1
|
Ahmad S, Deep G, Punzi HA, Su Y, Singh S, Kumar A, Mishra S, Saha AK, Wright KN, VonCannon JL, Dell'Italia LJ, Meredith WJ, Ferrario CM. Chymase Activity in Plasma and Urine Extracellular Vesicles in Primary Hypertension. KIDNEY360 2024; 5:1613-1622. [PMID: 39172521 DOI: 10.34067/kid.0000000000000555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024]
Abstract
Key Points
Blood and urine extracellular vesicles isolated from hypertensive patients possess high chymase enzymatic activity.Chymase activity was significantly higher in small extracellular vesicles obtained from hypertensive patients with suboptimal BP control.
Background
Circulating extracellular vesicles (EVs) carry protected cargoes of nucleic acids, proteins, and metabolites. In this study, we identified and validated the surface proteins and enzymatic activity of chymase, angiotensin converting enzymes 1 (ACE) and 2 (ACE2), and neprilysin (NEP) in EVs isolated from the blood and urine of primary hypertensive patients.
Methods
Peripheral venous blood and spot urine from 34 hypertensive patients were processed to isolate plasma and urinary EVs. Immunogold labeling and transmission electron microscopy validated the presence of the exosomal marker protein CD63 on the surface of plasma and urinary EVs. Flow cytometry characterized plasma and urinary EVs for CD63, CD9, and CD81 surface markers. In addition, exosomal CD63, TSG101, and Alix were analyzed in urine by western blotting. Urinary EVs did not express the endoplasmic reticulum protein calnexin and Golgi protein GM130. Chymase, ACE, ACE2, and NEP activities on 125I substrates—angiotensin-(1–12) (Ang-[1–12]) and angiotensin II—(1 nmol/L each) were quantified by HPLC. Data were analyzed based on whether the patient's BP was controlled (group 1: <140/80 mm Hg) or noncontrolled (group 2: ≥140/80 mm Hg).
Results
Chymase activity on Ang-(1–12) was significantly higher in plasma and urinary EVs than in ACE, ACE2, and NEP. In addition, chymase activity in urine EVs was more than three-fold higher than in plasma EVs. Chymase activity increased in plasma and urine EVs retrieved from group 2 patients. No comparable differences were found in the enzymatic activities of ACE, ACE2, and NEP urinary EVs between group 1 and group 2.
Conclusions
These studies reveal a differential enzymatic activity of renin angiotensin system enzymes in plasma and urine EVs isolated from hypertensive patients. Demonstrating a comparatively high chymase enzymatic activity in EVs expands a previously documented finding of increased plasma Ang-(1–12) in hypertensive patients.
Collapse
Affiliation(s)
- Sarfaraz Ahmad
- Laboratory of Translational Hypertension, Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Gagan Deep
- Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
- J Paul Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Henry A Punzi
- Punzi Medical Center, Carrollton, Texas
- UT Southwestern Medical Center, Dallas, Texas
| | - Yixin Su
- Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Sangeeta Singh
- Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Ashish Kumar
- Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Shalini Mishra
- Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Amit K Saha
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Kendra N Wright
- Laboratory of Translational Hypertension, Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Jessica L VonCannon
- Laboratory of Translational Hypertension, Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Louis J Dell'Italia
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
- Department of Veterans Affairs, Birmingham Veterans Affairs Health Care System, Birmingham, Alabama
| | - Wayne J Meredith
- Laboratory of Translational Hypertension, Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Carlos M Ferrario
- Laboratory of Translational Hypertension, Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina
| |
Collapse
|
2
|
Salati NA, Sharma M, Rao NN, Shetty SS, Radhakrishnan RA. Role of osteopontin in oral epithelial dysplasia, oral submucous fibrosis and oral squamous cell carcinoma. J Oral Maxillofac Pathol 2023; 27:706-714. [PMID: 38304518 PMCID: PMC10829450 DOI: 10.4103/jomfp.jomfp_492_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/27/2022] [Accepted: 03/06/2023] [Indexed: 02/03/2024] Open
Abstract
Background Inflammatory cells and cytokines in the chronically injured mucosa promote fibrosis in the oral submucous fibrosis (OSF) fibrotic milieu. Osteopontin (OPN) is a wound-healing mediator that upregulates the inflammatory response and is involved in the malignancy and fibrosis of multiple organ systems. Objectives We investigated the expression of OPN in oral potentially malignant disorders (OPMDs) and oral squamous cell carcinomas (OSCCs) to determine its role in the malignant transformation and fibrosis of oral tissues. The expression of OPN in OPMDs and OSCCs was compared and correlated, and the role of OPN as a fibrotic mediator in OSF was explained. Study Design A total of 30 cases of normal mucosa and OPMDs (mild dysplasia, severe dysplasia, OSF and OSCCs) were studied by purposive sampling. In these groups, OPN immunoreactivity was examined and correlated with clinical findings. Results In mild dysplasia, OPN expression was restricted to the basal cell layer with moderate staining intensity. In severe dysplasia, it was extremely intense and extended throughout the epithelium. In the OSF, OPN expression was moderate in the perinuclear areas of the basal cell layer. The expression of OPN was very strong in OSCC. A flow diagram explaining the profibrotic role of OPN in OSF has been provided. Conclusion A positive role of OPN in both pathogenesis and malignant transformation of OPMDs and OSCC has been demonstrated.
Collapse
Affiliation(s)
- Nasir A. Salati
- Department of Oral and Maxillofacial Pathology, Dr. Ziauddin Ahmad Dental College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohit Sharma
- Department of Oral Pathology, SGT Dental College Hospital and Research Institute, Gurugram, Haryana, India
| | - Nirmala N. Rao
- Former Dean, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Smitha S. Shetty
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Raghu A. Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
3
|
Nuri T, Jin D, Takai S, Ueda K. Tryptase-Positive Mast Cells Promote Adipose Fibrosis in Secondary Lymphedema through PDGF. Curr Issues Mol Biol 2023; 45:8027-8039. [PMID: 37886950 PMCID: PMC10605118 DOI: 10.3390/cimb45100507] [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: 09/05/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
Lymphedema is a chronic and progressive condition that causes physical disfigurement and psychological trauma due to the accumulation of lymphatic fluid in the interstitial space. Once it develops, lymphedema is difficult to treat because it leads to the fibrosis of adipose tissue. However, the mechanism behind this remains unclear. The purpose of this study was to investigate the involvement of mast cells (MCs) in the adipose tissues of patients with lymphedema. We found that fibrosis spread through blood vessels in the adipose tissues of lymphedema patients, and the expression of the collagen I and III genes was significantly increased compared to that of those in normal adipose tissue. Immunostaining of vimentin and α-smooth muscle actin showed that fibroblasts were the main cellular components in severely fibrotic regions. Toluidine blue staining confirmed a significant increase in the number of MCs in the adipose tissues of lymphedema patients, and immunostaining of serial sections of adipose tissue showed a significant increase in the number of tryptase-positive cells in lymphedema tissues compared with those in normal adipose tissues. Linear regression analyses revealed significant positive correlations between tryptase and the expressions of the TNF-α, platelet-derived growth factor (PDGF)-A, and PDGFR-α genes. PDGF-A-positive staining was observed in both fibroblasts and granules of tryptase-positive MCs. These results suggest that MC-derived tryptase plays a role in the fibrosis of adipose tissue due to lymphedema directly or in cooperation with other mediators.
