody fragments or (b) small molecules designed to take advantage of protease secondary binding sites (exosites) or allosteric sites. Clinical trials have been undertaken with several of these inhibitors, while others are in advanced pre-clinical stages.
Collapse
Affiliation(s)
- Sabrina Amar
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL, USA
| | - Dmitriy Minond
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Gregg B Fields
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL, USA.,Department of Chemistry, The Scripps Research Institute/Scripps Florida, Jupiter, FL, USA
| |
Collapse
|
36
|
Abstract
Polymorphonuclear neutrophils (PMNs) are innate immune system cells that play an essential role in eradicating invading pathogens. PMN migration to sites of infection/inflammation requires exiting the microcirculation and subsequent crossing of epithelial barriers in mucosa-lined organs such as the lungs and intestines. Although these processes usually occur without significant damage to surrounding host tissues, dysregulated/excessive PMN transmigration and resultant bystander-tissue damage are characteristic of numerous mucosal inflammatory disorders. Mechanisms controlling PMN extravasation have been well characterized, but the molecular details regarding regulation of PMN migration across mucosal epithelia are poorly understood. Given that PMN migration across mucosal epithelia is strongly correlated with disease symptoms in many inflammatory mucosal disorders, enhanced understanding of the mechanisms regulating PMN transepithelial migration should provide insights into clinically relevant tissue-targeted therapies aimed at ameliorating PMN-mediated bystander-tissue damage. This review will highlight current understanding of the molecular interactions between PMNs and mucosal epithelia and the associated functional consequences.
Collapse
Affiliation(s)
- Jennifer C Brazil
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
37
|
Abstract
Myeloid cells have diverse roles in regulating immunity, inflammation, and extracellular matrix turnover. To accomplish these tasks, myeloid cells carry an arsenal of metalloproteinases, which include the matrix metalloproteinases and the adamalysins. These enzymes have diverse substrate repertoires, and are thus involved in mediating proteolytic cascades, cell migration, and cell signaling. Dysregulation of metalloproteinases contributes to pathogenic processes, including inflammation, fibrosis, and cancer. Metalloproteinases also have important nonproteolytic functions in controlling cytoskeletal dynamics during macrophage fusion and enhancing transcription to promote antiviral immunity. This review highlights the diverse contributions of metalloproteinases to myeloid cell functions.
Collapse
|
38
|
Reyat JS, Chimen M, Noy PJ, Szyroka J, Rainger GE, Tomlinson MG. ADAM10-Interacting Tetraspanins Tspan5 and Tspan17 Regulate VE-Cadherin Expression and Promote T Lymphocyte Transmigration. THE JOURNAL OF IMMUNOLOGY 2017; 199:666-676. [PMID: 28600292 PMCID: PMC5502317 DOI: 10.4049/jimmunol.1600713] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 05/10/2017] [Indexed: 12/25/2022]
Abstract
The recruitment of blood leukocytes across the endothelium to sites of tissue infection is central to inflammation, but also promotes chronic inflammatory diseases. A disintegrin and metalloproteinase 10 (ADAM10) is a ubiquitous transmembrane molecular scissor that is implicated in leukocyte transmigration by proteolytically cleaving its endothelial substrates. These include VE-cadherin, a homotypic adhesion molecule that regulates endothelial barrier function, and transmembrane chemokines CX3CL1 and CXCL16, which have receptors on leukocytes. However, a definitive role for endothelial ADAM10 in transmigration of freshly isolated primary leukocytes under flow has not been demonstrated, and the relative importance of distinct ADAM10 substrates is unknown. Emerging evidence suggests that ADAM10 can be regarded as six different molecular scissors with different substrate specificities, depending on which of six TspanC8 tetraspanins it is associated with, but TspanC8s remain unstudied in leukocyte transmigration. In the current study, ADAM10 knockdown on primary HUVECs was found to impair transmigration of freshly isolated human peripheral blood T lymphocytes, but not neutrophils or B lymphocytes, in an in vitro flow assay. This impairment was due to delayed transmigration rather than a complete block, and was overcome in the presence of neutrophils. Transmigration of purified lymphocytes was dependent on ADAM10 regulation of VE-cadherin, but not CX3CL1 and CXCL16. Tspan5 and Tspan17, the two most closely related TspanC8s by sequence, were the only TspanC8s that regulated VE-cadherin expression and were required for lymphocyte transmigration. Therefore endothelial Tspan5- and Tspan17-ADAM10 complexes may regulate inflammation by maintaining normal VE-cadherin expression and promoting T lymphocyte transmigration.
