1
|
Xuan C, Chen D, Zhang S, Li C, Fang Q, Chen D, Liu J, Jiang X, Zhang Y, Shen W, Cai G, Chen X, Li P. Isoquercitrin Alleviates Diabetic Nephropathy by Inhibiting STAT3 Phosphorylation and Dimerization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2414587. [PMID: 40184310 DOI: 10.1002/advs.202414587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/10/2025] [Indexed: 04/06/2025]
Abstract
At the convergence point of multiple cytokine signals, signal transducer and activator of transcription 3 (STAT3) is a highly promising therapeutic target for diabetic nephropathy. Isoquercitrin, a natural small-molecule inhibitor of STAT3, may have beneficial effects on diabetic nephropathy; however, the underlying mechanism remains unclear. Isoquercitrin significantly mitigated renal inflammation and fibrosis by inhibiting STAT3 activity in mice with diabetic nephropathy. Moreover, STAT3 is a direct molecular target of isoquercitrin, which as corroborated by tight and stable noncovalent binding between them. This interaction is mechanistically supported by the affinity of isoquercitrin for the Ser668-Gln635-Gln633 region within the pY+1 binding pocket of the SH2 domain. This binding obstructs pivotal processes like STAT3 phosphorylation and dimerization, thereby suppressing its transcriptional function. Finally, a kidney-targeted nanocarrier, Iso@PEG-GK, is developed to load isoquercitrin, thus enhancing its therapeutic precision for diabetic nephropathy. Iso@PEG-GK significantly improved the absorption and renal distribution of isoquercitrin. This study is the first to demonstrate that isoquercitrin exerts a significant protective effect against diabetic nephropathy and may provide a novel therapeutic drug for this disease.
Collapse
Affiliation(s)
- Chen Xuan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| | - Donghui Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Shuangna Zhang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| | - Chaofan Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| | - Qingyun Fang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| | - Dinghua Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| | - Jiabao Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| | - Xin Jiang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| | - Yingjie Zhang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| | - Wanjun Shen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Medical Devices and Integrated Traditional Chinese and Western Drug Development for Severe Kidney Diseases, Beijing Key Laboratory of Digital Intelligent TCM for the Prevention and Treatment of Pan-vascular Diseases, Key Disciplines of National Administration of Traditional Chinese Medicine (zyyzdxk-2023310), Beijing, 100000, China
| |
Collapse
|
2
|
Gheitasi I, Akbari G, Savari F. Physiological and cellular mechanisms of ischemic preconditioning microRNAs-mediated in underlying of ischemia/reperfusion injury in different organs. Mol Cell Biochem 2025; 480:855-868. [PMID: 39001984 DOI: 10.1007/s11010-024-05052-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/10/2024] [Indexed: 07/15/2024]
Abstract
Ischemia-reperfusion (I/R) injury, as a pathological phenomenon, takes place when blood supply to an organ is disrupted and then aggravated during restoration of blood flow. Ischemic preconditioning (IPC) is a potent method for attenuating subsequent events of IR damage in numerous organs. IPC protocol is determined by a brief and sequential time periods of I/R before the main ischemia. MicroRNAs are endogenous non-coding RNAs that regulate post-transcriptionally target mRNA translation via degrading it and/or suppressing protein synthesis. This review introduces the physiological and cellular mechanisms of ischemic preconditioning microRNAs-mediated after I/R insult in different organs such as the liver, kidney, heart, brain, and intestine. Data of this review have been collected from the scientific articles published in databases such as Science Direct, Scopus, PubMed, Web of Science, and Scientific Information Database from 2000 to 2023. Based on these literature studies, IPC/IR intervention can affect cellular mechanisms including oxidative stress, apoptosis, angiogenesis, and inflammation through up-regulation or down-regulation of multiple microRNAs and their target genes.
Collapse
Affiliation(s)
- Izadpanah Gheitasi
- Department of Physiology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Ghaidafeh Akbari
- Department of Physiology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Feryal Savari
- Department of Medical Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| |
Collapse
|
3
|
Goeder D, Kröpfl JM, Angst T, Hanssen H, Hauser C, Infanger D, Maurer D, Oberhoffer-Fritz R, Schmidt-Trucksäss A, Königstein K. VascuFit: Aerobic exercise improves endothelial function independent of cardiovascular risk: A randomized-controlled trial. Atherosclerosis 2024; 399:118631. [PMID: 39536471 DOI: 10.1016/j.atherosclerosis.2024.118631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND AIMS Endothelial dysfunction predicts elevated cardiovascular (CV) risk in healthy individuals. Aerobic exercise reduces endothelial dysfunction in part by improving CV risk factors. Yet, this explains less than 50 % of the effect and a direct influence of exercise training on the endothelium is discussed as possible contributor. The VascuFit study applied non-linear periodized aerobic exercise (NLPE) training to assess its multilevel effects on endothelial function including potential epigenetic endothelial modifications by circulating micro-ribonucleic acids (endomiRs). METHODS Sedentary adults with elevated CV risk between 40 and 60 years were randomized 2:1 and engaged in an eight-week ergometer-based NLPE training (n = 30) or received standard exercise recommendations (n = 14). Macro-, microvascular, cellular and molecular adaptations were assessed via brachial-arterial flow-mediated dilation (baFMD), static retinal vessel analysis (SVA), flow cytometry, and endomiRs regulating key pathways of endothelial function. Statistics included ANCOVA, Principal Component Analysis (PCA), and regression analyses. RESULTS baFMD improved by 2.38 % (CI:0.70-4.06, p = 0.007) independent of CV risk, whereas SVA parameters and circulating endothelial (progenitor) cells did not significantly change in the NLPE group. The mean distance between baseline and follow-up PCA loadings of the endomiR dataset explaining 44.2 % of dataset variability was higher in the NLPE-group compared to the control group (2.71 ± 2.02 vs. 1.65 ± 0.93). However, regression analyses showed no evidence of endomiRs explaining the improvement of baFMD. CONCLUSIONS The improvement of macrovascular endothelial function by aerobic exercise training was independent from CV risk factors. Increased heterogeneity among endomiRs did not explain this effect, but suggests an adaptive response to the exercise stimulus on the epigenetic level.
Collapse
Affiliation(s)
- Daniel Goeder
- Department of Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Julia Maria Kröpfl
- Department of Sport, Exercise and Health, Division Sports and Exercise Medicine, University of Basel, Basel, Switzerland
| | - Thomas Angst
- Department of Sport, Exercise and Health, Division Sports and Exercise Medicine, University of Basel, Basel, Switzerland
| | - Henner Hanssen
- Department of Sport, Exercise and Health, Division Sports and Exercise Medicine, University of Basel, Basel, Switzerland
| | - Christoph Hauser
- Department of Sport, Exercise and Health, Division Sports and Exercise Medicine, University of Basel, Basel, Switzerland
| | - Denis Infanger
- Department of Sport, Exercise and Health, Division Sports and Exercise Medicine, University of Basel, Basel, Switzerland
| | - Debbie Maurer
- Department of Sport, Exercise and Health, Division Sports and Exercise Medicine, University of Basel, Basel, Switzerland
| | - Renate Oberhoffer-Fritz
- Department of Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Arno Schmidt-Trucksäss
- Department of Sport, Exercise and Health, Division Sports and Exercise Medicine, University of Basel, Basel, Switzerland
| | - Karsten Königstein
- Department of Sport, Exercise and Health, Division Sports and Exercise Medicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
4
|
Hou S, Zhang L, Ji H, Zhao T, Hu M, Jiang Y, Sun Q, Zhang M, Dou M. Development and evaluation of the model for acute kidney injury in patients with cardiac arrest after successful resuscitation. BMC Cardiovasc Disord 2024; 24:440. [PMID: 39180000 PMCID: PMC11342716 DOI: 10.1186/s12872-024-04110-8] [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: 02/22/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND This study aims to construct a clinical prediction model and create a visual line chart depicting the risk of acute kidney injury (AKI) following resuscitation in cardiac arrest (CA) patients. Additionally, the study aims to validate the clinical predictive accuracy of the developed model. METHODS Data were retrieved from the Dryad database, and publicly shared data were downloaded. This retrospective cohort study included 347 successfully resuscitated patients post-cardiac arrest from the Dryad database. Demographic and clinical data of patients in the database, along with their renal function during hospitalization, were included. Through data analysis, the study aimed to explore the relevant influencing factors of acute kidney injury (AKI) in patients after cardiopulmonary resuscitation. The study constructed a line chart prediction model using multivariate logistic regression analysis with post-resuscitation shock status (Post-resuscitation shock refers to the condition where, following successful cardiopulmonary resuscitation after cardiac arrest, some patients develop cardiogenic shock.), C reactive protein (CRP), Lactate dehydrogenase (LDH), and Alkaline phosphatase (ALP) identified as predictive factors. The predictive efficiency of the fitted model was evaluated by the area under the curve (AUC) of the receiver operating characteristic (ROC) curve. RESULTS Multivariate logistic regression analysis showed that post-resuscitation shock status, CRP, LDH, and PAL were the influencing factors of AKI after resuscitation in CA patients. The calibration curve test indicated that the prediction model was well-calibrated, and the results of the Decision Curve Analysis (DCA) demonstrated the clinical utility of the model constructed in this study. CONCLUSION Post-resuscitation shock status, CRP, LDH, and ALPare the influencing factors for AKI after resuscitation in CA patients. The clinical prediction model constructed based on the above indicators has good clinical discriminability and practicality.
Collapse
Affiliation(s)
- Shanbing Hou
- Department of Nursing, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Lixiang Zhang
- Department of Nursing, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China, Hefei, 230001, Anhui, China
| | - Hongzhi Ji
- Department of Nursing, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Tingting Zhao
- Department of Nursing, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Ming Hu
- Department of Nursing, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Ying Jiang
- Department of Nursing, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Quanquan Sun
- Department of Nursing, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Ming Zhang
- School of Innovation and Entrepreneurship, Wannan Medicine College, Wuhu, 241000, Anhui, China
| | - Min Dou
- Department of Nursing, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
5
|
Rathod ML, Aw WY, Huang S, Lu J, Doherty EL, Whithworth CP, Xi G, Roy-Chaudhury P, Polacheck WJ. Donor-Derived Engineered Microvessels for Cardiovascular Risk Stratification of Patients with Kidney Failure. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307901. [PMID: 38185718 PMCID: PMC11168887 DOI: 10.1002/smll.202307901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/28/2023] [Indexed: 01/09/2024]
Abstract
Cardiovascular disease is the cause of death in ≈50% of hemodialysis patients. Accumulation of uremic solutes in systemic circulation is thought to be a key driver of the endothelial dysfunction that underlies elevated cardiovascular events. A challenge in understanding the mechanisms relating chronic kidney disease to cardiovascular disease is the lack of in vitro models that allow screening of the effects of the uremic environment on the endothelium. Here, a method is described for microfabrication of human blood vessels from donor cells and perfused with donor serum. The resulting donor-derived microvessels are used to quantify vascular permeability, a hallmark of endothelial dysfunction, in response to serum spiked with pathophysiological levels of indoxyl sulfate, and in response to serum from patients with chronic kidney disease and from uremic pigs. The uremic environment has pronounced effects on microvascular integrity as demonstrated by irregular cell-cell junctions and increased permeability in comparison to cell culture media and healthy serum. Moreover, the engineered microvessels demonstrate an increase in sensitivity compared to traditional 2D assays. Thus, the devices and the methods presented here have the potential to be utilized to risk stratify and to direct personalized treatments for patients with chronic kidney disease.
Collapse
Affiliation(s)
- Mitesh L. Rathod
- Joint Department of Biomedical Engineering, University of
North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and
Raleigh, NC, United States of America
| | - Wen Yih Aw
- Joint Department of Biomedical Engineering, University of
North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and
Raleigh, NC, United States of America
| | - Stephanie Huang
- Joint Department of Biomedical Engineering, University of
North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and
Raleigh, NC, United States of America
| | - Jingming Lu
- Joint Department of Biomedical Engineering, University of
North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and
Raleigh, NC, United States of America
| | - Elizabeth L. Doherty
- Joint Department of Biomedical Engineering, University of
North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and
Raleigh, NC, United States of America
| | - Chloe P. Whithworth
- Department of Genetics, University of North Carolina at
Chapel Hill School of Medicine, Chapel Hill, NC, United States of America
| | - Gang Xi
- UNC Kidney Centre, University of North Carolina at Chapel
Hill, NC, United States of America
| | - Prabir Roy-Chaudhury
- UNC Kidney Centre, University of North Carolina at Chapel
Hill, NC, United States of America
- WG (Bill Hefner) Salisbury VA Medical Center, United States
of America
| | - William J. Polacheck
- Joint Department of Biomedical Engineering, University of
North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and
Raleigh, NC, United States of America
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- McAllister Heart Institute, University of North Carolina at
Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
6
|
Zhang K, Kan H, Mao A, Yu F, Geng L, Zhou T, Feng L, Ma X. Integrated Single-Cell Transcriptomic Atlas of Human Kidney Endothelial Cells. J Am Soc Nephrol 2024; 35:578-593. [PMID: 38351505 PMCID: PMC11149048 DOI: 10.1681/asn.0000000000000320] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/09/2024] [Indexed: 03/23/2024] Open
Abstract
Key Points We created a comprehensive reference atlas of normal human kidney endothelial cells. We confirmed that endothelial cell types in the human kidney were also highly conserved in the mouse kidney. Background Kidney endothelial cells are exposed to different microenvironmental conditions that support specific physiologic processes. However, the heterogeneity of human kidney endothelial cells has not yet been systematically described. Methods We reprocessed and integrated seven human kidney control single-cell/single-nucleus RNA sequencing datasets of >200,000 kidney cells in the same process. Results We identified five major cell types, 29,992 of which were endothelial cells. Endothelial cell reclustering identified seven subgroups that differed in molecular characteristics and physiologic functions. Mapping new data to a normal kidney endothelial cell atlas allows rapid data annotation and analysis. We confirmed that endothelial cell types in the human kidney were also highly conserved in the mouse kidney and identified endothelial marker genes that were conserved in humans and mice, as well as differentially expressed genes between corresponding subpopulations. Furthermore, combined analysis of single-cell transcriptome data with public genome-wide association study data showed a significant enrichment of endothelial cells, especially arterial endothelial cells, in BP heritability. Finally, we identified M1 and M12 from coexpression networks in endothelial cells that may be deeply involved in BP regulation. Conclusions We created a comprehensive reference atlas of normal human kidney endothelial cells that provides the molecular foundation for understanding how the identity and function of kidney endothelial cells are altered in disease, aging, and between species. Finally, we provide a publicly accessible online tool to explore the datasets described in this work (https://vascularmap.jiangnan.edu.cn ).