Collapse
Affiliation(s)
- Takashi Nuri
- Department of Plastic and Reconstructive Surgery, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Japan;
| | - Denan Jin
- Department of Innovative Medicine, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Japan; (D.J.); (S.T.)
| | - Shinji Takai
- Department of Innovative Medicine, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Japan; (D.J.); (S.T.)
| | - Koichi Ueda
- Department of Plastic and Reconstructive Surgery, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Japan;
| |
Collapse
|
4
|
Ferrario CM, Ahmad S, Speth R, Dell’Italia LJ. Is chymase 1 a therapeutic target in cardiovascular disease? Expert Opin Ther Targets 2023; 27:645-656. [PMID: 37565266 PMCID: PMC10529260 DOI: 10.1080/14728222.2023.2247561] [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: 03/13/2023] [Accepted: 08/09/2023] [Indexed: 08/12/2023]
Abstract
INTRODUCTION Non-angiotensin converting enzyme mechanisms of angiotensin II production remain underappreciated in part due to the success of current therapies to ameliorate the impact of primary hypertension and atherosclerotic diseases of the heart and the blood vessels. This review scrutinize the current literature to highlight chymase role as a critical participant in the pathogenesis of cardiovascular disease and heart failure. AREAS COVERED We review the contemporaneous understanding of circulating and tissue biotransformation mechanisms of the angiotensins focusing on the role of chymase as an alternate tissue generating pathway for angiotensin II pathological mechanisms of action. EXPERT OPINION While robust literature documents the singularity of chymase as an angiotensin II-forming enzyme, particularly when angiotensin converting enzyme is inhibited, this knowledge has not been fully recognized to clinical medicine. This review discusses the limitations of clinical trials' that explored the benefits of chymase inhibition in accounting for the failure to duplicate in humans what has been demonstrated in experimental animals.
Collapse
Affiliation(s)
- Carlos M Ferrario
- Laboratory of Translational Hypertension and Vascular Research, Department of Surgery, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Sarfaraz Ahmad
- Laboratory of Translational Hypertension and Vascular Research, Department of Surgery, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Robert Speth
- Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, Florida 33314
| | - Louis J Dell’Italia
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB), Birmingham AL 35294
- Birmingham Department of Veterans Affairs Health Care System, Birmingham AL 35233
| |
Collapse
|
5
|
van der Elst G, Varol H, Hermans M, Baan CC, Duong-van Huyen JP, Hesselink DA, Kramann R, Rabant M, Reinders MEJ, von der Thüsen JH, van den Bosch TPP, Clahsen-van Groningen MC. The mast cell: A Janus in kidney transplants. Front Immunol 2023; 14:1122409. [PMID: 36891297 PMCID: PMC9986315 DOI: 10.3389/fimmu.2023.1122409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
Mast cells (MCs) are innate immune cells with a versatile set of functionalities, enabling them to orchestrate immune responses in various ways. Aside from their known role in allergy, they also partake in both allograft tolerance and rejection through interaction with regulatory T cells, effector T cells, B cells and degranulation of cytokines and other mediators. MC mediators have both pro- and anti-inflammatory actions, but overall lean towards pro-fibrotic pathways. Paradoxically, they are also seen as having potential protective effects in tissue remodeling post-injury. This manuscript elaborates on current knowledge of the functional diversity of mast cells in kidney transplants, combining theory and practice into a MC model stipulating both protective and harmful capabilities in the kidney transplant setting.
Collapse
Affiliation(s)
- G van der Elst
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - H Varol
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - M Hermans
- Department of Internal Medicine, Division of Allergy and Clinical Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - C C Baan
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | - D A Hesselink
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - R Kramann
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany.,Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - M Rabant
- Department of Pathology, Necker Hospital, APHP, Paris, France
| | - M E J Reinders
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - J H von der Thüsen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - T P P van den Bosch
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - M C Clahsen-van Groningen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
6
|
Paul Owens E, Grania Healy H, Andrew Vesey D, Elizabeth Hoy W, Carolyn Gobe G. Targeted biomarkers of progression in chronic kidney disease. Clin Chim Acta 2022; 536:18-28. [PMID: 36041551 DOI: 10.1016/j.cca.2022.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Chronic kidney disease (CKD) is an increasingly significant health issue worldwide. Early stages of CKD can be asymptomatic and disease trajectory difficult to predict. Not everyone with CKD progresses to kidney failure, where kidney replacement therapy is the only life-sustaining therapy. Predicting which patients will progress to kidney failure would allow better use of targeted treatments and more effective allocation of health resources. Current diagnostic tests to identify patients with progressive disease perform poorly but there is a suite of new and emerging predictive biomarkers with great clinical promise. METHODS This narrative review describes new and emerging biomarkers of pathophysiologic processes of CKD development and progression, accessible in blood or urine liquid biopsies. Biomarkers were selected based on their reported pathobiological functions in kidney injury, inflammation, oxidative stress, repair and fibrosis. Biomarker function and evidence of involvement in CKD development and progression are reported. CONCLUSION Many biomarkers reviewed here have received little attention to date, perhaps because of conflicting conclusions of their utility in CKD. The functional roles of the selected biomarkers in the underlying pathobiology of progression of CKD are a powerful rationale for advancing and validating these molecules as prognosticators and predictors of CKD trajectory.
Collapse
Affiliation(s)
- Evan Paul Owens
- NHMRC CKD CRE (CKD.QLD), The University of Queensland, Brisbane 4072, Australia; Faculty of Medicine, The University of Queensland, Brisbane 4072, Australia; Kidney Disease Research Collaborative, Translational Research Institute, Brisbane 4102, Australia
| | - Helen Grania Healy
- NHMRC CKD CRE (CKD.QLD), The University of Queensland, Brisbane 4072, Australia; Faculty of Medicine, The University of Queensland, Brisbane 4072, Australia; Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane 4029, Australia
| | - David Andrew Vesey
- NHMRC CKD CRE (CKD.QLD), The University of Queensland, Brisbane 4072, Australia; Faculty of Medicine, The University of Queensland, Brisbane 4072, Australia
| | - Wendy Elizabeth Hoy
- NHMRC CKD CRE (CKD.QLD), The University of Queensland, Brisbane 4072, Australia; Faculty of Medicine, The University of Queensland, Brisbane 4072, Australia; Centre for Chronic Disease, The University of Queensland, Brisbane 4072, Australia
| | - Glenda Carolyn Gobe
- NHMRC CKD CRE (CKD.QLD), The University of Queensland, Brisbane 4072, Australia; Faculty of Medicine, The University of Queensland, Brisbane 4072, Australia; Kidney Disease Research Collaborative, Translational Research Institute, Brisbane 4102, Australia.
| |
Collapse
|
7
|
Toyama S, Tominaga M, Takamori K. Connections between Immune-Derived Mediators and Sensory Nerves for Itch Sensation. Int J Mol Sci 2021; 22:12365. [PMID: 34830245 PMCID: PMC8624544 DOI: 10.3390/ijms222212365] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/09/2021] [Accepted: 11/15/2021] [Indexed: 12/28/2022] Open
Abstract
Although histamine is a well-known itch mediator, histamine H1-receptor blockers often lack efficacy in chronic itch. Recent molecular and cellular based studies have shown that non-histaminergic mediators, such as proteases, neuropeptides and cytokines, along with their cognate receptors, are involved in evocation and modulation of itch sensation. Many of these molecules are produced and secreted by immune cells, which act on sensory nerve fibers distributed in the skin to cause itching and sensitization. This understanding of the connections between immune cell-derived mediators and sensory nerve fibers has led to the development of new treatments for itch. This review summarizes current knowledge of immune cell-derived itch mediators and neuronal response mechanisms, and discusses therapeutic agents that target these systems.
