1
|
Zhang L, Feng Q, Kong W. ECM Microenvironment in Vascular Homeostasis: New Targets for Atherosclerosis. Physiology (Bethesda) 2024; 39:0. [PMID: 38984789 DOI: 10.1152/physiol.00028.2023] [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: 11/20/2023] [Revised: 03/05/2024] [Accepted: 03/23/2024] [Indexed: 07/11/2024] Open
Abstract
Alterations in vascular extracellular matrix (ECM) components, interactions, and mechanical properties influence both the formation and stability of atherosclerotic plaques. This review discusses the contribution of the ECM microenvironment in vascular homeostasis and remodeling in atherosclerosis, highlighting Cartilage oligomeric matrix protein (COMP) and its degrading enzyme ADAMTS7 as examples, and proposes potential avenues for future research aimed at identifying novel therapeutic targets for atherosclerosis based on the ECM microenvironment.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qianqian Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
2
|
Rajalingam A, Ganjiwale A. Identification of common genetic factors and immune-related pathways associating more than two autoimmune disorders: implications on risk, diagnosis, and treatment. Genomics Inform 2024; 22:10. [PMID: 38956704 PMCID: PMC11221123 DOI: 10.1186/s44342-024-00004-5] [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: 09/21/2023] [Accepted: 12/22/2023] [Indexed: 07/04/2024] Open
Abstract
Autoimmune disorders (ADs) are chronic conditions resulting from failure or breakdown of immunological tolerance, resulting in the host immune system attacking its cells or tissues. Recent studies report shared effects, mechanisms, and evolutionary origins among ADs; however, the possible factors connecting them are unknown. This study attempts to identify gene signatures commonly shared between different autoimmune disorders and elucidate their molecular pathways linking the pathogenesis of these ADs using an integrated gene expression approach. We employed differential gene expression analysis across 19 datasets of whole blood/peripheral blood cell samples with five different autoimmune disorders (rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, Crohn's disease, and type 1 diabetes) to get nine key genes-EGR1, RUNX3, SMAD7, NAMPT, S100A9, S100A8, CYBB, GATA2, and MCEMP1 that were primarily involved in cell and leukocyte activation, leukocyte mediated immunity, IL-17, AGE-RAGE signaling in diabetic complications, prion disease, and NOD-like receptor signaling confirming its role in immune-related pathways. Combined with biological interpretations such as gene ontology (GO), pathway enrichment, and protein-protein interaction (PPI) network, our current study sheds light on the in-depth research on early detection, diagnosis, and prognosis of different ADs.
Collapse
Affiliation(s)
- Aruna Rajalingam
- Department of Life Science, Bangalore University, Bangalore, Karnataka, 560056, India
| | - Anjali Ganjiwale
- Department of Life Science, Bangalore University, Bangalore, Karnataka, 560056, India.
| |
Collapse
|
3
|
Liu Z, Wang J, Guo F, Xu T, Yu F, Deng Q, Tan W, Duan S, Song L, Wang Y, Sun J, Zhou L, Wang Y, Zhou X, Xia H, Jiang H. Role of S100β in Unstable Angina Pectoris: Insights from Quantitative Flow Ratio. Arch Med Res 2024; 55:103034. [PMID: 38972195 DOI: 10.1016/j.arcmed.2024.103034] [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: 11/24/2023] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND AND OBJECTIVE Disturbed autonomic nervous system (ANS) may promote inflammatory, immune, and oxidative stress responses, which may increase the risk of acute coronary events. S100β has been proposed as a biomarker of neuronal injury that would provide an insightful understanding of the crosstalk between the ANS, immune-inflammatory cells, and plaques that drive atherosclerosis. This study investigates the correlation between S100β, and functional coronary stenosis as determined by quantitative flow ratio (QFR). METHODS Patients with unstable angina pectoris (UAP) scheduled for coronary angiography and QFR were retrospectively enrolled. Serum S100β levels were determined by enzyme-linked immunosorbent assay. The Gensini score was used to estimate the extent of atherosclerotic lesions and the cumulative sum of three-vessel QFR (3V-QFR) was calculated to estimate the total atherosclerotic burden. RESULTS Two hundred thirty-three patients were included in this study. Receiver operator characteristic (ROC) curve indicated that S100β>33.28 pg/mL predicted functional ischemia in patients with UAP. Multivariate logistic analyses showed that a higher level of S100β was independently correlated with a functional ischemia-driven target vessel (QFR ≤0.8). This was also closely correlated with the severity of coronary lesions, as measured by the Gensini score (OR = 5.058, 95% CI: 2.912-8.793, p <0.001). According to 3V-QFR, S100β is inversely associated with total atherosclerosis burden (B = -0.002, p <0.001). CONCLUSIONS S100β was elevated in the functional ischemia stages of UAP. It was independently associated with coronary lesion severity as assessed by Gensini score and total atherosclerosis burden as estimated by 3V-QFR in patients with UAP.
Collapse
Affiliation(s)
- Zhihao Liu
- Department of Cardiology, Wuhan No.1 Hospital, Wuhan, Hubei, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Jun Wang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Fuding Guo
- Department of Cardiology, Yan'an Hospital, Kunming Medical University, Kunming, China
| | - Tianyou Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Fu Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Qiang Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Wuping Tan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Shoupeng Duan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Lingpeng Song
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Yijun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Ji Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Yueyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, Hubei, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China.
| |
Collapse
|
4
|
Reshadmanesh T, Behnoush AH, Farajollahi M, Khalaji A, Ghondaghsaz E, Ahangar H. Circulating Levels of Calprotectin as a Biomarker in Patients With Coronary Artery Disease: A Systematic Review and Meta-Analysis. Clin Cardiol 2024; 47:e24315. [PMID: 38961752 PMCID: PMC11222710 DOI: 10.1002/clc.24315] [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: 04/05/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Calprotectin, also known as MRP8/14, is generated by immune cells and is altered in several inflammatory diseases. Studies have assessed their levels in patients with coronary artery disease (CAD) and its subtypes (stable CAD and acute coronary syndrome [ACS]). Herein, we aimed to systematically investigate these associations through a systematic review and meta-analysis. METHODS A systematic search was conducted in four online databases, including PubMed, Scopus, Embase, and the Web of Science. Relevant studies were retrieved, screened, and extracted. Random-effect meta-analysis was performed for the calculation of standardized mean difference (SMD) and 95% confidence interval (CI). Blood calprotectin levels were compared between CAD patients and controls, as well as CAD subtypes. RESULTS A total of 20 studies were included in the systematic review and meta-analysis, comprising 3300 CAD patients and 1230 controls. Patients with CAD had significantly higher calprotectin levels (SMD 0.81, 95% CI 0.32-1.30, p < 0.01). Similarly, patients with ACS were reported to have higher levels compared to those with stable CAD. However, there was no significant difference in terms of blood calprotectin levels between stable CAD cases and healthy controls. Finally, studies have shown that calprotectin could be used as a diagnostic biomarker of CAD while also predicting major adverse events and mortality in these patients. CONCLUSION Based on our findings, calprotectin, as an inflammatory marker, could be used as a possible biomarker for patients with CAD and ACS. These suggest the possibility of pathophysiological pathways for this involvement and warrant further research on these associations as well as their clinical utility.
Collapse
Affiliation(s)
| | - Amir Hossein Behnoush
- School of MedicineTehran University of Medical SciencesTehranIran
- Non‐Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences InstituteTehran University of Medical SciencesTehranIran
| | | | - Amirmohammad Khalaji
- School of MedicineTehran University of Medical SciencesTehranIran
- Non‐Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Elina Ghondaghsaz
- Undergraduate Program in NeuroscienceUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Hassan Ahangar
- Department of Cardiology, School of Medicine, Mousavi HospitalZanjan University of Medical SciencesZanjanIran
| |
Collapse
|
5
|
Chittimalli K, Adkins S, Arora S, Singh J, Jarajapu YP. An Investigation of the Inflammatory Landscape in the Brain and Bone Marrow of the APP/PS1 Mouse. J Alzheimers Dis Rep 2024; 8:981-998. [PMID: 39114548 PMCID: PMC11305850 DOI: 10.3233/adr-240024] [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: 01/30/2024] [Accepted: 05/29/2024] [Indexed: 08/10/2024] Open
Abstract
Background The APP/PS1 mouse model recapitulates pathology of human Alzheimer's disease (AD). While amyloid-β peptide deposition and neurodegeneration are features of AD, the pathology may involve inflammation and impaired vascular regeneration. Objective This study evaluated inflammatory environments in the brain and bone marrow (BM), and the impact on brain microvascular density. Methods BM and frontal cortex from male nine-month-old APP/PS1 or the control C57Bl6/j mice were studied. Vascular density and inflammatory cells were evaluated in the sections of frontal cortex by immunohistochemistry. Different subsets of hematopoietic stem/progenitor cells (BM) and monocyte-macrophages were characterized by flow cytometry and by clonogenic assays. Myelopoietic or inflammatory factors were evaluated by real-time RT-PCR or by western blotting. Results CD34+ or CD31+ vascular structures were lower (p < 0.01, n = 6) in the frontal cortex that was associated with decreased number of Lin-Sca-1+cKit+ vasculogenic progenitor cells in the BM and circulation (p < 0.02, n = 6) compared to the control. Multipotent progenitor cells MPP4, common lymphoid, common myeloid and myeloid progenitor cells were higher in the APP/PS1-BM compared to the control, which agreed with increased numbers of monocytes and pro-inflammatory macrophages. The expression of pro-myelopoietic factors and alarmins was higher in the APP/PS1 BM-HSPCs or in the BM-supernatants compared to the control. Frontal cortices of APP/PS1 mice showed higher number of pro-inflammatory macrophages (CD11b+F4/80+ or CD80+) and microglia (OX42+Iba1+). Conclusions These findings show that AD pathology in APP/PS1 mice is associated with upregulated myelopoiesis, which contributes to the brain inflammation and decreased vascularity.
Collapse
Affiliation(s)
- Kishore Chittimalli
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Stephen Adkins
- School of Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Sanjay Arora
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Yagna P.R. Jarajapu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| |
Collapse
|
6
|
Zeng W, Gao Y, Wang Q, Chi J, Zhu Z, Diao Q, Li X, Wang Z, Qu M, Shi Y. Preliminary clinical analysis and pathway study of S100A8 as a biomarker for the diagnosis of acute deep vein thrombosis. Sci Rep 2024; 14:13298. [PMID: 38858401 PMCID: PMC11164926 DOI: 10.1038/s41598-024-61728-6] [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: 01/14/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024] Open
Abstract
Herein, we aimed to identify blood biomarkers that compensate for the poor specificity of D-dimer in the diagnosis of deep vein thrombosis (DVT). S100A8 was identified by conducting protein microarray analysis of blood samples from patients with and without DVT. We used ELISA to detect S100A8, VCAM-1, and ICAM-1 expression levels in human blood and evaluated their correlations. Additionally, we employed human recombinant protein S100A8 to induce human umbilical vein endothelial cells and examined the role of the TLR4/MAPK/VCAM-1 and ICAM-1 signaling axes in the pathogenic mechanism of S100A8. Simultaneously, we constructed a rat model of thrombosis induced by inferior vena cava stenosis and detected levels of S100A8, VCAM-1, and ICAM-1 in the blood of DVT rats using ELISA. The associations of thrombus tissue, neutrophils, and CD68-positive cells with S100A8 and p38MAPK, TLR4, and VCAM-1 expression levels in vein walls were explored. The results revealed that blood S100A8 was significantly upregulated during the acute phase of DVT and activated p38MAPK expression by combining with TLR4 to enhance the expression and secretion of VCAM-1 and ICAM-1, thereby affecting the occurrence and development of DVT. Therefore, S100A8 could be a potential biomarker for early diagnosis and screening of DVT.
Collapse
Affiliation(s)
- Wenjie Zeng
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Yangyang Gao
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Qitao Wang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Junyu Chi
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ziyan Zhu
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Qingfei Diao
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Xin Li
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Zhen Wang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ming Qu
- Vascular Gland Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China.
| | - Yongquan Shi
- Department of Clinical Laboratory Center, Shandong Second Provincial General Hospital, Jinan, Shandong, China
| |
Collapse
|
7
|
Plana E, Oto J, Herranz R, Medina P, Cana F, Miralles M. Calprotectin as a new inflammatory marker of abdominal aortic aneurysm: A pilot study. Vasc Med 2024; 29:189-199. [PMID: 38457311 DOI: 10.1177/1358863x241231494] [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] [Indexed: 03/10/2024]
Abstract
INTRODUCTION Abdominal aortic aneurysm (AAA) is a relevant clinical problem due to the risk of rupture of progressively dilated infrarenal aorta. It is characterized by degradation of elastic fibers, extracellular matrix, and inflammation of the arterial wall. Though neutrophil infiltration is a known feature of AAA, markers of neutrophil activation are scarcely analyzed; hence, the main objective of this study. METHODS Plasma levels of main neutrophil activation markers were quantified in patients with AAA and a double control group (CTL) formed by healthy volunteers (HV) and patients with severe atherosclerosis submitted for carotid endarterectomy (CE). Calprotectin, a cytoplasmic neutrophil protein, was quantified, by Western blot, in arterial tissue samples from patients with AAA and organ donors. Colocalization of calprotectin and neutrophil elastase was assessed by immunofluorescence. RESULTS Plasma calprotectin and IL-6 were both elevated in patients with AAA compared with CTL (p ⩽ 0.0001) and a strong correlation was found between both molecules (p < 0.001). This difference was maintained when comparing with HV and CE for calprotectin but only with HV for IL-6. Calprotectin was also elevated in arterial tissue samples from patients with AAA compared with organ donors (p < 0.0001), and colocalized with neutrophils in the arterial wall. CONCLUSIONS Circulating calprotectin could be a specific AAA marker and a potential therapeutical target. Calprotectin is related to inflammation and neutrophil activation in arterial wall and independent of other atherosclerotic events.
