1
|
Hossen F, Geng X, Sun GY, Yao X, Lee JC. Oligomeric Amyloid-β and Tau Alter Cell Adhesion Properties and Induce Inflammatory Responses in Cerebral Endothelial Cells Through the RhoA/ROCK Pathway. Mol Neurobiol 2024; 61:8759-8776. [PMID: 38561558 PMCID: PMC11445398 DOI: 10.1007/s12035-024-04138-z] [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: 07/03/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
Dysfunction of cerebral endothelial cells (CECs) has been implicated in the pathology of Alzheimer's disease (AD). Despite evidence showing cytotoxic effects of oligomeric amyloid-β (oAβ) and Tau (oTau) in the central nervous system, their direct effects on CECs have not been fully investigated. In this study, we examined the direct effects of oAβ, oTau, and their combination on cell adhesion properties and inflammatory responses in CECs. We found that both oAβ and oTau increased cell stiffness, as well as the p-selectin/Sialyl-LewisX (sLeX) bonding-mediated membrane tether force and probability of adhesion in CECs. Consistent with these biomechanical alterations, treatments with oAβ or oTau also increased actin polymerization and the expression of p-selectin at the cell surface. These toxic oligomeric peptides also triggered inflammatory responses, including upregulations of p-NF-kB p65, IL-1β, and TNF-α. In addition, they rapidly activated the RhoA/ROCK pathway. These biochemical and biomechanical changes were further enhanced by the treatment with the combination of oAβ and oTau, which were significantly suppressed by Fasudil, a specific inhibitor for the RhoA/ROCK pathway. In conclusion, our data suggest that oAβ, oTau, and their combination triggered subcellular mechanical alterations and inflammatory responses in CECs through the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Faruk Hossen
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Xue Geng
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Xincheng Yao
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - James C Lee
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
2
|
Brückner A, Brandtner A, Rieck S, Matthey M, Geisen C, Fels B, Stei M, Kusche-Vihrog K, Fleischmann BK, Wenzel D. Site-specific genetic and functional signatures of aortic endothelial cells at aneurysm predilection sites in healthy and AngII ApoE -/- mice. Angiogenesis 2024; 27:719-738. [PMID: 38965173 PMCID: PMC11564227 DOI: 10.1007/s10456-024-09933-9] [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: 12/18/2023] [Accepted: 06/16/2024] [Indexed: 07/06/2024]
Abstract
Aortic aneurysm is characterized by a pathological dilation at specific predilection sites of the vessel and potentially results in life-threatening vascular rupture. Herein, we established a modified "Häutchen method" for the local isolation of endothelial cells (ECs) from mouse aorta to analyze their spatial heterogeneity and potential role in site-specific disease development. When we compared ECs from aneurysm predilection sites of healthy mice with adjacent control segments we found regulation of genes related to extracellular matrix remodeling, angiogenesis and inflammation, all pathways playing a critical role in aneurysm development. We also detected enhanced cortical stiffness of the endothelium at these sites. Gene expression of ECs from aneurysms of the AngII ApoE-/- model when compared to sham animals mimicked expression patterns from predilection sites of healthy animals. Thus, this work highlights a striking genetic and functional regional heterogeneity in aortic ECs of healthy mice, which defines the location of aortic aneurysm formation in disease.
Collapse
Affiliation(s)
- Alexander Brückner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Adrian Brandtner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Sarah Rieck
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Michaela Matthey
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Caroline Geisen
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Benedikt Fels
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Marta Stei
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Kristina Kusche-Vihrog
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Bernd K Fleischmann
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Daniela Wenzel
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany.
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany.
| |
Collapse
|
3
|
Rooban S, Arul Senghor K, Vinodhini V, Kumar J. Adropin: A crucial regulator of cardiovascular health and metabolic balance. Metabol Open 2024; 23:100299. [PMID: 39045137 PMCID: PMC11263719 DOI: 10.1016/j.metop.2024.100299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
Adropin, a peptide discovered in 2008, has gained recognition as a key regulator of cardiovascular health and metabolic balance. Initially identified for its roles in energy balance, lipid metabolism, and glucose regulation, adropin has also been found to improve cardiovascular health by enhancing endothelial function, modulating lipid profiles, and reducing oxidative stress. These protective mechanisms suggest that adropin may be able to help prevent conditions such as atherosclerosis, hypertension, and other cardiovascular diseases. Research has established connections between adropin and cardiovascular risk factors, such as obesity, insulin resistance, and dyslipidemia, positioning it as a valuable biomarker for evaluating cardiovascular disease risk. New studies highlight adropin's diagnostic and prognostic significance, showing that higher levels are linked to better cardiovascular outcomes, while lower levels are associated with a higher risk of cardiovascular diseases. This review aims to summarize current knowledge on adropin, emphasizing its significance as a promising focus in the intersection of cardiovascular health and metabolic health. By summarizing the latest research findings, this review aims to offer insights into the potential applications of adropin in both clinical practice and research, leading to a deeper understanding of its role in maintaining cardiovascular and metabolic health.