Collapse
Affiliation(s)
- Sumika Toyama
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Tomioka, Chiba 279-0021, Japan; (S.T.); (M.T.)
| | - Mitsutoshi Tominaga
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Tomioka, Chiba 279-0021, Japan; (S.T.); (M.T.)
- Anti-Aging Skin Research Laboratory, Juntendo University Graduate School of Medicine, 2-1-1 Tomioka, Chiba 279-0021, Japan
| | - Kenji Takamori
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Tomioka, Chiba 279-0021, Japan; (S.T.); (M.T.)
- Anti-Aging Skin Research Laboratory, Juntendo University Graduate School of Medicine, 2-1-1 Tomioka, Chiba 279-0021, Japan
- Department of Dermatology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Chiba 279-0021, Japan
| |
Collapse
|
8
|
Yan H, Xu J, Xu Z, Yang B, Luo P, He Q. Defining therapeutic targets for renal fibrosis: Exploiting the biology of pathogenesis. Biomed Pharmacother 2021; 143:112115. [PMID: 34488081 DOI: 10.1016/j.biopha.2021.112115] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/11/2021] [Accepted: 08/19/2021] [Indexed: 01/14/2023] Open
Abstract
Renal fibrosis is a failed wound-healing process of the kidney tissue after chronic, sustained injury, which is a common pathway and pathological marker of virtually every type of chronic kidney disease (CKD), regardless of cause. However, there is a lack of effective treatment specifically targeting against renal fibrosis per se to date. The main pathological feature of renal fibrosis is the massive activation and proliferation of renal fibroblasts and the excessive synthesis and secretion of extracellular matrix (ECM) deposited in the renal interstitium, leading to structural damage, impairment of renal function, and eventually end-stage renal disease. In this review, we summarize recent advancements regarding the participation and interaction of many types of kidney residents and infiltrated cells during renal fibrosis, attempt to comprehensively discuss the mechanism of renal fibrosis from the cellular level and conclude by highlighting novel therapeutic targets and approaches for development of new treatments for patients with renal fibrosis.
Collapse
Affiliation(s)
- Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiangxin Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
9
|
Ferrario CM, Groban L, Wang H, Cheng CP, VonCannon JL, Wright KN, Sun X, Ahmad S. The Angiotensin-(1-12)/Chymase axis as an alternate component of the tissue renin angiotensin system. Mol Cell Endocrinol 2021; 529:111119. [PMID: 33309638 PMCID: PMC8127338 DOI: 10.1016/j.mce.2020.111119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/18/2020] [Accepted: 12/06/2020] [Indexed: 02/08/2023]
Abstract
The identification of an alternate extended form of angiotensin I composed of the first twelve amino acids at the N-terminal of angiotensinogen has generated new knowledge of the importance of noncanonical mechanisms for renin independent generation of angiotensins. The human sequence of the dodecapeptide angiotensin-(1-12) [N-Asp1-Arg2-Val3-Tyr4-Ile5-His6-Pro7-Phe8-His9-Leu10-Val1-Ile12-COOH] is an endogenous substrate that in the rat has been documented to be present in multiple organs including the heart, brain, kidney, gut, adrenal gland, and the bone marrow. Newer studies have confirmed the existence of Ang-(1-12) as an Ang II-forming substrate in the blood and heart of normal and diseased patients. Studies to-date document that angiotensin II generation from angiotensin-(1-12) does not require renin participation while chymase rather than angiotensin converting enzyme shows high catalytic activity in converting this tissue substrate into angiotensin II directly.
Collapse
Affiliation(s)
- Carlos M Ferrario
- Department of Surgery and Physiology-Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA.
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Che Ping Cheng
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Jessica L VonCannon
- Department of Surgery and Physiology-Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Kendra N Wright
- Department of Surgery and Physiology-Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Xuming Sun
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Sarfaraz Ahmad
- Department of Surgery and Physiology-Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| |
Collapse
|
10
|
Rossing P, Strand J, Avogaro A, Becka M, Kanefendt F, Otto C. Effects of the chymase inhibitor fulacimstat in diabetic kidney disease—results from the CADA DIA trial. Nephrol Dial Transplant 2020; 36:2263-2273. [DOI: 10.1093/ndt/gfaa299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Indexed: 12/25/2022] Open
Abstract
Abstract
Background
The protease chymase generates multiple factors involved in tissue remodelling including angiotensin II (Ang II) and has been implicated in the pathophysiology of diabetic kidney disease (DKD). This study investigated the effects of the chymase inhibitor fulacimstat on albuminuria in patients with Type II diabetes mellitus and a clinical diagnosis of DKD.
Methods
In this double-blind, randomized, placebo-controlled trial, patients were on the maximum tolerated dose of either an Ang II receptor blocker or an Ang-converting enzyme inhibitor since at least 3 months before the screening visit. Eligible patients were randomized in a 2:1 ratio to treatment with either 25 mg fulacimstat (n = 99) or placebo (n = 48) twice daily on top of standard of care.
Results
The randomized patients had a mean urine albumin–creatinine ratio (UACR) of 131 mg/g (range: 29–2429 mg) and a mean (standard deviation) estimated glomerular filtration rate of 60.8 ± 16.9 mL/min/1.73 m2 before treatment start. Fulacimstat was safe and well tolerated, and achieved mean total trough concentrations that were ∼9-fold higher than those predicted to be required for minimal therapeutic activity. UACR increased by 27.4% [coefficient of variation (CV) 86%] and 3% (CV 88.9%) after 24 weeks of treatment with placebo or fulacimstat, respectively. Analysis of covariance revealed a least square mean UACR ratio (fulacimstat/placebo) of 0.804 (90% CI 0.627–1.030, P = 0.1477), indicating a statistically non-significant UACR reduction of 19.6% after fulacimstat treatment compared with placebo.
Conclusions
Fulacimstat was safe and well tolerated but did not reduce albuminuria in patients with DKD. These findings do not support a therapeutic role for chymase inhibition in DKD.
Collapse
Affiliation(s)
- Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte and University of Copenhagen, Copenhagen, Denmark
| | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| | - Michael Becka
- Research and Clinical Sciences Statistics, Bayer AG, Wuppertal, Germany
| | | | - Christiane Otto
- Experimental Medicine Cardiovascular, Bayer AG, Wuppertal, Germany
| |
Collapse
|
11
|
Piotrowska A, Chmielewska M, Andrzejewski W, Dziegiel P, Podhorska-Okolow M. Influence of Angiotensin II on cell viability and apoptosis in rat renal proximal tubular epithelial cells in in vitro studies. J Renin Angiotensin Aldosterone Syst 2020; 21:1470320320949850. [PMID: 32962526 PMCID: PMC7649907 DOI: 10.1177/1470320320949850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Introduction: Angiotensin II (Ang II) is multifunctional peptide that plays an important role in blood pressure regulation and maintenance electrolyte homeostasis. It shows biological effects by activating two main receptors: AT1 and AT2. The aim of the present work was to investigate the effect of Ang II on NRK-52E cells in in vitro studies. Furthermore, an attempt was made to determine the effectiveness of the AT1 and AT2 receptor blocker activity (respectively, losartan and PD123319). Methods: The study was carried out using adherent NRK-52E cell line. Immunofluorescence and Western Blot method were used to confirm the presence of AT1 and AT2 receptors in the cells. The SRB and MTT tests showed decrease in the viability of NRK-52E cells incubated with Ang II in comparison to the control (without Ang II). Results: The blockade of the AT1 receptor caused an increase in cell viability in comparison to cells incubated with Ang II only. The blockade of AT2 receptor also triggered statistically significant increase in cell viability in comparison with cells only exposed to Ang II. Combined administration of blockers for both receptors (losartan and PD123319) decreased Ang II cytotoxicity against NRK-52E cell line. The apoptosis was only observed in cells incubated with Ang II in comparison with control cells. However, simultaneous use of both blockers caused statistically significant decrease in apoptosis. Conclusions: The result of our study indicates that Ang II causes damaging effect on NRK-52E cells by directing them to programmed cell death. It seems that not only does the AT2 receptor itself play an important role in the induction of apoptosis, but also its interaction with AT1 receptor does as well.