Collapse
Affiliation(s)
- Ka Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hao Kan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fan Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Li Geng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
7
|
Li L, Shen Y, Tang Z, Yang Y, Fu Z, Ni D, Cai X. Engineered nanodrug targeting oxidative stress for treatment of acute kidney injury. EXPLORATION (BEIJING, CHINA) 2023; 3:20220148. [PMID: 38264689 PMCID: PMC10742205 DOI: 10.1002/exp.20220148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 04/23/2023] [Indexed: 01/25/2024]
Abstract
Acute kidney injury (AKI) is a clinical syndrome characterized by a rapid decline in renal function, and is associated with a high risk of death. Many pathological changes happen in the process of AKI, including crucial alterations to oxidative stress levels. Numerous efforts have thus been made to develop effective medicines to scavenge excess reactive oxygen species (ROS). However, researchers have encountered several significant challenges, including unspecific biodistribution, high biotoxicity, and in vivo instability. To address these problems, engineered nanoparticles have been developed to target oxidative stress and treat AKI. This review thoroughly discusses the methods that empower nanodrugs to specifically target the glomerular filtration barrier and presents the latest achievements in engineering novel ROS-scavenging nanodrugs in clustered sections. The analysis of each study's breakthroughs and imperfections visualizes the progress made in developing effective nanodrugs with specific biodistribution and oxidative stress-targeting capabilities. This review fills the blank of a comprehensive outline over current progress in applying nanotechnology to treat AKI, providing potential insights for further research.
Collapse
Affiliation(s)
- Liwen Li
- Department of Ultrasound in MedicineShanghai Jiao Tong University School of Medicine Affiliated Sixth People's HospitalShanghaiPeople's Republic of China
| | - Yining Shen
- Department of Ultrasound in MedicineShanghai Jiao Tong University School of Medicine Affiliated Sixth People's HospitalShanghaiPeople's Republic of China
| | - Zhongmin Tang
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin‐MadisonWisconsinUSA
| | - Yuwen Yang
- Department of Ultrasound in MedicineShanghai Jiao Tong University School of Medicine Affiliated Sixth People's HospitalShanghaiPeople's Republic of China
| | - Zi Fu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Dalong Ni
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Xiaojun Cai
- Department of Ultrasound in MedicineShanghai Jiao Tong University School of Medicine Affiliated Sixth People's HospitalShanghaiPeople's Republic of China
| |
Collapse
|
8
|
Yung S, Chan TM. Endothelial cell activation and glycocalyx shedding - potential as biomarkers in patients with lupus nephritis. Front Immunol 2023; 14:1251876. [PMID: 37854589 PMCID: PMC10579905 DOI: 10.3389/fimmu.2023.1251876] [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: 07/02/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Lupus nephritis (LN) is a common and severe manifestation of systemic lupus erythematosus and an important cause of acute and chronic kidney injury. Early diagnosis of LN and preventing relapses are key to preserving renal reserve. However, due to the complexity and heterogeneity of the disease, clinical management remains challenging. Kidney biopsy remains the gold standard for confirming the diagnosis of LN and subsequent assessment of kidney histopathology, but it is invasive and cannot be repeated frequently. Current clinical indicators of kidney function such as proteinuria and serum creatinine level are non-specific and do not accurately reflect histopathological changes, while anti-dsDNA antibody and C3 levels reflect immunological status but not kidney injury. Identification of novel and specific biomarkers for LN is prerequisite to improve management. Renal function deterioration is associated with changes in the endothelial glycocalyx, a delicate gel-like layer located at the interface between the endothelium and bloodstream. Inflammation induces endothelial cell activation and shedding of glycocalyx constituents into the circulation. This review discusses the potential role of soluble glycocalyx components as biomarkers of active LN, especially in patients in whom conventional serological and biochemical markers do not appear helpful.
Collapse
Affiliation(s)
- Susan Yung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Tak Mao Chan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
9
|
Mause SF, Berger M, Lim HY, Vogt F, Brandenburg V, Stöhr R. Intravenous iron supplementation in heart failure patients induces temporary endothelial dysfunction with release of endothelial microvesicles. Front Immunol 2023; 13:1092704. [PMID: 36761158 PMCID: PMC9904167 DOI: 10.3389/fimmu.2022.1092704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/16/2022] [Indexed: 01/26/2023] Open
Abstract
Background Intravenous iron supplementation is an established therapy for patients with heart failure (HF) and concomitant iron deficiency reducing the risk of HF hospitalization. However, concerns persist regarding potential adverse vascular effects, since iron may induce oxidative stress, inflammation, and apoptosis of endothelial cells. To assess endothelial health following ferric carboxymaltose (FCM) administration, we analyzed the profile of circulating endothelial microvesicles (EMVs) and endothelial progenitor cells (EPCs) in a cohort of 23 HF patients using flow cytometry. Results Compared to healthy subjects, baseline levels of CD31+/CD41- EMVs were higher and EMVs featured a more apoptotic phenotype in HF patients. Following FCM administration, EMV levels showed a rapid but transient increase and displayed an altered phenotype profile with dominant augmentation of EMVs expressing inducible markers CD62E and CD54, indicating endothelial inflammatory activation and injury. Levels of circulating vasoregenerative CD45lowCD34+KDR+ EPCs were lower in HF patients and FCM application resulted in an early decrease of EPCs followed by substantial mobilization into the circulation after one week. Levels of EMVs and EPCs returned to baseline values within two and four weeks, respectively. HF patients with additional chronic kidney disease showed an elevated EMV/EPC ratio and diminished EPC mobilization, suggesting impaired vascular repair capacity. Providing a mechanistic link, in vitro experiments with cultured endothelial cells revealed that FCM dose-dependently promotes endothelial apoptosis, increases expression of adhesion molecules and CXCL12, and triggers generation of EMVs. Conclusion Intravenous iron supplementation with FCM in HF patients induces a biphasic response with initial increased release of CD62E+ and CD54+ enriched EMVs and subsequent mobilization of EPCs, indicating endothelial dysfunction upon FCM and suggesting consecutive engagement of a defense program aimed to reconstitute vascular health.
Collapse
Affiliation(s)
- Sebastian F. Mause
- Department of Internal Medicine I, Cardiology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany,*Correspondence: Sebastian F. Mause,
| | - Martin Berger
- Department of Internal Medicine I, Cardiology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Hwee Ying Lim
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology & Immunology, National University of Singapore, Singapore, Singapore,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Felix Vogt
- Department of Internal Medicine I, Cardiology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Vincent Brandenburg
- Department of Cardiology and Nephrology, Rhein-Maas Klinikum, Wuerselen, Germany
| | - Robert Stöhr
- Department of Internal Medicine I, Cardiology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
10
|
Abdelwahed OM, Aboulhoda BE, Awadallah MY, Gouda SAA, Abdallah H, Rashed L, Khaled M, Ghobrial EE, Alghabban HM, Sharawy N. Prediction of acute kidney injury using a combined model of inflammatory vascular endothelium biomarkers and ultrasound indices. Clin Hemorheol Microcirc 2023; 84:283-301. [PMID: 37212089 DOI: 10.3233/ch-231754] [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: 05/23/2023]
Abstract
BACKGROUND Acute kidney injury (AKI) is a common complication of sepsis, with the burden of long hospital admission. Early prediction of AKI is the most effective strategy for intervention and improvement of the outcomes. OBJECTIVE In our study, we aimed to investigate the predictive performance of the combined model using ultrasound indices (grayscale and Doppler indieces), endothelium injury (E-selectin, VCAM-1, ICAM1, Angiopoietin 2, syndecan-1, and eNOS) as well as inflammatory biomarkers (TNF-a, and IL-1β) to identify AKI. METHODS Sixty albino rats were divided into control and lipopolysaccharide (LPS) groups. Renal ultrasound, biochemical and immunohistological variables were recorded 6 hrs, 24 hrs, and 48 hrs after AKI. RESULTS Endothelium injury and inflammatory markers were found to be significantly increased early after AKI, and correlated significantly with kidney size reduction and renal resistance indices elevation. CONCLUSIONS Using area under the curve (AUC), the combined model was analyzed based on ultrasound and biochemical variables and provided the highest predictive value for renal injury.
Collapse
Affiliation(s)
| | | | - Maryse Youssef Awadallah
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Hend Abdallah
- Department of Anatomy, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Laila Rashed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mai Khaled
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Emad E Ghobrial
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hadel M Alghabban
- Department of Biochemistry and Molecular Medicine, College of Medicine, Taibah University, Medina, Saudi Arabia
| | - Nivin Sharawy
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
11
|
Zhong M, Huang J, Wu Z, Chan KG, Wang L, Li J, Lee LH, Law JWF. Potential Roles of Selectins in Periodontal Diseases and Associated Systemic Diseases: Could They Be Targets for Immunotherapy? Int J Mol Sci 2022; 23:14280. [PMID: 36430760 PMCID: PMC9698067 DOI: 10.3390/ijms232214280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/14/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022] Open
Abstract
Periodontal diseases are predisposing factors to the development of many systemic disorders, which is often initiated via leukocyte infiltration and vascular inflammation. These diseases could significantly affect human health and quality of life. Hence, it is vital to explore effective therapies to prevent disease progression. Periodontitis, which is characterized by gingival bleeding, disruption of the gingival capillary's integrity, and irreversible destruction of the periodontal supporting bone, appears to be caused by overexpression of selectins in periodontal tissues. Selectins (P-, L-, and E-selectins) are vital members of adhesion molecules regulating inflammatory and immune responses. They are mainly located in platelets, leukocytes, and endothelial cells. Furthermore, selectins are involved in the immunopathogenesis of vascular inflammatory diseases, such as cardiovascular disease, diabetes, cancers, and so on, by mediating leukocyte recruitment, platelet activation, and alteration of endothelial barrier permeability. Therefore, selectins could be new immunotherapeutic targets for periodontal disorders and their associated systemic diseases since they play a crucial role in immune regulation and endothelium dysfunction. However, the research on selectins and their association with periodontal and systemic diseases remains limited. This review aims to discuss the critical roles of selectins in periodontitis and associated systemic disorders and highlights the potential of selectins as therapeutic targets.
Collapse
Affiliation(s)
- Mei Zhong
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Jiangyong Huang
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Zhe Wu
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Kok-Gan Chan
- Division of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
- International Genome Centre, Jiangsu University, Zhenjiang 212013, China
| | - Lijing Wang
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Vascular Biology Research Institute, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiang Li
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
| | - Jodi Woan-Fei Law
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
12
|
Xu L, Lu LL, Wang YT, Zhou JB, Wang CX, Xin JD, Gao JD. Endothelial injury and inflammation in patients with hyperuricemic nephropathy at chronic kidney disease stages 1-2 and 3-4. World J Clin Cases 2022; 10:11766-11774. [PMID: 36405284 PMCID: PMC9669867 DOI: 10.12998/wjcc.v10.i32.11766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/15/2022] [Accepted: 10/17/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Endothelial injury and inflammation are the main pathological changes in hyperuricemic nephropathy (HN); however, they have not been assessed in patients in the early, middle, and late phases of HN. AIM To investigate endothelial injury and inflammatory conditions between patients with HN at chronic kidney disease (CKD) stages 3-4 and CKD 1-2. METHODS This study enrolled 80 patients (49 and 31 with HN at CKD stage 1-2 and 3-4, respectively) from the Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine between July 2021 and January 2022. Plasma levels of heparan sulfate, endocan, oxidized low-density lipoprotein (Ox-LDL), E-selectin, soluble intercellular adhesion molecule-1 (slCAM1), interleukin (IL)-1β, and IL-6 and urine levels of lipocalin-type prostaglandin D synthase (L-PGDS), IL-1β, and IL-6 were measured using enzyme-linked immunosorbnent assay. RESULTS Comparison between patients with HN at CKD 1-2 and those with HN at CKD 3-4 showed that age and disease course were significant factors (P < 0.001 and P < 0.010, respectively). There were no statistical differences in sex, heart rate, body mass index, and systolic and diastolic blood pressures. The incidence of hypertension was also significant (P = 0.03). Plasma levels of heparin sulfate (P < 0.001), endocan (P = 0.034), E-selectin (P < 0.001), slCAM1 (P < 0.001), IL-1β (P = 0.006), and IL-6 (P = 0.004) and the urine levels of L-PGDS (P < 0.001), IL-1β (P = 0.003), and IL-6 (P < 0.001) were high in patients with HN at CKD 3-4 than in those with HN at CKD 1-2. The difference in plasma Ox-LDL levels was not significant (P = 0.078). CONCLUSION Vascular endothelial injury and inflammation were higher in patients with HN at CKD3-4 than at CKD 1-2. Plasma heparin sulfate and slCAM1 levels are synergistic factors for CKD staging in HN.