Collapse
Affiliation(s)
- S. Rooban
- Department of Biochemistry, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| | - K.A. Arul Senghor
- Department of Biochemistry, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| | - V.M. Vinodhini
- Department of Biochemistry, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| | - J.S. Kumar
- Department of General Medicine, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| |
Collapse
|
4
|
Liu Q, Zhang Y, Han B, Wang M, Hu H, Ning J, Hu W, Chen M, Pang Y, Chen Y, Bao L, Niu Y, Zhang R. circRNAs deregulation in exosomes derived from BEAS-2B cells is associated with vascular stiffness induced by PM 2.5. J Environ Sci (China) 2024; 137:527-539. [PMID: 37980036 DOI: 10.1016/j.jes.2023.02.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 11/20/2023]
Abstract
As an environmental pollutant, ambient fine particulate matter (PM2.5) was linked to cardiovascular diseases. The molecular mechanisms underlying PM2.5-induced extrapulmonary disease has not been elucidated clearly. In this study the ambient PM2.5 exposure mice model we established was to explore adverse effects of vessel and potential mechanisms. Long-term PM2.5 exposure caused reduced lung function and vascular stiffness in mice. And chronic PM2.5 induced migration and epithelial-mesenchymal transition (EMT) phenotype in BEAS-2B cells. After PM2.5 treatment, the circRNAs and mRNAs levels of exosomes released by BEAS-2B cells were detected by competing endogenous RNA (ceRNA) array, which contained 1664 differentially expressed circRNAs (DE-circRNAs) and 308 differentially expressed mRNAs (DE-mRNAs). By bioinformatics analysis on host genes of DE-circRNAs, vascular diseases and some pathways related to vascular diseases including focal adhesion, tight junction and adherens junction were enriched. Then, ceRNA network was constructed, and DE-mRNAs in ceRNA network were conducted functional enrichment analysis by Ingenuity Pathway Analysis, which indicated that hsa_circ_0012627, hsa_circ_0053261 and hsa_circ_0052810 were related to vascular endothelial dysfunction. Furthermore, it was verified experimentally that ExoPM2.5 could induce endothelial dysfunction by increased endothelial permeability and decreased relaxation in vitro. In present study, we investigated in-depth knowledge into the molecule events related to PM2.5 toxicity and pathogenesis of vascular diseases.
Collapse
Affiliation(s)
- Qingping Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yaling Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Bin Han
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China; State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mengruo Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Huaifang Hu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Jie Ning
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Wentao Hu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Meiyu Chen
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yaxian Pang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yuanyuan Chen
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Lei Bao
- Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Yujie Niu
- Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Rong Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
5
|
Han WM, Hong YX, Xiao GS, Wang RY, Li G. NMDARs activation regulates endothelial ferroptosis via the PP2A-AMPK-HMGB1 axis. Cell Death Discov 2024; 10:34. [PMID: 38233385 PMCID: PMC10794209 DOI: 10.1038/s41420-023-01794-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 12/08/2023] [Accepted: 12/21/2023] [Indexed: 01/19/2024] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are ligand-gated, voltage-dependent channels of the ionotropic glutamate receptor family. The present study explored whether NMDAR activation induced ferroptosis in vascular endothelial cells and its complicated mechanisms in vivo and in vitro. Various detection approaches were used to determine the ferroptosis-related cellular iron content, lipid reactive oxygen species (LOS), siRNA molecules, RNA-sequence, MDA, GSH, and western blotting. The AMPK activator Acadesine (AICAR), HMGB1 inhibitor glycyrrhizin (GLY), PP2A inhibitor LB-100, and NMDAR inhibitor MK801 were used to investigate the involved in vivo and in vitro pathways. The activation of NMDAR with L-glutamic acid (GLU) or NMDA significantly promoted cellular ferroptosis, iron content, MDA, and the PTGS2 expression, while decreasing GPX4 expression and GSH concentration in human umbilical vein endothelial cells (HUVECs), which was reversed by ferroptosis inhibitors Ferrostatin-1(Fer-1), Liproxstatin-1 (Lip-1), or Deferoxamine (DFO). RNA-seq revealed that ferroptosis and SLC7A11 participate in NMDA or GLU-mediated NMDAR activation. The PP2A-AMPK-HMGB1 pathway was majorly associated with NMDAR activation-induced ferroptosis, validated using the PP2A inhibitor LB-100, AMPK activator AICAR, or HMGB1 siRNA. The role of NMDAR in ferroptosis was validated in HUVECs induced with the ferroptosis activator errasin or RSL3 and counteracted by the NMDAR inhibitor MK-801. The in vivo results showed that NMDA- or GLU-induced ferroptosis and LOS production was reversed by MK-801, LB-100, AICAR, MK-801, and GLY, confirming that the PP2A-AMPK-HMGB1 pathway is involved in NMDAR activation-induced vascular endothelium ferroptosis. In conclusion, the present study demonstrated a novel role of NMDAR in endothelial cell injury by regulating ferroptosis via the PP2A-AMPK-HMGB1 pathway.
Collapse
Affiliation(s)
- Wei-Min Han
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361008, China
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, Fujian, 361008, China
| | - Yi-Xiang Hong
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361008, China
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, Fujian, 361008, China
| | - Guo-Sheng Xiao
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361008, China.
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, Fujian, 361008, China.
| | - Rui-Ying Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361008, China.
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, Fujian, 361008, China.
| | - Gang Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361008, China.
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, Fujian, 361008, China.