Collapse
Affiliation(s)
- Emma Plana
- Angiology and Vascular Surgery Service, La Fe University and Polytechnic Hospital, Valencia, Spain
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Julia Oto
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Raquel Herranz
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Pilar Medina
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Fernando Cana
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Manuel Miralles
- Angiology and Vascular Surgery Service, La Fe University and Polytechnic Hospital, Valencia, Spain
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
- Department of Surgery, University of Valencia, Valencia, Spain
| |
Collapse
|
8
|
Zhou Y, Nomigni MT, Gaigneaux A, Tolle F, Wright HL, Bueb JL, Bréchard S. miRNA-132-5p mediates a negative feedback regulation of IL-8 secretion through S100A8/A9 downregulation in neutrophil-like HL-60 cells. Front Immunol 2024; 14:1274378. [PMID: 38292491 PMCID: PMC10824955 DOI: 10.3389/fimmu.2023.1274378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/19/2023] [Indexed: 02/01/2024] Open
Abstract
Background Neutrophils are an important source of pro-inflammatory and immunomodulatory cytokines. This makes neutrophils efficient drivers of interactions with immune and non-immune cells to maintain homeostasis and modulate the inflammatory process by notably regulating the release of cytokines. Ca2+-dependent regulatory mechanism encompassing cytokine secretion by neutrophils are not still identified. In this context, we propose to define new insights on the role of Ca2+-binding proteins S100A8/A9 and on the regulatory role of miRNA-132-5p, which was identified as a regulator of S100A8/A9 expression, on IL-8 secretion. Methods Differentiated HL-60 cells, a human promyelocytic leukemia cell line that can be induced to differentiate into neutrophil-like cells, were used as a model of human neutrophils and treated with N- formyl-methionyl-leucyl-phenylalanine (fMLF), a bacterial peptide that activates neutrophils. shRNA knockdown was used to define the role of selected targets (S100A8/A9 and miRNA-132-5p) on IL-8 secretion. Results and discussion Different types of cytokines engage different signaling pathways in the secretion process. IL-8 release is tightly regulated by Ca2+ binding proteins S100A8/A9. miRNA-132-5p is up-regulated over time upon fMLF stimulation and decreases S100A8/A9 expression and IL-8 secretion. Conclusion These findings reveal a novel regulatory loop involving S100A8/A9 and miRNA-132-5p that modulates IL-8 secretion by neutrophils in inflammatory conditions. This loop could be a potential target for therapeutic intervention in inflammatory diseases.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Milène Tetsi Nomigni
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Anthoula Gaigneaux
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Fabrice Tolle
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Helen L. Wright
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Jean-Luc Bueb
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sabrina Bréchard
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
9
|
Wang Q, Long G, Luo H, Zhu X, Han Y, Shang Y, Zhang D, Gong R. S100A8/A9: An emerging player in sepsis and sepsis-induced organ injury. Biomed Pharmacother 2023; 168:115674. [PMID: 37812889 DOI: 10.1016/j.biopha.2023.115674] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/11/2023] Open
Abstract
Sepsis, the foremost contributor to mortality in intensive care unit patients, arises from an uncontrolled systemic response to invading infections, resulting in extensive harm across multiple organs and systems. Recently, S100A8/A9 has emerged as a promising biomarker for sepsis and sepsis-induced organ injury, and targeting S100A8/A9 appeared to ameliorate inflammation-induced tissue damage and improve adverse outcomes. S100A8/A9, a calcium-binding heterodimer mainly found in neutrophils and monocytes, serves as a causative molecule with pro-inflammatory and immunosuppressive properties, which are vital in the pathogenesis of sepsis. Therefore, improving our comprehension of how S100A8/A9 acts as a pathological player in the development of sepsis is imperative for advancing research on sepsis. Our review is the first-to the best of our knowledge-to discuss the biology of S100A8/A9 and its release mechanisms, summarize recent advances concerning the vital roles of S100A8/A9 in sepsis and the consequential organ damage, and underscore its potential as a promising diagnostic biomarker and therapeutic target for sepsis.
Collapse
Affiliation(s)
- Qian Wang
- Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430023, China
| | - Gangyu Long
- Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430023, China
| | - Hong Luo
- Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430023, China
| | - Xiqun Zhu
- Hubei Cancer Hospital, Tongji Medical College, HUST, Wuhan 430079, China
| | - Yang Han
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Wuhan 430023, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HUST, Wuhan 430030, China.
| | - Dingyu Zhang
- Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430023, China; Hubei Clinical Research Center for Infectious Diseases, Wuhan 430023, China; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan 430023, China; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan 430023, China.
| | - Rui Gong
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
10
|
Ramineni P, Kamath SP, Manjrekar P, Kamath P, Mithra P, Kulkarni V. Serum calprotectin as a marker of neonatal sepsis: a hospital-based cross-sectional diagnostic study. F1000Res 2023; 12:626. [PMID: 37600908 PMCID: PMC10432886 DOI: 10.12688/f1000research.132099.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/07/2023] [Indexed: 08/22/2023] Open
Abstract
Background Despite significant advances in neonatal care, neonatal sepsis remains a major contributor to mortality, morbidity, and protracted hospitalization. The development of early possible diagnostic indicators for newborn sepsis is critical. Since calprotectin participates in major biological processes, it could be a diagnostic marker for infection/inflammation. This study aimed to estimate serum calprotectin in neonates with clinical sepsis. In addition, we compared serum calprotectin with standard sepsis markers and serum procalcitonin to evaluate its diagnostic accuracy. Methods A hospital-based cross-sectional diagnostic study of neonates identified with clinical sepsis using standard criteria was carried out. We compared estimated serum calprotectin levels to serum procalcitonin levels and conventional sepsis markers (leucocyte count, blood culture, immature to total neutrophil ratio, and C- reactive protein). We used SPSS version 25 to analyze the data. To examine diagnostic accuracy and determine a cut-off value for serum calprotectin, we used the receiver operating characteristics (ROC) curve. Results Of the 83 subjects included, 36.5% (30/83) had blood culture positive status, the median value of serum calprotectin being 0.93 ng/ml (0.67 to 1.3). Respiratory, cardiovascular, and gastrointestinal instabilities were present in 67.5% (56/83), 59% (49/83), and 50.1% (42/83) cases, respectively. The median values of serum calprotectin, procalcitonin, TLC, and I/T ratio between neonates withpositive blood culturesand negative culturesdid not differ significantly.. On ROC, calprotectin was not predictive for blood culture positivity (sensitivity: 50%; specificity: 44% at 0.83 ng/ml of serum calprotectin) and C-reactive protein (CRP) levels (sensitivity: 57%; specificity: 67% at serum calprotectin levels of 0.89 ng/ml). However, compared with serum procalcitonin, serum calprotectin at 1.2 ng/ml had sensitivity and specificity of 60% and 73%, respectively. Conclusions Serum calprotectin did not show a distinct advantage over the existing sepsis markers. Serum calprotectin level at 1.2 ng/ml had a sensitivity and specificity of 60% and 73%, respectively, compared to serum procalcitonin in detecting neonatal sepsis.
Collapse
Affiliation(s)
- Pardha Ramineni
- Department of Pediatrics, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Sowmini Padmanabh Kamath
- Department of Pediatrics, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Poornima Manjrekar
- Department of Biochemistry, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Padmanabh Kamath
- Department of Cardiology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Prasanna Mithra
- Department of Community Medicine, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Vaman Kulkarni
- Department of Community Medicine and Family Medicine, All India Institute of Medical Sciences (AIIMS), Bibinagar, Telangana, India
| |
Collapse
|
11
|
Basova LV, Lindsey A, McGovern A, Rosander A, Delorme-Walker V, ElShamy WM, Pendyala VV, Gaskill PJ, Ellis RJ, Cherner M, Iudicello JE, Marcondes MCG. MRP8/14 Is a Molecular Signature Triggered by Dopamine in HIV Latent Myeloid Targets That Increases HIV Transcription and Distinguishes HIV+ Methamphetamine Users with Detectable CSF Viral Load and Brain Pathology. Viruses 2023; 15:1363. [PMID: 37376663 PMCID: PMC10304659 DOI: 10.3390/v15061363] [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: 04/12/2023] [Revised: 06/01/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
There is a significant overlap between HIV infection and substance-use disorders. Dopamine (DA) is the most abundantly upregulated neurotransmitter in methamphetamine abuse, with receptors (DRD1-5) that are expressed by neurons as well as by a large diversity of cell types, including innate immune cells that are the targets of HIV infection, making them responsive to the hyperdopaminergic environment that is characteristic of stimulant drugs. Therefore, the presence of high levels of dopamine may affect the pathogenesis of HIV, particularly in the brain. The stimulation of HIV latently infected U1 promonocytes with DA significantly increased viral p24 levels in the supernatant at 24 h, suggesting effects on activation and replication. Using selective agonists to different DRDs, we found that DRD1 played a major role in activating viral transcription, followed by DRD4, which increased p24 with a slower kinetic rate compared to DRD1. Transcriptome and systems biology analyses led to the identification of a cluster of genes responsive to DA, where S100A8 and S100A9 were most significantly correlated with the early increase in p24 levels following DA stimulation. Conversely, DA increased the expression of these genes' transcripts at the protein level, MRP8 and MRP14, respectively, which form a complex also known as calprotectin. Interestingly, MRP8/14 was able to stimulate HIV transcription in latent U1 cells, and this occurred via binding of the complex to the receptor for an advanced glycosylation end-product (RAGE). Using selective agonists, both DRD1 and DRD4 increased MRP8/14 on the surface, in the cytoplasm, as well as secreted in the supernatants. On the other hand, while DRD1/5 did not affect the expression of RAGE, DRD4 stimulation caused its downregulation, offering a mechanism for the delayed effect via DRD4 on the p24 increase. To cross-validate MRP8/14 as a DA signature with a biomarker value, we tested its expression in HIV+ Meth users' postmortem brain specimens and peripheral cells. MRP8/14+ cells were more frequently identified in mesolimbic areas such as the basal ganglia of HIV+ Meth+ cases compared to HIV+ non-Meth users or to controls. Likewise, MRP8/14+ CD11b+ monocytes were more frequent in HIV+ Meth users, particularly in specimens from participants with a detectable viral load in the CSF. Overall, our results suggest that the MRP8 and MRP14 complex may serve as a signature to distinguish subjects using addictive substances in the context of HIV, and that this may play a role in aggravating HIV pathology by promoting viral replication in people with HIV who use Meth.
Collapse
Affiliation(s)
- Liana V. Basova
- San Diego Biomedical Research Institute, San Diego, CA 92121, USA
| | | | | | - Ashley Rosander
- San Diego Biomedical Research Institute, San Diego, CA 92121, USA
- Human Biology Program BISP, University of California San Diego, San Diego, CA 92037, USA
| | | | - Wael M. ElShamy
- San Diego Biomedical Research Institute, San Diego, CA 92121, USA
| | | | | | - Ronald J. Ellis
- HIV Neurobehavioral Research Program, University of California San Diego, San Diego, CA 92103, USA
| | - Mariana Cherner
- HIV Neurobehavioral Research Program, University of California San Diego, San Diego, CA 92103, USA
| | - Jennifer E. Iudicello
- HIV Neurobehavioral Research Program, University of California San Diego, San Diego, CA 92103, USA
| | | |
Collapse
|
12
|
Chittimalli K, Jahan J, Sakamuri A, Weyrick H, Winkle W, Adkins S, Vetter SW, Jarajapu YPR. Reversal of aging-associated increase in myelopoiesis and expression of alarmins by angiotensin-(1-7). Sci Rep 2023; 13:2543. [PMID: 36782016 PMCID: PMC9925828 DOI: 10.1038/s41598-023-29853-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 02/11/2023] [Indexed: 02/15/2023] Open
Abstract
Aging is associated with chronic systemic inflammation largely due to increased myelopoiesis, which in turn increases risk for vascular disease. We have previously shown evidence for the therapeutic potential of Angiotensin-(1-7) (Ang-(1-7)) in reversing vasoreparative dysfunction in aging. This study tested the hypothesis that ischemic vascular repair in aging by Ang-(1-7) involves attenuation of myelopoietic potential in the bone marrow and decreased mobilization of inflammatory cells. Young or Old male mice of age 3-4 and 22-24 months, respectively, received Ang-(1-7) (1 µg/kg/min, s.c.) for four weeks. Myelopoiesis was evaluated in the bone marrow (BM) cells by carrying out the colony forming unit (CFU-GM) assay followed by flow cytometry of monocyte-macrophages. Expression of pro-myelopoietic factors and alarmins in the hematopoietic progenitor-enriched BM cells was evaluated. Hindlimb ischemia (HLI) was induced by femoral ligation, and mobilization of monocytes into the blood stream was determined. Blood flow recovery was monitored by Laser Doppler imaging and infiltration of inflammatory cells was evaluated by immunohistochemistry. BM cells from Old mice generated a higher number of monocytes (Ly6G-CD11b+Ly6Chi) and M1 macrophages (Ly6ChiF4/80+) compared to that of Young, which was reversed by Ang-(1-7). Gene expression of selected myelopoietic factors, alarmins (S100A8, S100A9, S100A14 and HMGb1) and the receptor for alarmins, RAGE, was higher in the Old hematopoietic progenitor-enriched BM cells compared to the Young. Increased expressions of these factors were decreased by Ang-(1-7). Ischemia-induced mobilization of monocytes was higher in Old mice with decreased blood flow recovery and increased infiltration of monocyte-macrophages compared to the Young, all of which were reversed by Ang-(1-7). Enhanced ischemic vascular repair by Ang-(1-7) in aging is largely by decreasing the generation and recruitment of inflammatory monocyte-macrophages to the areas of ischemic injury. This is associated with decreased alarmin signaling in the BM-hematopoietic progenitor cells.