Collapse
Affiliation(s)
- Li Xu
- Department of Nephrology, Shuguang Hospital Affiliated to the Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Li-Li Lu
- Department of Nephrology, Shuguang Hospital Affiliated to the Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Ya-Ting Wang
- Department of Nephrology, Shuguang Hospital Affiliated to the Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Jia-Bao Zhou
- Department of Nephrology, Shuguang Hospital Affiliated to the Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Chuan-Xu Wang
- Department of Nephrology, Shuguang Hospital Affiliated to the Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Jia-Dong Xin
- Department of Nephrology, Shuguang Hospital Affiliated to the Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Jian-Dong Gao
- Department of Nephrology, Shuguang Hospital Affiliated to the Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| |
Collapse
|
13
|
Kröpfl JM, Schmid M, Schön P, Greutmann M, Spengler CM. Correspondence to: "Preclinical atherosclerosis and cardiovascular events: Do we have a consensus about the role of preclinical atherosclerosis in the prediction of cardiovascular events?". Atherosclerosis 2022; 361:47-49. [PMID: 36261306 DOI: 10.1016/j.atherosclerosis.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Julia M Kröpfl
- Division of Sport and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Michelle Schmid
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Patrick Schön
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Matthias Greutmann
- Division of Cardiology, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Christina M Spengler
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| |
Collapse
|
14
|
Königstein K, Meier J, Angst T, Maurer DJ, Kröpfl JM, Carrard J, Infanger D, Baumann S, Bischofsberger I, Harder M, Jäggi Y, Wettach S, Hanssen H, Schmidt-Trucksäss A. VascuFit: vascular effects of non-linear periodized exercise training in sedentary adults with elevated cardiovascular risk - protocol for a randomized controlled trial. BMC Cardiovasc Disord 2022; 22:449. [PMID: 36303113 PMCID: PMC9615395 DOI: 10.1186/s12872-022-02905-1] [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: 08/16/2022] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
Background Early vascular aging (EVA) is increasingly prevalent in the general population. Exercise is important for primary cardiovascular prevention, but often insufficient due to ineffective training methods and a lack of biomarkers suitable to monitor its vascular effects. VascuFit will assess the effectiveness of non-linear periodized aerobic exercise (NLPE) in a non-athletic sedentary population to improve both established and promising biomarkers of EVA. Methods Forty-three sedentary adults, aged 40–60 years, with elevated cardiovascular risk will either engage in 8 weeks of ergometer-based NLPE (n = 28) or receive standard exercise recommendations (n = 15). The primary outcome will be the change of brachial-arterial flow-mediated dilation (baFMD) after versus before the intervention. Secondary outcomes will be the change in static vessel analysis (SVA; clinical biomarker of microvascular endothelial function), endomiRs (microRNAs regulating key molecular pathways of endothelial cell homeostasis) and circulating cellular markers of endothelial function (mature endothelial cells, endothelial progenitor cells). Tertiary outcomes will be the change in sphingolipidome, maximum oxygen capacity, and traditional cardiovascular risk factors (blood pressure, triglycerides, cholesterol, fasting glucose, high-sensitivity C-reactive protein). Discussion We expect an improvement of baFMD of at least 2.6% and significant pre-post intervention differences of SVA and endomiRs as well as of the tertiary outcomes in the intervention group. VascuFit may demonstrate the effectiveness of NLPE to improve endothelial function, thus vascular health, in the general sedentary population. Furthermore, this project might demonstrate the potential of selected molecular and cellular biomarkers to monitor endothelial adaptations to aerobic exercise. Trial registration The trial was registered on www.clinicaltrials.gov (NCT05235958) in February 11th 2022.
Collapse
Affiliation(s)
- Karsten Königstein
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland. .,Clinic for Children and Adolescent Medicine, Staedtisches Klinikum Karlsruhe, Karlsruhe, Germany.
| | - Jennifer Meier
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Thomas Angst
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Debbie J Maurer
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland.,Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
| | - Julia M Kröpfl
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Justin Carrard
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Denis Infanger
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Sandra Baumann
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Imerio Bischofsberger
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Marc Harder
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Yves Jäggi
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Sabrina Wettach
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Henner Hanssen
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| | - Arno Schmidt-Trucksäss
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052, Basel, Switzerland
| |
Collapse
|
15
|
Juncos LA, Wieruszewski PM, Kashani K. Pathophysiology of Acute Kidney Injury in Critical Illness: A Narrative Review. Compr Physiol 2022; 12:3767-3780. [PMID: 36073750 DOI: 10.1002/cphy.c210028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Acute kidney injury (AKI) is a syndrome that entails a rapid decline in kidney function with or without injury. The consequences of AKI among acutely ill patients are dire and lead to higher mortality, morbidity, and healthcare cost. To prevent AKI and its short and long-term repercussions, understanding its pathophysiology is essential. Depending on the baseline kidney histology and function reserves, the number of kidney insults, and the intensity of each insult, the clinical presentation of AKI may differ. While many factors are capable of inducing renal injury, they can be categorized into a few processes. The three primary processes reported in the literature are hemodynamic changes, inflammatory reactions, and nephrotoxicity. The majority of patients with AKI will suffer from more than one during their development and/or progression of AKI. Moreover, the development of one usually leads to the instigation of another. Thus, the interactions and progression between these mechanisms may determine the severity and duration of the AKI. Other factors such as organ crosstalk and how our concurrent therapies interact with these mechanisms complicate the pathophysiology of the progression of the AKI even further. In this narrative review article, we describe these three main pathophysiological processes that lead to the development and progression of AKI. © 2022 American Physiological Society. Compr Physiol 12: 1-14, 2022.
Collapse
Affiliation(s)
- Luis A Juncos
- Division of Nephrology, Central Arkansas Veterans' Healthcare System, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Patrick M Wieruszewski
- Division of Hospital Pharmacy, Department of Pharmacy, Mayo Clinic, Rochester, Minnesota, USA
| | - Kianoush Kashani
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
16
|
Cheng S, Wu D, Yuan L, Liu H, Ei-Seedi HR, Du M. Crassostrea gigas-Based Bioactive Peptide Protected Thrombin-Treated Endothelial Cells against Thrombosis and Cell Barrier Dysfunction. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:9664-9673. [PMID: 35900011 DOI: 10.1021/acs.jafc.2c02435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The activation of thrombin-treated endothelial cells resulted in disruption of the vascular tissues. A novel oyster-derived bioactive dodecapeptide (IEELEELEAER, P-2-CG) was reported to protect the human umbilical vein endothelial cells and their barrier function via the decrease of VE-cadherin disruption and the restoration of the F-actin arrangement. The promotion of the extrinsic pathway in this case triggers the release of tissue factors that occurs on the surface of the endothelial cells, thus changing the antithrombotic to prothrombotic. P-2-CG induced accordingly a prolongation of plasma clotting time and thrombin generation time, following the alteration of the antithrombotic phenotype. Furthermore, the antithrombotic activity of P-2-CG was also supported by the reduction of FXa and the inhibition of other factors release, for instance, inflammation factors, ROS, etc. In addition to its antithrombogenic role, P-2-CG displayed anti-inflammatory and antioxidant properties via the mitogen-activated protein kinase cascades and central signaling pathways as shown in an in vitro model of endothelial dysfunction.
Collapse
Affiliation(s)
- Shuzhen Cheng
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Di Wu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Lushun Yuan
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Nephrology, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Hanxiong Liu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Hesham R Ei-Seedi
- Pharmacognosy Group, Department of Medicinal Chemistry, Uppsala University, Biomedical Centre, Uppsala 75123, Sweden
| | - Ming Du
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
17
|
Baek S, Jang MG, Kim JW, Ko HC, Nam MH, Hur SP, Park SA, Kim SJ. Polymethoxyflavone-rich Fraction from Citrus sunki Leaves Alleviates Renal Dysfunction in Mice with Unilateral Ureteral Obstruction. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221109412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Polymethoxyflavones (PMFs) are flavonoid compounds present in citrus plants that are proposed to be advantageous to human health. However, the advantageous effects of PMFs in the context of renal dysfunction are unclear. In this study, we made a PMF-rich fraction (PRF) from the leaves of Citrus sunki Hort ex. Tanaka and identified its components using liquid chromatography and mass spectrometry. We then investigated the effect of PRF—comprising 9 types of PMF—on renal dysfunction induced by unilateral ureteral obstruction (UUO) in mice. Animals were divided into four experimental groups ( n = 7 per group): I) sham-operated group (Sham); II) UUO group (UUO); III) UUO + Enalapril 0.1 mg/1 mL (UUO + Enap); IV) UUO + PRF 100 mg/kg/day (UUO + PRF). All mice were orally administered with the drugs once a day from 7 days before UUO to 1 week after UUO. After the experiments were over, serum and tissues were taken for biochemical and histological analysis. PRF promoted the recovery of body weight in the background of UUO. Biochemical and histological analysis revealed that PRF ameliorated UUO-induced renal dysfunction and moderately reversed inflammation and tubulointerstitial fibrosis. Further, PRF inhibited the expression of endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), transforming growth factor-β (TGF-β), collagen I (Col-I), and collagen IV (Col-IV). These results suggest that PRF improves UUO-induced renal dysfunction by regulating the expression of inflammatory and fibrotic response-related genes.
Collapse
Affiliation(s)
- Songyee Baek
- Department of Biology, Jeju National University, Jeju, Korea
- Biotech Regional Innovation Center, Jeju National University, Jeju, Korea
| | - Mi Gyeong Jang
- Biotech Regional Innovation Center, Jeju National University, Jeju, Korea
| | - Jae-Won Kim
- Biotech Regional Innovation Center, Jeju National University, Jeju, Korea
| | - Hee Chul Ko
- Jeju Institute of Korean Medicine, Jeju, Korea
| | - Mi Hyun Nam
- Jeju Institute of Korean Medicine, Jeju, Korea
| | - Sung-Pyo Hur
- Korea Institute of Ocean Science & Technology, Jeju, Korea
| | - Soo Ah Park
- In Vivo Research Center, Central Research Facilities, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Se-Jae Kim
- Department of Biology, Jeju National University, Jeju, Korea
- Biotech Regional Innovation Center, Jeju National University, Jeju, Korea
| |
Collapse
|
18
|
Differential effects of cyclo-oxygenase 1 and 2 inhibition on angiogenesis inhibitor-induced hypertension and kidney damage. Clin Sci (Lond) 2022; 136:675-694. [PMID: 35441670 PMCID: PMC9093150 DOI: 10.1042/cs20220182] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022]
Abstract
Vascular endothelial growth factor antagonism with angiogenesis inhibitors in cancer patients induces a ‘preeclampsia-like’ syndrome including hypertension, proteinuria and elevated endothelin (ET)-1. Cyclo-oxygenase (COX) inhibition with aspirin is known to prevent the onset of preeclampsia in high-risk patients. In the present study, we hypothesised that treatment with aspirin would prevent the development of angiogenesis inhibitor-induced hypertension and kidney damage. Our aims were to compare the effects of low-dose (COX-1 inhibition) and high-dose (dual COX-1 and COX-2 inhibition) aspirin on blood pressure, vascular function, oxidative stress, ET-1 and prostanoid levels and kidney damage during angiogenesis-inhibitor therapy in rodents. To this end, Wistar Kyoto rats were treated with vehicle, angiogenesis inhibitor (sunitinib) alone or in combination with low- or high-dose aspirin for 8 days (n=5–7/group). Our results demonstrated that prostacyclin (PGI2) and ET-1 were increased during angiogenesis-inhibitor therapy, while thromboxane (TXA2) was unchanged. Both low- and high-dose aspirin blunted angiogenesis inhibitor-induced hypertension and vascular superoxide production to a similar extent, whereas only high-dose aspirin prevented albuminuria. While circulating TXA2 and prostaglandin F2α levels were reduced by both low- and high-dose aspirin, circulating and urinary levels PGI2 were only reduced by high-dose aspirin. Lastly, treatment with aspirin did not significantly affect ET-1 or vascular function. Collectively our findings suggest that prostanoids contribute to the development of angiogenesis inhibitor-induced hypertension and renal damage and that targeting the prostanoid pathway could be an effective strategy to mitigate the unwanted cardiovascular and renal toxicities associated with angiogenesis inhibitors.