| |
Collapse
|
6
|
Lai A, Zhou Y, Thurgood P, Chheang C, Chandra Sekar N, Nguyen N, Peter K, Khoshmanesh K, Baratchi S. Endothelial Response to the Combined Biomechanics of Vessel Stiffness and Shear Stress Is Regulated via Piezo1. ACS APPLIED MATERIALS & INTERFACES 2023; 15:59103-59116. [PMID: 38073418 DOI: 10.1021/acsami.3c07756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
How endothelial cells sense and respond to dynamic changes in their biophysical surroundings as we age is not fully understood. Vascular stiffness is clearly a contributing factor not only in several cardiovascular diseases but also in physiological processes such as aging and vascular dementia. To address this gap, we utilized a microfluidic model to explore how substrate stiffness in the presence of shear stress affects endothelial morphology, senescence, proliferation, and inflammation. We also studied the role of mechanosensitive ion channel Piezo1 in endothelial responses under the combined effect of shear stress and substrate stiffness. To do so, we cultured endothelial cells inside microfluidic channels covered with fibronectin-coated elastomer with elastic moduli of 40 and 200 kPa, respectively, mimicking the stiffness of the vessel walls in young and aged arteries. The endothelial cells were exposed to atheroprotective and atherogenic shear stress levels of 10 and 2 dyn/cm2, respectively. Our findings show that substrate stiffness affects senescence under atheroprotective flow conditions and cytoskeleton remodeling, senescence, and inflammation under atherogenic flow conditions. Additionally, we found that the expression of Piezo1 plays a crucial role in endothelial adaptation to flow and regulation of inflammation under both atheroprotective and atherogenic shear stress levels. However, Piezo1 contribution to endothelial senescence was limited to the soft substrate and atheroprotective shear stress level. Overall, our study characterizes the response of endothelial cells to the combined effect of shear stress and substrate stiffness and reveals a previously unidentified role of Piezo1 in endothelial response to vessel stiffening, which potentially can be therapeutically targeted to alleviate endothelial dysfunction in aging adults.
Collapse
Affiliation(s)
- Austin Lai
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria 3082, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Ying Zhou
- Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Peter Thurgood
- Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
- School of Engineering, RMIT University, Melbourne, Victoria 3000, Australia
| | - Chanly Chheang
- Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Nadia Chandra Sekar
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria 3082, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Ngan Nguyen
- School of Engineering, RMIT University, Melbourne, Victoria 3000, Australia
- Medical Technology Victoria, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Khashayar Khoshmanesh
- Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
- School of Engineering, RMIT University, Melbourne, Victoria 3000, Australia
| | - Sara Baratchi
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria 3082, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
7
|
Sun L, Wang L, Ye KX, Wang S, Zhang R, Juan Z, Feng L, Min S. Endothelial Glycocalyx in Aging and Age-related Diseases. Aging Dis 2023; 14:1606-1617. [PMID: 37196119 PMCID: PMC10529737 DOI: 10.14336/ad.2023.0131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/31/2023] [Indexed: 05/19/2023] Open
Abstract
The worldwide population is aging exponentially, creating burdens to patients, their families and society. Increasing age is associated with higher risk of a wide range of chronic diseases, and aging of the vascular system is closely linked to the development of many age-related diseases. Endothelial glycocalyx is a layer of proteoglycan polymers on the surface of the inner lumen of blood vessels. It plays an important role in maintaining vascular homeostasis and protecting various organ functions. Endothelial glycocalyx loss happens through the aging process and repairing the endothelial glycocalyx may alleviate the symptoms of age-related diseases. Given the important role of the glycocalyx and its regenerative properties, it is posited that the endothelial glycocalyx may be a potential therapeutic target for aging and age-related diseases and repairing endothelial glycocalyx could play a role in the promotion of healthy aging and longevity. Here, we review the composition, function, shedding, and manifestation of the endothelial glycocalyx in aging and age-related diseases, as well as regeneration of endothelial glycocalyx.
Collapse
Affiliation(s)
- Lina Sun
- School of Anesthesiology, Weifang Medical University, Weifang, China.
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Lingyan Wang
- School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Kaisy Xinhong Ye
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Shoushi Wang
- School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Rui Zhang
- School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Zhaodong Juan
- School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Lei Feng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Su Min
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
8
|
Zhang L, Li J, Chen J, Lei J, Yuan Z, Zhang J, Liu Z, Yu C, Ma L. Oscillatory shear stress-mediated aberrant O-GlcNAc SIRT3 accelerates glycocalyx inflammatory injury via LKB1/p47 phox/Hyal2 signaling. Cell Signal 2023:110790. [PMID: 37392860 DOI: 10.1016/j.cellsig.2023.110790] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/07/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023]
Abstract
Glycocalyx coating on endothelial surface layer helps to sense shear forces and maintain endothelial function. However, the underlying mechanism of endothelial glycocalyx degradation upon disordered shear stress stimulation is not fully understood. SIRT3, a major NAD+-dependent protein deacetylases, is required for protein stability during vascular homeostasis and partly involved in atherosclerotic process. While few studies showed that SIRT3 is responsible for endothelial glycocalyx homeostasis under shear stress, the underlying mechanisms remain largely unknown. Here, we demonstrated that oscillatory shear stress (OSS) induces glycocalyx injury by activating LKB1/p47phox/Hyal2 axis both in vivo and in vitro. And O-GlcNAc modification served to prolong SIRT3 deacetylase activity and stabilized p47/Hyal2 complex. OSS could decrease SIRT3 O-GlcNAcylation to activate LKB1, further accelerated endothelial glycocalyx injury in inflammatory microenvironment. SIRT3Ser329 mutation or inhibition of SIRT3 O-GlcNAcylation strongly promoted glycocalyx degradation. On the contrary, overexpression of SIRT3 reverse glycocalyx damage upon OSS treatment. Together, our findings indicated that targeting O-GlcNAcylation of SIRT3 could prevent and/or treat diseases whereby glycocalyx injured.