Collapse
Affiliation(s)
- Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Poland
| | - Magdalena Chmielewska
- Amphibian Biology Group, Department of Evolutionary Biology and Conservation of Vertebrates, Faculty of Biological Sciences, University of Wroclaw, Poland
| | - Waldemar Andrzejewski
- Department of Physiotherapy, Wroclaw University School of Physical Education, Poland
| | - Piotr Dziegiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Poland.,Department of Physiotherapy, Wroclaw University School of Physical Education, Poland
| | | |
Collapse
|
12
|
Stanchev S, Landzhov B, Kotov G, Stamenov N, Dikov T, Iliev A. The potential role of mast cells and fibroblast growth factor-2 in the development of hypertension-induced renal damage. Acta Histochem 2020; 122:151599. [PMID: 32778237 DOI: 10.1016/j.acthis.2020.151599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/21/2022]
Abstract
Hypertension-induced renal injury is a multifactorial process which plays a crucial role in the development of chronic kidney disease. Multiple studies have demonstrated that interstitial rather than glomerular changes correlate better with renal functional capacity. Recent evidence indicates that mast cells and cell signaling proteins such as fibroblast growth factor-2 may contribute to the progression of interstitial changes under hypertensive conditions. The aim of our study was to determine the localization of mast cells in the renal cortex and report on the changes in their number, to analyze the distribution of fibroblast growth factor-2, to assess the extent of renal fibrosis and to evaluate renal damage and correlate it with the changes in the number of mast cells in a model of hypertension-induced renal injury by comparing two age groups of spontaneously hypertensive rats. We used 6- and 12-month-old animals. A light microscopic study was conducted on sections stained with hematoxylin and eosin, periodic acid-Schiff stain, Mallory's trichrome method and toluidine blue. For the immunohistochemical study we used monoclonal antibodies against mast cell tryptase and fibroblast growth factor-2 and a polyclonal antibody against c-kit. The expression of fibroblast growth factor-2 was assessed semi-quantitatively. The number of mast cells was evaluated on toluidine blue-, tryptase- and c-kit-stained sections, as well as double-stained sections and a comparative statistical analysis with the Mann-Whitney test was conducted between the two age groups. Our results showed that mast cells were located mainly in the peritubular and perivascular areas and were absent in the region of the renal corpuscles. Their number increased significantly in 12-month-old animals. Immunostaining for tryptase, c-kit and double staining for both molecules yielded identical results. The immunohistochemical expression of fibroblast growth factor-2 increased in the kidneys of older animals, as did the percentage of collagen fibers. In addition, we described more severe renal damage in 12-month-old spontaneously hypertensive rats and noted a positive correlation in both age groups between the number of mast cells on the one hand and glomerular sclerosis index and tubulointerstitial damage index, on the other. The results obtained in the present study support the pivotal role of mast cells in the development of hypertension-induced kidney damage.
Collapse
|
13
|
Kaltenecker CC, Domenig O, Kopecky C, Antlanger M, Poglitsch M, Berlakovich G, Kain R, Stegbauer J, Rahman M, Hellinger R, Gruber C, Grobe N, Fajkovic H, Eskandary F, Böhmig GA, Säemann MD, Kovarik JJ. Critical Role of Neprilysin in Kidney Angiotensin Metabolism. Circ Res 2020; 127:593-606. [PMID: 32418507 DOI: 10.1161/circresaha.119.316151] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RATIONALE Kidney homeostasis is critically determined by the coordinated activity of the renin-angiotensin system (RAS), including the balanced synthesis of its main effector peptides Ang (angiotensin) II and Ang (1-7). The condition of enzymatic overproduction of Ang II relative to Ang (1-7) is termed RAS dysregulation and leads to cellular signals, which promote hypertension and organ damage, and ultimately progressive kidney failure. ACE2 (angiotensin-converting enzyme 2) and NEP (neprilysin) induce the alternative, and potentially reno-protective axis by enhancing Ang (1-7) production. However, their individual contribution to baseline RAS balance and whether their activities change in chronic kidney disease (CKD) has not yet been elucidated. OBJECTIVE To examine whether NEP-mediated Ang (1-7) generation exceeds Ang II formation in the healthy kidney compared with diseased kidney. METHODS AND RESULTS In this exploratory study, we used liquid chromatography-tandem mass spectrometry to measure Ang II and Ang (1-7) synthesis rates of ACE, chymase and NEP, ACE2, PEP (prolyl-endopeptidase), PCP (prolyl-carboxypeptidase) in kidney biopsy homogenates in 11 healthy living kidney donors, and 12 patients with CKD. The spatial expression of RAS enzymes was determined by immunohistochemistry. Healthy kidneys showed higher NEP-mediated Ang (1-7) synthesis than Ang II formation, thus displaying a strong preference towards the reno-protective alternative RAS axis. In contrast, in CKD kidneys higher levels of Ang II were recorded, which originated from mast cell chymase activity. CONCLUSIONS Ang (1-7) is the dominant RAS peptide in healthy human kidneys with NEP rather than ACE2 being essential for its generation. Severe RAS dysregulation is present in CKD dictated by high chymase-mediated Ang II formation. Kidney RAS enzyme analysis might lead to novel therapeutic approaches for CKD.
Collapse
Affiliation(s)
- Christopher C Kaltenecker
- From the Division of Nephrology and Dialysis, Department of Internal Medicine III (C.C.K., F.E., G.A.B., J.J.K.), Medical University of Vienna, Austria
| | - Oliver Domenig
- Attoquant Diagnostics GmbH, Vienna, Austria (O.D., M.P.)
| | - Chantal Kopecky
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia (C.K.)
| | - Marlies Antlanger
- 2nd Department of Internal Medicine, Kepler University Hospital, Med Campus III, Linz, Austria (M.A.)
| | | | - Gabriela Berlakovich
- Division of Transplantation, Department of Surgery (G.B.), Medical University of Vienna, Austria
| | - Renate Kain
- Department of Pathology (R.K.), Medical University of Vienna, Austria
| | - Johannes Stegbauer
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany (J.S., M.R.)
| | - Masudur Rahman
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany (J.S., M.R.)
| | - Roland Hellinger
- Center for Physiology and Pharmacology (R.H., C.G.), Medical University of Vienna, Austria
| | - Christian Gruber
- Center for Physiology and Pharmacology (R.H., C.G.), Medical University of Vienna, Austria
| | - Nadja Grobe
- Renal Research Institute, New York, NY (N.G.)
| | - Harun Fajkovic
- Department of Urology (H.F.), Medical University of Vienna, Austria
| | - Farsad Eskandary
- From the Division of Nephrology and Dialysis, Department of Internal Medicine III (C.C.K., F.E., G.A.B., J.J.K.), Medical University of Vienna, Austria
| | - Georg A Böhmig
- From the Division of Nephrology and Dialysis, Department of Internal Medicine III (C.C.K., F.E., G.A.B., J.J.K.), Medical University of Vienna, Austria
| | - Marcus D Säemann
- 6th Medical Department with Nephrology and Dialysis, Wilhelminenhospital, Vienna, Austria (M.D.S.).,Sigmund-Freud University, Vienna, Austria (M.D.S.)