Collapse
|
19
|
Junaid A, van Duinen V, Stam W, Dólleman S, Yang W, de Rijke Y, Endeman H, van Kooten C, Mashaghi A, de Boer H, van Gils J, Hankemeier T, van Zonneveld AJ. A Microfluidics-Based Screening Tool to Assess the Impact of Blood Plasma Factors on Microvascular Integrity. Adv Biol (Weinh) 2021; 5:e2100954. [PMID: 34590440 DOI: 10.1002/adbi.202100954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/07/2021] [Indexed: 11/07/2022]
Abstract
This study provides a method to assess the impact of circulating plasma factors on microvascular integrity by using a recently developed microvessel-on-a-chip platform featuring the human endothelium that is partly surrounded by the extracellular matrix. The system is high-throughput, which allows parallel analysis of organ-level microvessel pathophysiology, including vascular leakage. Ethylenediaminetetraacetic acid plasma samples are mixed with inhibitors for recalcification of the plasma samples to avoid activation of the coagulation- or complement system. Moreover, the assay is validated by spiking vascular endothelial growth factor, histamine, or tumor necrosis factor alpha to recalcified plasma and confirms their modulation of microvessel barrier function at physiologically relevant concentrations. Finally, this study shows that perfusing the microvessels with recalcified plasma samples of coronavirus disease-2019 patients, with a confirmed proinflammatory profile, results in markedly increased leakage of the microvessels. The assay provides opportunities for diagnostic screening of inflammatory or endothelial disrupting plasma factors associated with endothelial dysfunction.
Collapse
Affiliation(s)
- Abidemi Junaid
- A. Junaid, W. Yang, A. Mashaghi, T. Hankemeier, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, 2333 CC, The Netherlands
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Vincent van Duinen
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Wendy Stam
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Sophie Dólleman
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Wei Yang
- A. Junaid, W. Yang, A. Mashaghi, T. Hankemeier, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, 2333 CC, The Netherlands
| | - Yolanda de Rijke
- Y. de Rijke, Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Hendrik Endeman
- H. Endeman, Department of Intensive Care, Erasmus MC, University Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Cees van Kooten
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Alireza Mashaghi
- A. Junaid, W. Yang, A. Mashaghi, T. Hankemeier, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, 2333 CC, The Netherlands
| | - Hetty de Boer
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Janine van Gils
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Thomas Hankemeier
- A. Junaid, W. Yang, A. Mashaghi, T. Hankemeier, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, 2333 CC, The Netherlands
| | - Anton Jan van Zonneveld
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
- A. Junaid, V. van Duinen, W. Stam, S. Dólleman, C. van Kooten, H. de Boer, J. van Gils, A. J. van Zonneveld, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| |
Collapse
|
20
|
Caccuri F, Bugatti A, Zani A, De Palma A, Di Silvestre D, Manocha E, Filippini F, Messali S, Chiodelli P, Campisi G, Fiorentini S, Facchetti F, Mauri P, Caruso A. SARS-CoV-2 Infection Remodels the Phenotype and Promotes Angiogenesis of Primary Human Lung Endothelial Cells. Microorganisms 2021; 9:1438. [PMID: 34361874 PMCID: PMC8305478 DOI: 10.3390/microorganisms9071438] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2-associated acute respiratory distress syndrome (ARDS) and acute lung injury are life-threatening manifestations of severe viral infection. The pathogenic mechanisms that lead to respiratory complications, such as endothelialitis, intussusceptive angiogenesis, and vascular leakage remain unclear. In this study, by using an immunofluorescence assay and in situ RNA-hybridization, we demonstrate the capability of SARS-CoV-2 to infect human primary lung microvascular endothelial cells (HL-mECs) in the absence of cytopathic effects and release of infectious particles. Preliminary data point to the role of integrins in SARS-CoV-2 entry into HL-mECs in the absence of detectable ACE2 expression. Following infection, HL-mECs were found to release a plethora of pro-inflammatory and pro-angiogenic molecules, as assessed by microarray analyses. This conditioned microenvironment stimulated HL-mECs to acquire an angiogenic phenotype. Proteome analysis confirmed a remodeling of SARS-CoV-2-infected HL-mECs to inflammatory and angiogenic responses and highlighted the expression of antiviral molecules as annexin A6 and MX1. These results support the hypothesis of a direct role of SARS-CoV-2-infected HL-mECs in sustaining vascular dysfunction during the early phases of infection. The construction of virus-host interactomes will be instrumental to identify potential therapeutic targets for COVID-19 aimed to inhibit HL-mEC-sustained inflammation and angiogenesis upon SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.B.); (A.Z.); (E.M.); (F.F.); (S.M.); (G.C.); (S.F.)
| | - Antonella Bugatti
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.B.); (A.Z.); (E.M.); (F.F.); (S.M.); (G.C.); (S.F.)
| | - Alberto Zani
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.B.); (A.Z.); (E.M.); (F.F.); (S.M.); (G.C.); (S.F.)
| | - Antonella De Palma
- Proteomic and Metabolomic Laboratory, Institute of Biomedical Technologies, National Research Council (ITB-CNR), 20054 Segrate, Italy; (A.D.P.); (D.D.S.); (P.M.)
| | - Dario Di Silvestre
- Proteomic and Metabolomic Laboratory, Institute of Biomedical Technologies, National Research Council (ITB-CNR), 20054 Segrate, Italy; (A.D.P.); (D.D.S.); (P.M.)
| | - Ekta Manocha
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.B.); (A.Z.); (E.M.); (F.F.); (S.M.); (G.C.); (S.F.)
| | - Federica Filippini
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.B.); (A.Z.); (E.M.); (F.F.); (S.M.); (G.C.); (S.F.)
| | - Serena Messali
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.B.); (A.Z.); (E.M.); (F.F.); (S.M.); (G.C.); (S.F.)
| | - Paola Chiodelli
- Section of General Pathology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy;
| | - Giovanni Campisi
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.B.); (A.Z.); (E.M.); (F.F.); (S.M.); (G.C.); (S.F.)
| | - Simona Fiorentini
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.B.); (A.Z.); (E.M.); (F.F.); (S.M.); (G.C.); (S.F.)
| | - Fabio Facchetti
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy;
| | - Pierluigi Mauri
- Proteomic and Metabolomic Laboratory, Institute of Biomedical Technologies, National Research Council (ITB-CNR), 20054 Segrate, Italy; (A.D.P.); (D.D.S.); (P.M.)
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.B.); (A.Z.); (E.M.); (F.F.); (S.M.); (G.C.); (S.F.)
| |
Collapse
|
21
|
Yu KYC, Yung S, Chau MKM, Tang CSO, Yap DYH, Tang AHN, Ying SKY, Lee CK, Chan TM. Serum syndecan-1, hyaluronan and thrombomodulin levels in patients with lupus nephritis. Rheumatology (Oxford) 2021; 60:737-750. [PMID: 32793966 DOI: 10.1093/rheumatology/keaa370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/28/2020] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES We investigated circulating syndecan-1, HA and thrombomodulin levels in patients with biopsy-proven Class III/IV ± V LN and their clinico-pathological associations. Patients with non-renal SLE or non-lupus chronic kidney disease, and healthy subjects served as controls. METHODS Serum syndecan-1, HA and thrombomodulin levels were determined by ELISAs. RESULTS Syndecan-1, HA and thrombomodulin levels were significantly higher during active LN compared with remission (P < 0.01, for all), and correlated with the level of proteinuria, estimated glomerular filtration rate, anti-dsDNA antibodies, complement 3 and serum creatinine. Longitudinal studies showed that syndecan-1 and thrombomodulin levels increased prior to clinical renal flare by 3.6 months, while HA level increased at the time of nephritic flare, and the levels decreased in parallel with treatment response. Receiver operating characteristic curve analysis showed that syndecan-1 and thrombomodulin levels distinguished patients with active LN from healthy subjects, LN patients in remission, patients with active non-renal lupus and patients with non-lupus chronic kidney disease (receiver operating characteristic area under curve of 0.98, 0.91, 0.82 and 0.95, respectively, for syndecan-1; and area under curve of 1.00, 0.84, 0.97 and 0.79, respectively, for thrombomodulin). HA level distinguished active LN from healthy subjects, LN patients in remission and non-lupus chronic kidney disease (receiver operating characteristic area under curve of 0.82, 0.71 and 0.90, respectively) but did not distinguish between renal vs non-renal lupus. Syndecan-1 and thrombomodulin levels correlated with the severity of interstitial inflammation, while HA level correlated with chronicity grading in kidney biopsies of active LN. CONCLUSION Our findings suggest potential utility of serum syndecan-1, thrombomodulin and HA levels in clinical management, and their potential contribution to LN pathogenesis.
Collapse
Affiliation(s)
- Kelvin Y C Yu
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Susan Yung
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Mel K M Chau
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Colin S O Tang
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Desmond Y H Yap
- Department of Medicine, The University of Hong Kong, Hong Kong
| | | | - Shirley K Y Ying
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong
| | | | - Tak Mao Chan
- Department of Medicine, The University of Hong Kong, Hong Kong
| |
Collapse
|
22
|
Kumboyono K, Nurwidyaningtyas W, Chomsy IN, Wihastuti TA. Early Detection of Negative Smoking Impacts: Vascular Adaptation Deviation Based on Quantification of Circulated Endothelial Activation Markers. Vasc Health Risk Manag 2021; 17:103-109. [PMID: 33790567 PMCID: PMC8001718 DOI: 10.2147/vhrm.s296293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/03/2021] [Indexed: 12/04/2022] Open
Abstract
Introduction Smoking can cause vascular damage in the form of an inflammatory reaction characterized by endothelial activation. Endothelial activation forms a pathological adaptation pattern so that it can induce the atherogenesis process. Several markers, such as E-selectin, platelet-derived micro particles (PMPs) and hematopoietic stem cell (HSC) can identify the activation of endothelial in circulating blood. Therefore, the deviation of vascular adaptation due to smoking can be detected early through the feedback mechanism between E-selectin, PMPs, and HSC. Purpose This study aims to analyze the initial picture of the negative impact of smoking on vascular adaptation by measuring E-selectin, PMPs, and HSC in the peripheral blood circulation. Participant criteria and methods: Peripheral blood samples (5 mL) were taken from each participant, both the smoking group (n = 30) and the non-smoker group (n = 31) to obtain peripheral blood mononuclear cells (PBMNC). PBMNC was isolated using ficoll-based gradient centrifugation. The flow cytometry assay method used to measure the E-selectin, PMPs and hematopoietic stem cells. Results The mean of circulating E-selectin in smokers was higher than that of non-smokers. On the other hand, the average number of PMPs and HSCs in smokers was lower than non-smokers. Conclusion Smoking increases the risk of accelerated vascular block formation, as indicated by an increase in the amount of circulating E-selectin. The increase in E-selectin in the blood vessels mediates the increased adhesion of PMPs in the vascular area so that the number of circulating PMPs in smokers decreases. The decrease in circulating PMPs decreases the signal of vascular repair, which is characterized by a decline in the number of HSCs.
Collapse
Affiliation(s)
- Kumboyono Kumboyono
- Nursing Department, Faculty of Medicine, University of Brawijaya, Malang, 65145, Indonesia
| | - Wiwit Nurwidyaningtyas
- Doctoral Program of Medical Science, Faculty of Medicine, University of Brawijaya, Malang, 65145, Indonesia
| | - Indah Nur Chomsy
- Doctoral Program of Medical Science, Faculty of Medicine, University of Brawijaya, Malang, 65145, Indonesia
| | - Titin Andri Wihastuti
- Department of Biomedicine, Faculty of Medicine, University of Brawijaya, Malang, 65145, Indonesia
| |
Collapse
|
23
|
Kröpfl JM, Beltrami FG, Rehm M, Gruber HJ, Stelzer I, Spengler CM. Acute exercise-induced glycocalyx shedding does not differ between exercise modalities, but is associated with total antioxidative capacity. J Sci Med Sport 2021; 24:689-695. [PMID: 33632661 DOI: 10.1016/j.jsams.2021.01.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/02/2021] [Accepted: 01/28/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVES Regular physical exercise is known to protect endothelial integrity. It has been proposed that acute exercise-induced changes of the (anti-)oxidative system influence early (glycocalyx shedding) and sustained endothelial activation (shedding of endothelial cells, ECs) as well as endothelial-cell repair by circulating hematopoietic stem and progenitor cells (HPCs). However, results are not conclusive and data in trained participants performing different exercise modalities is lacking. DESIGN Eighteen healthy, well-trained participants (9 runners, 9 cyclists; age: 29.7 ± 4.2 yrs) performed a strenuous acute exercise session consisting of 4 bouts of 4-min high-intensity with decreasing power profile and 3-min low-intensity in-between. METHODS Average power/speed of intense phases was 85% of the peak achieved in a previous incremental test. Before and shortly after exercise, total oxidative and antioxidative capacities (TAC), shedding of syndecan-1, heparan sulfate, hyaluronan, ECs, and circulating HPCs were investigated. RESULTS TAC decreased from 1.81 ± 0.42 nmol/L to 1.47 ± 0.23 nmol/L post-exercise (p = 0.010) only in runners. Exercise-induced early and sustained endothelial activation were enhanced post-exercise- syndecan-1: 103.2 ± 63.3 ng/mL to 111.3 ± 71.3 ng/mL, heparan sulfate: from 2637.9 ± 800.1 ng/mL to 3197.1 ± 1416.3 ng/mL, both p < 0.05; hyaluronan: 84.3 ± 21.8 ng/mL to 121.4 ± 29.4 ng/mL, ECs: from 6.6 ± 4.5 cells/μL to 9.5 ± 6.2 cells/μL, both p < 0.01; results were not different between exercise modalities and negatively related to TAC concentrations post-exercise. HPC proportions and self-renewal ability were negatively, while EC concentrations were positively associated with circulating hyaluronan concentrations. CONCLUSIONS These results highlight the importance of the antioxidative system to prevent the endothelium from acute exercise-induced vascular injury - independent of exercise modality - in well-trained participants. Endothelial-cell repair is associated with hyluronan signaling, possibly a similar mechanism as in wound repair.