Collapse
Affiliation(s)
- Lei Zhang
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jiajia Li
- Hechuan District People's Hospital, Chongqing, China
| | - Jun Chen
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jin Lei
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Zhiyi Yuan
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jun Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Zhaohong Liu
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Chao Yu
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China.
| | - Limei Ma
- Chongqing Key Research Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
9
|
Vahldieck C, Cianflone E, Fels B, Löning S, Depelmann P, Sabatino J, Salerno N, Karsten CM, Torella D, Weil J, Sun D, Goligorsky MS, Kusche-Vihrog K. Endothelial Glycocalyx and Cardiomyocyte Damage Is Prevented by Recombinant Syndecan-1 in Acute Myocardial Infarction. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:474-492. [PMID: 36669683 PMCID: PMC10123521 DOI: 10.1016/j.ajpath.2022.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/24/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023]
Abstract
The outer layer of endothelial cells (ECs), consisting of the endothelial glycocalyx (eGC) and the cortex (CTX), provides a protective barrier against vascular diseases. Structural and functional impairments of their mechanical properties are recognized as hallmarks of endothelial dysfunction and can lead to cardiovascular events, such as acute myocardial infarction (AMI). This study investigated the effects of AMI on endothelial nanomechanics and function and the use of exogenous recombinant syndecan-1 (rSyn-1), a major component of the eGC, as recovering agent. ECs were exposed in vitro to serum samples collected from patients with AMI. In addition, in situ ECs of ex vivo aorta preparations derived from a mouse model for AMI were employed. Effects were quantified by using atomic force microscopy-based nanoindentation measurements, fluorescence staining, and histologic examination of the mouse hearts. AMI serum samples damaged eGC/CTX and augmented monocyte adhesion to the endothelial surface. In particular, the anaphylatoxins C3a and C5a played an important role in these processes. The impairment of endothelial function could be prevented by rSyn-1 treatment. In the mouse model of myocardial infarction, pretreatment with rSyn-1 alleviated eGC/CTX deterioration and reduced cardiomyocyte damage in histologic analyses. However, echocardiographic measurements did not indicate a functional benefit. These results provide new insights into the underlying mechanisms of AMI-induced endothelial dysfunction and perspectives for future studies on the benefit of rSyn-1 in post-AMI treatment.
Collapse
Affiliation(s)
- Carl Vahldieck
- Institute of Physiology, University of Luebeck, Luebeck, Germany; Department of Anesthesiology and Intensive Care Medicine, University Medical Centre Schleswig-Holstein Campus Luebeck, University of Luebeck, Luebeck, Germany.
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Benedikt Fels
- Institute of Physiology, University of Luebeck, Luebeck, Germany; DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Luebeck/Kiel, Luebeck, Germany
| | - Samuel Löning
- Institute of Physiology, University of Luebeck, Luebeck, Germany
| | - Patrik Depelmann
- Institute of Physiology, University of Luebeck, Luebeck, Germany
| | - Jolanda Sabatino
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy; Division of Pediatric Cardiology, Department of Women's and Children's Health, University Hospital Padua, Padua, Italy; Pediatric Research Institute "Città della Speranza", Padua, Italy
| | - Nadia Salerno
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Christian M Karsten
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Germany
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Joachim Weil
- Medizinische Klinik II, Sana Kliniken Luebeck, Luebeck, Germany
| | - Dong Sun
- Renal Research Institute and Departments of Medicine, Pharmacology and Physiology, New York Medical College, Valhalla, New York
| | - Michael S Goligorsky
- Renal Research Institute and Departments of Medicine, Pharmacology and Physiology, New York Medical College, Valhalla, New York
| | - Kristina Kusche-Vihrog
- Institute of Physiology, University of Luebeck, Luebeck, Germany; DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Luebeck/Kiel, Luebeck, Germany
| |
Collapse
|
10
|
Schulz A, Drost CC, Hesse B, Beul K, Boeckel GR, Lukasz A, Pavenstädt H, Brand M, Di Marco GS. The Endothelial Glycocalyx as a Target of Excess Soluble Fms-like Tyrosine Kinase-1. Int J Mol Sci 2023; 24:ijms24065380. [PMID: 36982455 PMCID: PMC10049398 DOI: 10.3390/ijms24065380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Soluble fms-like tyrosine kinase-1 (sFlt-1) is a secreted protein that binds heparan sulfate expressed on the endothelial glycocalyx (eGC). In this paper we analyze how excess sFlt-1 causes conformational changes in the eGC, leading to monocyte adhesion, a key event triggering vascular dysfunction. In vitro exposure of primary human umbilical vein endothelial cells to excess sFlt-1 decreased eGC height and increased stiffness as determined by atomic force microscopy (AFM). Yet, structural loss of the eGC components was not observed, as indicated by Ulex europaeus agglutinin I and wheat germ agglutinin staining. Moreover, the conformation observed under excess sFlt-1, a collapsed eGC, is flat and stiff with unchanged coverage and sustained content. Functionally, this conformation increased the endothelial adhesiveness to THP-1 monocytes by about 35%. Heparin blocked all these effects, but the vascular endothelial growth factor did not. In vivo administration of sFlt-1 in mice also resulted in the collapse of the eGC in isolated aorta analyzed ex vivo by AFM. Our findings show that excess sFlt-1 causes the collapse of the eGC and favors leukocyte adhesion. This study provides an additional mechanism of action by which sFlt-1 may cause endothelial dysfunction and injury.