Collapse
Affiliation(s)
- Kishore Chittimalli
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Sudro-16, Albrecht Blvd., Fargo, ND, 58108, USA
| | - Jesmin Jahan
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Sudro-16, Albrecht Blvd., Fargo, ND, 58108, USA
| | - Anil Sakamuri
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Sudro-16, Albrecht Blvd., Fargo, ND, 58108, USA
| | - Hope Weyrick
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Sudro-16, Albrecht Blvd., Fargo, ND, 58108, USA
| | - Wink Winkle
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Sudro-16, Albrecht Blvd., Fargo, ND, 58108, USA
| | - Steven Adkins
- School of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Stefan W Vetter
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Sudro-16, Albrecht Blvd., Fargo, ND, 58108, USA
| | - Yagna P R Jarajapu
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Sudro-16, Albrecht Blvd., Fargo, ND, 58108, USA.
| |
Collapse
|
13
|
Circulating Calprotectin (cCLP) in autoimmune diseases. Autoimmun Rev 2023; 22:103295. [PMID: 36781037 DOI: 10.1016/j.autrev.2023.103295] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 02/09/2023] [Indexed: 02/13/2023]
Abstract
BACKGROUND AND AIM Calprotectin (CLP) is a heterodimeric complex formed by two S100 proteins (S100A8/A9), which plays a pivotal role in innate immunity. Due to its intrinsic cytotoxic and proinflammatory properties, CLP controls cell differentiation, proliferation and NETosis and has been associated with a wide range of rheumatic diseases. Our review summarizes the widespread interest in circulating CLP (cCLP) as a biomarker of neutrophil-related inflammation, in autoimmune rheumatic disease (ARD) and non-ARD. METHODS A thorough literature review was performed using PubMed and EMBASE databases searching for circulating calprotectin and synonyms S100A8/A9, myeloid-related protein 8/14 (MRP8/MRP14), calgranulin A/B and L1 protein in addition to specific ARDs and autoimmune non-rheumatic diseases. We selected only English-language articles and excluded abstracts without the main text. RESULTS High cCLP serum levels are associated with worse structural outcomes in rheumatoid arthritis and to a lesser extent, in spondyloarthritis. In addition, cCLP can predict disease relapse in some autoimmune diseases including systemic lupus erythematosus (SLE), anti-neutrophil cytoplasmic antibodies-associated vasculitis (AAV) and some severe manifestations of connective tissue diseases, such as glomerulonephritis in SLE, AAV, juvenile idiopathic arthritis, adult-onset Still's disease and lung fibrosis in systemic sclerosis. Therefore, cCLP levels enable the identification of patients who need an accurate and tight follow-up. The clinical usefulness of cCLP as an inflammatory marker has been suggested for inflammatory/autoimmune non-rheumatic diseases, and especially for the monitoring of the inflammatory bowel diseases patients. Currently, there are only a few studies that evaluated the cCLP efficacy as a clinical biomarker in inflammatory/autoimmune non-rheumatic diseases with controversial results. Future studies are warranted to better clarify the role of cCLP in relation to the disease severity in myasthenia gravis, multiple sclerosis, chronic inflammatory demyelinating polyneuropathy, Graves' orbitopathy, autoimmune bullous diseases and uveitis. CONCLUSION Our literature review supports a relevant role of cCLP as potential prognostic biomarker mirroring local or systemic inflammation, especially in chronic inflammatory rheumatic diseases.
Collapse
|
14
|
Colicchia M, Perrella G, Gant P, Rayes J. Novel mechanisms of thrombo-inflammation during infection: spotlight on neutrophil extracellular trap-mediated platelet activation. Res Pract Thromb Haemost 2023; 7:100116. [PMID: 37063765 PMCID: PMC10099327 DOI: 10.1016/j.rpth.2023.100116] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/21/2023] [Accepted: 02/10/2023] [Indexed: 03/13/2023] Open
Abstract
A state-of-the-art lecture titled "novel mechanisms of thrombo-inflammation during infection" was presented at the ISTH Congress in 2022. Platelet, neutrophil, and endothelial cell activation coordinate the development, progression, and resolution of thrombo-inflammatory events during infection. Activated platelets and neutrophil extracellular traps (NETs) are frequently observed in patients with sepsis and COVID-19, and high levels of NET-derived damage-associated molecular patterns (DAMPs) correlate with thrombotic complications. NET-associated DAMPs induce direct and indirect platelet activation, which in return potentiates neutrophil activation and NET formation. These coordinated interactions involve multiple receptors and signaling pathways contributing to vascular and organ damage exacerbating disease severity. This state-of-the-art review describes the main mechanisms by which platelets support NETosis and the key mechanisms by which NET-derived DAMPs trigger platelet activation and the formation of procoagulant platelets leading to thrombosis. We report how these DAMPs act through multiple receptors and signaling pathways differentially regulating cell activation and disease outcome, focusing on histones and S100A8/A9 and their contribution to the pathogenesis of sepsis and COVID-19. We further discuss the complexity of platelet activation during NETosis and the potential benefit of targeting selective or multiple NET-associated DAMPs to limit thrombo-inflammation during infection. Finally, we summarize relevant new data on this topic presented during the 2022 ISTH Congress.
Collapse
Affiliation(s)
- Martina Colicchia
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, U.K
| | - Gina Perrella
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, U.K
| | - Poppy Gant
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, U.K
| | - Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, U.K
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, U.K
| |
Collapse
|
15
|
Du L, Chen Y, Shi J, Yu X, Zhou J, Wang X, Xu L, Liu J, Gao J, Gu X, Wang T, Yin Z, Li C, Yan M, Wang J, Yin X, Lu Q. Inhibition of S100A8/A9 ameliorates renal interstitial fibrosis in diabetic nephropathy. Metabolism 2022:155376. [PMID: 36521551 DOI: 10.1016/j.metabol.2022.155376] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Renal interstitial fibrosis (RIF) is one of the main features of diabetic nephropathy (DN), but the molecular mechanisms mediating RIF in DN has yet been fully understood. S100A8 and S100A9 are the proteins associated with immune and inflammation response. Here we reported the expression of S100A8 and S100A9 were significantly increased on tubular epithelial cells in diabetic kidneys through a proteomic analysis. METHODS We detected the expression of S100A8/A9 in diabetic kidneys by using immunoblotting, real-time PCR and immunostaining. RNA silencing and overexpression were performed by using S100A8/A9 expression/knockdown lentivirus to investigate the connection between S100A8/A9 and epithelial to mesenchymal transition (EMT) process. We also identify the expression of TLR4/NFκB pathway-related molecules in the case mentioned above. Afterwards a CO-IP assay was used to verify that compound AB38b ameliorates the EMT by interfering S100A8/A9 expression. RESULTS The expression of S100A8 and S100A9 were significantly increased on tubular epithelial cells in diabetic kidneys. S100A8/A9 knocking-down alleviate and over-expression promote the renal interstitial fibrosis of diabetic mice. Mechanically, high levels of S100A8/A9 expression in tubular epithelial cells during diabetic condition activated the TLR4/NF-κB signal pathway which promoted the EMT process and finally led to RIF progression. S100A8/A9 knockdown ameliorated RIF of diabetic mice. Further experiments revealed that compound AB38b inhibited the EMT progression of tubular epithelial cells induced by S100A8/A9 through interfering the expressions of S100A8/A9. CONCLUSIONS Our study suggest that abnormal expression of S100A8/A9 in the disease condition promotes EMT process and RIF through TLR4/NF-κB signal pathway. Using small molecular inhibitor AB38b to inhibit the abnormal expressions of S100A8/A9 might be a novel therapeutic strategy in treating DN.
Collapse
Affiliation(s)
- Lei Du
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Yibing Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Jiasen Shi
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Xiujuan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Jieling Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Xue Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Liu Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Junjie Liu
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, China
| | - Jian Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Xiaoke Gu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Tao Wang
- Department of Clinical Pharmacy, Affiliated Hospital of Xuzhou Medical University, China
| | - Zeyuan Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China; Department of Clinical Medicine, Xuzhou Medical University, China
| | - Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Meng Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Jianyun Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Qian Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China.
| |
Collapse
|
16
|
Zhou Y, Bréchard S. Neutrophil Extracellular Vesicles: A Delicate Balance between Pro-Inflammatory Responses and Anti-Inflammatory Therapies. Cells 2022; 11:cells11203318. [PMID: 36291183 PMCID: PMC9600967 DOI: 10.3390/cells11203318] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) are released in the extracellular environment during cell activation or apoptosis. Working as signal transducers, EVs are important mediators of intercellular communication through the convoying of proteins, nucleic acids, lipids, and metabolites. Neutrophil extracellular vesicles (nEVs) contain molecules acting as key modulators of inflammation and immune responses. Due to their potential as therapeutic tools, studies about nEVs have been increasing in recent years. However, our knowledge about nEVs is still in its infancy. In this review, we summarize the current understanding of the role of nEVs in the framework of neutrophil inflammation functions and disease development. The therapeutic potential of nEVs as clinical treatment strategies is deeply discussed. Moreover, the promising research landscape of nEVs in the near future is also examined.
Collapse
|
17
|
Curran CS, Kopp JB. RAGE pathway activation and function in chronic kidney disease and COVID-19. Front Med (Lausanne) 2022; 9:970423. [PMID: 36017003 PMCID: PMC9395689 DOI: 10.3389/fmed.2022.970423] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/21/2022] [Indexed: 12/23/2022] Open
Abstract
The multi-ligand receptor for advanced glycation end-products (RAGE) and its ligands are contributing factors in autoimmunity, cancers, and infectious disease. RAGE activation is increased in chronic kidney disease (CKD) and coronavirus disease 2019 (COVID-19). CKD may increase the risk of COVID-19 severity and may also develop in the form of long COVID. RAGE is expressed in essentially all kidney cell types. Increased production of RAGE isoforms and RAGE ligands during CKD and COVID-19 promotes RAGE activity. The downstream effects include cellular dysfunction, tissue injury, fibrosis, and inflammation, which in turn contribute to a decline in kidney function, hypertension, thrombotic disorders, and cognitive impairment. In this review, we discuss the forms and mechanisms of RAGE and RAGE ligands in the kidney and COVID-19. Because various small molecules antagonize RAGE activity in animal models, targeting RAGE, its co-receptors, or its ligands may offer novel therapeutic approaches to slowing or halting progressive kidney disease, for which current therapies are often inadequate.
Collapse
Affiliation(s)
- Colleen S. Curran
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Jeffrey B. Kopp
- Kidney Disease Section, NIDDK (National Institute of Diabetes and Digestive and Kidney Diseases), National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
18
|
Alarmins S100A8/A9 promote intervertebral disc degeneration and inflammation-related pain in a rat model through toll-like receptor-4 and activation of the NF-κB signaling pathway. Osteoarthritis Cartilage 2022; 30:998-1011. [PMID: 35405347 DOI: 10.1016/j.joca.2022.03.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The molecules released from cells undergoing necrosis are recognized as alarmins, and S100A8/9, a typical alarmin, is associated with several inflammation-related diseases. This study was to investigate the molecular role of S100A8/A9 on the process of intervertebral disc degeneration (IVDD) and inflammation-related pain. METHODS The expression pattern of S100A8/A9 in different degenerated human nucleus pulposus (NP) tissues were measured by Real-time quantitative reverse transcription PCR (RT-qPCR) and immunohistochemical (IHC). The effects of S100A8/A9 on matrix production were assessed by RT-qPCR, western blotting, and cell immunofluorescence. Involvement of TLR4 and NF-κB signaling pathways were studied by pharmachemical inhibitors and small interfering RNAs (siRNAs). The development of degenerative and pain features in the IVDD model were examed by IHC and pain-behavior testing. RESULTS The expression of S100A8/A9 was significantly elevated in severely degenerated human NP tissue with similar expression pattern of TNF-α. In NP cells, S100A8/A9 increased MMP-3/13, TNF-α, IL-6 expression and inhibited aggrecan and collagen II expression. RT-qPCR and western blotting showed that the regulatory effects of S100A8/A9 on IVD were TLR4 dependent. Pharmacological inhibition or siRNA knockdown of the NF-κB signaling attenuated S100A8/A9-induced upregulation of MMP-3/13, TNF-α and IL-6. In vivo, S100A9 inhibitor treatment inhibited disc-puncture induced IVDD and inflammation-related pain. CONCLUSIONS This study showed that S100A8/A9 bound to TLR4 and increased the expression of MMPs, TNF-α, and IL-6 through NF-κB signaling pathways in NP cells. Furthermore, S100A8/A9 inhibitor could prevent development of IVDD and inflammation-related pain in the rat model.
Collapse
|
19
|
Chen L, Chen X, Wang Y, Li S, Huang S, Wu Z, He J, Chen S, Deng F, Zhu P, Zhong W, Zhao B, Ma G, Li Y. Polymorphisms of Calgranulin Genes and Ischemic Stroke in a Chinese Population. J Inflamm Res 2022; 15:3355-3368. [PMID: 35706528 PMCID: PMC9191198 DOI: 10.2147/jir.s360775] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
Background The S100/calgranulin gene appears to modulate neuroinflammation following cerebral ischemia and could be a valuable biomarker for stroke prognosis, according to growing research. This study aimed at evaluating the correlation between calgranulin gene variants and susceptibility to ischemic stroke (IS) in the Southern Chinese population. Methods Using an enhanced multi-temperature ligase detection reaction genotyping, 310 IS patients and 324 age-matched healthy controls were genotyped to identify five calgranulin gene variants. Results According to the obtained results, the S100A8 rs3795391, rs3806232, and S100A12 rs2916191 variants were linked to a higher risk of IS, while the S100A9 rs3014866 variant was associated with a lower risk of IS. Moreover, the T-T-C-A-T, T-T-C-G-T, or C-C-C-G-C haplotypes have been linked to a greater risk of developing IS, according to haplotype analysis. The occurrence of the variant C allele there in S100A8 rs3795391, rs3806232, and S100A12 rs2916191 variants may impart a greater risk of stroke in the LAA subtype, according to further stratification by IS subtypes, while the T allele of the S100A9 rs3014866 variant may be linked to a reduced risk of stroke of all subtypes. Furthermore, patients with the variant C allele of the S100A8 rs3795391, rs3806232, and S100A12 rs2916191 variants presented with increased circulating S100A8 and S100A12 levels and larger infarct volumes relative to those with the major TT genotype. Conclusion Our findings suggest that calgranulin gene variants are linked to IS susceptibility, implying that the calgranulin gene may be a potential biomarker for IS prevention and personalized treatment.