| | - Johannes J Kovarik
- From the Division of Nephrology and Dialysis, Department of Internal Medicine III (C.C.K., F.E., G.A.B., J.J.K.), Medical University of Vienna, Austria
| |
Collapse
|
14
|
Succar J, Giatsidis G, Yu N, Hassan K, Khouri R, Gurish MF, Pejler G, Åbrink M, Orgill DP. Mouse Mast Cell Protease-4 Recruits Leukocytes in the Inflammatory Phase of Surgically Wounded Skin. Adv Wound Care (New Rochelle) 2019; 8:469-475. [PMID: 31456904 DOI: 10.1089/wound.2018.0898] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/10/2019] [Indexed: 01/29/2023] Open
Abstract
Objective: Mouse mast cell protease-4 (mMCP-4, also known as chymase) has both pro- and anti-inflammatory roles depending on the disease model. However, its effects have not been studied in surgically wounded skin. Given the significant clinical applications of modulating the inflammatory response in wound healing, we examined the role of mMCP-4 and the effect of its inhibitor chymostatin on leukocyte and polymorphonuclear cell (PMN) recruitment in our skin model. Approach: Recruitment was assessed on day-1 postwounding of three groups of mice (n = 10 each): mMCP-4 null mice, wild-type (WT) mice treated with the mMCP-4 inhibitor chymostatin, and WT with no other intervention. Leukocytes were stained with CD-45 cell marker, and PMN cells were stained with chloroacetate esterase. Results: The WT mice had 27 ± 9 leukocytes per field compared with 11 ± 6 for the mMCP-4 nulls, a decrease of 60% (p = 0.03), whereas the chymostatin-injected group had a count comparable with the uninjected WT controls at 24 ± 9. The WT group had a PMN count of 96 ± 12 cells, compared with just 24 ± 8 in the mMCP-4 null group, a decrease of 75% (p = 0.001), whereas the chymostatin-treated group had 60 ± 18 cells, a decrease of 38% compared with the WT group (p = 0.03). Innovation: We showed that the inflammatory process can be influenced by impeding the arrival of PMNs into the surgically injured site using the mMCP-4 inhibitor chymostatin. Conclusion: Chymase contributes to the recruitment of white blood cells in surgically wounded skin.
Collapse
Affiliation(s)
- Julien Succar
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Giorgio Giatsidis
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Nanze Yu
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Kazi Hassan
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Roger Khouri
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Michael F. Gurish
- Human Immunology Center, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Magnus Åbrink
- Section of Immunology, Department of Biomedical Sciences & Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Dennis Paul Orgill
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Abstract
During degranulation, mast cells secrete a specific set of mediators defined as "secretome" including the preformed mediators that have already been synthesized by a cell and contained in the cytoplasmic granules. This group includes serine proteases, in particular, chymase and tryptase. Biological significance of chymase depends on the mechanisms of degranulation and is characterized by selective effects on the cellular and non-cellular components of the specific tissue microenvironment. Chymase is known to be closely involved in the mechanisms of inflammation and allergy, angiogenesis, and oncogenesis, remodeling of the extracellular matrix of the connective tissue and changes in organ histoarchitectonics. Number of chymase-positive mast cells in the intra-organ population, and the mechanisms of biogenesis and secretome degranulation appear to be the informative criteria for interpreting the state of the internal organs, characterizing not only the diagnostic efficacy but also the properties of targets of pharmacotherapy. In this review, we discussed the current state of knowledge about mast cell chymase as one of the mast cell secretome proteases. Main issues of the reviewed publications are highlighted with our microscopic images of mast cell chymase visualized using immunohistochemical staining.
Collapse
|
16
|
Kopel J, Brower GL. Impact of fossil fuel emissions and particulate matter on pulmonary health. Proc (Bayl Univ Med Cent) 2019; 32:636-638. [PMID: 31656449 DOI: 10.1080/08998280.2019.1641367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/02/2019] [Accepted: 07/05/2019] [Indexed: 01/23/2023] Open
Abstract
In recent decades, several national and international legislative efforts have aimed to improve air quality standards and limit major pollutants, such as carbon monoxide, sulfur dioxide, and nitrogen dioxide, linked to several public health problems. In recent years, particulate matter sources have become an important cause of several pulmonary and systemic diseases. Specifically, several studies examining cigarette smoke particulates have discovered the important contribution that mast cells play in the pathogenesis and progression of smoking-related lung disease and other particulate matter-related lung injury. By understanding the mechanisms of activation and signaling cascades involved in cigarette smoke and mast cell activation, novel pharmacological therapies for particulate matter-induced lung diseases could be developed.
Collapse
Affiliation(s)
- Jonathan Kopel
- School of Medicine, Texas Tech University Health Sciences CenterLubbockTexas
| | - Gregory L Brower
- Department of Medical Education, Texas Tech University Health Sciences CenterLubbockTexas
| |
Collapse
|
17
|
Hiroyasu S, Turner CT, Richardson KC, Granville DJ. Proteases in Pemphigoid Diseases. Front Immunol 2019; 10:1454. [PMID: 31297118 PMCID: PMC6607946 DOI: 10.3389/fimmu.2019.01454] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 06/10/2019] [Indexed: 12/28/2022] Open
Abstract
Pemphigoid diseases are a subgroup of autoimmune skin diseases characterized by widespread tense blisters. Standard of care typically involves immunosuppressive treatments, which may be insufficient and are often associated with significant adverse events. As such, a deeper understanding of the pathomechanism(s) of pemphigoid diseases is necessary in order to identify improved therapeutic approaches. A major initiator of pemphigoid diseases is the accumulation of autoantibodies against proteins at the dermal-epidermal junction (DEJ), followed by protease activation at the lesion. The contribution of proteases to pemphigoid disease pathogenesis has been investigated using a combination of in vitro and in vivo models. These studies suggest proteolytic degradation of anchoring proteins proximal to the DEJ is crucial for dermal-epidermal separation and blister formation. In addition, proteases can also augment inflammation, expose autoantigenic cryptic epitopes, and/or provoke autoantigen spreading, which are all important in pemphigoid disease pathology. The present review summarizes and critically evaluates the current understanding with respect to the role of proteases in pemphigoid diseases.