Collapse
|
11
|
The Soluble Fms-like Tyrosine Kinase-1 Contributes to Structural and Functional Changes in Endothelial Cells in Chronic Kidney Disease. Int J Mol Sci 2022; 23:ijms232416059. [PMID: 36555698 PMCID: PMC9787493 DOI: 10.3390/ijms232416059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Endothelial cells are a critical target of the soluble Fms-like tyrosine kinase-1 (sFlt-1), a soluble factor increased in different diseases with varying degrees of renal impairment and endothelial dysfunction, including chronic kidney disease (CKD). Although the mechanisms underlying endothelial dysfunction are multifactorial and complex, herein, we investigated the damaging effects of sFlt-1 on structural and functional changes in endothelial cells. Our results evidenced that sera from patients with CKD stiffen the endothelial cell cortex in vitro, an effect correlated with sFlt-1 levels and prevented by sFlt-1 neutralization. Besides, we could show that recombinant sFlt-1 leads to endothelial stiffening in vitro and in vivo. This was accompanied by cytoskeleton reorganization and changes in the endothelial barrier function, as observed by increased actin polymerization and endothelial cell permeability, respectively. These results depended on the activation of the p38 MAPK and were blocked by the specific inhibitor SB203580. However, sFlt-1 only minimally affected the expression of stiffness-sensitive genes. These findings bring new insight into the mechanism of action of sFlt-1 and its biological effects that cannot be exclusively ascribed to the regulation of angiogenesis.
Collapse
|
12
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
13
|
Jurrissen TJ, Ramirez-Perez FI, Cabral-Amador FJ, Soares RN, Pettit-Mee RJ, Betancourt-Cortes EE, McMillan NJ, Sharma N, Rocha HNM, Fujie S, Morales-Quinones M, Lazo-Fernandez Y, Butler AA, Banerjee S, Sacks HS, Ibdah JA, Parks EJ, Rector RS, Manrique-Acevedo C, Martinez-Lemus LA, Padilla J. Role of adropin in arterial stiffening associated with obesity and type 2 diabetes. Am J Physiol Heart Circ Physiol 2022; 323:H879-H891. [PMID: 36083795 PMCID: PMC9602697 DOI: 10.1152/ajpheart.00385.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 01/16/2023]
Abstract
Adropin is a peptide largely secreted by the liver and known to regulate energy homeostasis; however, it also exerts cardiovascular effects. Herein, we tested the hypothesis that low circulating levels of adropin in obesity and type 2 diabetes (T2D) contribute to arterial stiffening. In support of this hypothesis, we report that obesity and T2D are associated with reduced levels of adropin (in liver and plasma) and increased arterial stiffness in mice and humans. Establishing causation, we show that mesenteric arteries from adropin knockout mice are also stiffer, relative to arteries from wild-type counterparts, thus recapitulating the stiffening phenotype observed in T2D db/db mice. Given the above, we performed a set of follow-up experiments, in which we found that 1) exposure of endothelial cells or isolated mesenteric arteries from db/db mice to adropin reduces filamentous actin (F-actin) stress fibers and stiffness, 2) adropin-induced reduction of F-actin and stiffness in endothelial cells and db/db mesenteric arteries is abrogated by inhibition of nitric oxide (NO) synthase, and 3) stimulation of smooth muscle cells or db/db mesenteric arteries with a NO mimetic reduces stiffness. Lastly, we demonstrated that in vivo treatment of db/db mice with adropin for 4 wk reduces stiffness in mesenteric arteries. Collectively, these findings indicate that adropin can regulate arterial stiffness, likely via endothelium-derived NO, and thus support the notion that "hypoadropinemia" should be considered as a putative target for the prevention and treatment of arterial stiffening in obesity and T2D.NEW & NOTEWORTHY Arterial stiffening, a characteristic feature of obesity and type 2 diabetes (T2D), contributes to the development and progression of cardiovascular diseases. Herein we establish that adropin is decreased in obese and T2D models and furthermore provide evidence that reduced adropin may directly contribute to arterial stiffening. Collectively, findings from this work support the notion that "hypoadropinemia" should be considered as a putative target for the prevention and treatment of arterial stiffening in obesity and T2D.
Collapse
Affiliation(s)
- Thomas J Jurrissen
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | | | | | - Rogerio N Soares
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
| | - Ryan J Pettit-Mee
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | | | - Neil J McMillan
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Neekun Sharma
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
| | - Helena N M Rocha
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Physiology and Pharmacology, Fluminense Federal University, Niteroi, Brazil
| | - Shumpei Fujie
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| | - Mariana Morales-Quinones
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
| | - Yoskaly Lazo-Fernandez
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
| | - Andrew A Butler
- Department of Pharmacology and Physiological Sciences, Saint Louis University, Saint Louis, Missouri
| | - Subhashis Banerjee
- Department of Pharmacology and Physiological Sciences, Saint Louis University, Saint Louis, Missouri
| | - Harold S Sacks
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Jamal A Ibdah
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Elizabeth J Parks
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
| |
Collapse
|
14
|
Pacia MZ, Chorazy N, Sternak M, Fels B, Pacia M, Kepczynski M, Kusche-Vihrog K, Chlopicki S. Rac1 regulates lipid droplets formation, nanomechanical, and nanostructural changes induced by TNF in vascular endothelium in the isolated murine aorta. Cell Mol Life Sci 2022; 79:317. [PMID: 35622139 PMCID: PMC9142475 DOI: 10.1007/s00018-022-04362-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/21/2022] [Accepted: 05/09/2022] [Indexed: 11/24/2022]
Abstract
Endothelial inflammation is recognized as a critical condition in the development of cardiovascular diseases. TNF-induced inflammation of endothelial cells is linked to the formation of lipid droplets, augmented cortical stiffness, and nanostructural endothelial plasma membrane remodelling, but the insight into the mechanism linking these responses is missing. In the present work, we determined the formation of lipid droplets (LDs), nanomechanical, and nanostructural responses in the model of TNF-activated vascular inflammation in the isolated murine aorta using Raman spectroscopy, fluorescence imaging, atomic force microscopy (AFM), and scanning electron microscopy (SEM). We analysed the possible role of Rac1, a major regulator of cytoskeletal organization, in TNF-induced vascular inflammation. We demonstrated that the formation of LDs, polymerization of F-actin, alterations in cortical stiffness, and nanostructural protuberances in endothelial plasma membrane were mediated by the Rac1. In particular, we revealed a significant role for Rac1 in the regulation of the formation of highly unsaturated LDs formed in response to TNF. Inhibition of Rac1 also downregulated the overexpression of ICAM-1 induced by TNF, supporting the role of Rac1 in vascular inflammation. Altogether, our results demonstrate that LDs formation, an integral component of vascular inflammation, is activated by Rac1 that also regulates nanomechanical and nanostructural alterations linked to vascular inflammation.