Collapse
Affiliation(s)
- Linfa Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Department of Neurology, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, People's Republic of China
| | - Xinglan Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Yajun Wang
- Maternal and Children's Health Research Institute, Shunde Maternal and Children's Hospital, Guangdong Medical University, Shunde, People's Republic of China
| | - Shengnan Li
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Shaoting Huang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Zhaochun Wu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Jiawen He
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Shaofeng Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Fu Deng
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Peiyi Zhu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Wangtao Zhong
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Bin Zhao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Guoda Ma
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Maternal and Children's Health Research Institute, Shunde Maternal and Children's Hospital, Guangdong Medical University, Shunde, People's Republic of China
| | - You Li
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.,Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| |
Collapse
|
20
|
Yamazaki Y, Wake H, Nishinaka T, Hatipoglu OF, Liu K, Watanabe M, Toyomura T, Mori S, Yoshino T, Nishibori M, Takahashi H. Involvement of multiple scavenger receptors in advanced glycation end product-induced vessel tube formation in endothelial cells. Exp Cell Res 2021; 408:112857. [PMID: 34600900 DOI: 10.1016/j.yexcr.2021.112857] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/16/2021] [Accepted: 09/28/2021] [Indexed: 01/01/2023]
Abstract
Toxic advanced glycation end products (toxic AGEs) derived from glycolaldehyde (AGE3) have been implicated in the development of diabetic vascular complications such as retinopathy characterised by excessive angiogenesis. Different receptor types, such as receptor for AGEs (RAGE), Toll like receptor-4 and scavenger receptors, are expressed in endothelial cells and contribute to AGE-elicited alteration of cell function. In the present study, we examined the involvement of AGE-related receptors on AGE-induced angiogenesis in endothelial cells. The effects of pharmacological inhibitors or receptor neutralizing antibodies on AGE3-induced tube formation were investigated using the in vitro Matrigel tube formation assay in b.End5 cells (mouse endothelial cells). AGE3-induced signalling pathways and receptor expression changes were analysed by Western blot analysis and flow cytometry, respectively. Both FPS-ZM1, a RAGE inhibitor, and fucoidan, a ligand for scavenger receptors, suppressed AGE3-induced tube formation. Cocktails of neutralizing antibodies against the scavenger receptors CD36, CD163 and LOX-1 prevented AGE3-induced tube formation. AGE3 activated mTOR signalling, resulting in facilitation of tube formation. Activation of the AGE-RAGE pathway also led to the upregulation of scavenger receptors. Taken together, our findings suggest that the scavenger receptors CD36, CD163 and LOX-1 in conjunction with the RAGE receptor work together to mediate toxic AGE-induced facilitation of angiogenesis.
Collapse
Affiliation(s)
- Yui Yamazaki
- Department of Pharmacology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Hidenori Wake
- Department of Pharmacology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Takashi Nishinaka
- Department of Pharmacology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan.
| | - Omer Faruk Hatipoglu
- Department of Pharmacology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | | | - Masahiro Watanabe
- Department of Pharmacology, School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama, Japan
| | - Takao Toyomura
- Department of Pharmacology, School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama, Japan
| | - Shuji Mori
- Department of Pharmacology, School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama, Japan
| | - Tadashi Yoshino
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, Japan
| | | | - Hideo Takahashi
- Department of Pharmacology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| |
Collapse
|
21
|
Role of S100A8/A9 for Cytokine Secretion, Revealed in Neutrophils Derived from ER-Hoxb8 Progenitors. Int J Mol Sci 2021; 22:ijms22168845. [PMID: 34445548 PMCID: PMC8396251 DOI: 10.3390/ijms22168845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022] Open
Abstract
S100A9, a Ca2+-binding protein, is tightly associated to neutrophil pro-inflammatory functions when forming a heterodimer with its S100A8 partner. Upon secretion into the extracellular environment, these proteins behave like damage-associated molecular pattern molecules, which actively participate in the amplification of the inflammation process by recruitment and activation of pro-inflammatory cells. Intracellular functions have also been attributed to the S100A8/A9 complex, notably its ability to regulate nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation. However, the complete functional spectrum of S100A8/A9 at the intracellular level is far from being understood. In this context, we here investigated the possibility that the absence of intracellular S100A8/A9 is involved in cytokine secretion. To overcome the difficulty of genetically modifying neutrophils, we used murine neutrophils derived from wild-type and S100A9−/− Hoxb8 immortalized myeloid progenitors. After confirming that differentiated Hoxb8 neutrophil-like cells are a suitable model to study neutrophil functions, our data show that absence of S100A8/A9 led to a dysregulation of cytokine secretion after lipopolysaccharide (LPS) stimulation. Furthermore, we demonstrate that S100A8/A9-induced cytokine secretion was regulated by the nuclear factor kappa B (NF-κB) pathway. These results were confirmed in human differentiated HL-60 cells, in which S100A9 was inhibited by shRNAs. Finally, our results indicate that the degranulation process could be involved in the regulation of cytokine secretion by S100A8/A9.
Collapse
|
22
|
Molecular Characteristics of RAGE and Advances in Small-Molecule Inhibitors. Int J Mol Sci 2021; 22:ijms22136904. [PMID: 34199060 PMCID: PMC8268101 DOI: 10.3390/ijms22136904] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/16/2022] Open
Abstract
Receptor for advanced glycation end-products (RAGE) is a member of the immunoglobulin superfamily. RAGE binds and mediates cellular responses to a range of DAMPs (damage-associated molecular pattern molecules), such as AGEs, HMGB1, and S100/calgranulins, and as an innate immune sensor, can recognize microbial PAMPs (pathogen-associated molecular pattern molecules), including bacterial LPS, bacterial DNA, and viral and parasitic proteins. RAGE and its ligands stimulate the activations of diverse pathways, such as p38MAPK, ERK1/2, Cdc42/Rac, and JNK, and trigger cascades of diverse signaling events that are involved in a wide spectrum of diseases, including diabetes mellitus, inflammatory, vascular and neurodegenerative diseases, atherothrombosis, and cancer. Thus, the targeted inhibition of RAGE or its ligands is considered an important strategy for the treatment of cancer and chronic inflammatory diseases.
Collapse
|
23
|
Wang A, Guo B, Jia Q, Chen Y, Gao X, Xu S. S100A9-containing serum exosomes of burn injury patients promote permeability of pulmonary microvascular endothelial cells. J Biosci 2021. [DOI: 10.1007/s12038-021-00151-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
24
|
Cheng M, Su X, Liu D, Tian X, Yan C, Zhang X, Han Y. Role of Neutrophil-Derived S100B in Acute Myocardial Infarction Patients From the Han Chinese Population. Front Cardiovasc Med 2021; 7:595446. [PMID: 33796567 PMCID: PMC8008063 DOI: 10.3389/fcvm.2020.595446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 12/16/2020] [Indexed: 11/16/2022] Open
Abstract
Objective: This study aimed to clarify the novel role of homeostatic calmodulin S100B and determined whether S100B genetic variants affected atherosclerosis progression in acute myocardial infarction (AMI) patients. Methods: Plasma levels of S100B were measured systemically in AMI patients, stable angina pectoris patients, and control subjects. S100B was obtained from the human coronary artery thrombi using a thrombectomy catheter and quantified via immunohistochemical analysis, qRT-PCR and Western blot analyse. We also screened for S100B variations (rs9722, rs9984765, rs2839356, rs1051169, and rs2186358) via direct sequencing, and investigated the relationship between these variants and AMI patients in the Chinese Han population. Results: Plasma S100B levels increased significantly in AMI patients compared to the levels in stable angina pectoris patients and control subjects (119.45 ± 62.46, 161.96 ± 73.30, and 312.91 ± 127.59 pg/ml, respectively). Immunohistochemical staining results showed that S100B expression was increased in the neutrophils of coronary artery thrombi obtained from AMI patients, as compared to that in normal blood clot, and S100B expression was significantly increased in fresh thrombi tissues, as compared to that in organized thrombi tissues. Western blot and qRT-PCR analysis showed that S100B expression increased in coronary artery thrombi, as compared to that in normal blood clots. After pre-treating the neutrophils with siRAGE, the neutrophils migration induced by S100B were abolished through the NFκB-IL1β/IL6 signaling pathway. Compared to their corresponding wild-type genotypes, the S100B rs9722 variant was associated with increased susceptibility to AMI (OR = 1.35, 95%CI: 1.12–1.65, P = 0.02). Individuals with the S100B 9722 A allele had higher plasma S100B levels than those with the G allele in control subjects and AMI patients (141.70 ± 76.69 vs. 107.31 ± 56.05 and 347.13 ± 148.94 vs. 273.05 ± 133.62, respectively). Conclusions: Levels of neutrophil-derived S100B, a novel homeostatic calmodulin, were elevated in the early stages of myocardial infarction. The S100B rs9722 allele was independently associated with AMI patients in the Han Chinese population.
Collapse
Affiliation(s)
- Minghui Cheng
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Xu Su
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Dan Liu
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaoxiang Tian
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaolin Zhang
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Yaling Han
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
25
|
Stahl LS, Roth J, Rudack C, McNally A, Weber J, Vogl T, Spiekermann C. Evaluation of the QUANTUM BLUE sCAL rapid test as a point of care tool to identify patients with peritonsillar abscess. Sci Rep 2021; 11:4497. [PMID: 33627801 PMCID: PMC7904841 DOI: 10.1038/s41598-021-84027-w] [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: 11/14/2020] [Accepted: 02/08/2021] [Indexed: 11/30/2022] Open
Abstract
S100A8/A9 (Calprotectin) serves as a biomarker for various inflammatory diseases, such as for peritonsillar abscess (PTA). Recently, the PTA score was developed for reliable PTA identification. It uses a combination of characteristic clinical symptoms and elevated calprotectin levels in serum and saliva to determine this score. Although well-established point-of-care tests (POCT) to determine serum or faecal calprotectin levels exist, a reliable and rapid tool to analyse salivary calprotectin has not yet been described. In this study, we analysed the potential of the QUANTUM BLUE sCAL Test (QBT, BÜHLMANN Laboratories AG, Switzerland) to determine S100A8/A9 levels during outpatient management. These QBT measurements are combined with other clinical factors to determine the PTA score. Significantly higher calprotectin levels were determined by QBT in patients with PTA compared to healthy controls. The receiver operating characteristic (ROC) curves for the QBT revealed cut-off values of 2940 ng/ml (sensitivity = 0.88, specificity = 0.78) in serum and 5310 ng/ml (sensitivity = 0.80, specificity = 0.50) in saliva. By adding the QBT results to determine PTA values, a ROC analysis provided a statistical cut-off score of 2.5 points to identify the existence of a PTA with a sensitivity of 100% and a specificity of 89.3%. The QUANTUM BLUE sCAL Test (QBT) is an appropriate POCT to determine serum and salivary calprotectin levels. Thus, PTA scores can be determined within a short time frame by applying the QBT during outpatient management.
Collapse
Affiliation(s)
- Lea-Sophie Stahl
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Münster, 48149, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University Hospital Münster, Röntgenstr. 21, 48149, Münster, Germany
| | - Claudia Rudack
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Münster, 48149, Münster, Germany
| | - Annika McNally
- Institute of Immunology, University Hospital Münster, Röntgenstr. 21, 48149, Münster, Germany
| | - Jakob Weber
- BÜHLMANN Laboratories AG, 4124, Schönenbuch, Switzerland
| | - Thomas Vogl
- Institute of Immunology, University Hospital Münster, Röntgenstr. 21, 48149, Münster, Germany
| | - Christoph Spiekermann
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Münster, 48149, Münster, Germany. .,Institute of Immunology, University Hospital Münster, Röntgenstr. 21, 48149, Münster, Germany.
| |
Collapse
|
26
|
Cai Z, Xie Q, Hu T, Yao Q, Zhao J, Wu Q, Tang Q. S100A8/A9 in Myocardial Infarction: A Promising Biomarker and Therapeutic Target. Front Cell Dev Biol 2020; 8:603902. [PMID: 33282877 PMCID: PMC7688918 DOI: 10.3389/fcell.2020.603902] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/21/2020] [Indexed: 12/21/2022] Open
Abstract
Myocardial infarction (MI), the main cause of cardiovascular-related deaths worldwide, has long been a hot topic because of its threat to public health. S100A8/A9 has recently attracted an increasing amount of interest as a crucial alarmin that regulates the pathogenesis of cardiovascular disease after its release from myeloid cells. However, the role of S100A8/A9 in the etiology of MI is not well understood. Here, we elaborate on the critical roles and potential mechanisms of S100A8/A9 driving the pathogenesis of MI. First, cellular source of S100A8/A9 in infarcted heart is discussed. Then we highlight the effect of S100A8/A9 heterodimer in the early inflammatory period and the late reparative period of MI as well as myocardial ischemia/reperfusion (I/R) injury. Moreover, the predictive value of S100A8/A9 for the risk of recurrence of cardiovascular events is elucidated. Therefore, this review focuses on the molecular mechanisms of S100A8/A9 in MI pathogenesis to provide a promising biomarker and therapeutic target for MI.
Collapse
Affiliation(s)
- ZhuLan Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qingwen Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Tongtong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jinhua Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qingqing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| |
Collapse
|
27
|
Guo D, Zhu Z, Xu T, Zhong C, Wang A, Xie X, Peng Y, Peng H, Li Q, Ju Z, Geng D, Chen J, Liu L, Wang Y, Zhang Y, He J. Plasma S100A8/A9 Concentrations and Clinical Outcomes of Ischemic Stroke in 2 Independent Multicenter Cohorts. Clin Chem 2020; 66:706-717. [PMID: 32285094 DOI: 10.1093/clinchem/hvaa069] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 01/30/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND S100A8/A9 is implicated in inflammation mechanisms related to atherosclerosis and plaque vulnerability, but it remains unclear whether S100A8/A9 is associated with the prognosis of ischemic stroke. The aim of this study was to investigate these associations in 2 independent multicenter cohorts. METHODS Plasma S100A8/A9 concentrations at baseline were measured among 4785 patients with ischemic stroke from 2 independent cohorts: Infectious Factors, Inflammatory Markers, and Prognosis of Acute Ischemic Stroke (IIPAIS) and China Antihypertensive Trial in Acute Ischemic Stroke (CATIS). The primary outcome was a composite outcome of death or major disability at 3 months after ischemic stroke. Secondary outcomes were major disability, death, and a composite outcome of death or vascular events. RESULTS Among the combined participants of IIPAIS and CATIS, the adjusted odds ratios associated with the highest quartile of plasma S100A8/A9 were 2.11 (95% CI, 1.66-2.68) for the primary outcome and 1.62 (95% CI, 1.27-2.07) for the secondary outcome of major disability; adjusted hazard ratios were 4.14 (95% CI, 2.10-8.15) for the secondary outcome of death and 2.08 (95% CI, 1.38-3.13) for the composite outcome of death or vascular events. Each SD increase of log-transformed S100A8/A9 was associated with 28% (95% CI, 18%-39%; P < 0.001) increased risk of the primary outcome. Multivariable-adjusted spline regression analyses showed a linear association between plasma S100A8/A9 concentrations and primary outcome (P < 0.001 for linearity). Subgroup analyses further confirmed these associations. CONCLUSIONS High plasma S100A8/A9 concentrations at baseline were independently associated with increased risks of adverse clinical outcomes at 3 months after ischemic stroke, suggesting that S100A8/A9 might have a role as a prognostic marker of ischemic stroke.