Collapse
Affiliation(s)
- Sho Hiroyasu
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| | - Christopher T. Turner
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| | - Katlyn C. Richardson
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| | - David J. Granville
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| |
Collapse
|
18
|
Frossi B, Mion F, Sibilano R, Danelli L, Pucillo CEM. Is it time for a new classification of mast cells? What do we know about mast cell heterogeneity? Immunol Rev 2019; 282:35-46. [PMID: 29431204 DOI: 10.1111/imr.12636] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mast cells (MCs) are derived from committed precursors that leave the hematopoietic tissue, migrate in the blood, and colonize peripheral tissues where they terminally differentiate under microenvironment stimuli. They are distributed in almost all vascularized tissues where they act both as immune effectors and housekeeping cells, contributing to tissue homeostasis. Historically, MCs were classified into 2 subtypes, according to tryptic enzymes expression. However, MCs display a striking heterogeneity that reflects a complex interplay between different microenvironmental signals delivered by various tissues, and a differentiation program that decides their identity. Moreover, tissue-specific MCs show a trained memory, which contributes to shape their function in a specific microenvironment. In this review, we summarize the current state of our understanding of MC heterogeneity that reflects their different tissue experiences. We describe the discovery of unique cell molecules that can be used to distinguish specific MC subsets in vivo, and discuss how the improved ability to recognize these subsets provided new insights into the biology of MCs. These recent advances will be helpful for the understanding of the specific role of individual MC subsets in the control of tissue homeostasis, and in the regulation of pathological conditions such as infection, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Barbara Frossi
- Department of Medicine, University of Udine, Udine, Italy
| | - Francesca Mion
- Department of Medicine, University of Udine, Udine, Italy
| | - Riccardo Sibilano
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Luca Danelli
- Retroviral Immunology, The Francis Crick Institute, London, UK
| | | |
Collapse
|
19
|
Hsieh WY, Chang TH, Chang HF, Chuang WH, Lu LC, Yang CW, Lin CS, Chang CC. Renal chymase-dependent pathway for angiotensin II formation mediated acute kidney injury in a mouse model of aristolochic acid I-induced acute nephropathy. PLoS One 2019; 14:e0210656. [PMID: 30633770 PMCID: PMC6329531 DOI: 10.1371/journal.pone.0210656] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/28/2018] [Indexed: 01/20/2023] Open
Abstract
Angiotensin-converting enzyme (ACE) is the primary enzyme that converts angiotensin I (Ang I) to angiotensin II (Ang II) in the renin-angiotensin system (RAS). However, chymase hydrates Ang I to Ang II independently of ACE in some kidney diseases, and it may play an important role. The present study investigated whether chymase played a crucial role in aristolochic acid I (AAI)-induced nephropathy. C57BL/6 mice were treated with AAI via intraperitoneal injection for an accumulated AAI dosage of 45 mg/kg body weight (BW) (15 mg/kg BW per day for 3 days). The animals were sacrificed after acute kidney injury development, and blood, urine and kidneys were harvested for biochemical and molecular assays. Mice exhibited increased serum creatinine, BUN and urinary protein after the AAI challenge. Significant infiltrating inflammatory cells and tubular atrophy were observed in the kidneys, and high immunocytokine levels were detected. Renal RAS-related enzyme activities were measured, and a significantly increased chymase activity and slightly decreased ACE activity were observed in the AAI-treated mice. The renal Ang II level reflected the altered profile of RAS enzymes and was significantly increased in AAI-treated mice. Treatment of AAI-induced nephropathic mice with an ACE inhibitor (ACEI) or chymase inhibitor (CI; chymostatin) reduced renal Ang II levels. The combination of ACEI and CI (ACEI+CI) treatment significantly reversed the AAI-induced changes of Ang II levels and kidney inflammation and injuries. AAI treatment significantly increased renal p-MEK without increasing p-STAT3 and p-Smad3 levels, and p-MEK/p-ERK1/2 signalling pathway was significantly activated. CI and ACEI+CI treatments reduced this AAI-activated signaling pathway. AAI-induced nephropathy progression was significantly mitigated with CI and ACEI+CI treatment. This study elucidates the role of RAS in the pathogenesis of AAI-induced nephropathy.
Collapse
Affiliation(s)
- Wen-Yeh Hsieh
- Division of Pulmonary Medicine, Department of Internal Medicine, Hsinchu Mackay Memorial Hospital, Hsinchu, Taiwan
| | - Teng-Hsiang Chang
- Division of Nephrology, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Hui-Fang Chang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Division of Endocrinology, Department of Internal Medicine, Hsinchu Mackay Memorial Hospital, Hsinchu, Taiwan
| | - Wan-Hsuan Chuang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Li-Che Lu
- Division of Nephrology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Chung-Wei Yang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Chih-Sheng Lin
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu, Taiwan
| | - Chia-Chu Chang
- Division of Nephrology, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
- School of Medicine, Chung-Shan Medical University, Taichung, Taiwan
- Department of Environmental and Precision Medicine Laboratory, Changhua Christian Hospital, Changhua, Taiwan
- Department of Nutrition, Hungkuang University, Taichung, Taiwan
- Department of Internal Medicine, Kuang Tien General Hospital, Taichung, Taiwan
| |
Collapse
|
20
|
Meng XM. Inflammatory Mediators and Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:381-406. [PMID: 31399975 DOI: 10.1007/978-981-13-8871-2_18] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Renal inflammation is the initial, healthy response to renal injury. However, prolonged inflammation promotes the fibrosis process, which leads to chronic pathology and eventually end-stage kidney disease. There are two major sources of inflammatory cells: first, bone marrow-derived leukocytes that include neutrophils, macrophages, fibrocytes and mast cells, and second, locally activated kidney cells such as mesangial cells, podocytes, tubular epithelial cells, endothelial cells and fibroblasts. These activated cells produce many profibrotic cytokines and growth factors that cause accumulation and activation of myofibroblasts, and enhance the production of the extracellular matrix. In particular, activated macrophages are key mediators that drive acute inflammation into chronic kidney disease. They produce large amounts of profibrotic factors and modify the microenvironment via a paracrine effect, and they also transdifferentiate to myofibroblasts directly, although the origin of myofibroblasts in the fibrosing kidney remains controversial. Collectively, understanding inflammatory cell functions and mechanisms during renal fibrosis is paramount to improving diagnosis and treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Xiao-Ming Meng
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
21
|
Ansary TM, Urushihara M, Fujisawa Y, Nagata S, Urata H, Nakano D, Hirofumi H, Kitamura K, Kagami S, Nishiyama A. Effects of the selective chymase inhibitor TEI-F00806 on the intrarenal renin-angiotensin system in salt-treated angiotensin I-infused hypertensive mice. Exp Physiol 2018; 103:1524-1531. [PMID: 30137655 DOI: 10.1113/ep087209] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022]
Abstract
NEW FINDINGS What is the central question of this study? Can chymase inhibition prevent angiotensin I-induced hypertension through inhibiting the conversion of angiotensin I to angiotensin II in the kidney? What is the main finding and its importance? Treatment with TEI-F00806 decreased angiotensin II content of the kidney, renal cortical angiotensinogen protein levels and chymase mRNA expression, and attenuated the development of hypertension. ABSTRACT The effects of the selective chymase inhibitor TEI-F00806 were examined on angiotensin I (Ang I)-induced hypertension and intrarenal angiotensin II (Ang II) production in salt-treated mice. Twelve-week-old C57BL male mice were given a high-salt diet (4% NaCl + saline (0.9% NaCl)), and divided into three groups: (1) sham + vehicle (5% acetic acid in saline), (2) Ang I (1 μg kg-1 min-1 , s.c.) + vehicle, and (3) Ang I + TEI-F00806 (100 mg kg-1 day-1 , p.o.) (n = 8-10 per group). Systolic blood pressure was measured weekly using a tail-cuff method. Kidney Ang II content was measured by radioimmunoassay. Chronic infusion of Ang I resulted in the development of hypertension (P < 0.001), and augmented intrarenal chymase gene expression (P < 0.05), angiotensinogen protein level (P < 0.001) and Ang II content (P < 0.01) in salt-treated mice. Treatment with TEI-F00806 attenuated the development of hypertension (P < 0.001) and decreased Ang II content of the kidney (P < 0.05), which was associated with reductions in renal cortical angiotensinogen protein levels (P < 0.001) and chymase mRNA expression (P < 0.05). These data suggest that a chymase inhibitor decreases intrarenal renin-angiotensin activity, thereby reducing salt-dependent hypertension.
