1
|
Kumar S, Petschner P, Gecse K, Torok D, Juhasz G. Acute neuroendocrine challenge elicits enhanced cortisol response and parallel transcriptomic changes in patients with migraine. Pain Rep 2025; 10:e1254. [PMID: 40322023 PMCID: PMC12047896 DOI: 10.1097/pr9.0000000000001254] [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: 08/30/2024] [Revised: 12/18/2024] [Accepted: 01/10/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Migraine is a neurological disorder with recurrent attacks characterized by headaches and sensitivity to stimuli. Stress is a significant trigger for attacks; however, molecular mechanisms of the connection are poorly understood. Objectives To better characterize such mechanisms, we performed a placebo-controlled, double-blind crossover study with 51 participants (21 patients with migraine without aura and 30 healthy controls). Methods Participants received a low-dose citalopram- or placebo challenge on 2 separate days. Prechallenge and postchallenge assessment of cortisol concentrations and transcriptomic changes using RNA-seq was performed from whole blood samples. Analysis of an accidental attack following the citalopram challenge was also conducted. Results Neuroendocrine challenge elicited elevated cortisol concentrations at 30 (P-value = 0.1355) and 70 minutes (P-value = 0.07292) postchallenge in patients with migraine compared with controls. Gene expression analysis showed 10 differentially expressed genes (2 down- and 8 upregulated, P-value ≤ 0.005) and 10 dysregulated gene sets (P-value ≤ 0.005). Among them, dysregulated IKBKGP1 and NKRF genes and upregulated protein synthesis and translation, carbohydrate metabolism, and, attack-related, glycosylation can be highlighted. Conclusion Patients with migraine without aura showed an enhanced cortisol response to a neuroendocrine challenge. This was accompanied by a probable suppression of NFκB activity through dysregulation of NKRF and an altered immune function. Upregulated carbohydrate metabolism may reflect the elevated cortisol concentrations' stimulating effects on endothelial glycocalyx, playing a role in NO-induced vasodilation, a trigger for migraine attacks. The results suggest the elevated cortisol response may trigger migraine attacks through altered glycocalyx and immune functions.
Collapse
Affiliation(s)
- Sahel Kumar
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, Budapest, Hungary
| | - Peter Petschner
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, Budapest, Hungary
| | - Kinga Gecse
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Dora Torok
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, Budapest, Hungary
| | - Gabriella Juhasz
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| |
Collapse
|
2
|
Nielsen SF, Duus CL, Buus NH, Bech JN, Mose FH. The effects of empagliflozin on systemic haemodynamic function: three randomized, placebo-controlled trials. J Hypertens 2025; 43:1021-1029. [PMID: 40156337 DOI: 10.1097/hjh.0000000000004007] [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: 10/22/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Sodium glucose cotransporter 2 inhibitors lower blood pressure. The underlying mechanisms are multifactorial and include effects on vascular function. We examined the systemic hemodynamic effects of empagliflozin in patients with type 2 diabetes mellitus (DM2) with and without chronic kidney disease (CKD) and in patients with nondiabetic CKD. METHODS Three double-blinded, randomized, placebo-controlled cross-over trials, including patients with DM2 and preserved renal function ( n = 16), DM2 and CKD ( n = 17) and nondiabetic CKD ( n = 16). Participants were randomized to 4 weeks of empagliflozin 10 mg or placebo and crossed over after a 2-week washout. We measured brachial and central 24-h ambulatory blood pressure (ABP), pulse wave velocity (PWV), augmentation index (AIx@75), markers of nitric oxide and erythrocyte sodium sensitivity (ESS), a marker of endothelial glycocalyx function. RESULTS Empagliflozin reduced PWV [-0.16 m/s, 95% confidence interval (95% CI): -0.26; -0.06, P = 0.002], AIx@75 (-2.17%, 95% CI: -3.31; -1.02, P < 0.001) and brachial and central ABP in the combined study population ( n = 49). Changes in PWV and AIx@75 correlated to changes in systolic brachial ABP. Markers of nitric oxide did not increase, but empagliflozin decreased ESS, which was correlated to an increase in haematocrit. CONCLUSION Empagliflozin decreased arterial stiffness, mediated partly by a decrease in brachial ABP. We found no increase in nitric oxide activity, but ESS decreased. While this may be explained partly by a change in haematocrit, it could indicate an improvement in endothelial glycocalyx function. TRIAL REGISTRATION EU Clinical Trials Register 2019-004303-12, 2019-004447-80 and 2019-004467-50.