Collapse
Affiliation(s)
- Daoxia Guo
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, Tulane University, School of Public Health and Tropical Medicine, New Orleans, LA
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, Tulane University, School of Public Health and Tropical Medicine, New Orleans, LA
| | - Tan Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Chongke Zhong
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Aili Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Xuewei Xie
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yanbo Peng
- Department of Neurology, Affiliated Hospital of North China University of Science and Technology, Hebei, China
| | - Hao Peng
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Qunwei Li
- Department of Epidemiology, School of Public Health, Taishan Medical College, Shandong, China
| | - Zhong Ju
- Department of Neurology, Kerqin District First People's Hospital of Tongliao City, Inner Mongolia, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Jing Chen
- Department of Epidemiology, Tulane University, School of Public Health and Tropical Medicine, New Orleans, LA.,Department of Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Liping Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Jiang He
- Department of Epidemiology, Tulane University, School of Public Health and Tropical Medicine, New Orleans, LA.,Department of Medicine, Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
28
|
Sreejit G, Abdel Latif A, Murphy AJ, Nagareddy PR. Emerging roles of neutrophil-borne S100A8/A9 in cardiovascular inflammation. Pharmacol Res 2020; 161:105212. [PMID: 32991974 DOI: 10.1016/j.phrs.2020.105212] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Elevated neutrophil count is associated with higher risk of major adverse cardiac events including myocardial infarction and early development of heart failure. Neutrophils contribute to cardiac damage through a number of mechanisms, including attraction of other immune cells and release of inflammatory mediators. Recently, a number of independent studies have reported a causal role for neutrophil-derived alarmins (i.e. S100A8/A9) in inducing inflammation and cardiac injury following myocardial infarction (MI). Furthermore, a positive correlation between serum S100A8/A9 levels and major adverse cardiac events (MACE) in MI patients was also observed implying that targeting neutrophils or their inflammatory cargo could be beneficial in reducing heart failure. However, contradictory to this idea, neutrophils and neutrophil-derived S100A8/A9 also seem to play a vital role in the resolution of inflammation. Thus, a better understanding of how neutrophils balance these seemingly contrasting functions would allow us to develop effective therapies that preserve the inflammation-resolving function while restricting the damage caused by inflammation. In this review, we specifically discuss the mechanisms behind neutrophil-derived S100A8/A9 in promoting inflammation and resolution in the context of MI. We also provide a perspective on how neutrophils could be potentially targeted to ameliorate cardiac inflammation and the ensuing damage.
Collapse
Affiliation(s)
- Gopalkrishna Sreejit
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ahmed Abdel Latif
- Division of Cardiovascular Medicine, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia
| | - Prabhakara R Nagareddy
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
29
|
Yano T, Hagiwara Y, Ando A, Kanazawa K, Koide M, Sekiguchi T, Itaya N, Onoki T, Suzuki K, Tsuchiya M, Sogi Y, Yabe Y, Itoi E. RAGE-dependent NF-kB inflammation processes in the capsule of frozen shoulders. J Shoulder Elbow Surg 2020; 29:1884-1891. [PMID: 32279986 DOI: 10.1016/j.jse.2020.01.076] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/30/2019] [Accepted: 01/01/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND The etiology of frozen shoulder (FS) remains uncertain. Advanced glycation end-products (AGEs) cause the cross-linking and stabilization of collagen and are increased in FS. The purpose of this study was to elucidate the pathogenesis of FS by evaluating the receptor of AGE (RAGE)-dependent pathways. METHODS Tissue samples of the coracohumeral ligament (CHL) and anterior inferior glenohumeral ligament (IGHL) were collected from 33 patients with FS, with severe stiffness, and 25 with rotator cuff tears (RCTs) as controls. Gene expression levels of RAGE, high-mobility group box 1 (HMGB1), Toll-like receptor 2 (TLR2), TLR4, nuclear factor-kappa B (NF-kB), and cytokines were evaluated using a quantitative real-time polymerase chain reaction. The immunoreactivities of carboxymethyllysine (CML), pentosidine, and RAGE were also evaluated. CML and pentosidine were further evaluated using high-performance liquid chromatography. RESULTS Gene expression levels of RAGE, HMGB1, TLR2, TLR4, and NF-kB were significantly greater in the CHLs and IGHLs from the FS group than in those from the RCT group. Immunoreactivities of RAGE and CML were stronger in the CHLs and IGHLs from the FS group than in those from the RCT group. Pentosidine was weakly immunostained in the CHLs and IGHLs from the FS group. CML using high-performance liquid chromatography was significantly greater in the CHLs and IGHLs from the FS group than in those from the RCT group. CONCLUSIONS AGEs and HMGB1 might play important roles in the pathogenesis of FS by binding to RAGE and activating NF-kB signaling pathways. Suppression of these pathways could be a treatment option for FS.
Collapse
Affiliation(s)
- Toshihisa Yano
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Yoshihiro Hagiwara
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan.
| | - Akira Ando
- Department of Orthopaedic Surgery, Matsuda Hospital, Sendai, Japan
| | - Kenji Kanazawa
- Department of Orthopaedic Surgery, South Miyagi Medical Center, Ogawara, Japan
| | - Masashi Koide
- Department of Orthopaedic Surgery, Matsuda Hospital, Sendai, Japan
| | - Takuya Sekiguchi
- Department of Orthopaedic Surgery, Iwate Prefectural Central Hospital, Morioka, Japan
| | - Nobuyuki Itaya
- Department of Orthopaedic Surgery, Sendai Hospital of East Japan Railway Company, Sendai, Japan
| | - Takahiro Onoki
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Kazuaki Suzuki
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | | | - Yasuhito Sogi
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Yutaka Yabe
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Eiji Itoi
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| |
Collapse
|
30
|
Shi H, Zuo Y, Yalavarthi S, Gockman K, Zuo M, Madison JA, Blair C, Woodward W, Lezak SP, Lugogo NL, Woods RJ, Lood C, Knight JS, Kanthi Y. Neutrophil calprotectin identifies severe pulmonary disease in COVID-19. J Leukoc Biol 2020; 109:67-72. [PMID: 32869342 DOI: 10.1002/jlb.3covcra0720-359r] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/15/2020] [Accepted: 07/30/2020] [Indexed: 12/15/2022] Open
Abstract
Severe cases of coronavirus disease 2019 (COVID-19) are regularly complicated by respiratory failure. Although it has been suggested that elevated levels of blood neutrophils associate with worsening oxygenation in COVID-19, it is unknown whether neutrophils are drivers of the thrombo-inflammatory storm or simple bystanders. To better understand the potential role of neutrophils in COVID-19, we measured levels of the neutrophil activation marker S100A8/A9 (calprotectin) in hospitalized patients and determined its relationship to severity of illness and respiratory status. Patients with COVID-19 (n = 172) had markedly elevated levels of calprotectin in their blood. Calprotectin tracked with other acute phase reactants including C-reactive protein, ferritin, lactate dehydrogenase, and absolute neutrophil count, but was superior in identifying patients requiring mechanical ventilation. In longitudinal samples, calprotectin rose as oxygenation worsened. When tested on day 1 or 2 of hospitalization (n = 94 patients), calprotectin levels were significantly higher in patients who progressed to severe COVID-19 requiring mechanical ventilation (8039 ± 7031 ng/ml, n = 32) as compared to those who remained free of intubation (3365 ± 3146, P < 0.0001). In summary, serum calprotectin levels track closely with current and future COVID-19 severity, implicating neutrophils as potential perpetuators of inflammation and respiratory compromise in COVID-19.
Collapse
Affiliation(s)
- Hui Shi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Division of Rheumatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zuo
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Srilakshmi Yalavarthi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kelsey Gockman
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Melanie Zuo
- Division of Geriatric and Palliative Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jacqueline A Madison
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Blair
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Wrenn Woodward
- Michigan Clinical Research Unit, University of Michigan, Ann Arbor, Michigan, USA
| | - Sean P Lezak
- Michigan Clinical Research Unit, University of Michigan, Ann Arbor, Michigan, USA
| | - Njira L Lugogo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Robert J Woods
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christian Lood
- Division of Rheumatology, University of Washington, Department of Medicine, Seattle, Washington, USA
| | - Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yogendra Kanthi
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
31
|
Zhang W, Kong Y, Wang L, Song L, Tan L, Xue X. Prognostic value of serum calprotectin level in elderly diabetic patients with acute coronary syndrome undergoing percutaneous coronary intervention: A Cohort study. Medicine (Baltimore) 2020; 99:e20805. [PMID: 32871971 PMCID: PMC7437750 DOI: 10.1097/md.0000000000020805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Patients with acute coronary syndrome (ACS) have an increased serum level of calprotectin. The purpose of present study was to analyze the prognostic significance of serum calprotectin levels in elderly diabetic patients underwent percutaneous coronary intervention (PCI) due to ACS.A total of 273 consecutive elderly diabetic patients underwent PCI for primary ACS were enrolled. Serum calprotectin levels were measured before PCI, and baseline clinical characteristics of all patients were collected. All patients were followed up at regular interval for major adverse cardiovascular events (MACEs) during 1 year after PCI. MACEs include cardiovascular death, nonfatal myocardial infarction, and target vessel revascularization (TVR). The predicting value of serum calprotectin for MACEs was analyzed by using univariate and multivariate analysis and receiver-operating characteristic curve (ROC).At the endpoint of this study, 47 patients of all 273 patients had MACEs. According to optimal cutoff value of calprotectin for predicting MACEs by ROC analysis, all patients were stratified into a high calprotectin group and a low calprotectin group. The incidence rate of MACEs and TVR in high calprotectin group was prominently higher than that in low calprotectin group (21.9% vs 11.5%, P = .02). In multivariable COX regression analysis adjusting for potential confounders, serum calprotectin level remains as an independent risk predictor of MACE (hazard ratio, 1.56; 95% confidence interval [CI]: 1.08-4.62; P = .01).In diabetic patients with a comorbidity of ACS, a high serum level of calprotectin is associated to a higher MACE rate after PCI.
Collapse
|
32
|
Neutrophil calprotectin identifies severe pulmonary disease in COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020. [PMID: 32511540 DOI: 10.1101/2020.05.06.20093070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Severe cases of coronavirus disease 2019 (COVID-19) are regularly complicated by respiratory failure. While it has been suggested that elevated levels of blood neutrophils associate with worsening oxygenation in COVID-19, it is unknown whether neutrophils are drivers of the thrombo-inflammatory storm or simple bystanders. To better understand the potential role of neutrophils in COVID-19, we measured levels of the neutrophil activation marker S100A8/A9 (calprotectin) in hospitalized patients and determined its relationship to severity of illness and respiratory status. Patients with COVID-19 (n=172) had markedly elevated levels of calprotectin in their blood. Calprotectin tracked with other acute phase reactants including C-reactive protein, ferritin, lactate dehydrogenase, and absolute neutrophil count, but was superior in identifying patients requiring mechanical ventilation. In longitudinal samples, calprotectin rose as oxygenation worsened. When tested on day 1 or 2 of hospitalization (n=94 patients), calprotectin levels were significantly higher in patients who progressed to severe COVID-19 requiring mechanical ventilation (8039 +/- 7031 ng/ml, n=32) as compared to those who remained free of intubation (3365 +/- 3146, p<0.0001). In summary, serum calprotectin levels track closely with current and future COVID-19 severity, implicating neutrophils as potential perpetuators of inflammation and respiratory compromise in COVID-19.
Collapse
|
33
|
Scott NR, Swanson RV, Al-Hammadi N, Domingo-Gonzalez R, Rangel-Moreno J, Kriel BA, Bucsan AN, Das S, Ahmed M, Mehra S, Treerat P, Cruz-Lagunas A, Jimenez-Alvarez L, Muñoz-Torrico M, Bobadilla-Lozoya K, Vogl T, Walzl G, du Plessis N, Kaushal D, Scriba TJ, Zúñiga J, Khader SA. S100A8/A9 regulates CD11b expression and neutrophil recruitment during chronic tuberculosis. J Clin Invest 2020; 130:3098-3112. [PMID: 32134742 PMCID: PMC7259997 DOI: 10.1172/jci130546] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 02/20/2020] [Indexed: 01/01/2023] Open
Abstract
Neutrophil accumulation is associated with lung pathology during active tuberculosis (ATB). However, the molecular mechanism or mechanisms by which neutrophils accumulate in the lung and contribute to TB immunopathology are not fully delineated. Using the well-established mouse model of TB, our new data provide evidence that the alarmin S100A8/A9 mediates neutrophil accumulation during progression to chronic TB. Depletion of neutrophils or S100A8/A9 deficiency resulted in improved Mycobacterium tuberculosis (Mtb) control during chronic but not acute TB. Mechanistically, we demonstrate that, following Mtb infection, S100A8/A9 expression is required for upregulation of the integrin molecule CD11b specifically on neutrophils, mediating their accumulation during chronic TB disease. These findings are further substantiated by increased expression of S100A8 and S100A9 mRNA in whole blood in human TB progressors when compared with nonprogressors and rapidly decreased S100A8/A9 protein levels in the serum upon TB treatment. Furthermore, we demonstrate that S100A8/A9 serum levels along with chemokines are useful in distinguishing between ATB and asymptomatic Mtb-infected latent individuals. Thus, our results support targeting S100A8/A9 pathways as host-directed therapy for TB.