Collapse
Affiliation(s)
- Jasmeet S Reyat
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and.,Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Myriam Chimen
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Peter J Noy
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Justyna Szyroka
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - G Ed Rainger
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Michael G Tomlinson
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| |
Collapse
|
39
|
Dreymueller D, Pruessmeyer J, Schumacher J, Fellendorf S, Hess FM, Seifert A, Babendreyer A, Bartsch JW, Ludwig A. The metalloproteinase ADAM8 promotes leukocyte recruitment in vitro and in acute lung inflammation. Am J Physiol Lung Cell Mol Physiol 2017; 313:L602-L614. [PMID: 28596294 DOI: 10.1152/ajplung.00444.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 12/20/2022] Open
Abstract
Alveolar leukocyte recruitment is a hallmark of acute lung inflammation and involves transmigration of leukocytes through endothelial and epithelial layers. The disintegrin and metalloproteinase (ADAM) 8 is expressed on human isolated leukocytic cells and can be further upregulated on cultured endothelial and epithelial cells by proinflammatory cytokines. By shRNA-mediated knockdown we show that leukocytic ADAM8 is required on monocytic THP-1 cells for chemokine-induced chemotaxis as well as transendothelial and transepithelial migration. Furthermore, ADAM8 promotes αL-integrin upregulation and THP-1 cell adhesion to endothelial cells. On endothelial cells ADAM8 enhances transendothelial migration and increases cytokine-induced permeability. On epithelial cells the protease facilitates migration in a wound closure assay but does not affect transepithelial leukocyte migration. Blood leukocytes and bone marrow-derived macrophages (BMDM) from ADAM8-deficient mice show suppressed chemotactic response. Intranasal application of LPS to mice is accompanied with ADAM8 upregulation in the lung. In this model of acute lung inflammation ADAM8-deficient mice are protected against leukocyte infiltration. Finally, transfer experiments of BMDM in mice indicate that ADAM8 exerts a promigratory function predominantly on leukocytes. Our study provides in vitro and in vivo evidence that ADAM8 on leukocytes holds a proinflammatory function in acute lung inflammation by promoting alveolar leukocyte recruitment.
Collapse
Affiliation(s)
- Daniela Dreymueller
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany; and
| | - Jessica Pruessmeyer
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany; and
| | - Julian Schumacher
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany; and
| | - Sandra Fellendorf
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany; and
| | - Franz Martin Hess
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany; and
| | - Anke Seifert
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany; and
| | - Aaron Babendreyer
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany; and
| | - Jörg W Bartsch
- Department of Neurosurgery, Philipps University Marburg, University Hospital Marburg, Marburg, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany; and
| |
Collapse
|
40
|
Buchanan PC, Boylan KLM, Walcheck B, Heinze R, Geller MA, Argenta PA, Skubitz APN. Ectodomain shedding of the cell adhesion molecule Nectin-4 in ovarian cancer is mediated by ADAM10 and ADAM17. J Biol Chem 2017; 292:6339-6351. [PMID: 28232483 PMCID: PMC5391762 DOI: 10.1074/jbc.m116.746859] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 02/14/2017] [Indexed: 11/06/2022] Open
Abstract
We previously showed that the cell adhesion molecule Nectin-4 is overexpressed in ovarian cancer tumors, and its cleaved extracellular domain can be detected in the serum of ovarian cancer patients. The ADAM (adisintegrin and metalloproteinase) proteases are involved in ectodomain cleavage of transmembrane proteins, and ADAM17 is known to cleave Nectin-4 in breast cancer. However, the mechanism of Nectin-4 cleavage in ovarian cancer has not yet been determined. Analysis of ovarian cancer gene microarray data showed that higher expression of Nectin-4, ADAM10, and ADAM17 is associated with significantly decreased progression-free survival. We quantified Nectin-4 shedding from the