Collapse
Affiliation(s)
- Steffen F Nielsen
- University Clinic in Nephrology and Hypertension, Gødstrup Hospital, Herning
- Department of Clinical Medicine, Aarhus University
| | - Camilla L Duus
- University Clinic in Nephrology and Hypertension, Gødstrup Hospital, Herning
- Department of Clinical Medicine, Aarhus University
| | - Niels Henrik Buus
- Department of Clinical Medicine, Aarhus University
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Jesper N Bech
- University Clinic in Nephrology and Hypertension, Gødstrup Hospital, Herning
- Department of Clinical Medicine, Aarhus University
| | - Frank H Mose
- University Clinic in Nephrology and Hypertension, Gødstrup Hospital, Herning
- Department of Clinical Medicine, Aarhus University
| |
Collapse
|
3
|
Li H, Wen H, Liu J, Luo X, Pei B, Ge P, Sun Z, Liu J, Wang J, Chen H. The glycocalyx: a key target for treatment of severe acute pancreatitis-associated multiple organ dysfunction syndrome. Hum Cell 2025; 38:107. [PMID: 40411704 PMCID: PMC12103372 DOI: 10.1007/s13577-025-01227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/28/2025] [Indexed: 05/26/2025]
Abstract
The endothelial glycocalyx is a dynamic brush-like layer composed of proteoglycans and glycosaminoglycans, including heparan sulfate (HS) and hyaluronic acid (HA), and is an important regulator of vascular homeostasis. Its structure (thickness ranges from 20 to 6450 nm in different species) not only provides a charge-selective barrier but also serves to anchor mechanosensors such as the glypican-1 (GPC-1)/caveolin-1 (CAV-1) complex and buffers shear stress. In severe acute pancreatitis (SAP), inflammatory factors promote the expression of matrix metalloproteinases (MMPs) and heparinases, which degrade syndecan-1 (SDC-1) and HS, while oxidative stress disrupts HA-CD44 binding, leading to increased capillary leakage and neutrophil adhesion. This degradation process occurs before the onset of multiple organ dysfunction syndrome (MODS), highlighting the potential of the glycocalyx as an early biomarker. More importantly, the regeneration of glycocalyx through endothelial cell synthesis of glycosaminoglycans (GAGs) and shear stress-driven SDC recycling provides therapeutic prospects. This review redefines the pathophysiology of severe acute pancreatitis-associated multiple organ dysfunction (SAP-MODS) by exploring the glycocalyx's central mechanistic role and proposes stabilizing glycocalyx structure as a potential strategy to prevent microcirculatory failure.
Collapse
Affiliation(s)
- Huijuan Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
| | - Haiyun Wen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
| | - Jie Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
| | - Xinyu Luo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
| | - Boliang Pei
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
| | - Peng Ge
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
| | - Zhenxuan Sun
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
| | - Jin Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China
| | - Junjie Wang
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China.
| | - Hailong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China.
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China.
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China.
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, People's Republic of China.