Collapse
Affiliation(s)
| | | | - Noor Al-Hammadi
- Division of Biostatistics, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Belinda A. Kriel
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, South African Medical Research Council (SAMRC) Centre for Tuberculosis Research, DST-NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Stellenbosch, South Africa
| | - Allison N. Bucsan
- Division of Bacteriology and
- Division of Parasitology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | | | | | - Smriti Mehra
- Division of Bacteriology and
- Division of Parasitology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | | | - Alfredo Cruz-Lagunas
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Luis Jimenez-Alvarez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Marcela Muñoz-Torrico
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Karen Bobadilla-Lozoya
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Thomas Vogl
- Institute of Immunology and
- Interdisciplinary Center for Clinical Research, University of Münster, Münster, Germany
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, South African Medical Research Council (SAMRC) Centre for Tuberculosis Research, DST-NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Stellenbosch, South Africa
| | - Nelita du Plessis
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, South African Medical Research Council (SAMRC) Centre for Tuberculosis Research, DST-NRF Centre of Excellence for Biomedical TB Research, Stellenbosch University, Stellenbosch, South Africa
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative and
- Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Joaquín Zúñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | | |
Collapse
|
34
|
Freeberg MAT, Easa A, Lillis JA, Benoit DS, van Wijnen AJ, Awad HA. Transcriptomic Analysis of Cellular Pathways in Healing Flexor Tendons of Plasminogen Activator Inhibitor 1 (PAI-1/Serpine1) Null Mice. J Orthop Res 2020; 38:43-58. [PMID: 31424116 PMCID: PMC7364818 DOI: 10.1002/jor.24448] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 08/07/2019] [Indexed: 02/04/2023]
Abstract
Injuries to flexor tendons can be complicated by fibrotic adhesions, which severely impair the function of the hand. Plasminogen activator inhibitor 1 (PAI-1/SERPINE1), a master suppressor of fibrinolysis and protease activity, is associated with adhesions. Here, we used next-generation RNA sequencing (RNA-Seq) to assess genome-wide differences in messenger RNA expression due to PAI-1 deficiency after zone II flexor tendon injury. We used the ingenuity pathway analysis to characterize molecular pathways and biological drivers associated with differentially expressed genes (DEG). Analysis of hundreds of overlapping and DEG in PAI-1 knockout (KO) and wild-type mice (C57Bl/6J) during tendon healing revealed common and distinct biological processes. Pathway analysis identified cell proliferation, survival, and senescence, as well as chronic inflammation as potential drivers of fibrotic healing and adhesions in injured tendons. Importantly, we identified the activation of PTEN signaling and the inhibition of FOXO1-associated biological processes as unique transcriptional signatures of the healing tendon in the PAI-1/Serpine1 KO mice. Further, transcriptomic differences due to the genetic deletion of PAI-1 were mechanistically linked to PI3K/Akt/mTOR, PKC, and MAPK signaling cascades. These transcriptional observations provide novel insights into the biological roles of PAI-1 in tendon healing and could identify therapeutic targets to achieve scar-free regenerative healing of tendons. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:43-58, 2020.
Collapse
Affiliation(s)
- Margaret A. T. Freeberg
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States,Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
| | - Anas Easa
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
| | - Jacquelyn A. Lillis
- Genomics Research Center, University of Rochester, Rochester, NY, United States
| | - Danielle S.W. Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States,Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
| | | | - Hani A. Awad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States,Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States,Department of Orthopedics, University of Rochester, Rochester, NY, United States
| |
Collapse
|
35
|
Sreejit G, Flynn MC, Patil M, Krishnamurthy P, Murphy AJ, Nagareddy PR. S100 family proteins in inflammation and beyond. Adv Clin Chem 2020; 98:173-231. [PMID: 32564786 DOI: 10.1016/bs.acc.2020.02.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The S100 family proteins possess a variety of intracellular and extracellular functions. They interact with multiple receptors and signal transducers to regulate pathways that govern inflammation, cell differentiation, proliferation, energy metabolism, apoptosis, calcium homeostasis, cell cytoskeleton and microbial resistance. S100 proteins are also emerging as novel diagnostic markers for identifying and monitoring various diseases. Strategies aimed at targeting S100-mediated signaling pathways hold a great potential in developing novel therapeutics for multiple diseases. In this chapter, we aim to summarize the current knowledge about the role of S100 family proteins in health and disease with a major focus on their role in inflammatory conditions.
Collapse
Affiliation(s)
| | - Michelle C Flynn
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mallikarjun Patil
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Immunology, Monash University, Melbourne, VIC, Australia
| | | |
Collapse
|
36
|
Li C, Ma Y, Fei F, Zheng M, Li Z, Zhao Q, Du J, Liu K, Lu R, Zhang S. Critical role and its underlying molecular events of the plasminogen receptor, S100A10 in malignant tumor and non-tumor diseases. J Cancer 2020; 11:826-836. [PMID: 31949486 PMCID: PMC6959022 DOI: 10.7150/jca.36203] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/13/2019] [Indexed: 12/28/2022] Open
Abstract
S100A10 is a small molecular weight protein expressed in the cytoplasm of many cells and one of the members of the S100 protein family that binds calcium and forms the largest subgroup of EF-hand proteins. The regulatory processes of S100A10 are complicated. S100A10 participates in the regulation of a variety of tumor and non-tumor diseases through cascade reactions with multitudinous signaling molecules. In malignant tumors, such as acute promyelocytic leukemia (APL) and lung cancer, S100A10 is likely involved in their progression, including invasion and metastasis through the regulation of plasmin production and subsequent plasmin-dependent stimulation of other proteases, such as matrix metalloproteinase (MMP)-2 and -9. Both the plasmin and MMPs are capable of inducing degradation of the extracellular matrix (ECM) and basement membrane, which is a critical step for tumor progression. In non-tumor diseases, the distribution of S100A10 in the brain and its interaction with 5-hydroxytryptamine 1B (5-HT1B) receptor, an important mediator in the central nervous system that maintains a dynamic balance of the neurotransmitters, correlates with depression-like behavior. S100A10 also participates in inflammatory responses through the regulation of peripheral macrophage migration to the inflammatory sites, which depends on the generation of plasmin and other proteinases at the surface of macrophages. Considerable attention should be paid to understand the significant role of S100A10 in the modulation of malignant tumor and non-tumor diseases.
Collapse
Affiliation(s)
- Chunyuan Li
- Department of Pathology, Tianjin Union Medical Center, Tianjin, P.R. China
| | - Yi Ma
- Department of ophthalmology, Tianjin Union Medical Center, Tianjin, P.R. China
| | - Fei Fei
- Department of Pathology, Tianjin Union Medical Center, Tianjin, P.R. China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, P.R. China
| | - Zugui Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Qi Zhao
- Tianjin Medical University, Tianjin, P.R. China
| | - Jiaxing Du
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Kai Liu
- Tianjin Medical University, Tianjin, P.R. China
| | - Rui Lu
- Tianjin Medical University, Tianjin, P.R. China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, P.R. China
| |
Collapse
|
37
|
Duvvuri B, Pachman LM, Morgan G, Khojah AM, Klein-Gitelman M, Curran ML, Doty S, Lood C. Neutrophil Extracellular Traps in Tissue and Periphery in Juvenile Dermatomyositis. Arthritis Rheumatol 2020; 72:348-358. [PMID: 31403247 DOI: 10.1002/art.41078] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/08/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Neutrophils are key immune cells participating in host defense through several mechanisms, including the formation of neutrophil extracellular traps (NETs). This study was undertaken to investigate the role of neutrophils in juvenile dermatomyositis (JDM). METHODS Electron microscopy was used to identify neutrophils in tissue. NETs were also imaged using fluorescence microscopy and quantified using a myeloperoxidase-DNA enzyme-linked immunosorbent assay (ELISA) in plasma obtained from healthy children (n = 20), disease controls (n = 29), JDM patients (n = 66), and JDM patients with history of calcifications (n = 20). Clinical data included disease activity scores and complement C4 levels. Levels of immune complexes (ICs) and calprotectin were analyzed using ELISA. RESULTS Using electron microscopy, neutrophils were found to infiltrate affected muscle tissue, engulfing deposited calcium crystals. Uptake of the crystals led to neutrophil activation (P < 0.01) and subsequent phosphatidylinositol 3-kinase- and NADPH oxidase-dependent but peptidylarginine deiminase 4-independent formation of NETs, which contained mitochondrial DNA (P < 0.05), as confirmed in vivo (P < 0.001) and in vitro (P < 0.01). Peripheral NET levels were associated with calcinosis (P = 0.01), ICs (P = 0.008), and interleukin-8 levels (P = 0.004). Children with JDM had impaired NET clearance (P = 0.01), associated with autoantibody profiles including melanoma differentiation-associated protein 5 (P = 0.005), and depressed complement C4 levels (r = -0.72, P = 0.002). Furthermore, children with JDM showed evidence of neutrophil activation, with elevated levels of peroxidase activity (P = 0.02) and calprotectin (P < 0.01), which were associated with disease activity (P = 0.007), and dyslipidemia (odds ratio 4.7, P < 0.05). CONCLUSION We found novel mechanisms of both calcium crystal-mediated neutrophil activation and cell death in JDM pathophysiology. Targeting this pathway may reduce the frequency and extent of calcinosis, as well as prevent long-term development of comorbidities, including atherosclerosis.
Collapse
Affiliation(s)
| | - Lauren M Pachman
- Cure JM Center of Excellence, Ann & Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Gabrielle Morgan
- Cure JM Center of Excellence, Ann & Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Amer M Khojah
- Cure JM Center of Excellence, Ann & Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Marisa Klein-Gitelman
- Ann & Robert H. Lurie Children's Hospital of Chicago and Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Stephen Doty
- Hospital for Special Surgery, New York, New York
| | | |
Collapse
|
38
|
Diklić M, Mitrović-Ajtić O, Subotički T, Djikić D, Kovačić M, Bjelica S, Beleslin-Čokić B, Tošić M, Leković D, Gotić M, Santibanez JF, Čokić VP. IL6 inhibition of inflammatory S100A8/9 proteins is NF-κB mediated in essential thrombocythemia. Cell Biochem Funct 2019; 38:362-372. [PMID: 31885098 DOI: 10.1002/cbf.3482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/24/2019] [Accepted: 12/15/2019] [Indexed: 11/07/2022]
Abstract
This study has been performed to determine the mechanism of activation of the myeloid related S100A proteins by inflammatory cytokines in myeloproliferative neoplasm (MPN). Besides microarray analysis of MPN-derived CD34+ cells, we analysed the pro-inflammatory IL6 and anti-inflammatory IL10 dependence of NF-κB, PI3K-AKT, and JAK-STAT signalling during induction of S100A proteins in mononuclear cells of MPN, by immunoblotting and flow cytometry. We observed the reduced gene expression linked to NF-κB and inflammation signalling in MPN-derived CD34+ cells. Both IL6 and IL10 reduced S100A8 and 100A9 protein levels mediated via NF-κB and PI3K signalling, respectively, in mononuclear cells of essential thrombocythemia (ET). We also determined the increased percentage of S100A8 and S100A9 positive granulocytes in ET and primary myelofibrosis, upgraded by the JAK2V617F mutant allele burden. S100A8/9 heterodimer induced JAK1/2-dependent mitotic arrest of the ET-derived granulocytes. SIGNIFICANCE OF THE STUDY: We demonstrated that inflammation reduced the myeloid related S100A8/9 proteins by negative feedback mechanism in ET. S100A8/9 can be a diagnostic marker of inflammation in MPN, supported by the concomitant NF-κB and JAK1/2 signalling inhibition in regulation of myeloproliferation and therapy of MPN.
Collapse
Affiliation(s)
- Miloš Diklić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Olivera Mitrović-Ajtić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Tijana Subotički
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Dragoslava Djikić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Marijana Kovačić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Sunčica Bjelica
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Bojana Beleslin-Čokić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Genetic Laboratory, Clinical Center of Serbia, Belgrade, Serbia
| | - Milica Tošić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Danijela Leković
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia.,School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Mirjana Gotić
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia.,School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Juan F Santibanez
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| | - Vladan P Čokić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgade, Serbia
| |
Collapse
|
39
|
Wang C, Kong Y, Ding Y, Sun J, Chen T. Serum Calprotectin Levels and Outcome Following Percutaneous Coronary Intervention in Patients with Diabetes and Acute Coronary Syndrome. Med Sci Monit 2019; 25:9517-9523. [PMID: 31834876 PMCID: PMC6929552 DOI: 10.12659/msm.918126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background A retrospective study of data from a prospective clinical registry was conducted to evaluate the prognostic role of serum calprotectin in patients with diabetes who underwent percutaneous coronary intervention (PCI) for acute coronary syndrome (ACS). Material/Methods Data were retrieved for 273 patients with diabetes mellitus who underwent PCI for primary ACS in a single center. Serum calprotectin levels were measured before PCI. Baseline clinical data included the Global Registry of Acute Coronary Events (GRACE) risk score for ACS. All patients underwent regular follow-up for major adverse cardiovascular events (MACE) during 12 months after PCI, including target vessel revascularization (TVR), defined as the need for an unplanned repeat PCI or coronary artery procedure. The predictive value of serum calprotectin for MACE was analyzed by using univariate and multivariate analysis and receiver operating characteristic (ROC) curve analysis. Results At the final follow-up, 47 of the 273 patients studies experienced MACE. Optimal cutoff values for serum calprotectin levels predictive for MACE stratified patients into a high calprotectin group and a low calprotectin group. The incidence of MACE and TVR in the high calprotectin group was significantly greater than in the low calprotectin group (21.9% vs. 11.5%; P=0.02). Multivariate analysis, adjusted for confounders, showed that the serum level of calprotectin was an independent risk factor for MACE (HR, 1.56; 95% CI, 1.08–4.62; P=0.01). Conclusions In patients with diabetes and the co-morbidity of ACS, a high serum level of calprotectin was associated with a significantly increased risk for MACE following PCI.