surface of ovarian cancer cells after stimulation with lysophosphatidic acid. We report that ADAM17 and ADAM10 cleave Nectin-4 and release soluble Nectin-4 (sN4). Small molecule inhibitors and siRNA knockdown of both ADAM proteases confirmed these results. In matched samples from 11 high-grade serous ovarian cancer patients, we detected 2-20-fold more sN4 in ascites fluid than serum. Co-incubation of ovarian cancer cells with ascites fluid significantly increased sN4 shedding, which could be blocked using a dual inhibitor of ADAM10 and ADAM17. Furthermore, we detected RNA for Nectin-4, ADAM10, and ADAM17 in primary ovarian carcinoma tumors, secondary omental metastases, and ascites cells isolated from serous ovarian cancer patients. In a signaling pathway screen, lysophosphatidic acid increased phosphorylation of AKT, EGF receptor, ERK1/2, JNK1/2/3, and c-Jun. Understanding the function of Nectin-4 shedding in ovarian cancer progression is critical to facilitate its development as both a serum biomarker and a therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
| | | | - Bruce Walcheck
- From the Departments of Laboratory Medicine and Pathology
- Veterinary and Biomedical Sciences, and
| | - Rachel Heinze
- From the Departments of Laboratory Medicine and Pathology
| | - Melissa A Geller
- Obstetrics, Gynecology, and Women's Health, University of Minnesota, Minneapolis, Minnesota 55455
| | - Peter A Argenta
- Obstetrics, Gynecology, and Women's Health, University of Minnesota, Minneapolis, Minnesota 55455
| | | |
Collapse
|
41
|
Fine Tuning Cell Migration by a Disintegrin and Metalloproteinases. Mediators Inflamm 2017; 2017:9621724. [PMID: 28260841 PMCID: PMC5316459 DOI: 10.1155/2017/9621724] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/22/2016] [Indexed: 02/07/2023] Open
Abstract
Cell migration is an instrumental process involved in organ development, tissue homeostasis, and various physiological processes and also in numerous pathologies. Both basic cell migration and migration towards chemotactic stimulus consist of changes in cell polarity and cytoskeletal rearrangement, cell detachment from, invasion through, and reattachment to their neighboring cells, and numerous interactions with the extracellular matrix. The different steps of immune cell, tissue cell, or cancer cell migration are tightly coordinated in time and place by growth factors, cytokines/chemokines, adhesion molecules, and receptors for these ligands. This review describes how a disintegrin and metalloproteinases interfere with several steps of cell migration, either by proteolytic cleavage of such molecules or by functions independent of proteolytic activity.
Collapse
|
42
|
Discovery of an enzyme and substrate selective inhibitor of ADAM10 using an exosite-binding glycosylated substrate. Sci Rep 2016; 6:11. [PMID: 28442704 PMCID: PMC5431342 DOI: 10.1038/s41598-016-0013-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/12/2016] [Indexed: 02/01/2023] Open
Abstract
ADAM10 and ADAM17 have been shown to contribute to the acquired drug resistance of HER2-positive breast cancer in response to trastuzumab. The majority of ADAM10 and ADAM17 inhibitor development has been focused on the discovery of compounds that bind the active site zinc, however, in recent years, there has been a shift from active site to secondary substrate binding site (exosite) inhibitor discovery in order to identify non-zinc-binding molecules. In the present work a glycosylated, exosite-binding substrate of ADAM10 and ADAM17 was utilized to screen 370,276 compounds from the MLPCN collection. As a result of this uHTS effort, a selective, time-dependent, non-zinc-binding inhibitor of ADAM10 with Ki = 883 nM was discovered. This compound exhibited low cell toxicity and was able to selectively inhibit shedding of known ADAM10 substrates in several cell-based models. We hypothesize that differential glycosylation of these cognate substrates is the source of selectivity of our novel inhibitor. The data indicate that this novel inhibitor can be used as an in vitro and, potentially, in vivo, probe of ADAM10 activity. Additionally, results of the present and prior studies strongly suggest that glycosylated substrate are applicable as screening agents for discovery of selective ADAM probes and therapeutics.