Collapse
Affiliation(s)
- Tuba M Ansary
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Maki Urushihara
- Department of Pediatrics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yoshihide Fujisawa
- Life Science Research Center, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Sayaka Nagata
- Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hidenori Urata
- Department of Cardiovascular Diseases, Fukuoka University Chikushi Hospital, Fukuoka, Japan
| | - Daisuke Nakano
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hitomi Hirofumi
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kazuo Kitamura
- Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Shoji Kagami
- Department of Pediatrics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
22
|
Qiu M, Li S, Jin L, Feng P, Kong Y, Zhao X, Lin Y, Xu Y, Li C, Wang W. Combination of Chymostatin and Aliskiren attenuates ER stress induced by lipid overload in kidney tubular cells. Lipids Health Dis 2018; 17:183. [PMID: 30064425 PMCID: PMC6069859 DOI: 10.1186/s12944-018-0818-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/09/2018] [Indexed: 12/24/2022] Open
Abstract
Background Lipotoxicity plays an important role in the pathogenesis of kidney injury. Our previous study demonstrated that activation of local renin-angiotensin system (RAS) was involved in saturated free fatty acids palmitic acid (PA)-induced tubular cell injuries. The current study aims to investigate whether suppression of RAS by combination of direct renin inhibitor aliskiren and noncanonical RAS pathway chymase inhibitor chymostatin attenuates PA or cholesterol induced-endoplasmic reticulum stress (ER stress) and apopotosis in cultured human proximal tubular HK2 cells. Methods HK2 cells were treated with saturated fatty acid PA (0.6 mM) for 24 h or cholesterol (10 μg/ml) for 6d with or without chymostatin and/or aliskiren. Expressions of the ER stress associated proteins and apoptosis markers were detected by western blotting. The mRNA levels of RAS components were measured by real-time qPCR. Results Combination treatment of chymostatin and aliskiren markedly suppressed PA or cholesterol-induced ER stress, as reflected by increased BiP, IRE1α, phosphorylated-eIF2α and ATF4 as well as proapoptotic transcription factor CHOP. The ratio of Bax/Bcl-2 and cleaved caspase-3, two markers of apoptosis were upregulated by PA or cholesterol treatment. PA treatment was also associated with increased levels of angiotensinogen and angiotensin type 1 receptor (AT1R) mRNA expression. Combination treatment of chymostatin and aliskiren markedly suppressed PA or cholesterol-induced ER stress and apoptosis. The protective effect of two inhibitors was also observed in primary cultured cortical tubular cells treated with PA. In contrast, chymostatin and/or aliskiren failed to prevent ER stress induced by tunicamycin. Conclusions These results suggested that combination treatment of chymostatin and aliskiren attenuates lipid-induced renal tubular cell injury, likely through suppressing activation of intracellular RAS. Electronic supplementary material The online version of this article (10.1186/s12944-018-0818-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Miaojuan Qiu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, 74# Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Suchun Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, 74# Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Lizi Jin
- Department of Cardiology, The 5th Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Pinning Feng
- Department of Clinical Laboratory, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yonglun Kong
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, 74# Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Xiaoduo Zhao
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, 74# Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Yu Lin
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yunyun Xu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, 74# Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Chunling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, 74# Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Weidong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, 74# Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|
23
|
Chang HF, Sun YL, Yeh FY, Tseng IH, Chang CC, Lin CS. Detection of chymase activity using a specific peptide probe conjugated onto gold nanoparticles. RSC Adv 2018; 8:29013-29021. [PMID: 35547971 PMCID: PMC9084417 DOI: 10.1039/c8ra04322a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/27/2018] [Indexed: 02/03/2023] Open
Abstract
The gold nanoparticles (AuNPs) peptide probe functionalized with specific peptide sequences was developed for the sensitive and efficient detection of chymase activity.
Collapse
Affiliation(s)
- Hui-Fang Chang
- Department of Biological Science and Technology
- National Chiao Tung University
- Hsinchu
- Taiwan
- Division of Endocrinology
| | - Yu-Ling Sun
- Aquatic Technology Laboratories
- Agricultural Technology Research Institute
- Hsinchu
- Taiwan
| | - Fang-Yuan Yeh
- Department of Biological Science and Technology
- National Chiao Tung University
- Hsinchu
- Taiwan
| | - I-Hua Tseng
- Department of Biological Science and Technology
- National Chiao Tung University
- Hsinchu
- Taiwan
| | - Chia-Chu Chang
- Department of Internal Medicine
- Changhua Christian Hospital
- Changhua
- Taiwan
- Department of Environmental and Precision Medicine Laboratory
| | - Chih-Sheng Lin
- Department of Biological Science and Technology
- National Chiao Tung University
- Hsinchu
- Taiwan
| |
Collapse
|
24
|
Di S, Ziyou Y, Liu NF. Pathological Changes of Lymphedematous Skin: Increased Mast Cells, Related Proteases, and Activated Transforming Growth Factor-β1. Lymphat Res Biol 2016; 14:162-71. [PMID: 27599355 DOI: 10.1089/lrb.2016.0010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Sun Di
- Department of Plastic and Reconstructive Surgery, Lymphology Centre, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Ziyou
- Department of Plastic and Reconstructive Surgery, Lymphology Centre, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning-Fei Liu
- Department of Plastic and Reconstructive Surgery, Lymphology Centre, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Karnik SS, Unal H, Kemp JR, Tirupula KC, Eguchi S, Vanderheyden PML, Thomas WG. International Union of Basic and Clinical Pharmacology. XCIX. Angiotensin Receptors: Interpreters of Pathophysiological Angiotensinergic Stimuli [corrected]. Pharmacol Rev 2015; 67:754-819. [PMID: 26315714 PMCID: PMC4630565 DOI: 10.1124/pr.114.010454] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The renin angiotensin system (RAS) produced hormone peptides regulate many vital body functions. Dysfunctional signaling by receptors for RAS peptides leads to pathologic states. Nearly half of humanity today would likely benefit from modern drugs targeting these receptors. The receptors for RAS peptides consist of three G-protein-coupled receptors—the angiotensin II type 1 receptor (AT1 receptor), the angiotensin II type 2 receptor (AT2 receptor), the MAS receptor—and a type II trans-membrane zinc protein—the candidate angiotensin IV receptor (AngIV binding site). The prorenin receptor is a relatively new contender for consideration, but is not included here because the role of prorenin receptor as an independent endocrine mediator is presently unclear. The full spectrum of biologic characteristics of these receptors is still evolving, but there is evidence establishing unique roles of each receptor in cardiovascular, hemodynamic, neurologic, renal, and endothelial functions, as well as in cell proliferation, survival, matrix-cell interaction, and inflammation. Therapeutic agents targeted to these receptors are either in active use in clinical intervention of major common diseases or under evaluation for repurposing in many other disorders. Broad-spectrum influence these receptors produce in complex pathophysiological context in our body highlights their role as precise interpreters of distinctive angiotensinergic peptide cues. This review article summarizes findings published in the last 15 years on the structure, pharmacology, signaling, physiology, and disease states related to angiotensin receptors. We also discuss the challenges the pharmacologist presently faces in formally accepting newer members as established angiotensin receptors and emphasize necessary future developments.
Collapse
Affiliation(s)
- Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Hamiyet Unal
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Jacqueline R Kemp
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Kalyan C Tirupula
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Satoru Eguchi
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Patrick M L Vanderheyden
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Walter G Thomas
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| |
Collapse
|
26
|
Wang R, Chen J, Zhang Z, Cen Y. Role of chymase in the local renin-angiotensin system in keloids: inhibition of chymase may be an effective therapeutic approach to treat keloids. Drug Des Devel Ther 2015; 9:4979-88. [PMID: 26357464 PMCID: PMC4560513 DOI: 10.2147/dddt.s87842] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background Histologically, keloids contain excess fibroblasts and an overabundance of dermal collagen. Recently, it was reported that chymase induced a profibrotic response via transforming growth factor-β1 (TGF-β1)/Smad activation in keloid fibroblasts (KFs). However, the role of chymase in the local renin-angiotensin system (RAS) in keloids has not been elucidated. This study aims to determine whether chymase plays an important role in the local RAS in keloids. Methods We compared the expression and activity of chymase in keloids and normal skin tissues using Western blotting and radioimmunoassay, and studied the expression of TGF-β1, interleukin-1β, collagen I, hydroxyproline, and angiotensin II in KFs after chymase and inhibitors’ treatment. Results The results revealed an increased activity of chymase in keloid tissues, and that chymase enhanced the expression of angiotensin II, collagen I, TGF-β1, and interleukin-1β in KFs. Blockade of the chymase pathway involved in the local RAS lowered the expression of these signaling factors. Conclusion This research suggests that inhibition of chymase might be an effective therapeutic approach to improve the clinical treatment of keloids.