Collapse
Affiliation(s)
- Chengji Wang
- Fifth Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Yu Kong
- Fifth Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Yuanyuan Ding
- Department of Gastroenterology, Jining No. 1 People's Hospital, Jining, Shandong, China (mainland)
| | - Jingzhi Sun
- Fifth Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Tao Chen
- Department of Critical Care Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| |
Collapse
|
40
|
Hemolysis Derived Products Toxicity and Endothelium: Model of the Second Hit. Toxins (Basel) 2019; 11:toxins11110660. [PMID: 31766155 PMCID: PMC6891750 DOI: 10.3390/toxins11110660] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/02/2019] [Accepted: 11/06/2019] [Indexed: 12/16/2022] Open
Abstract
Vascular diseases are multifactorial, often requiring multiple challenges, or ‘hits’, for their initiation. Intra-vascular hemolysis illustrates well the multiple-hit theory where a first event lyses red blood cells, releasing hemolysis-derived products, in particular cell-free heme which is highly toxic for the endothelium. Physiologically, hemolysis derived-products are rapidly neutralized by numerous defense systems, including haptoglobin and hemopexin which scavenge hemoglobin and heme, respectively. Likewise, cellular defense mechanisms are involved, including heme-oxygenase 1 upregulation which metabolizes heme. However, in cases of intra-vascular hemolysis, those systems are overwhelmed. Heme exerts toxic effects by acting as a damage-associated molecular pattern and promoting, together with hemoglobin, nitric oxide scavenging and ROS production. In addition, it activates the complement and the coagulation systems. Together, these processes lead to endothelial cell injury which triggers pro-thrombotic and pro-inflammatory phenotypes. Moreover, among endothelial cells, glomerular ones display a particular susceptibility explained by a weaker capacity to counteract hemolysis injury. In this review, we illustrate the ‘multiple-hit’ theory through the example of intra-vascular hemolysis, with a particular focus on cell-free heme, and we advance hypotheses explaining the glomerular susceptibility observed in hemolytic diseases. Finally, we describe therapeutic options for reducing endothelial injury in hemolytic diseases.
Collapse
|
41
|
Ganta VC, Choi M, Farber CR, Annex BH. Antiangiogenic VEGF 165b Regulates Macrophage Polarization via S100A8/S100A9 in Peripheral Artery Disease. Circulation 2019; 139:226-242. [PMID: 30586702 DOI: 10.1161/circulationaha.118.034165] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Atherosclerotic occlusions decrease blood flow to the lower limbs, causing ischemia and tissue loss in patients with peripheral artery disease (PAD). No effective medical therapies are currently available to induce angiogenesis and promote perfusion recovery in patients with severe PAD. Clinical trials aimed at inducing vascular endothelial growth factor (VEGF)-A levels, a potent proangiogenic growth factor to induce angiogenesis, and perfusion recovery were not successful. Alternate splicing in the exon-8 of VEGF-A results in the formation of VEGFxxxa (VEGF165a) and VEGFxxxb (VEGF165b) isoforms with existing literature focusing on VEGF165b's role in inhibiting vascular endothelial growth factor receptor 2-dependent angiogenesis. However, we have recently shown that VEGF165b blocks VEGF-A-induced endothelial vascular endothelial growth factor receptor 1 (VEGFR1) activation in ischemic muscle to impair perfusion recovery. Because macrophage-secreted VEGF165b has been shown to decrease angiogenesis in peripheral artery disease, and macrophages were well known to play important roles in regulating ischemic muscle vascular remodeling, we examined the role of VEGF165b in regulating macrophage function in PAD. METHODS Femoral artery ligation and resection were used as an in vivo preclinical PAD model, and hypoxia serum starvation was used as an in vitro model for PAD. Experiments including laser-Doppler perfusion imaging, adoptive cell transfer to ischemic muscle, immunoblot analysis, ELISAs, immunostainings, flow cytometry, quantitative polymerase chain reaction analysis, and RNA sequencing were performed to determine a role of VEGF165b in regulating macrophage phenotype and function in PAD. RESULTS First, we found increased VEGF165b expression with increased M1-like macrophages in PAD versus non-PAD (controls) muscle biopsies. Next, using in vitro hypoxia serum starvation, in vivo pre clinical PAD models, and adoptive transfer of VEGF165b-expressing bone marrow-derived macrophages or VEGFR1+/- bone marrow-derived macrophages (M1-like phenotype), we demonstrate that VEGF165b inhibits VEGFR1 activation to induce an M1-like phenotype that impairs ischemic muscle neovascularization. Subsequently, we found S100A8/S100A9 as VEGFR1 downstream regulators of macrophage polarization by RNA-Seq analysis of hypoxia serum starvation-VEGFR1+/+ versus hypoxia serum starvation-VEGFR1+/- bone marrow-derived macrophages. CONCLUSIONS In our current study, we demonstrate that increased VEGF165b expression in macrophages induces an antiangiogenic M1-like phenotype that directly impairs angiogenesis. VEGFR1 inhibition by VEGF165b results in S100A8/S100A9-mediated calcium influx to induce an M1-like phenotype that impairs ischemic muscle revascularization and perfusion recovery.
Collapse
Affiliation(s)
- Vijay Chaitanya Ganta
- Robert M. Berne Cardiovascular Research Center (V.C.G., M.C., B.H.A.), University of Virginia, Charlottesville.,Division Cardiovascular Medicine, Department of Medicine (V.C.G., B.H.A.), University of Virginia, Charlottesville
| | - Min Choi
- Robert M. Berne Cardiovascular Research Center (V.C.G., M.C., B.H.A.), University of Virginia, Charlottesville
| | - Charles R Farber
- Department of Public Health Sciences (C.R.F.), University of Virginia, Charlottesville
| | - Brian H Annex
- Robert M. Berne Cardiovascular Research Center (V.C.G., M.C., B.H.A.), University of Virginia, Charlottesville.,Division Cardiovascular Medicine, Department of Medicine (V.C.G., B.H.A.), University of Virginia, Charlottesville
| |
Collapse
|
42
|
Advanced Glycation End Products of Bovine Serum Albumin Suppressed Th1/Th2 Cytokine but Enhanced Monocyte IL-6 Gene Expression via MAPK-ERK and MyD88 Transduced NF-κB p50 Signaling Pathways. Molecules 2019; 24:molecules24132461. [PMID: 31277476 PMCID: PMC6652144 DOI: 10.3390/molecules24132461] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 02/06/2023] Open
Abstract
Advanced glycation end products (AGE), the most known aging biomarker, may cause “inflamm-aging” (i.e., chronic low-grade inflammation that develops with aging) in both aged and diabetes groups. However, the molecular bases of inflamm-aging remain obscure. We prepared AGE by incubating BSA (0.0746 mmol/L) + glucose (0.5 mol/L) at 37 °C in 5% CO2–95% air for 1–180 days. The lysine glycation in BSA–AGE reached 77% on day 30 and 100% after day 130, whereas the glycation of arginine and cysteine was minimal. The Nε-(carboxymethyl)-lysine content in BSA–AGE was also increased with increasing number of incubation days. The lectin-binding assay revealed that the glycation of BSA not only altered the conformational structure, but lost binding capacity with various lectins. An immunological functional assay showed that BSA–AGE > 8 μg/mL significantly suppressed normal human Th1 (IL-2 and IFN-γ) and Th2 (IL-10) mRNA expression, whereas AGE > 0.5 μg/mL enhanced monocyte IL-6 production irrelevant to cell apoptosis. The AGE-enhanced monocyte IL-6 production was via MAPK–ERK and MyD88-transduced NF-κBp50 signaling pathways. To elucidate the structure–function relationship of BSA–AGE-enhanced IL-6 production, we pre-preincubated BSA–AGE with different carbohydrate-degrading, protein-degrading, and glycoprotein-degrading enzymes. We found that trypsin and carboxypeptidase Y suppressed whereas β-galactosidase enhanced monocyte IL-6 production. In conclusion, BSA–AGE exerted both immunosuppressive and pro-inflammatory effects that are the molecular basis of inflamm-aging in aged and diabetes groups.
Collapse
|
43
|
Di Ceglie I, Blom AB, Davar R, Logie C, Martens JHA, Habibi E, Böttcher LM, Roth J, Vogl T, Goodyear CS, van der Kraan PM, van Lent PL, van den Bosch MH. The alarmin S100A9 hampers osteoclast differentiation from human circulating precursors by reducing the expression of RANK. FASEB J 2019; 33:10104-10115. [PMID: 31199668 DOI: 10.1096/fj.201802691rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The alarmin S100A8/A9 is implicated in sterile inflammation-induced bone resorption and has been shown to increase the bone-resorptive capacity of mature osteoclasts. Here, we investigated the effects of S100A9 on osteoclast differentiation from human CD14+ circulating precursors. Hereto, human CD14+ monocytes were isolated and differentiated toward osteoclasts with M-CSF and receptor activator of NF-κB (RANK) ligand (RANKL) in the presence or absence of S100A9. Tartrate-resistant acid phosphatase staining showed that exposure to S100A9 during monocyte-to-osteoclast differentiation strongly decreased the numbers of multinucleated osteoclasts. This was underlined by a decreased resorption of a hydroxyapatite-like coating. The thus differentiated cells showed a high mRNA and protein production of proinflammatory factors after 16 h of exposure. In contrast, at d 4, the cells showed a decreased production of the osteoclast-promoting protein TNF-α. Interestingly, S100A9 exposure during the first 16 h of culture only was sufficient to reduce osteoclastogenesis. Using fluorescently labeled RANKL, we showed that, within this time frame, S100A9 inhibited the M-CSF-mediated induction of RANK. Chromatin immunoprecipitation showed that this was associated with changes in various histone marks at the epigenetic level. This S100A9-induced reduction in RANK was in part recovered by blocking TNF-α but not IL-1. Together, our data show that S100A9 impedes monocyte-to-osteoclast differentiation, probably via a reduction in RANK expression.-Di Ceglie, I., Blom, A. B., Davar, R., Logie, C., Martens, J. H. A., Habibi, E., Böttcher, L.-M., Roth, J., Vogl, T., Goodyear, C. S., van der Kraan, P. M., van Lent, P. L., van den Bosch, M. H. The alarmin S100A9 hampers osteoclast differentiation from human circulating precursors by reducing the expression of RANK.
Collapse
Affiliation(s)
- Irene Di Ceglie
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arjen B Blom
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robab Davar
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Colin Logie
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Ehsan Habibi
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Lisa-Marie Böttcher
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Carl S Goodyear
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Peter M van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter L van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | |
Collapse
|
44
|
Kashirina DN, Percy AJ, Pastushkova LK, Borchers CH, Kireev KS, Ivanisenko VA, Kononikhin AS, Nikolaev EN, Larina IM. The molecular mechanisms driving physiological changes after long duration space flights revealed by quantitative analysis of human blood proteins. BMC Med Genomics 2019; 12:45. [PMID: 30871558 PMCID: PMC6416832 DOI: 10.1186/s12920-019-0490-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The conditions of space flight have a significant effect on the physiological processes in the human body, yet the molecular mechanisms driving physiological changes remain unknown. METHODS Blood samples of 18 Russian cosmonauts who had conducted long-duration missions to the International Space Station were collected 30 days before launch and on the first and seventh days after landing. RESULTS A panel of 125 proteins in the blood plasma was quantitated by a well-established and highly regarded targeted mass spectrometry approach. This method involves the monitoring of multiple reactions in conjunction with stable isotope-labeled standards at the University of Victoria - Genome BC Proteomics Centre. CONCLUSIONS Reduction of circulating plasma volume during space flight and activation of fluid retention at the final stage of the flight affect the changes in plasma protein concentrations present in the first days after landing. Using an ANOVA approach, it was revealed that only 1 protein (S100A9) reliably responded to space flight conditions. This protein plays an important role in the functioning of the endothelium and can serve as a marker for activation of inflammatory reactions. Concentrations of the proteins of complement, coagulation cascades, and acute phase reactants increase in the blood of cosmonauts as measured the first day after landing. Most of these proteins' concentrations continue to increase by the 7th day after space flight. Similar dynamics are observed for proteases and their inhibitors. Thus, there is a shift in proteolytic blood systems, which is necessary for the restoration of muscle tissue and maintenance of oncotic homeostasis.
Collapse
Affiliation(s)
- Daria N. Kashirina
- Institute for Biomedical Problems – Russian Federation State Scientific Research Center of RAS, Moscow, Russia
| | - Andrew J. Percy
- Genome British Columbia Proteomics Centre, University of Victoria, Victoria, BC Canada
| | - Liudmila Kh. Pastushkova
- Institute for Biomedical Problems – Russian Federation State Scientific Research Center of RAS, Moscow, Russia
| | - Christoph H. Borchers
- Genome British Columbia Proteomics Centre, University of Victoria, Victoria, BC Canada
| | - Kirill S. Kireev
- Yu.A.Gagarin Research and Test Cosmonaut Training Center, Star City, Moscow Region, Russia
| | - Vladimir A. Ivanisenko
- Institute of Cytology and Genetics of SB RAS, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Alexey S. Kononikhin
- Institute for Biomedical Problems – Russian Federation State Scientific Research Center of RAS, Moscow, Russia
- V.L. Talrose Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow region, Russia
| | - Eugene N. Nikolaev
- V.L. Talrose Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow region, Russia
- Skolkovo Institute of Science and Technology, Skoltech, Moscow region, Russia
| | - Irina M. Larina
- Institute for Biomedical Problems – Russian Federation State Scientific Research Center of RAS, Moscow, Russia
| |
Collapse
|
45
|
Zheng X, Huo X, Zhang Y, Wang Q, Zhang Y, Xu X. Cardiovascular endothelial inflammation by chronic coexposure to lead (Pb) and polycyclic aromatic hydrocarbons from preschool children in an e-waste recycling area. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 246:587-596. [PMID: 30597391 DOI: 10.1016/j.envpol.2018.12.055] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 11/22/2018] [Accepted: 12/17/2018] [Indexed: 02/05/2023]
Abstract
Lead (Pb) and polycyclic aromatic hydrocarbon (PAH) exposure is positively associated with cardiovascular disease (CVD), and the possible potential mechanism may be caused by damage to the endothelium by modulation of inflammatory processes. No comprehensive research shows co-exposure of Pb and PAH on cardiovascular endothelial inflammation in electronic waste (e-waste) exposed populations. Given this, the aim of this study is to provide evidence for a relationship between Pb and PAH co-exposure and cardiovascular endothelial inflammation, in an e-waste-exposed population, to delineate the link between a potential mechanism for CVD and environmental exposure. We recruited 203 preschool children (3-7 years) were enrolled from Guiyu (e-waste-exposed group, n = 105) and Haojiang (reference group, n = 98). Blood Pb levels and urinary PAH metabolites were measured. Percentages of T cells, CD4+ T cells and CD8+ T cells, complete blood counts, endothelial inflammation biomarker (serum S100A8/A9), and other inflammatory biomarkers [serum interleukin (IL)-6, IL-12p70, gamma interferon-inducible protein 10 (IP-10)] levels were evaluated. Blood Pb, total urinary hydroxylated PAH (ΣOHPAH), total hydroxynaphthalene (ΣOHNap) and total hydroxyfluorene (ΣOHFlu) levels, S100A8/A9, IL-6, IL-12p70 and IP-10 concentrations, absolute counts of monocytes, neutrophils, and leukocytes, as well as CD4+ T cell percentages were significantly higher in exposed children. Elevated blood Pb, urinary 2-hydroxynaphthalene (2-OHNap) and ΣOHFlu levels were associated with higher levels of IL-6, IL-12p70, IP-10, CD4+ T cell percentages, neutrophil and monocyte counts. Mediator models indicated that neutrophils exert the significant mediation effect on the relationship between blood Pb levels and S100A8/A9. IL-6 exerts a significant mediation effect on the relationship between blood Pb levels and IP-10, as well as the relationship between urinary ΣOHFlu levels and IP-10. Our results indicate that children with elevated exposure levels of Pb and PAHs have exacerbated vascular endothelial inflammation, which may indicate future CVD risk in e-waste recycling areas.