Collapse
Affiliation(s)
- Marta Z Pacia
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Krakow, Poland.
| | - Natalia Chorazy
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Krakow, Poland
| | - Magdalena Sternak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Krakow, Poland
| | - Benedikt Fels
- Institute of Physiology, University of Luebeck, 160 Ratzeburger Allee, 23562, Luebeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Michal Pacia
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387, Krakow, Poland
| | - Mariusz Kepczynski
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387, Krakow, Poland
| | - Kristina Kusche-Vihrog
- Institute of Physiology, University of Luebeck, 160 Ratzeburger Allee, 23562, Luebeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Krakow, Poland
- Chair of Pharmacology, Jagiellonian University, 16 Grzegorzecka Str., 31-531, Krakow, Poland
| |
Collapse
|
15
|
Lei J, Xiang P, Zeng S, Chen L, Zhang L, Yuan Z, Zhang J, Wang T, Yu R, Zhang W, Ibrahim II, Ma L, Yu C. Tetramethylpyrazine Alleviates Endothelial Glycocalyx Degradation and Promotes Glycocalyx Restoration via TLR4/NF-κB/HPSE1 Signaling Pathway During Inflammation. Front Pharmacol 2022; 12:791841. [PMID: 35185540 PMCID: PMC8850260 DOI: 10.3389/fphar.2021.791841] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Tetramethylpyrazine (TMP), a Chinese traditional herbal extraction widely used in treating cardiovascular diseases, could attenuate vascular endothelial injuries, but the underlying mechanism remains incomprehensive. Vascular glycocalyx coating on the endothelium would be damaged and caused endothelial dysfunction in the inflammatory microenvironment, which was the initial factor of morbidity of many vascular diseases, such as atherosclerosis (AS). Here, we thoroughly investigated the molecular mechanism of TMP on vascular endothelial glycocalyx in the LPS-induced inflammatory model both in vitro and in vivo. Results showed that pretreatment with TMP significantly inhibited glycocalyx degradation and monocytes adhesion to the endothelial process. Moreover, TMP pretreatment inhibited the expression of HPSE1 (a major degrading enzyme of endothelial glycocalyx), Toll-like receptor 4 (TLR4), and the translocation of nuclear factor kappa B p65 (NF-κB p65). We were utilized withTLR4 siRNA, NF-κB inhibitor, and HPSE1 overexpression analysis confirmed TMP's protection on endothelial glycocalyx injury, which further contributed to the monocyte-endothelial adhesion process. It was indicated that TMP might suppress glycocalyx degradation through TLR4/NF-κB/HPSE1 signaling pathway. Taken together, our results enriched the occurrence molecular mechanism of glycocalyx shedding and molecular regulation mechanism of TMP in protecting integrity of the glycocalyx structure during inflammation. As TMP is currently used in clinical applications, it may be considered a novel strategy against atherosclerosis through its ability to protect endothelial glycocalyx.
Collapse
Affiliation(s)
- Jin Lei
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Peng Xiang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Shengmei Zeng
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Le Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Lei Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Zhiyi Yuan
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Jun Zhang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.,Institute of Life Sciences, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Tingting Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Ruihong Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Wanping Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Issa Issoufou Ibrahim
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Limei Ma
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| |
Collapse
|
16
|
Xie Z, Hou L, Shen S, Wu Y, Wang J, Jie Z, Zhao X, Li X, Zhang X, Chen J, Xu W, Ning L, Ma Q, Wang S, Wang H, Yuan P, Fang X, Qin A, Fan S. Mechanical force promotes dimethylarginine dimethylaminohydrolase 1-mediated hydrolysis of the metabolite asymmetric dimethylarginine to enhance bone formation. Nat Commun 2022; 13:50. [PMID: 35013196 PMCID: PMC8748781 DOI: 10.1038/s41467-021-27629-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/30/2021] [Indexed: 12/30/2022] Open
Abstract
Mechanical force is critical for the development and remodeling of bone. Here we report that mechanical force regulates the production of the metabolite asymmetric dimethylarginine (ADMA) via regulating the hydrolytic enzyme dimethylarginine dimethylaminohydrolase 1 (Ddah1) expression in osteoblasts. The presence of -394 4 N del/ins polymorphism of Ddah1 and higher serum ADMA concentration are negatively associated with bone mineral density. Global or osteoblast-specific deletion of Ddah1 leads to increased ADMA level but reduced bone formation. Further molecular study unveils that mechanical stimulation enhances TAZ/SMAD4-induced Ddah1 transcription. Deletion of Ddah1 in osteoblast-lineage cells fails to respond to mechanical stimulus-associated bone formation. Taken together, the study reveals mechanical force is capable of down-regulating ADMA to enhance bone formation.