Collapse
|
43
|
Stimulated release and functional activity of surface expressed metalloproteinase ADAM17 in exosomes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2795-2808. [DOI: 10.1016/j.bbamcr.2016.09.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 08/02/2016] [Accepted: 09/02/2016] [Indexed: 12/15/2022]
|
44
|
Schumacher N, Schmidt S, Schwarz J, Dohr D, Lokau J, Scheller J, Garbers C, Chalaris A, Rose-John S, Rabe B. Circulating Soluble IL-6R but Not ADAM17 Activation Drives Mononuclear Cell Migration in Tissue Inflammation. THE JOURNAL OF IMMUNOLOGY 2016; 197:3705-3715. [PMID: 27698010 DOI: 10.4049/jimmunol.1600909] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/31/2016] [Indexed: 01/09/2023]
Abstract
Neutrophil and mononuclear cell infiltration during inflammatory processes is highly regulated. The first cells at the site of infection or inflammation are neutrophils, followed by mononuclear cells. IL-6 plays an important role during inflammatory states. It has been shown in several models that the soluble form of IL-6R (sIL-6R) is involved in the recruitment of mononuclear cells by a mechanism called IL-6 trans-signaling. It had been speculated that sIL-6R was generated at the site of inflammation by shedding from neutrophils via activation of the metalloprotease ADAM17. Attempts to genetically delete the floxed ADAM17 gene selectively in myeloid cells infiltrating an air pouch cavity upon injection of carrageenan failed because in transgenic mice, LysMcre did not lead to appreciable loss of the ADAM17 protein in these cells. We therefore used ADAM17 hypomorphic mice, which only express ∼5% of ADAM17 wild-type levels in all tissues and show virtually no shedding of all tested ADAM17 substrates, to clarify the role of ADAM17 during local inflammation in the murine air pouch model. In the present study, we demonstrate that although IL-6 and the trans-signaling mechanism is mandatory for cellular infiltration in this model, it is not ADAM17-mediated shedding of IL-6R within the pouch that orchestrates this inflammatory process. Instead, we demonstrate that sIL-6R is infiltrating from the circulation in an ADAM17-independent process. Our data suggest that this infiltrating sIL-6R, which is needed for IL-6 trans-signaling, is involved in the controlled resolution of an acute inflammatory episode.
Collapse
Affiliation(s)
- Neele Schumacher
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, 24098 Kiel, Germany; and
| | - Stefanie Schmidt
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, 24098 Kiel, Germany; and
| | - Jeanette Schwarz
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, 24098 Kiel, Germany; and
| | - Dana Dohr
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, 24098 Kiel, Germany; and
| | - Juliane Lokau
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, 24098 Kiel, Germany; and
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Christoph Garbers
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, 24098 Kiel, Germany; and
| | - Athena Chalaris
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, 24098 Kiel, Germany; and
| | - Stefan Rose-John
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, 24098 Kiel, Germany; and
| | - Björn Rabe
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, 24098 Kiel, Germany; and
| |
Collapse
|
45
|
Dreymueller D, Ludwig A. Considerations on inhibition approaches for proinflammatory functions of ADAM proteases. Platelets 2016; 28:354-361. [PMID: 27460023 DOI: 10.1080/09537104.2016.1203396] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Proteases of the disintegrin and metalloproteinase (ADAM) family mediate the proteolytic shedding of various surface molecules including cytokine precursors, adhesion molecules, growth factors, and receptors. Within the vasculature ADAM10 and ADAM17 regulate endothelial permeability, transendothelial leukocyte migration, and the adhesion of leukocytes and platelets. In vivo studies show that both proteases are implicated in several inflammatory pathologies, for example, edema formation, leukocyte infiltration, and thrombosis. However, both proteases also contribute to developmental and regenerative processes. Thus, although ADAMs can be regarded as valuable drug targets in many aspects, the danger of severe side effects is clearly visible. To circumvent these side effects, traditional inhibition approaches have to be improved to target ADAMs at the right time in the right place. Moreover, the inhibitors need to be more selective for the target protease and if possible also for the substrate. Antibodies recognizing the active conformation of ADAMs or small molecules blocking exosites of ADAM proteases may represent inhibitors with the desired selectivities.