Collapse
Affiliation(s)
- Xiangbin Zheng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Yu Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, 9713, GZ, the Netherlands
| | - Qihua Wang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yuling Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
46
|
Sanders KA, Delker DA, Huecksteadt T, Beck E, Wuren T, Chen Y, Zhang Y, Hazel MW, Hoidal JR. RAGE is a Critical Mediator of Pulmonary Oxidative Stress, Alveolar Macrophage Activation and Emphysema in Response to Cigarette Smoke. Sci Rep 2019; 9:231. [PMID: 30659203 PMCID: PMC6338799 DOI: 10.1038/s41598-018-36163-z] [Citation(s) in RCA: 41] [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: 04/18/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023] Open
Abstract
The receptor for advanced glycation end products (RAGE), a cell membrane receptor, recognizes ligands produced by cigarette smoke (CS) and has been implicated in the pathogenesis of COPD. We demonstrate that deletion or pharmacologic inhibition of RAGE prevents development of CS-induced emphysema. To identify molecular pathways by which RAGE mediates smoking related lung injury we performed unbiased gene expression profiling of alveolar macrophages (AM) obtained from RAGE null and C57BL/6 WT mice exposed to CS for one week or four months. Pathway analysis of RNA expression identified a number of genes integral to the pathogenesis of COPD impacted by the absence of RAGE. Altered expression of antioxidant response genes and lung protein 4-HNE immunostaining suggest attenuated oxidative stress in the RAGE null mice despite comparable CS exposure and lung leukocyte burden as the WT mice. Reduced endoplasmic reticulum stress in response to CS exposure also was observed in the AM from RAGE null mice. These findings provide novel insight into the sources of oxidative stress, macrophage activation, and the pathogenesis of lung disease due to CS exposure.
Collapse
Affiliation(s)
- Karl A Sanders
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Don A Delker
- Division of Gastroenterology, Hepatology, and Nutrition, University of Utah, Salt Lake City, Utah, USA
| | - Tom Huecksteadt
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Emily Beck
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Tanna Wuren
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Yuntian Chen
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mark W Hazel
- Division of Gastroenterology, Hepatology, and Nutrition, University of Utah, Salt Lake City, Utah, USA
| | - John R Hoidal
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA.
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA.
| |
Collapse
|
47
|
Kubo F, Ariestanti DM, Oki S, Fukuzawa T, Demizu R, Sato T, Sabirin RM, Hirose S, Nakamura N. Loss of the adhesion G-protein coupled receptor ADGRF5 in mice induces airway inflammation and the expression of CCL2 in lung endothelial cells. Respir Res 2019; 20:11. [PMID: 30654796 PMCID: PMC6337809 DOI: 10.1186/s12931-019-0973-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 01/02/2019] [Indexed: 01/09/2023] Open
Abstract
Background Adhesion G-protein coupled receptor F5 (ADGRF5) was recently identified as an essential regulator of pulmonary surfactant homeostasis in alveolar type II cells. We previously showed that in addition to abnormal surfactant accumulation, Adgrf5-deficient (Adgrf5−/−) mice exhibit emphysema-like signs, suggesting a possible role for ADGRF5 in immune regulation. Here, we extended the phenotypic analysis of Adgrf5−/− mice to help understand its biological role in the lung, and especially in immune regulation. Methods Histological features of lungs were evaluated by Alcian blue and Masson’s trichrome staining. Quantitative real-time PCR (qPCR) and western blot analyses were performed to analyze the differential expression of genes/proteins related to airway inflammation in lungs between wildtype and Adgrf5−/− mice. Acid–base status was assessed by performing blood gas tests and urine pH measurements. Inflammatory cell counting was performed using Giemsa-stained bronchoalveolar lavage cells. Serum IgE concentrations were determined by enzyme-linked immunosorbent assay. The expression of Ccl2, S100a8, S100a9, and Saa3 in primary lung endothelial cells (ECs) was determined by qPCR and/or western blotting. Finally, the effect of administrating RS504393 to 2-week-old Adgrf5−/− mice on gene expression in the lungs was analyzed by qPCR. Results Adgrf5−/− mice exhibited several features of chronic airway inflammation (mucous cell metaplasia, mucus hyperproduction, subepithelial fibrosis, respiratory acidosis, high serum IgE, mast cell accumulation, and neutrophilia) in parallel with elevated expression of genes involved in mucous cell metaplasia (Muc5ac, Muc5b, Slc26a4, and Clca1), fibrosis (Tgfb1, Col1a1, Fn1, and Tnc), and type 2 immune response (Il4, Il5, Il13, IL-25, and IL-33) at 12 and/or 30 weeks of age. In contrast, mRNA expression of Ccl2, S100a8, and S100a9 was upregulated in embryonic or neonatal Adgrf5−/− lungs as well as in lung ECs of Adgrf5−/− mice at 1 week of age. RS504393 treatment suppressed the upregulation of S100a8, S100a9, Slc26a4, and Il5 in Adgrf5−/− lungs. Conclusions Targeted disruption of ADGRF5 results in the development of airway inflammation, which is likely mediated by the type 2 immune response and possibly CCL2-mediated inflammation. ADGRF5 also has a potential role in the regulation of genes encoding CCL2 in lung ECs, thereby maintaining immune homeostasis. Electronic supplementary material The online version of this article (10.1186/s12931-019-0973-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fumimasa Kubo
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Donna Maretta Ariestanti
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Souta Oki
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Taku Fukuzawa
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Ryotaro Demizu
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Tomoya Sato
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Rahmaningsih Mara Sabirin
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan.,Department of Physiology, Faculty of Medicine, Public Health and Nursing, Gadjah Mada University, JI.Farmako Sekip Utara, Yogyakarta, 55281, Indonesia
| | - Shigehisa Hirose
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Nobuhiro Nakamura
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan.
| |
Collapse
|
48
|
Zhou QY, Lin W, Zhu XX, Xu SL, Ying MX, Shi L, Lin BJ. Increased Plasma Levels of S100A8, S100A9, and S100A12 in Chronic Spontaneous Urticaria. Indian J Dermatol 2019; 64:441-446. [PMID: 31896840 PMCID: PMC6862366 DOI: 10.4103/ijd.ijd_375_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background: Chronic spontaneous urticaria (CSU) is a skin disorder with an important immunologic profile. S100A8, S100A9, and S100A12 are the members of S100 family that have been reported to play important role in autoimmune diseases, but the characteristics of these three S100 members have not been defined in CSU. Aims: This study was performed to investigate the levels of these three S100s in patients with CSU and to study whether they were associated with the severity and clinical characteristics of CSU. Materials and Methods: The levels of plasma S100A8, S100A9, and S100A12 were measured in 51 CSU patients and 20 healthy controls using enzyme linked immunosorbent assay kits. The values in the patient group and that of the healthy controls were statistically compared. The relationships between the different markers were evaluated by correlation analysis. Results: The plasma levels of S100A8, S100A9, and S100A12 were significantly higher in CSU patients than those in controls. Interestingly, the level of S100A12 was significantly correlated with S100A8 and S100A9 in CSU patients (P < 0.05 and P < 0.001, respectively). In addition, S100A8, S100A9, and S100A12 were all significantly inversely correlated with blood basophil percentage. Conclusions: Plasma S100A8, S100A9, and S100A12 levels were elevated in CSU patients. They might be useful biomarkers of CSU, with the potential role in the pathogenesis of CSU.
Collapse
Affiliation(s)
- Qiong-Yan Zhou
- Department of Dermatology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, China
| | - Wei Lin
- Department of Pharmacy, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, China
| | - Xiao-Xia Zhu
- Department of Dermatology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Su-Ling Xu
- Department of Dermatology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, China
| | - Meng-Xia Ying
- Department of Dermatology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, China
| | - Lei Shi
- Department of Dermatology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, China
| | - Bing-Jiang Lin
- Department of Dermatology, Ningbo First Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
49
|
Fang X, Dorcely B, Ding XP, Yin S, Son NH, Hu SL, Goldberg IJ. Glycemic reduction alters white blood cell counts and inflammatory gene expression in diabetes. J Diabetes Complications 2018; 32:1027-1034. [PMID: 30197161 PMCID: PMC6174091 DOI: 10.1016/j.jdiacomp.2018.08.003] [Citation(s) in RCA: 8] [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: 06/13/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Systemic inflammation contributes to cardiovascular disease in patients with type 2 diabetes, and elevated white blood cell (WBC) counts are an established risk factor. Our goal is to describe changes in WBCs and inflammatory markers after glycemic reductions in diabetes. RESEARCH DESIGN AND METHODS This study enrolled 63 subjects with poorly controlled diabetes, defined as hemoglobin A1c (HbA1c) ≥8% [64 mmol/mol]. Circulating granulocytes and mononuclear cells were separated by histopaque double-density protocol. Inflammatory markers from these isolated WBCs were assessed at baseline and after 3 months of medical management. RESULTS After 3 months, significant glycemic reduction, defined as a decrease in HbA1c ≥ 1.5%, occurred in 42 subjects. Fasting plasma glucose decreased by 47% (165.6 mg/dL), and HbA1c decreased from 10.2 ± 1.8 to 6.8 ± 0.9. Glycemic reductions were associated with a 9.4% decrease in total WBC counts, 10.96% decrease in neutrophils, and 21.74% decrease in monocytes. The mRNA levels of inflammatory markers from granulocytes and mononuclear cells decreased, including receptor for advanced glycation endproducts; S100 calcium binding proteins A8, A9, A12; krüppel-like factor 5; and IL-1. Also, circulating levels of IL-1β and C-reactive protein decreased. Insulin dose was a mediator between HbA1c and both total WBC and neutrophil counts, but not changes in WBC inflammatory markers. In contrast, the 17 subjects without significant glycemic reductions showed no significant differences in their WBC counts and proteins of inflammatory genes. CONCLUSION Significant glycemic reduction in subjects with poorly controlled diabetes led to reduced circulating WBC counts and inflammatory gene expression.
Collapse
Affiliation(s)
- Xiang Fang
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, No. 17, Lujiang Road, Hefei City 230001, Anhui Province, China; Gerontology Institute of Anhui Province, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, Anhui, China
| | - Brenda Dorcely
- Division of Endocrinology, Diabetes and Metabolism, NYU Langone Health, New York, NY 10016, United States of America
| | - Xi-Ping Ding
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, No. 17, Lujiang Road, Hefei City 230001, Anhui Province, China
| | - Shi Yin
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, No. 17, Lujiang Road, Hefei City 230001, Anhui Province, China
| | - Ni-Huiping Son
- Division of Endocrinology, Diabetes and Metabolism, NYU Langone Health, New York, NY 10016, United States of America
| | - Shi-Lian Hu
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, No. 17, Lujiang Road, Hefei City 230001, Anhui Province, China; Gerontology Institute of Anhui Province, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, Anhui, China
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, NYU Langone Health, New York, NY 10016, United States of America.
| |
Collapse
|
50
|
Tammaro A, Florquin S, Brok M, Claessen N, Butter LM, Teske GJD, de Boer OJ, Vogl T, Leemans JC, Dessing MC. S100A8/A9 promotes parenchymal damage and renal fibrosis in obstructive nephropathy. Clin Exp Immunol 2018; 193:361-375. [PMID: 29746703 PMCID: PMC6150262 DOI: 10.1111/cei.13154] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2018] [Indexed: 12/11/2022] Open
Abstract
Despite advances in our understanding of the mechanisms underlying the progression of chronic kidney disease and the development of fibrosis, only limited efficacious therapies exist. The calcium binding protein S100A8/A9 is a damage-associated molecular pattern which can activate Toll-like receptor (TLR)-4 or receptor for advanced glycation end-products (RAGE). Activation of these receptors is involved in the progression of renal fibrosis; however, the role of S100A8/A9 herein remains unknown. Therefore, we analysed S100A8/A9 expression in patients and mice with obstructive nephropathy and subjected wild-type and S100A9 knock-out mice lacking the heterodimer S100A8/A9 to unilateral ureteral obstruction (UUO). We found profound S100A8/A9 expression in granulocytes that infiltrated human and murine kidney, together with enhanced renal expression over time, following UUO. S100A9 KO mice were protected from UUO-induced renal fibrosis, independently of leucocyte infiltration and inflammation. Loss of S100A8/A9 protected tubular epithelial cells from UUO-induced apoptosis and critical epithelial-mesenchymal transition steps. In-vitro studies revealed S100A8/A9 as a novel mediator of epithelial cell injury through loss of cell polarity, cell cycle arrest and subsequent cell death. In conclusion, we demonstrate that S100A8/A9 mediates renal damage and fibrosis, presumably through loss of tubular epithelial cell contacts and irreversible damage. Suppression of S100A8/A9 could be a therapeutic strategy to halt renal fibrosis in patients with chronic kidney disease.
Collapse
Affiliation(s)
- A. Tammaro
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - S. Florquin
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - M. Brok
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - N. Claessen
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - L. M. Butter
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - G. J. D. Teske
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - O. J. de Boer
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - T. Vogl
- Institute of ImmunologyUniversity of MünsterMünsterGermany
| | - J. C. Leemans
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - M. C. Dessing
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| |
Collapse
|