Collapse
Affiliation(s)
- Ziang Xie
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Lei Hou
- Department of Cardiology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shuying Shen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Yizheng Wu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jian Wang
- Department of Orthopaedics, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zhiwei Jie
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangde Zhao
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiang Li
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xuyang Zhang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Junxin Chen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Wenbin Xu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Lei Ning
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Qingliang Ma
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Shiyu Wang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Haoming Wang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Putao Yuan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangqian Fang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - An Qin
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Shunwu Fan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
17
|
Stoddart P, Satchell SC, Ramnath R. Cerebral microvascular endothelial glycocalyx damage, its implications on the blood-brain barrier and a possible contributor to cognitive impairment. Brain Res 2022; 1780:147804. [DOI: 10.1016/j.brainres.2022.147804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/31/2022]
|
18
|
Starodubtseva MN, Nadyrov EA, Shkliarava NM, Tsukanava AU, Starodubtsev IE, Kondrachyk AN, Matveyenkau MV, Nedoseikina MS. Heterogeneity of nanomechanical properties of the human umbilical vein endothelial cell surface. Microvasc Res 2021; 136:104168. [PMID: 33845104 DOI: 10.1016/j.mvr.2021.104168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/13/2021] [Accepted: 03/30/2021] [Indexed: 11/26/2022]
Abstract
Endothelial cells, due to heterogeneity in the cell structure, can potentially form an inhomogeneous on structural and mechanical properties of the inner layer of the capillaries. Using quantitative nanomechanical mapping mode of atomic force microscopy, the parameters of the structural, elastic, and adhesive properties of the cell surface for living and glutaraldehyde-fixed human umbilical vein endothelial cells were studied. A significant difference in the studied parameters for three cell surface zones (peripheral, perinuclear, and nuclear zones) was established. The perinuclear zone appeared to be the softest zone of the endothelial cell surface. The heterogeneity of the endothelial cell mechanical properties at the nanoscale level can be an important mechanism in regulating the endothelium functions in blood vessels.
Collapse
Affiliation(s)
- Maria N Starodubtseva
- Institute of Radiobiology of NAS of Belarus, 4 Fedyuninskogo str., Gomel BY-246007, Belarus; Gomel State Medical University, 5 Lange str., Gomel BY-246000, Belarus.
| | - Eldar A Nadyrov
- Gomel State Medical University, 5 Lange str., Gomel BY-246000, Belarus
| | - Nastassia M Shkliarava
- Institute of Radiobiology of NAS of Belarus, 4 Fedyuninskogo str., Gomel BY-246007, Belarus
| | - Alena U Tsukanava
- Institute of Radiobiology of NAS of Belarus, 4 Fedyuninskogo str., Gomel BY-246007, Belarus
| | | | | | - Matsvei V Matveyenkau
- Institute of Radiobiology of NAS of Belarus, 4 Fedyuninskogo str., Gomel BY-246007, Belarus
| | | |
Collapse
|
19
|
Daiber A, Chlopicki S. Revisiting pharmacology of oxidative stress and endothelial dysfunction in cardiovascular disease: Evidence for redox-based therapies. Free Radic Biol Med 2020; 157:15-37. [PMID: 32131026 DOI: 10.1016/j.freeradbiomed.2020.02.026] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/05/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
According to the latest Global Burden of Disease Study data, non-communicable diseases in general and cardiovascular disease (CVD) in particular are the leading cause of premature death and reduced quality of life. Demographic shifts, unhealthy lifestyles and a higher burden of adverse environmental factors provide an explanation for these findings. The expected growing prevalence of CVD requires enhanced research efforts for identification and characterisation of novel therapeutic targets and strategies. Cardiovascular risk factors including classical (e.g. hypertension, diabetes, hypercholesterolaemia) and non-classical (e.g. environmental stress) factors induce the development of endothelial dysfunction, which is closely associated with oxidant stress and vascular inflammation and results in CVD, particularly in older adults. Most classically successful therapies for CVD display vasoprotective, antioxidant and anti-inflammatory effects, but were originally designed with other therapeutic aims. So far, only a few 'redox drugs' are in clinical use and many antioxidant strategies have not met expectations. With the present review, we summarise the actual knowledge on CVD pathomechanisms, with special emphasis on endothelial dysfunction, adverse redox signalling and oxidative stress, highlighting the preclinical and clinical evidence. In addition, we provide a brief overview of established CVD therapies and their relation to endothelial dysfunction and oxidative stress. Finally, we discuss novel strategies for redox-based CVD therapies trying to explain why, despite a clear link between endothelial dysfunction and adverse redox signalling and oxidative stress, redox- and oxidative stress-based therapies have not yet provided a breakthrough in the treatment of endothelial dysfunction and CVD.
Collapse
Affiliation(s)
- Andreas Daiber
- The Center for Cardiology, Department of Cardiology 1, Laboratory of Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany; The Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Stefan Chlopicki
- The Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland; Jagiellonian University Medical College, Grzegorzecka 16, 31-531, Krakow, Poland.
| |
Collapse
|
20
|
Khalyfa A, Castro-Grattoni AL, Gozal D. Cardiovascular morbidities of obstructive sleep apnea and the role of circulating extracellular vesicles. Ther Adv Respir Dis 2020; 13:1753466619895229. [PMID: 31852426 PMCID: PMC6923690 DOI: 10.1177/1753466619895229] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Obstructive sleep apnea (OSA) is characterized by recurrent upper airway collapse
during sleep resulting in impaired blood gas exchange, namely intermittent
hypoxia (IH) and hypercapnia, fragmented sleep (SF), increased oxidative stress
and systemic inflammation. Among a myriad of potential associated morbidities,
OSA has been particularly implicated as mechanistically contributing to the
prevalence and severity of cardiovascular diseases (CVD). However, the benefits
of continuous positive airway pressure (CPAP), which is generally employed in
OSA treatment, to either prevent or improve CVD outcomes remain unconvincing,
suggesting that the pathophysiological mechanisms underlying the incremental CVD
risk associated with OSA are not clearly understood. One of the challenges in
development of non-invasive diagnostic assays is the ability to identify
clinically and mechanistically relevant biomarkers. Circulating extracellular
vesicles (EVs) and their cargos reflect underlying changes in cellular
homeostasis and can provide insights into how cells and systems cope with
physiological perturbations by virtue of the identity and abundance of miRNAs,
mRNAs, proteins, and lipids that are packaged in the EVs under normal as well as
diseased states, such as OSA. EVs can not only provide unique insights into
coordinated cellular responses at the organ or systemic level, but can also
serve as reporters of the effects of OSA in CVD, either by their properties
enabling regeneration and repair of injured vascular cells or by damaging them.