Collapse
Affiliation(s)
- Julia M Kröpfl
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Switzerland
| | - Fernando G Beltrami
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Switzerland
| | - Markus Rehm
- Department of Anaesthesiology, Ludwig-Maximilians-University Munich, Germany
| | - Hans-Jürgen Gruber
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Ingeborg Stelzer
- Institute of Medical and Chemical Laboratory Diagnostics, LKH Hochsteiermark, Austria
| | - Christina M Spengler
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland.
| |
Collapse
|
24
|
Hajizadeh-Sharafabad F, Sharifi Zahabi E. Role of alpha-lipoic acid in vascular function: A systematic review of human intervention studies. Crit Rev Food Sci Nutr 2020; 62:2928-2941. [DOI: 10.1080/10408398.2020.1861425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Fatemeh Hajizadeh-Sharafabad
- Nutrition Research Center, Department of Clinical Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Sharifi Zahabi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Schmid M, Gruber HJ, Kröpfl JM, Spengler CM. Acute Exercise-Induced Oxidative Stress Does Not Affect Immediate or Delayed Precursor Cell Mobilization in Healthy Young Males. Front Physiol 2020; 11:577540. [PMID: 33192581 PMCID: PMC7606978 DOI: 10.3389/fphys.2020.577540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/23/2020] [Indexed: 11/24/2022] Open
Abstract
Exercise is known to acutely and transiently mobilize precursor cells to the peripheral blood. To date, the underlying mechanisms have not yet been fully elucidated and we hypothesized that exercise-induced oxidative stress could be a mobilizing agent, either directly or via circulating apoptotic cells as mediators. The aim of the study was to assess the effect of acute exercise-induced oxidative stress on numbers of circulating angiogenic precursor cells (CACs), circulating non-angiogenic precursor cells (nCACs), mesenchymal precursor cells (MPCs), mature endothelial cells (ECs), and mononuclear cells (MNCs), as well as their apoptotic subsets. Healthy, young males (n = 18, age: 24.2 ± 3.5 years) completed two identical, standardized incremental cycling tests. The first, un-supplemented control test was followed by a 7-day-long supplementation of vitamin C (1,000 mg/day) and E (400 I.U./day), immediately preceding the second test. Blood samples were collected before, directly after, 30, 90, 180, and 270 min after exercise, and aforementioned circulating cell numbers were determined by flow cytometry and a hematology analyzer. Additionally, total oxidative capacity (TOC) and total antioxidative capacity (TAC) were measured in serum at all timepoints. Antioxidative supplementation abolished the exercise-induced increase in the oxidative stress index (TOC/TAC), and reduced baseline concentrations of TOC and TOC/TAC. However, it did not have any effect on CACs, nCACs, and MPC numbers or the increase in apoptotic MNCs following exercise. Our results indicate that exercise-induced oxidative stress is neither a main driver of lymphocyte and monocyte apoptosis, nor one of the mechanisms involved in the immediate or delayed mobilization of precursor cells.
Collapse
Affiliation(s)
- Michelle Schmid
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Hans-Jürgen Gruber
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Julia M Kröpfl
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Christina M Spengler
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
Wei H, Tan T, Cheng L, Liu J, Song H, Li L, Zhang K. MRI tracing of ultrasmall superparamagnetic iron oxide nanoparticle‑labeled endothelial progenitor cells for repairing atherosclerotic vessels in rabbits. Mol Med Rep 2020; 22:3327-3337. [PMID: 32945451 PMCID: PMC7453557 DOI: 10.3892/mmr.2020.11431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 03/30/2020] [Indexed: 12/14/2022] Open
Abstract
Endothelial progenitor cells (EPCs) have been discovered to be relevant to the prognosis of cardiovascular diseases. Previous research has demonstrated that EPCs serve vital roles in the occurrence and development of atherosclerosis. Significant improvements have been made in MRI technology and in the experimental use of EPCs for therapeutic angiogenesis and vascular repair. Nevertheless, the migratory, adhesive, proliferative and angiogenic properties of EPCs remain unknown. The aims of the present study were to investigate the potential of using non-invasive monitoring with ultrasmall superparamagnetic iron oxide nanoparticle (USPION)-labeled endothelial progenitor cells (EPCs) after transplantation, and to assess the treatment outcomes in an atherosclerotic rabbit model. EPCs derived from rabbit peripheral blood samples were labeled with USPION-poly-l-lysine (USPION-PLL). The morphology, proliferation, adhesive ability and labeling efficiency of the EPCs were determined by optical and electron microscopy. Moreover, biological activity was assessed by flow cytometry. In addition, T2-weighted image fast spin-echo MRI was used to detect cell labeling. USPION content in the labeled EPCs was determined by Prussian blue staining and scanning electron microscopy. Rabbit atherosclerosis model was established using a high-fat diet. USPION-labeled EPCs were transplanted into rabbits, and in vivo MRI was performed 1 and 7 days after transplantation. It was found that EPCs cultured on Matrigel formed capillary-like structures, and expressed the surface markers CD133, CD31, CD34 and vascular endothelial growth factor receptor 2 (VEGFR2). The optimal USPION concentration was 32 µg/ml, as determined by adhesion and proliferation assays. It was identified that USPION-PLL nanoparticles were 10–20 nm in diameter. Histopathological analysis results indicated that 1 day after transplantation of the labeled EPCs, blue-stained granules were observed in the intima of vascular lesions in rabbit models after Prussian blue staining. Therefore, the present results suggest that USPION-labeled EPCs may play a role in repairing endothelial injury and preventing atherosclerosis in vivo.
Collapse
Affiliation(s)
- Hongxia Wei
- Department of Laboratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Tingting Tan
- Department of Laboratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Li Cheng
- Department of Laboratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Jiapeng Liu
- Department of Medical Imaging, Shanghai Jiahui International Hospital, Shanghai 200233, P.R. China
| | - Hongyan Song
- Department of Laboratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Lei Li
- Department of Laboratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Kui Zhang
- Department of Laboratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
27
|
Junaid A, Schoeman J, Yang W, Stam W, Mashaghi A, van Zonneveld AJ, Hankemeier T. Metabolic response of blood vessels to TNFα. eLife 2020; 9:54754. [PMID: 32749215 PMCID: PMC7476757 DOI: 10.7554/elife.54754] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 08/02/2020] [Indexed: 12/11/2022] Open
Abstract
TNFα signaling in the vascular endothelium elicits multiple inflammatory responses that drive vascular destabilization and leakage. Bioactive lipids are main drivers of these processes. In vitro mechanistic studies of bioactive lipids have been largely based on two-dimensional endothelial cell cultures that, due to lack of laminar flow and the growth of the cells on non-compliant stiff substrates, often display a pro-inflammatory phenotype. This complicates the assessment of inflammatory processes. Three-dimensional microvessels-on-a-chip models provide a unique opportunity to generate endothelial microvessels in a more physiological environment. Using an optimized targeted liquid chromatography–tandem mass spectrometry measurements of a panel of pro- and anti-inflammatory bioactive lipids, we measure the profile changes upon administration of TNFα. We demonstrate that bioactive lipid profiles can be readily detected from three-dimensional microvessels-on-a-chip and display a more dynamic, less inflammatory response to TNFα, that resembles more the human situation, compared to classical two-dimensional endothelial cell cultures. In a range of conditions called autoimmune diseases, the immune system attacks the body rather than foreign elements. This can cause inflammation that is harmful for many organs. In particular, immune cells can produce excessive amounts of a chemical messenger called tumor necrosis factor alpha (TNFα for short), which can lead to the release of fatty molecules that damage blood vessels. This process is normally studied in blood vessels cells that are grown on a dish, without any blood movement. However, in this rigid 2D environment, the cells become ‘stressed’ and show higher levels of inflammation than in the body. This makes it difficult to assess the exact role that TNFα plays in disease. A new technology is addressing this issue by enabling scientist to culture blood vessels cells in dishes coated with gelatin. This allows the cells to organize themselves in 3D, creating tiny blood vessels in which fluids can flow. However, it was unclear whether these ‘microvessels-on-a-chip’ were better models to study the role of TNFα compared to cells grown on a plate. Here, Junaid et al. compared the levels of inflammation in blood vessels cells grown in the two environments, showing that cells are less inflamed when they are cultured in 3D. In addition, when the artificial 3D-blood vessels were exposed to TNFα, they responded more like real blood vessels than the 2D models. Finally, experiments showed that it was possible to monitor the release of fatty molecules in this environment. Together, this work suggests that microvessels-on-a-chip are better models to study how TNFα harms blood vessels. Next, systems and protocols could be develop to allow automated mass drug testing in microvessels-on-a-chip. This would help scientists to quickly screen thousands of drugs and find candidates that can protect blood vessels from TNFα.
Collapse
Affiliation(s)
- Abidemi Junaid
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands.,Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Johannes Schoeman
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Wei Yang
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Wendy Stam
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Alireza Mashaghi
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Thomas Hankemeier
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| |
Collapse
|
28
|
Gomelsky A, Abreo K, Khater N, Abreo A, Amin B, Craig MK, Prabhakar A, Cornett EM, Urman RD, Kaye AD. Perioperative acute kidney injury: Stratification and risk reduction strategies. Best Pract Res Clin Anaesthesiol 2020; 34:167-182. [PMID: 32711827 DOI: 10.1016/j.bpa.2020.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 04/07/2020] [Indexed: 12/29/2022]
Abstract
Perioperative acute kidney injury (AKI) is associated with increased morbidity and mortality. Patient comorbidities, the type of surgery, timing of surgery, and exposure to nephrotoxins are important contributors for developing acute kidney injury. Urgent or emergent surgery, cardiac, and organ transplantation procedures are associated with a higher risk of acute kidney injury. Nephrotoxic drugs, contrast dye, and diuretics can worsen preexisting kidney dysfunction or act as an additive and/or synergistic insult to perioperative injury. A history of preoperative chronic kidney disease is the main risk factor for developing AKI, conferring as much as a 10-fold risk. However, beyond the preoperative renal function, the development of AKI is a complex phenomenon that involves a combination of patient-related and surgery-related factors.
Collapse
Affiliation(s)
- Alexander Gomelsky
- Department of Urology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| | - Kenneth Abreo
- Department of Urology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA; Department of Nephrology and Hypertension, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| | - Nazih Khater
- Department of Urology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| | - Adrian Abreo
- Division of Nephrology, Clerkship Director, Internal Medicine Clerkship, Associate Program Director, Adrian AbreoA, 71103, USA.
| | - Bakhtiar Amin
- Department of Nephrology and Hypertension, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| | - Madelyn K Craig
- Department of Anesthesiology, LSU Health Science Center New Orleans, 1542 Tulane Avenue, New Orleans, LA, 70112, USA.
| | - Amit Prabhakar
- Department of Anesthesiology, Division of Critical Care, Emory University School of Medicine, Atlanta, GA, USA.
| | - Elyse M Cornett
- Department of Anesthesiology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| | - Richard D Urman
- Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Medical School, Brigham and Women's Hospital, 75 Francis St, Boston, MA, 02115, USA.
| | - Alan D Kaye
- Departments of Anesthesiology and Pharmacology, Toxicology, and Neurosciences; Provost, Chief Academic Officer, and Vice Chancellor of Academic Affairs, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| |
Collapse
|
29
|
Kröpfl JM, Kammerer T, Faihs V, Gruber HJ, Stutz J, Rehm M, Stelzer I, Schäfer ST, Spengler CM. Acute Exercise in Hypobaric Hypoxia Attenuates Endothelial Shedding in Subjects Unacclimatized to High Altitudes. Front Physiol 2020; 10:1632. [PMID: 32116736 PMCID: PMC7010936 DOI: 10.3389/fphys.2019.01632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/26/2019] [Indexed: 11/13/2022] Open
Abstract
Travel of unacclimatized subjects to a high altitude has been growing in popularity. Changes in endothelial shedding [circulating endothelial cells (ECs)] and hematopoietic stem and progenitor cells (CPCs) during physical exercise in hypobaric hypoxia, however, are not well understood. We investigated the change in ECs and CPCs when exposed to high altitude, after acute exercise therein, and after an overnight stay in hypobaric hypoxia in 11 healthy unacclimatized subjects. Blood withdrawal was done at baseline (520 m a.s.l.; baseline), after passive ascent to 3,883 m a.s.l. (arrival), after acute physical exercise (±400 m, postexercise) and after an overnight stay at 3,883 m a.s.l. (24 h). Mature blood cells, ECs, and CPCs were assessed by a hematology analyzer and flow cytometry, respectively. The presence of matrix metalloproteinases (MMPs), their activity, and hematopoietic cytokines were assessed in serum and plasma. EC and CPC concentrations significantly decreased after exercise (p = 0.019, p = 0.007, respectively). CPCs remained low until the next morning (24 h, p = 0.002), while EC concentrations returned back to baseline. MMP-9 decreased at arrival (p = 0.021), stayed low postexercise (p = 0.033), and returned to baseline at 24 h (p = 0.035 to postexercise). MMP-activity did not change throughout the study. Circulating MMP-9 concentrations, but not MMP-activity, were associated with EC concentrations (rrm = 0.48, p = 0.010). CPC concentrations were not linked to hematopoietic cytokines. Acute exercise at high altitude attenuated endothelial shedding, but did not enhance regenerative CPCs. Results were not linked to endothelial matrix remodeling or CPC mobilization. These results provide information to better understand the endothelium and immature immune system during an active, short-term sojourn at high altitude.