Collapse
Affiliation(s)
- Daniela Dreymueller
- a Institute of Pharmacology and Toxicology , RWTH Aachen University , Aachen , Germany
| | - Andreas Ludwig
- a Institute of Pharmacology and Toxicology , RWTH Aachen University , Aachen , Germany
| |
Collapse
|
46
|
Felix K, Gaida MM. Neutrophil-Derived Proteases in the Microenvironment of Pancreatic Cancer -Active Players in Tumor Progression. Int J Biol Sci 2016; 12:302-13. [PMID: 26929737 PMCID: PMC4753159 DOI: 10.7150/ijbs.14996] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A hallmark of pancreatic ductal adenocarcinoma (PDAC) is the fibro-inflammatory microenvironment, consisting of activated pancreatic stellate cells, extracellular matrix proteins, and a variety of inflammatory cells, such as T cells, macrophages, or neutrophils. Tumor-infiltrating immune cells, which are found in nearly all cancers, including PDAC, often fail to eliminate the tumor, but conversely can promote its progression by altering the tumor microenvironment. Pancreatic cancer cells are able to attract polymorphonuclear neutrophils (PMN) via tumor secreted chemokines and in human PDAC, PMN infiltrates can be observed in the vicinity of tumor cells and in the desmoplastic tumor stroma, which correlate with undifferentiated tumor growth and poor prognosis. The behavior of tumor-infiltrating neutrophils in the tumor micromilieu is not yet understood at a mechanistic level. It has been shown that PMN have the potential to kill tumor cells, either directly or by antibody-dependent cell-mediated cytotoxicity, but on the other side various adverse effects of PMN, such as promotion of aggressive tumor growth with epithelial-to-mesenchymal transition and increased metastatic potential, have been described. Recent therapeutic approaches for PDAC focus not only the tumor cell itself, but also elements of the tumor microenvironment. Therefore, the role of PMN and their derived products (e.g. cytokines, proteases) as a new vein for a therapeutic target should be critically evaluated in this context. This review summarizes the current understanding of the interplay between proteases of tumor-infiltrating neutrophils and pancreatic tumor cells and elements of the desmoplastic stroma.
Collapse
Affiliation(s)
- Klaus Felix
- 1. Department of General Surgery, University of Heidelberg, INF 110, Heidelberg, Germany
| | - Matthias M Gaida
- 2. Institute of Pathology, University of Heidelberg, INF 224, Heidelberg, Germany
| |
Collapse
|
47
|
Goldmann O, Tuchscherr L, Rohde M, Medina E. α-Hemolysin enhances Staphylococcus aureus internalization and survival within mast cells by modulating the expression of β1 integrin. Cell Microbiol 2016; 18:807-19. [PMID: 26595647 DOI: 10.1111/cmi.12550] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 11/06/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022]
Abstract
Mast cells (MCs) are important sentinels of the host defence against invading pathogens. We previously reported that Staphylococcus aureus evaded the extracellular antimicrobial activities of MCs by promoting its internalization within these cells via β1 integrins. Here, we investigated the molecular mechanisms governing this process. We found that S. aureus responded to the antimicrobial mediators released by MCs by up-regulating the expression of α-hemolysin (Hla), fibronectin-binding protein A and several regulatory systems. We also found that S. aureus induced the up-regulation of β1 integrin expression on MCs and that this effect was mediated by Hla-ADAM10 (a disintegrin and metalloproteinase 10) interaction. Thus, deletion of Hla or inhibition of Hla-ADAM10 interaction significantly impaired S. aureus internalization within MCs. Furthermore, purified Hla but not the inactive HlaH35L induced up-regulation of β1 integrin expression in MCs in a dose-dependent manner. Our data support a model in which S. aureus counter-reacts the extracellular microbicidal mechanisms of MCs by increasing expression of fibronectin-binding proteins and by inducing Hla-ADAM10-mediated up-regulation of β1 integrin in MCs. The up-regulation of bacterial fibronectin-binding proteins, concomitantly with the increased expression of its receptor β1 integrin on the MCs, resulted in enhanced S. aureus internalization through the binding of fibronectin-binding proteins to integrin β1 via fibronectin.
Collapse
Affiliation(s)
- Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Lorena Tuchscherr
- Institute of Medical Microbiology, University Hospital of Jena, Erlanger Allee 101, 07747, Jena, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| |
Collapse
|
48
|
Lee IJ, Hilliard BA, Ulas M, Yu D, Vangala C, Rao S, Lee J, Gadegbeku CA, Cohen PL. Monocyte and plasma expression of TAM ligand and receptor in renal failure: Links to unregulated immunity and chronic inflammation. Clin Immunol 2015; 158:231-41. [PMID: 25814173 DOI: 10.1016/j.clim.2015.01.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 01/30/2015] [Accepted: 01/31/2015] [Indexed: 12/29/2022]
Abstract
Chronic inflammation is increased in patients with chronic kidney disease (CKD) and contributes to cardiovascular morbidity and mortality. Specific immune mechanisms and pathways that drive and maintain chronic inflammation in CKD are not well described. The TAM ligands (Gas6 and protein S) and receptors (Axl and Mer) have been recently recognized as playing a prominent role in immune regulation. The receptors exist in both soluble and cell-bound forms; the soluble receptors (sAxl and sMer) are believed to compete with the bound receptors and thus inhibit their function. In this study, we determined the expression of cell-bound and soluble TAM proteins in patients with CKD. CKD patients had significantly lower expression of Mer in monocytes, yet increased expression of soluble TAM receptors sAxl and sMer in plasma compared to controls. The metalloproteinase ADAM 17, responsible for cleavage of Mer to its soluble form, was increased in patient monocytes. Elevated levels of soluble TAM receptors were more evident in patients with progressive renal failure. These observations suggest that functional deficiency of TAM receptor-mediated regulation of inflammation may contribute to chronic inflammation in patients with CKD.