Here, we highlight recent progress in the pathological CVD consequences of OSA,
and explore the putative roles of EVs in OSA-associated CVD, along with emerging
diagnostic and therapeutic opportunities. The reviews of this paper are available via the supplemental material
section.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA
| | - Anabel L Castro-Grattoni
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA
| | - David Gozal
- Department of Child Health and MU Women's and Children's Hospital, University of Missouri School of Medicine, 400 N. Keene Street, Suite 010, Columbia, MO 65201, USA
| |
Collapse
|
21
|
Cosgun ZC, Fels B, Kusche-Vihrog K. Nanomechanics of the Endothelial Glycocalyx. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:732-741. [DOI: 10.1016/j.ajpath.2019.07.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/10/2019] [Accepted: 07/30/2019] [Indexed: 12/12/2022]
|
22
|
It takes more than two to tango: mechanosignaling of the endothelial surface. Pflugers Arch 2020; 472:419-433. [PMID: 32239285 PMCID: PMC7165135 DOI: 10.1007/s00424-020-02369-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023]
Abstract
The endothelial surface is a highly flexible signaling hub which is able to sense the hemodynamic forces of the streaming blood. The subsequent mechanosignaling is basically mediated by specific structures, like the endothelial glycocalyx building the top surface layer of endothelial cells as well as mechanosensitive ion channels within the endothelial plasma membrane. The mechanical properties of the endothelial cell surface are characterized by the dynamics of cytoskeletal proteins and play a key role in the process of signal transmission from the outside (lumen of the blood vessel) to the interior of the cell. Thus, the cell mechanics directly interact with the function of mechanosensitive structures and ion channels. To precisely maintain the vascular tone, a coordinated functional interdependency between endothelial cells and vascular smooth muscle cells is necessary. This is given by the fact that mechanosensitive ion channels are expressed in both cell types and that signals are transmitted via autocrine/paracrine mechanisms from layer to layer. Thus, the outer layer of the endothelial cells can be seen as important functional mechanosensitive and reactive cellular compartment. This review aims to describe the known mechanosensitive structures of the vessel building a bridge between the important role of physiological mechanosignaling and the proper vascular function. Since mutations and dysfunction of mechanosensitive proteins are linked to vascular pathologies such as hypertension, they play a potent role in the field of channelopathies and mechanomedicine.
Collapse
|
23
|
Jannatbabaei A, Tafazzoli‐Shadpour M, Seyedjafari E. Effects of substrate mechanics on angiogenic capacity and nitric oxide release in human endothelial cells. Ann N Y Acad Sci 2020; 1470:31-43. [DOI: 10.1111/nyas.14326] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/15/2019] [Accepted: 02/13/2020] [Indexed: 01/06/2023]
Affiliation(s)
- Atefeh Jannatbabaei
- Department of Biomedical EngineeringAmirkabir University of Technology Tehran Iran
| | | | - Ehsan Seyedjafari
- Department of Biotechnology, College of ScienceUniversity of Tehran Tehran Iran
| |
Collapse
|
24
|
Prystopiuk V, Fels B, Simon CS, Liashkovich I, Pasrednik D, Kronlage C, Wedlich-Söldner R, Oberleithner H, Fels J. A two-phase response of endothelial cells to hydrostatic pressure. J Cell Sci 2018; 131:jcs.206920. [DOI: 10.1242/jcs.206920] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 05/10/2018] [Indexed: 01/15/2023] Open
Abstract
The vascular endothelium is exposed to three types of mechanical forces: blood flow-mediated shear stress, vessel-diameter dependent wall tension and hydrostatic pressure. Despite considerable variations of blood pressure in normal and pathological physiology, little is known about the acute molecular and cellular effects of hydrostatic pressure on endothelial cells. Here, we used a combination of quantitative fluorescence microscopy, atomic force microscopy and molecular perturbations to characterize the specific response of endothelial cells to pressure application. We identified a two-phase response of endothelial cells to acute (1 h) vs. chronic (24 h) pressure application (100 mmHg). While both regimes induce cortical stiffening, the acute response is linked to calcium-mediated myosin activation, whereas the chronic cell response is dominated by increased cortical actin density and a loss in endothelial barrier function. GsMTx-4 and amiloride inhibit the acute pressure response, which suggest the sodium channel ENaC as key player in endothelial pressure sensing. The described two-phase pressure response may participate in the differential effects of transient changes in blood pressure and hypertension.
Collapse
Affiliation(s)
- Valeria Prystopiuk
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
- current address: Institute of Life Sciences, Université Catholique de Louvain, Croix du Sud, 4-5, bte L7.07.06, Louvain-la-Neuve B-1348, Belgium
| | - Benedikt Fels
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Caroline Sophie Simon
- Institute of Cell Dynamics and Imaging, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Ivan Liashkovich
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Dzmitry Pasrednik
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Cornelius Kronlage
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Roland Wedlich-Söldner
- Institute of Cell Dynamics and Imaging, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Hans Oberleithner
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Johannes Fels
- Institute of Cell Dynamics and Imaging, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| |
Collapse
|