Collapse
Affiliation(s)
- Julia M Kröpfl
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zürich, Zurich, Switzerland
| | - Tobias Kammerer
- Department of Anaesthesiology, Ludwig Maximilian University of Munich, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University of Munich, Munich, Germany.,Institute of Anesthesiology, Heart and Diabetes Center NRW, Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Valentina Faihs
- Department of Anaesthesiology, Ludwig Maximilian University of Munich, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University of Munich, Munich, Germany
| | - Hans-Jürgen Gruber
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Jan Stutz
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zürich, Zurich, Switzerland
| | - Markus Rehm
- Department of Anaesthesiology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Ingeborg Stelzer
- Institute of Medical and Chemical Laboratory Diagnostics, LKH Hochsteiermark, Leoben, Austria
| | - Simon T Schäfer
- Department of Anaesthesiology, Ludwig Maximilian University of Munich, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University of Munich, Munich, Germany
| | - Christina M Spengler
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zürich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
30
|
Hueso M, Casas A, Mallén A, de Ramón L, Bolaños N, Varela C, Cruzado JM, Torras J, Navarro E. The double edge of anti-CD40 siRNA therapy: It increases renal microcapillar density but favours the generation of an inflammatory milieu in the kidneys of ApoE -/- mice. JOURNAL OF INFLAMMATION-LONDON 2019; 16:25. [PMID: 31889910 PMCID: PMC6916081 DOI: 10.1186/s12950-019-0228-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/27/2019] [Indexed: 12/12/2022]
Abstract
Background Chronic kidney disease (CKD) is associated with endothelial dysfunctions thus prompting links between microcirculation (MC), inflammation and major cardiovascular risk factors. Purpose of the study We have previously reported that siRNA-silencing of CD40 (siCD40) reduced atherosclerosis (ATH) progression. Here, we have deepened on the effects of the siCD40 treatment by evaluating retrospectively, in stored kidneys from the siCD40 treated ApoE−/− mice, the renal microcirculation (measured as the density of peritubular capillaries), macrophage infiltration and NF-κB activation. Methods Kidneys were isolated after 16 weeks of treatment with the anti-CD40 siRNA (siCD40), with a scrambled control siRNA (siSC) or with PBS (Veh. group). Renal endothelium, infiltrating macrophages and activated NF-κB in endothelium were identified by immunohistochemistry, while the density of stained peritubular capillaries was quantified by image analysis. Results ATH was associated with a reduction in renal MC, an effect reversed by the anti-CD40 siRNA treatment (3.8 ± 2.7% in siCD40; vs. 1.8 ± 0.1% in siSC; or 1.9 ± 1.6% in Veh.; p < 0.0001). Furthermore, siCD40 treatment reduced the number of infiltrating macrophages compared to the SC group (14.1 ± 5.9 cells/field in siCD40; vs. 37.1 ± 17.8 cells/field in siSC; and 1.3 ± 1.7 cells/field in Veh.; p = 0.001). NF-κB activation also peaked in the siSC group, showing lower levels in the siCD40 and Veh. groups (63 ± 60 positive cells/section in siCD40; vs. 152 ± 44 positive cells/section in siSC; or 26 ± 29 positive cells/section in veh.; p = 0.014). Lastly, serum creatinine was also increased in the siCD40 (3.4 ± 3.3 mg/dL) and siSC (4.6 ± 3.0 mg/dL) groups when compared with Veh. (1.1 ± 0.9 mg/dL, p = 0.1). Conclusions Anti-CD40 siRNA therapy significantly increased the density of peritubular capillaries and decreased renal inflammation in the ATH model. These data provide a physiological basis for the development of renal diseases in patients with ATH. Furthermore, our results also highligth renal off-target effects of the siRNA treatment which are discussed. Graphical abstract ![]()
Collapse
Affiliation(s)
- Miguel Hueso
- 1Department of Nephrology, Hospital Universitari Bellvitge, and Bellvitge Research Institute (IDIBELL). L'Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Angela Casas
- 1Department of Nephrology, Hospital Universitari Bellvitge, and Bellvitge Research Institute (IDIBELL). L'Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Adrian Mallén
- 2Laboratori de Nefrología Experimental, Bellvitge Research Institute (IDIBELL). L'Hospitalet de Llobregat, Barcelona, Spain
| | - Laura de Ramón
- 2Laboratori de Nefrología Experimental, Bellvitge Research Institute (IDIBELL). L'Hospitalet de Llobregat, Barcelona, Spain
| | - Nuria Bolaños
- 2Laboratori de Nefrología Experimental, Bellvitge Research Institute (IDIBELL). L'Hospitalet de Llobregat, Barcelona, Spain
| | - Cristian Varela
- 2Laboratori de Nefrología Experimental, Bellvitge Research Institute (IDIBELL). L'Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M Cruzado
- 1Department of Nephrology, Hospital Universitari Bellvitge, and Bellvitge Research Institute (IDIBELL). L'Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Joan Torras
- 1Department of Nephrology, Hospital Universitari Bellvitge, and Bellvitge Research Institute (IDIBELL). L'Hospitalet de Llobregat, 08907 Barcelona, Spain
| | | |
Collapse
|
31
|
Abstract
The kidney harbours different types of endothelia, each with specific structural and functional characteristics. The glomerular endothelium, which is highly fenestrated and covered by a rich glycocalyx, participates in the sieving properties of the glomerular filtration barrier and in the maintenance of podocyte structure. The microvascular endothelium in peritubular capillaries, which is also fenestrated, transports reabsorbed components and participates in epithelial cell function. The endothelium of large and small vessels supports the renal vasculature. These renal endothelia are protected by regulators of thrombosis, inflammation and complement, but endothelial injury (for example, induced by toxins, antibodies, immune cells or inflammatory cytokines) or defects in factors that provide endothelial protection (for example, regulators of complement or angiogenesis) can lead to acute or chronic renal injury. Moreover, renal endothelial cells can transition towards a mesenchymal phenotype, favouring renal fibrosis and the development of chronic kidney disease. Thus, the renal endothelium is both a target and a driver of kidney and systemic cardiovascular complications. Emerging therapeutic strategies that target the renal endothelium may lead to improved outcomes for both rare and common renal diseases.
Collapse
|
32
|
Florijn BW, Duijs JMGJ, Levels JH, Dallinga-Thie GM, Wang Y, Boing AN, Yuana Y, Stam W, Limpens RWAL, Au YW, Nieuwland R, Rabelink TJ, Reinders MEJ, van Zonneveld AJ, Bijkerk R. Diabetic Nephropathy Alters the Distribution of Circulating Angiogenic MicroRNAs Among Extracellular Vesicles, HDL, and Ago-2. Diabetes 2019; 68:2287-2300. [PMID: 31506344 DOI: 10.2337/db18-1360] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 08/31/2019] [Indexed: 11/13/2022]
Abstract
Previously, we identified plasma microRNA (miR) profiles that associate with markers of microvascular injury in patients with diabetic nephropathy (DN). However, miRs circulate in extracellular vesicles (EVs) or in association with HDL or the RNA-binding protein argonaute-2 (Ago-2). Given that the EV- and HDL-mediated miR transfer toward endothelial cells (ECs) regulates cellular quiescence and inflammation, we hypothesized that the distribution of miRs among carriers affects microvascular homeostasis in DN. Therefore, we determined the miR expression in EV, HDL, and Ago-2 fractions isolated from EDTA plasma of healthy control subjects, patients with diabetes mellitus (DM) with or without early DN (estimated glomerular filtration rate [eGFR] >30 mL/min/1.73 m2), and patients with DN (eGFR <30 mL/min/1.73 m2). Consistent with our hypothesis, we observed alterations in miR carrier distribution in plasma of patients with DM and DN compared with healthy control subjects. Both miR-21 and miR-126 increased in EVs of patients with DN, whereas miR-660 increased in the Ago-2 fraction and miR-132 decreased in the HDL fraction. Moreover, in vitro, differentially expressed miRs improved EC barrier formation (EV-miR-21) and rescued the angiogenic potential (HDL-miR-132) of ECs cultured in serum from patients with DM and DN. In conclusion, miR measurement in EVs, HDL, and Ago-2 may improve the biomarker sensitivity of these miRs for microvascular injury in DN, while carrier-specific miRs can improve endothelial barrier formation (EV-miR-21/126) or exert a proangiogenic response (HDL-miR-132).
Collapse
Affiliation(s)
- Barend W Florijn
- Department of Internal Medicine (Nephrology), Amsterdam University Medical Center, Amsterdam, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Jacques M G J Duijs
- Department of Internal Medicine (Nephrology), Amsterdam University Medical Center, Amsterdam, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Johannes H Levels
- Department of Vascular Biology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Geesje M Dallinga-Thie
- Department of Vascular Biology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Yanan Wang
- Department of Internal Medicine (Endocrinology), Leiden University Medical Center, Leiden, the Netherlands
| | - Anita N Boing
- Laboratory of Experimental Clinical Chemistry, Department of Clinical Chemistry, and Vesicle Observation Center, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Yuana Yuana
- Laboratory of Experimental Clinical Chemistry, Department of Clinical Chemistry, and Vesicle Observation Center, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Wendy Stam
- Department of Internal Medicine (Nephrology), Amsterdam University Medical Center, Amsterdam, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Ronald W A L Limpens
- Section Electron Microscopy, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Yu Wah Au
- Department of Internal Medicine (Nephrology), Amsterdam University Medical Center, Amsterdam, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Department of Clinical Chemistry, and Vesicle Observation Center, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine (Nephrology), Amsterdam University Medical Center, Amsterdam, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine (Nephrology), Amsterdam University Medical Center, Amsterdam, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology), Amsterdam University Medical Center, Amsterdam, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Roel Bijkerk
- Department of Internal Medicine (Nephrology), Amsterdam University Medical Center, Amsterdam, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Amsterdam University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
33
|
Serum levels of the endothelial glycocalyx constituents and the early failure of forearm autologous arteriovenous fistulas in end-stage renal disease patients: a prospective cohort study. Int Urol Nephrol 2019; 52:169-177. [DOI: 10.1007/s11255-019-02317-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/10/2019] [Indexed: 10/25/2022]
|
34
|
Microparticles and autophagy: a new frontier in the understanding of atherosclerosis in rheumatoid arthritis. Immunol Res 2019; 66:655-662. [PMID: 30574665 DOI: 10.1007/s12026-018-9053-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Microparticles (MPs) are small membrane vesicles released by many cell types under physiological and pathological conditions. In the last years, these particles were considered as inert cell debris, but recently many studies have demonstrated they could have a role in intercellular communication. Increased levels of MPs have been reported in various pathological conditions including infections, malignancies, and autoimmune diseases, such as rheumatoid arthritis (RA). RA is an autoimmune systemic inflammatory disease characterized by chronic synovial inflammation, resulting in cartilage and bone damage with accelerated atherosclerosis increasing mortality. According to the literature data, also MPs could have a role in endothelial dysfunction, contributing to atherosclerosis in RA patients. Moreover many researchers have shown that a dysregulated autophagy seems to be involved in endothelial dysfunction. Autophagy is a reparative process by which cytoplasmic components are sequestered in double-membrane vesicles and degraded on fusion with lysosomal compartments. It has been shown in many works that basal autophagy is essential to proper vascular function. Taking into account these considerations, we hypothesized that in RA patients MPs could contribute to atherosclerosis process by dysregulation of endothelial autophagy process.