Collapse
Affiliation(s)
- Iris J Lee
- Section of Nephrology, Department of Medicine, Temple University, Philadelphia, PA, USA.
| | - Brendan A Hilliard
- Section of Rheumatology, Department of Medicine, Temple University, Philadelphia, PA, USA
| | - Mehriban Ulas
- Section of Rheumatology, Department of Medicine, Temple University, Philadelphia, PA, USA
| | - Daohai Yu
- Department of Clinical Sciences, Temple University, Philadelphia, PA, USA
| | - Chandan Vangala
- School of Medicine, Temple University, Philadelphia, PA, USA
| | - Swati Rao
- Section of Nephrology, Department of Medicine, Temple University, Philadelphia, PA, USA
| | - Jean Lee
- Section of Nephrology, Department of Medicine, Temple University, Philadelphia, PA, USA
| | - Crystal A Gadegbeku
- Section of Nephrology, Department of Medicine, Temple University, Philadelphia, PA, USA
| | - Philip L Cohen
- Section of Rheumatology, Department of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
49
|
Amendola RS, Martin ACBM, Selistre-de-Araújo HS, Paula-Neto HA, Saldanha-Gama R, Barja-Fidalgo C. ADAM9 disintegrin domain activates human neutrophils through an autocrine circuit involving integrins and CXCR2. J Leukoc Biol 2015; 97:951-962. [PMID: 25765677 DOI: 10.1189/jlb.3a0914-455r] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 02/04/2015] [Accepted: 02/18/2015] [Indexed: 12/30/2022] Open
Abstract
ADAM9 is a member of the ADAM family whose expression positively correlates with tumor progression. Besides the metalloprotease activity, ADAM9D interacts with different integrins, modulating cell-adhesion events. Previous studies pointed to an important role for neutrophils in tumor development, as the inhibition of neutrophil migration or depletion of this immune cell impairs tumor growth. However, our understanding of the molecular mechanisms involved in this process, as well as the main key players acting on neutrophils, is very limited. Here, we investigated the possible modulatory effects of ADAM9D on human neutrophil functions. Our results show that ADAM9D promotes neutrophil activation and chemotaxis in a process that depends on the engagement of αvβ3 and α9β1 integrins and on the activation of PI3K/Akt and MAPK signaling pathway. ADAM9D impairs migration of neutrophils toward fMLP, LTB4, and IL-8 as classic chemoattractants. This effect is blocked by PTX, a G(i)PCR inhibitor. Furthermore, CXCR2 antagonists RPTX and SB225002 also impaired neutrophil chemotaxis in response to ADAM9D, suggesting a hierarchical cross-talk of integrins with CXCR2. Our results indicate that ADAM9D activates neutrophil functions and may be implicated in the inflammatory events associated with cancer and other disorders.