Collapse
Affiliation(s)
- Ru Wang
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Junjie Chen
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Zhenyu Zhang
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Ying Cen
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
27
|
Kritikou E, Kuiper J, Kovanen PT, Bot I. The impact of mast cells on cardiovascular diseases. Eur J Pharmacol 2015; 778:103-15. [PMID: 25959384 DOI: 10.1016/j.ejphar.2015.04.050] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 04/10/2015] [Accepted: 04/21/2015] [Indexed: 12/30/2022]
Abstract
Mast cells comprise an innate immune cell population, which accumulates in tissues proximal to the outside environment and, upon activation, augments the progression of immunological reactions through the release and diffusion of either pre-formed or newly generated mediators. The released products of mast cells include histamine, proteases, as well as a variety of cytokines, chemokines and growth factors, which act on the surrounding microenvironment thereby shaping the immune responses triggered in various diseased states. Mast cells have also been detected in the arterial wall and are implicated in the onset and progression of numerous cardiovascular diseases. Notably, modulation of distinct mast cell actions using genetic and pharmacological approaches highlights the crucial role of this cell type in cardiovascular syndromes. The acquired evidence renders mast cells and their mediators as potential prognostic markers and therapeutic targets in a broad spectrum of pathophysiological conditions related to cardiovascular diseases.
Collapse
Affiliation(s)
- Eva Kritikou
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Johan Kuiper
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Ilze Bot
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
28
|
DeBruin EJ, Gold M, Lo BC, Snyder K, Cait A, Lasic N, Lopez M, McNagny KM, Hughes MR. Mast cells in human health and disease. Methods Mol Biol 2015; 1220:93-119. [PMID: 25388247 DOI: 10.1007/978-1-4939-1568-2_7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Mast cells are primarily known for their role in defense against pathogens, particularly bacteria; neutralization of venom toxins; and for triggering allergic responses and anaphylaxis. In addition to these direct effector functions, activated mast cells rapidly recruit other innate and adaptive immune cells and can participate in "tuning" the immune response. In this review we touch briefly on these important functions and then focus on some of the less-appreciated roles of mast cells in human disease including cancer, autoimmune inflammation, organ transplant, and fibrosis. Although it is difficult to formally assign causal roles to mast cells in human disease, we offer a general review of data that correlate the presence and activation of mast cells with exacerbated inflammation and disease progression. Conversely, in some restricted contexts, mast cells may offer protective roles. For example, the presence of mast cells in some malignant or cardiovascular diseases is associated with favorable prognosis. In these cases, specific localization of mast cells within the tissue and whether they express chymase or tryptase (or both) are diagnostically important considerations. Finally, we review experimental animal models that imply a causal role for mast cells in disease and discuss important caveats and controversies of these findings.
Collapse
Affiliation(s)
- Erin J DeBruin
- Department of Experimental Medicine, The Biomedical Research Centre, The University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Inflammatory and neuroinflammatory processes are increasingly recognized as critical pathophysiologic steps in the development of multiple chronic diseases and in the etiology of persistent pain and depression. Mast cells are immune cells now viewed as cellular sensors in inflammation and immunity. When stimulated, mast cells release an array of mediators to orchestrate an inflammatory response. These mediators can directly initiate tissue responses on resident cells, and may also regulate the activity of other immune cells, including central microglia. New evidence supports the involvement of peripheral and central mast cells in the development of pain processes as well as in the transition from acute, to chronic and neuropathic pain. That behavioral and endocrine states can increase the number and activation of peripheral and brain mast cells suggests that mast cells represent the immune cells that peripherally and centrally coordinate inflammatory processes in neuropsychiatric diseases such as depression and anxiety which are associated with chronic pelvic pain. Given that increasing evidence supports the activated mast cell as a director of common inflammatory pathways/mechanisms contributing to chronic and neuropathic pelvic pain and comorbid neuropsychiatric diseases, mast cells may be considered a viable target for the multifactorial management of both pain and depression.
Collapse
|
30
|
Michaud A, Acharya KR, Masuyer G, Quenech'du N, Gribouval O, Morinière V, Gubler MC, Corvol P. Absence of cell surface expression of human ACE leads to perinatal death. Hum Mol Genet 2014; 23:1479-91. [PMID: 24163131 PMCID: PMC3929087 DOI: 10.1093/hmg/ddt535] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 10/22/2013] [Indexed: 11/14/2022] Open
Abstract
Renal tubular dysgenesis (RTD) is a recessive autosomal disease characterized most often by perinatal death. It is due to the inactivation of any of the major genes of the renin-angiotensin system (RAS), one of which is the angiotensin I-converting enzyme (ACE). ACE is present as a tissue-bound enzyme and circulates in plasma after its solubilization. In this report, we present the effect of different ACE mutations associated with RTD on ACE intracellular trafficking, secretion and enzymatic activity. One truncated mutant, R762X, responsible for neonatal death was found to be an enzymatically active, secreted form, not inserted in the plasma membrane. In contrast, another mutant, R1180P, was compatible with life after transient neonatal renal insufficiency. This mutant was located at the plasma membrane and rapidly secreted. These results highlight the importance of tissue-bound ACE versus circulating ACE and show that the total absence of cell surface expression of ACE is incompatible with life. In addition, two missense mutants (W594R and R828H) and two truncated mutants (Q1136X and G1145AX) were also studied. These mutants were neither inserted in the plasma membrane nor secreted. Finally, the structural implications of these ACE mutations were examined by molecular modelling, which suggested some important structural alterations such as disruption of intra-molecular non-covalent interactions (e.g. salt bridges).
Collapse
Affiliation(s)
- Annie Michaud
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), 11 Place Marcelin Berthelot, Paris F-75005, France
- INSERM U 1050, Paris F-75005, France
- MEMOLIFE Laboratory of Excellence and Paris Sciences Lettres and
| | - K. Ravi Acharya
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Geoffrey Masuyer
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Nicole Quenech'du
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), 11 Place Marcelin Berthelot, Paris F-75005, France
- INSERM U 1050, Paris F-75005, France
- MEMOLIFE Laboratory of Excellence and Paris Sciences Lettres and
| | - Olivier Gribouval
- INSERM U983, Hôpital Necker-Enfants Malades, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Vincent Morinière
- AP-HP, Département de Génétique, Centre de Référence MARHEA, Hôpital Necker-Enfants Malades, Paris, France and
- Centre de Référence des Maladies Rénales Héréditaires de L'Enfant et de L'Adulte, Paris, France
| | - Marie-Claire Gubler
- INSERM U983, Hôpital Necker-Enfants Malades, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de L'Enfant et de L'Adulte, Paris, France
| | - Pierre Corvol
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), 11 Place Marcelin Berthelot, Paris F-75005, France
- INSERM U 1050, Paris F-75005, France
- MEMOLIFE Laboratory of Excellence and Paris Sciences Lettres and
| |
Collapse
|