Collapse
|
35
|
Bernardi L, Giampietro C, Marina V, Genta M, Mazza E, Ferrari A. Adaptive reorientation of endothelial collectives in response to strain. Integr Biol (Camb) 2019; 10:527-538. [PMID: 30112523 DOI: 10.1039/c8ib00092a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mature epithelial monolayers share the ability to coherently respond to external mechanical stimuli. Tissue remodeling requires cell shape changes and coordinated movements. Human endothelia provide an exquisite example of such emerging collective activities. As part of their function in maintaining body homeostasis under variable hemodynamic loadings, endothelial ensembles must dynamically adapt to wall shear stress and cyclic deformation. While the alignment of several types of cells, including fibroblasts, osteoblasts and epithelial tissues, in response to various flow conditions or wall shear stress levels has been described in detail, less is known about collective endothelial remodeling under pure wall deformation. Here, using a custom-developed bioreactor, we exposed mature human endothelia to two distinct physiological levels of cyclic loading, generating overlapping gradients of strain. Endothelial cells remodeled depending on the level of imposed strain yielding local variations of cell density. In particular, a collective cell orientation orthogonal to the main direction of strain was observed at low levels of wall deformation, while cells reoriented parallel to the main direction of strain at high levels of wall deformation. The tissue adaptation depended on the establishment of mature adherens junctions, which were reinforced by the polarized recruitment of the adaptor protein vinculin. The pivotal role of cell-to-cell junctions was confirmed by the biochemical inhibition of vascular endothelial cadherin homotypic contacts, which impaired the collective remodeling. Together, our data establish wall deformation as an independent determinant of endothelial architecture with direct implications in vascular physiopathology.
Collapse
Affiliation(s)
- Laura Bernardi
- ETH Zurich, Institute for Mechanical Systems, 8092 Zürich, Switzerland.
| | | | | | | | | | | |
Collapse
|
36
|
Endothelial factors in the pathogenesis and treatment of chronic kidney disease Part I: General mechanisms: a joint consensus statement from the European Society of Hypertension Working Group on Endothelin and Endothelial Factors and The Japanese Society of Hypertension. J Hypertens 2019; 36:451-461. [PMID: 29120962 DOI: 10.1097/hjh.0000000000001599] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
: Kidney damage is a common consequence of arterial hypertension, but is also a cause of atherogenesis. Dysfunction and/or harm of the endothelium in glomeruli and tubular interstitium damage the function of these structures and translates into dynamic changes of filtration fraction, with progressive reduction in glomerular filtration rate, expansion of extracellular fluid volume, abnormal ion balance, and hypoxia, ultimately leading to chronic kidney disease. Considering the key role played by endothelial dysfunction in chronic kidney disease, the Working Group on Endothelin and Endothelial Factors of the European Society of Hypertension and the Japanese Society of Hypertension have critically reviewed available knowledge on the mechanisms underlying endothelial cell injury. This resulted into two articles: in the first, we herein examine the mechanisms by which endothelial factors induce vascular remodeling and the role of different players, including endothelin-1, the renin-angiotensin-aldosterone system and their interactions, and of oxidative stress; in the second, we discuss the role of endothelial dysfunction in the major disease conditions that affect the kidney.
Collapse
|
37
|
Zhu T, Wang H, Wang L, Zhong X, Huang W, Deng X, Guo H, Xiong J, Xu Y, Fan J. Ginsenoside Rg1 attenuates high glucose-induced endothelial barrier dysfunction in human umbilical vein endothelial cells by protecting the endothelial glycocalyx. Exp Ther Med 2019; 17:3727-3733. [PMID: 30988758 DOI: 10.3892/etm.2019.7378] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023] Open
Abstract
Disruption of the endothelial barrier is essential for vascular complications associated with diabetes mellitus, and damage to the endothelial glycocalyx has been demonstrated to participate in this process. Ginsenoside Rg1 (Rg1), the major active component isolated from Panax notoginseng, is widely applied for the protection against vascular injury. The present study aimed to analyze the effect of high glucose on endothelial barrier function and its association with endothelial glycocalyx in human umbilical vein endothelial cells (HUVECs), and explore the potential benefits of Rg1 in protecting endothelial barrier function from high glucose-induced injury. The results indicated that high glucose induced a disorder of the endothelial glycocalyx and increased heparanase mRNA expression in HUVECs, which was reversed by Rg1 treatment. In addition, Rg1 treatment reduced transendothelial electrical resistance and transendothelial albumin passage after high-glucose stimulation. The present study suggested that high glucose caused a disruption in the endothelial glycocalyx and increased heparanase expression, which finally resulted in endothelial barrier dysfunction in HUVECs. Of note, Rg1 has a protective effect on high glucose-induced endothelial barrier dysfunction by attenuating the associated increase in heparanase expression.
Collapse
Affiliation(s)
- Tingting Zhu
- Department of Traditional Chinese and Western Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China.,Department of Nephrology, The First Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Nephrology, The Research Center of Combined Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Honglian Wang
- Department of Nephrology, The Research Center of Combined Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li Wang
- Department of Nephrology, The Research Center of Combined Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xia Zhong
- Department of Nephrology, The Research Center of Combined Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wei Huang
- Department of Traditional Chinese and Western Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China
| | - Xian Deng
- Department of Traditional Chinese and Western Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China
| | - Hengli Guo
- Department of Traditional Chinese and Western Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China
| | - Jianfeng Xiong
- Department of Traditional Chinese and Western Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China
| | - Youhua Xu
- Department of Traditional Chinese and Western Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China
| | - Junming Fan
- Department of Traditional Chinese and Western Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, P.R. China.,Department of Nephrology, The Research Center of Combined Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Department of Traditional Chinese and Western Medicine, Chengdu Medical College, Chengdu, Sichuan 610000, P.R. China
| |
Collapse
|
38
|
Stenosis Indicators Applied to Patient-Specific Renal Arteries without and with Stenosis. FLUIDS 2019. [DOI: 10.3390/fluids4010026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Pulsatile flow in the abdominal aorta and the renal arteries of three patients was studied numerically. Two of the patients had renal artery stenosis. The aim of the study was to assess the use of four types of indicators for determining the risk of new stenosis after revascularization of the affected arteries. The four indicators considered include the time averaged wall shear stress (TAWSS), the oscillatory shear index (OSI), the relative reference time (RRT) and a power law model based in platelet activation modeling but applied to the endothelium, named endothelium activation indicator (EAI). The results show that the indicators can detect the existing stenosis but are less successful in the revascularized cases. The TAWSS and, more clearly, the EAI approach seem to be better in predicting the risk for stenosis relapse at the original location and close to the post-stenotic dilatation. The shortcomings of the respective indicators are discussed along with potential improvements to endothelial activation modeling and its use as an indicator for risks of restenosis.
Collapse
|
39
|
McVey MJ, Kuebler WM. Extracellular vesicles: biomarkers and regulators of vascular function during extracorporeal circulation. Oncotarget 2018; 9:37229-37251. [PMID: 30647856 PMCID: PMC6324688 DOI: 10.18632/oncotarget.26433] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are generated at increased rates from parenchymal and circulating blood cells during exposure of the circulation to abnormal flow conditions and foreign materials associated with extracorporeal circuits (ExCors). This review describes types of EVs produced in different ExCors and extracorporeal life support (ECLS) systems including cardiopulmonary bypass circuits, extracorporeal membrane oxygenation (ECMO), extracorporeal carbon dioxide removal (ECCO2R), apheresis, dialysis and ventricular assist devices. Roles of EVs not only as biomarkers of adverse events during ExCor/ECLS use, but also as mediators of vascular dysfunction are explored. Manipulation of the number or subtypes of circulating EVs may prove a means of improving vascular function for individuals requiring ExCor/ECLS support. Strategies for therapeutic manipulation of EVs during ExCor/ECLS use are discussed such as accelerating their clearance, preventing their genesis or pharmacologic options to reduce or select which and how many EVs circulate. Strategies to reduce or select for specific types of EVs may prove beneficial in preventing or treating other EV-related diseases such as cancer.
Collapse
Affiliation(s)
- Mark J McVey
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Anesthesia, University of Toronto, Toronto, ON, Canada.,Department of Anesthesia and Pain Medicine, SickKids, Toronto, ON, Canada
| | - Wolfgang M Kuebler
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Heart Institute, Berlin, Germany
| |
Collapse
|
40
|
Chronic kidney failure mineral bone disorder leads to a permanent loss of hematopoietic stem cells through dysfunction of the stem cell niche. Sci Rep 2018; 8:15385. [PMID: 30337617 PMCID: PMC6194087 DOI: 10.1038/s41598-018-33979-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/04/2018] [Indexed: 01/11/2023] Open
Abstract
In chronic kidney disease (CKD), endothelial injury, is associated with disease progression and an increased risk for cardiovascular complications. Circulating cells with vascular reparative functions are hematopoietic and also reduced in CKD. To explore the mechanistic basis behind these observations, we have investigated hematopoietic stem cell (HSC) homeostasis in a mouse model for non-progressive CKD-mineral and bone disorder with experimentally induced chronic renal failure (CRF). In mice subjected to 12 weeks of CRF, bone marrow HSC frequencies were decreased and transplantation of bone marrow cells from CRF donors showed a decrease in long-term HSC repopulation compared to controls. This loss was directly associated with a CRF-induced defect in the HSC niche affecting the cell cycle status of HSC and could not be restored by the PTH-reducing agent cinacalcet. In CRF, frequencies of quiescent (G0) HSC were decreased coinciding with an increase in hematopoietic progenitor cells (HPC) in the S-and G2-phases of cell cycle. Moreover, in CRF mice, HSC-niche supporting macrophages were decreased compared to controls concomitant to impaired B lymphopoiesis. Our data point to a permanent loss of HSC and may provide insight into the root cause of the loss of homeostatic potential in CKD.
Collapse
|
41
|
Cellular and molecular mechanisms of kidney fibrosis. Mol Aspects Med 2018; 65:16-36. [PMID: 29909119 DOI: 10.1016/j.mam.2018.06.002] [Citation(s) in RCA: 320] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/12/2018] [Indexed: 12/14/2022]
Abstract
Renal fibrosis is the final pathological process common to any ongoing, chronic kidney injury or maladaptive repair. It is considered as the underlying pathological process of chronic kidney disease (CKD), which affects more than 10% of world population and for which treatment options are limited. Renal fibrosis is defined by excessive deposition of extracellular matrix, which disrupts and replaces the functional parenchyma that leads to organ failure. Kidney's histological structure can be divided into three main compartments, all of which can be affected by fibrosis, specifically termed glomerulosclerosis in glomeruli, interstitial fibrosis in tubulointerstitium and arteriosclerosis and perivascular fibrosis in vasculature. In this review, we summarized the different appearance, cellular origin and major emerging processes and mediators of fibrosis in each compartment. We also depicted and discussed the challenges in translation of anti-fibrotic treatment to clinical practice and discuss possible solutions and future directions.
Collapse
|
42
|
17β-Estradiol Accelerated Renal Tubule Regeneration in Male Rats After Ischemia/Reperfusion-Induced Acute Kidney Injury. Shock 2018; 46:158-63. [PMID: 26849629 DOI: 10.1097/shk.0000000000000586] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ischemic/reperfusion injury (IRI) is the most common cause of acute kidney injury (AKI). Murine studies report that pretreatment with 17β-estradiol protects against AKI using multiple mechanisms, but how 17β-estradiol is involved in regenerating tubular cells is unknown. To visualize the kidney injury and repair, we used 17β-estradiol to treat rats with postischemic acute kidney injury. AKI was induced by clamping the renal pedicle for 90 minutes 2 weeks after a unilateral nephrectomy. Rats were treated with an intravenous injection of 17β-estradiol or vehicle immediately after reperfusion. Kidney injury was assessed by measuring biochemical and histopathological changes. Immunohistochemical staining of vimentin, proliferating cell nuclear antigen (PCNA), and E-cadherin were used to assess dedifferentiation, proliferation, and redifferentiation. Rats treated with 17β-estradiol had less kidney injury than did vehicle-treated rats post-IRI day 1. The number of PCNA-positive (PCNA) cells was significantly higher in post-IRI kidneys on day 1 in 17β-estradiol-treated rats. Moreover, vimentin and E-cadherin cells, which were interpreted as regeneration markers, were expressed earlier and significantly more copiously in 17β-estradiol-treated rats. We hypothesize that 17β-estradiol attenuates IRI-induced AKI by reducing inflammation and accelerating injured tubular cell regeneration.
Collapse
|
43
|
Guo Y, Wei F, Wang J, Zhao Y, Sun S, Zhang H, Liu Z. Carotid artery wall shear stress is independently correlated with renal function in the elderly. Oncotarget 2018; 9:5251-5262. [PMID: 29435176 PMCID: PMC5797047 DOI: 10.18632/oncotarget.23825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 12/27/2017] [Indexed: 11/25/2022] Open
Abstract
Hemodynamic has increasingly been regarded as an important factor of renal function. However, the relationship between carotid artery wall shear stress (WSS) and renal function is not clarified. To investigate the relationship between carotid WSS and renal function, we recruited 761 older subjects aged 60 years and over from community-dwelling in the Shandong area, China. Carotid WSS, endothelial function, and estimated glomerular filtration rate (eGFR) were assessed in all subjects. Subjects were grouped by the interquartile of the carotid artery mean WSS. We found that the eGFRs derived from serum creatinine and/or cystatin C using three CKD-EPI equations were significantly higher and albumin/creatinine ratio was lower in the higher interquartile groups than in the lower interquartile groups (P <0.05). The mean WSS was independently correlated with eGFRs even after adjustment for confounders. Similar findings were found between carotid artery peak WSS and eGFRs and albumin/creatinine ratio. In addition, we found that endothelial function was strongly related to carotid WSS and renal function after adjustment for confounders. In conclusion, there is an independent correlation of carotid WSS with renal function in the elderly. The local rheologic forces may play an important role in renal function changing. The correlation may be mediated by regulation of endothelial function.