Collapse
Affiliation(s)
- Rafael S Amendola
- *Laboratório de Farmacologia Celular e Molecular, Departamento de Biologia Celular, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil; and Laboratório de Bioquímica e Biologia Molecular, Departamento de Ciências Fisiológicas, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Ana Carolina B M Martin
- *Laboratório de Farmacologia Celular e Molecular, Departamento de Biologia Celular, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil; and Laboratório de Bioquímica e Biologia Molecular, Departamento de Ciências Fisiológicas, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Heloísa S Selistre-de-Araújo
- *Laboratório de Farmacologia Celular e Molecular, Departamento de Biologia Celular, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil; and Laboratório de Bioquímica e Biologia Molecular, Departamento de Ciências Fisiológicas, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Heitor A Paula-Neto
- *Laboratório de Farmacologia Celular e Molecular, Departamento de Biologia Celular, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil; and Laboratório de Bioquímica e Biologia Molecular, Departamento de Ciências Fisiológicas, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Roberta Saldanha-Gama
- *Laboratório de Farmacologia Celular e Molecular, Departamento de Biologia Celular, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil; and Laboratório de Bioquímica e Biologia Molecular, Departamento de Ciências Fisiológicas, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Christina Barja-Fidalgo
- *Laboratório de Farmacologia Celular e Molecular, Departamento de Biologia Celular, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil; and Laboratório de Bioquímica e Biologia Molecular, Departamento de Ciências Fisiológicas, Universidade Federal de São Carlos, São Carlos, Brazil
| |
Collapse
|
50
|
Cui L, Gao Y, Xie Y, Wang Y, Cai Y, Shao X, Ma X, Li Y, Ma G, Liu G, Cheng W, Liu Y, Liu T, Pan Q, Tao H, Liu Z, Zhao B, Shao Y, Li K. An ADAM10 promoter polymorphism is a functional variant in severe sepsis patients and confers susceptibility to the development of sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2015; 19:73. [PMID: 25888255 PMCID: PMC4373036 DOI: 10.1186/s13054-015-0796-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 02/09/2015] [Indexed: 12/17/2022]
Abstract
Introduction Although genetic variants of the A disintegrin and metalloproteinase 10 (ADAM10) gene have been shown to be associated with susceptibility to several inflammatory-related diseases, to date little is known about the clinical relationship in the development of sepsis. Methods Two genetic variants in the promoter of ADAM10 were selected to analyze the potential association with the risk of sepsis. A total of 440 sepsis patients and 450 matched healthy individuals in two independent Chinese Han population were enrolled. Pyrosequencing and polymerase chain reaction-length polymorphism was used to determine the genotypes of the rs514049 and rs653765. A real-time qPCR method was used to detect the mRNA level of ADAM10. Enzyme-linked immunosorbent assay was used to measure the expression levels of substrates CX3CL1, interleukin (IL)-6R, tumor necrosis factor alpha (TNF-α), and the pro-inflammatory cytokines IL-1β and IL-6. Luciferase assay was used to analyze the activities of the promoter haplotypes of ADAM10. Results No statistically significant differences between sepsis cases and controls in the genotype or allele frequencies were observed, suggesting that ADAM10 single nucleotide polymorphisms (SNPs) may not be risk factors for the occurrence of sepsis. A significant difference in the genotype and allele frequencies of the rs653765 SNP between patients with sepsis subtype and severe sepsis (P = 0.0014) or severe sepsis/sepsis shock (P = 0.0037) were observed. Moreover, the rs653765 CC genotype in severe sepsis showed a higher ADAM10 level compared to healthy groups, and the rs653765 CC polymorphism had a strong impact on the production of the ADAM10 substrates CX3CL1, IL-6R and TNF-α. Furthermore, the functional assay showed that ADAM10 C-A haplotype carriers exhibited significantly higher reporter activity compared with the T-A carriers and T-C carriers in human acute monocytic leukemia cell line. Conclusions Our data initially indicated the ADAM10 rs653765 polymorphism was associated with the development of severe sepsis; the risk CC genotype could functionally affect the expression level of ADAM10 mRNA and was accompanied by the up-regulation of its substrates. Thus, ADAM10 might be clinically important and play a critical role in the pathogenesis of the development of sepsis, with potentially important therapeutic implications. Electronic supplementary material The online version of this article (doi:10.1186/s13054-015-0796-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Yan Gao
- The Intensive Care Unit, the Forth Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Yuliu Xie
- The Intensive Care Unit, Affiliated Hospital of Guangdong Medical College, Zhanjiang, PR China.
| | - Yan Wang
- Clinical Research Center of Guangdong Medical College, Affiliated Hospital of Guangdong Medical College, Zhanjiang, PR China.
| | - Yujie Cai
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Xin Shao
- The Intensive Care Unit, Affiliated Hospital of Guangdong Medical College, Zhanjiang, PR China.
| | - Xiaotang Ma
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - You Li
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Guoda Ma
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Gen Liu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Wanwen Cheng
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Yu Liu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Tingting Liu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Qunwen Pan
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Hua Tao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Zhou Liu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Bin Zhao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| | - Yiming Shao
- The Intensive Care Unit, Affiliated Hospital of Guangdong Medical College, Zhanjiang, PR China.
| | - Keshen Li
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Renmin street south 57, Xiashan district, Zhanjiang City, 524001, Guangdong Province, PR China.
| |
Collapse
|