Collapse
Affiliation(s)
- Yuqi Guo
- Cardio-Cerebrovascular Control and Research Center, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Fang Wei
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, China
| | - Juan Wang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250000, China
| | - Yingxin Zhao
- Cardio-Cerebrovascular Control and Research Center, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Shangwen Sun
- Cardio-Cerebrovascular Control and Research Center, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Hua Zhang
- Cardio-Cerebrovascular Control and Research Center, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Zhendong Liu
- Cardio-Cerebrovascular Control and Research Center, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| |
Collapse
|
44
|
Saboia Z, Meneses G, Martins A, Daher E, Silva G. Association between syndecan-1 and renal function in adolescents with excess weight: evidence of subclinical kidney disease and endothelial dysfunction. Braz J Med Biol Res 2018; 51:e7174. [PMID: 29340529 PMCID: PMC5769763 DOI: 10.1590/1414-431x20177174] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 10/26/2017] [Indexed: 01/30/2023] Open
Abstract
Excess weight (overweight and obesity) is associated with kidney and cardiovascular disease. The aim of this study was to investigate the association between syndecan-1 and renal function among adolescents with excess weight. A total of 56 students from a public school at Fortaleza, CE, Brazil, were investigated. The adolescents were submitted to anthropometric evaluation, including weight, height, blood pressure and body mass index. Blood and urine samples were collected for the determination of serum lipids (total cholesterol, high density lipoprotein cholesterol, low density lipoprotein cholesterol, triglycerides), and the endothelial injury biomarker syndecan-1. Participants' mean age was 16±1 years (range 14-19 years), and 68% were females. Overweight was observed in 4 cases (7.1%) and obesity in 7 (12.5%). Changes in serum lipid levels were more frequent in the overweight group. A positive correlation between syndecan-1 and serum creatinine (r=0.5, P=0.001) and triglycerides (r=0.37, P=0.004), and a negative correlation with glomerular filtration rate (r=-0.33, P=0.02) were found. These findings suggest that adolescents with excess weight present incipient changes at the cellular level that make them more vulnerable to the development of kidney and cardiovascular diseases.
Collapse
Affiliation(s)
- Z.M.R.M. Saboia
- Programa de Pós-Graduação em Saúde Coletiva, Centro de Ciências da Saúde, Universidade de Fortaleza, Fortaleza, CE, Brasil
- Coordenadoria do Serviço de Saúde, Instituto Federal de Educação, Ciência e Tecnologia do Ceará, Fortaleza, CE, Brasil
| | - G.C. Meneses
- Programa de Pós-Graduação em Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A.M.C. Martins
- Programa de Pós-Graduação em Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - E.F. Daher
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - G.B. Silva
- Programa de Pós-Graduação em Saúde Coletiva, Centro de Ciências da Saúde, Universidade de Fortaleza, Fortaleza, CE, Brasil
- Coordenadoria do Serviço de Saúde, Instituto Federal de Educação, Ciência e Tecnologia do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
45
|
Raup-Konsavage WM, Wang Y, Wang WW, Feliers D, Ruan H, Reeves WB. Neutrophil peptidyl arginine deiminase-4 has a pivotal role in ischemia/reperfusion-induced acute kidney injury. Kidney Int 2017; 93:365-374. [PMID: 29061334 DOI: 10.1016/j.kint.2017.08.014] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 07/27/2017] [Accepted: 08/17/2017] [Indexed: 12/13/2022]
Abstract
Ischemia/reperfusion is a common cause of acute kidney injury (AKI). However, mechanisms underlying the sudden loss in kidney function and tissue injury remain to be fully elucidated. Here, we investigated the role of peptidyl arginine deiminase-4 (PAD4), which converts arginine to citrulline and plays a role in epigenetic regulation and inflammation, in renal ischemia/reperfusion injury. PAD4 expression was highly induced in infiltrating leukocytes 24 hours following renal ischemia and reperfusion. This induction was accompanied by citrullination of histone H3 and formation of neutrophil extracellular traps in kidneys of wild-type mice. By contrast, PAD4-deficient mice did not form neutrophil extracellular traps, expressed lower levels of pro-inflammatory cytokines and were partially protected from renal ischemia/reperfusion-induced AKI. Furthermore, PAD4-deficient mice recovered kidney function 48 hours after ischemia/reperfusion, whereas kidney function in the wild-type mice progressively worsened. Administration of DNase I, which degrades neutrophil extracellular traps or the PAD-specific inhibitor YW3-56 before ischemia, partially prevented renal ischemia/reperfusion-induced AKI. Notably, transfer of neutrophils from wild-type, but not from PAD4-deficient mice, was sufficient to restore renal neutrophil extracellular trap formation and impair kidney function following renal ischemia/reperfusion. Thus, neutrophil PAD4 plays a pivotal role in renal ischemia/reperfusion-induced AKI.
Collapse
Affiliation(s)
| | - Yanming Wang
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Wei Wei Wang
- Department of Medicine, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Denis Feliers
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Hong Ruan
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - W Brian Reeves
- Department of Medicine, Penn State College of Medicine, Hershey, Pennsylvania, USA; Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA.
| |
Collapse
|
46
|
Grudyanov AI, Tkacheva ON, Khatagov AT. [The impact of surgical and conservative treatment of periodontal disease on structural and functional condition of major blood vessels]. STOMATOLOGII︠A︡ 2017; 96:12-16. [PMID: 28514340 DOI: 10.17116/stomat201796212-16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The study included 55 patients (24 men and 31 women aged 30-70 years) with chronic periodontal disease and atherosclerosis of brachiocephalic arteries (BCA). Patients in group 1 received only conservative treatment for periodontal disease, while in group 2 both conservative and surgical treatment were performed. The values were evaluated at baseline and 6 months after treatment. Capillaroscopy and laser Doppler flowmetry showed an improvement in periodontal regional vascular network. The level of IL-6 decreased by 3.7 pg/ml and 5.9 pg/ml, C-reactive protein - by 2.0 mg/l and 3.4 mg/l in groups 1 and 2 correspondingly. Dark-field microscopy showed the decrease in the thickness of perfused boundary region by 0.05 mm and 0.28 mm, and the increase of red blood cells filling by 1.2% and 7.8% correspondingly. According to the results of ultrasound scanning of BCA after 6 months the number of atherosclerotic plaques and lumen of the vessels in the area of the plaques has not changed. BCA tone has not changed in group 1 and increased by 1-2% in group 2. Intima-media complex thickness decreased in group 1 by 0.02-0.03 mm, in group 2 - by 0.04 mm. The obtained data prove correlation of vascular status of periodontal and major blood vessels.
Collapse
Affiliation(s)
- A I Grudyanov
- Central Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
| | - O N Tkacheva
- Central Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
| | - A T Khatagov
- Central Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
| |
Collapse
|
47
|
Gomaraschi M, Ossoli A, Castelnuovo S, Simonelli S, Pavanello C, Balzarotti G, Arca M, Di Costanzo A, Sampietro T, Vaudo G, Baldassarre D, Veglia F, Franceschini G, Calabresi L. Depletion in LpA-I:A-II particles enhances HDL-mediated endothelial protection in familial LCAT deficiency. J Lipid Res 2017; 58:994-1001. [PMID: 28351888 DOI: 10.1194/jlr.p072371] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 03/04/2017] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to evaluate the vasoprotective effects of HDL isolated from carriers of LCAT deficiency, which are characterized by a selective depletion of LpA-I:A-II particles and predominance of preβ migrating HDL. HDLs were isolated from LCAT-deficient carriers and tested in vitro for their capacity to promote NO production and to inhibit vascular cell adhesion molecule-1 (VCAM-1) expression in cultured endothelial cells. HDLs from carriers were more effective than control HDLs in promoting eNOS activation with a gene-dose-dependent effect (PTrend = 0.048). As a consequence, NO production induced by HDL from carriers was significantly higher than that promoted by control HDL (1.63 ± 0.24-fold vs. 1.34 ± 0.07-fold, P = 0.031). HDLs from carriers were also more effective than control HDLs in inhibiting the expression of VCAM-1 (homozygotes, 65.0 ± 8.6%; heterozygotes, 53.1 ± 7.2%; controls, 44.4 ± 4.1%; PTrend = 0.0003). The increased efficiency of carrier HDL was likely due to the depletion in LpA-I:A-II particles. The in vitro findings might explain why carriers of LCAT deficiency showed flow-mediated vasodilation and plasma-soluble cell adhesion molecule concentrations comparable to controls, despite low HDL-cholesterol levels. These results indicate that selective depletion of apoA-II-containing HDL, as observed in carriers of LCAT deficiency, leads to an increased capacity of HDL to stimulate endothelial NO production, suggesting that changes in HDL apolipoprotein composition may be the target of therapeutic interventions designed to improve HDL functionality.
Collapse
Affiliation(s)
- Monica Gomaraschi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari Università degli Studi di Milano, Milano, Italy
| | - Alice Ossoli
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari Università degli Studi di Milano, Milano, Italy
| | - Samuela Castelnuovo
- Centro Dislipidemie, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Sara Simonelli
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari Università degli Studi di Milano, Milano, Italy
| | - Chiara Pavanello
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari Università degli Studi di Milano, Milano, Italy
| | - Gloria Balzarotti
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari Università degli Studi di Milano, Milano, Italy
| | - Marcello Arca
- Atherosclerosis Center, Department of Internal Medicine and Allied Sciences, Sapienza University of Rome, Rome, Italy
| | - Alessia Di Costanzo
- Atherosclerosis Center, Department of Internal Medicine and Allied Sciences, Sapienza University of Rome, Rome, Italy
| | | | - Gaetano Vaudo
- Department of Medicine, University of Perugia, Perugia, Italy
| | | | | | - Guido Franceschini
- Section of Chemical and Biomolecular Sciences, DeFENS, Università degli Studi di Milano, Milano, Italy
| | - Laura Calabresi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
48
|
Grudyanov AI, Tkacheva ON, Khatagov AT, Mustafina FN, Gorshkov AY. [The impact of periodontal disease treatment on endothelium of sublingual microvessels]. STOMATOLOGII︠A︡ 2016; 95:9-12. [PMID: 27636753 DOI: 10.17116/stomat20169549-12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The intactness degree of glycocalyx and the thickness of endothelium layer in sublingual microvessels was determined by means of darkfield microscopy and GlycoCheck RI software in 35 patients (19 men and 16 women, aged 35 to 65 years) with chronic generalized severe periodontitis before and after anti-inflammatory and surgical treatment. The patients were divided into two groups. The first group recieved only local anti-inflammatory therapy while patients of group 2 underwent surgical intervention on the periodontium in addition to local anti-inflammatory therapy. In both groups the changes of parameters of endothelial glycocalyx (EGC) in the early stages of treatment were not significant. In long terms the changes were not revealed in group 1, but in group 2 6 months after surgical treatment in all patients the decrease in the thickness of the permeable glycocalyx decreased and increase of the volume of red blood cells filling was observed.
Collapse
Affiliation(s)
- A I Grudyanov
- Central Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
| | - O N Tkacheva
- Central Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
| | - A T Khatagov
- Central Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
| | - F N Mustafina
- Central Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
| | - A Yu Gorshkov
- Central Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
| |
Collapse
|
49
|
Flamant S, Tamarat R. Extracellular Vesicles and Vascular Injury: New Insights for Radiation Exposure. Radiat Res 2016; 186:203-18. [PMID: 27459703 DOI: 10.1667/rr14482.1] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This article reviews our current knowledge about cell-derived extracellular vesicles (EVs), including microparticles and exosomes, and their emergence as mediators of a new important mechanism of cell-to-cell communication. Particular emphasis has been given to the increasing involvement of EVs in the field of radiation-induced vascular injury. Although EVs have been considered for a long time as cell "dust", they in fact precisely reflect the physiological state of the cells. The role of microparticles and exosomes in mediating vascular dysfunction suggests that they may represent novel pathways in short- or long-distance paracrine intercellular signaling in vascular environment. In this article, the mechanisms involved in the biogenesis of microparticles and exosomes, their composition and participation in the pathogenesis of vascular dysfunction are discussed. Furthermore, this article highlights the concept of EVs as potent vectors of biological information and protagonists of an intercellular communication network. Special emphasis is made on EV-mediated microRNA transfer and on the principal consequences of such signal exchange on vascular injury and radiation-induced nontargeted effect. The recent progress in elucidating the biology of EVs has provided new insights for the field of radiation, advancing their use as diagnostic biomarkers or in therapeutic interventions.
Collapse
Affiliation(s)
- Stéphane Flamant
- Institute for Radiological Protection and Nuclear Safety (IRSN) PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| | - Radia Tamarat
- Institute for Radiological Protection and Nuclear Safety (IRSN) PRP-HOM/SRBE/LR2I, Fontenay-aux-Roses, France
| |
Collapse
|
50
|
Cellular response of the blood-brain barrier to injury: Potential biomarkers and therapeutic targets for brain regeneration. Neurobiol Dis 2016; 91:262-73. [DOI: 10.1016/j.nbd.2016.03.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/09/2016] [Accepted: 03/16/2016] [Indexed: 02/07/2023] Open
|