| |
Collapse
|
4
|
Wang C, Fan M, Heo SC, Adams SM, Li T, Liu Y, Li Q, Loebel C, Burdick JA, Lu XL, Birk DE, Alisafaei F, Mauck RL, Han L. Structure, Mechanics, and Mechanobiology of Fibrocartilage Pericellular Matrix Mediated by Type V Collagen. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e14750. [PMID: 40407177 DOI: 10.1002/advs.202414750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 05/01/2025] [Indexed: 05/28/2025]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding cells in various tissues, regulating matrix turnover, cell-matrix interactions, and disease. This study elucidates the structure-mechanical properties and mechanobiology of the PCM in fibrocartilage, using the murine meniscus as the model. The fibrocartilage PCM is comprised of thin, randomly oriented collagen fibrils that entrap proteoglycans, contrasting with the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). Compared to the ECM, the PCM exhibits lower modulus and greater isotropy, but has similar relative viscoelastic properties. In Col5a1+/- menisci, the reduction of collagen V results in thicker, more heterogeneous collagen fibrils, reduced modulus, loss of isotropy and faster viscoelastic relaxation in the PCM. Such altered PCM leads to impaired matrix-to-cell strain transmission, and in turn, disrupts mechanotransduction of meniscal cells, as illustrated by reduced calcium signaling activities and alters expression of matrix genes. In vitro, Col5a1+/- cells produce a weakened PCM with inferior properties and reduced protection of cells against tensile stretch. These findings highlight the PCM as a distinctive microstructure in fibrocartilage mechanobiology, underscoring a pivotal role of collagen V in PCM function. Targeting the PCM or its constituents offers potential for improving meniscus regeneration, osteoarthritis intervention and broader fibrocartilage-related therapies.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Su Chin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sheila M Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Claudia Loebel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA, 19104, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| |
Collapse
|
5
|
Wadowski PP, Hülsmann M, Lang IM, Schörgenhofer C, Pultar J, Weikert C, Gremmel T, Steiner S, Koppensteiner R, Kopp CW, Jilma B. Glycocalyx Disintegration Is Associated with Mortality in Chronic Heart Failure. J Clin Med 2025; 14:3571. [PMID: 40429565 PMCID: PMC12112248 DOI: 10.3390/jcm14103571] [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: 04/02/2025] [Revised: 05/04/2025] [Accepted: 05/18/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Glycocalyx disintegration is associated with adverse outcomes in patients with trauma or sepsis. As microvascular dysfunction has an impact on disease progression in chronic heart failure (CHF) patients, we hypothesized that changes in microcirculation might be associated with mortality. Methods: Fifty patients with ischemic and non-ischemic cardiomyopathy and conservative treatment with baseline measurements of the sublingual microcirculation (via Sidestream Darkfield videomicroscopy) were followed up for two years. Glycocalyx thickness was assessed indirectly by calculation of the perfused boundary region (PBR). Results: Loss of glycocalyx was pronounced in non-survivors after one, n = 10, and two years, n = 16; PBR: 2.05 μm (1.88-2.15 μm) vs. 1.87 μm (1.66-2.03 μm) and 2.04 (1.93-2.11) vs. 1.84 (1.62-1.97); p = 0.042 and p = 0.003, respectively. Area under the ROC curve for the analysis of the predictive value of PBR on two-year mortality was 0.77 (p = 0.003; SE: 0.07, CI (95%): 0.63-0.91). ROC curve analysis determined a PBR of 1.9 μm as the best predictor for two-year mortality (sensitivity: 0.81; specificity: 0.59). Moreover, multivariate regression analysis revealed PBR and functional capillary density as significant predictors of two-year mortality, p = 0.036 and p = 0.048, respectively. Conclusions: Glycocalyx disintegration is related to poor overall survival in CHF patients.
Collapse
Affiliation(s)
- Patricia P. Wadowski
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
| | - Martin Hülsmann
- Department of Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (M.H.); (I.M.L.)
| | - Irene M. Lang
- Department of Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (M.H.); (I.M.L.)
| | - Christian Schörgenhofer
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (C.S.)
| | - Joseph Pultar
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
- Department of Anesthesia and Intensive Care Medicine, Universitätsklinikum St. Pölten, 3100 St. Pölten, Austria
| | - Constantin Weikert
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
| | - Thomas Gremmel
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
- Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria
- Institute of Cardiovascular Pharmacotherapy and Interventional Cardiology (Karl Landsteiner Society), 3100 St. Pölten, Austria
- Karl Landsteiner University of Health Sciences, 3500 Krems an der Donau, Austria
| | - Sabine Steiner
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
| | - Renate Koppensteiner
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
| | - Christoph W. Kopp
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (C.S.)
| |
Collapse
|
6
|
Sei Y, Kishino T, Shibasaki S, Harashima K, Sakata K, Ohnishi H, Watanabe T. Development of lower leg edema in the late afternoon may be associated with decreased blood flow in the leg in women in the luteal phase. Phlebology 2025:2683555251341395. [PMID: 40354192 DOI: 10.1177/02683555251341395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
ObjectivesLower leg edema is common in the late afternoon even in healthy individuals and could involve venous blood flows. Women appear more likely to develop lower leg edema, possibly due to the menstrual cycle. However, this phenomenon has not been quantitatively investigated using imaging. This study therefore used sonography to investigate sex-dependent impacts on physiological lower leg edema, in relation to venous blood flows in the legs and the menstrual cycle.MethodsParticipants comprised 54 healthy young adults (30 men, 24 women; mean age, 21 ± 1 years). Thickness of the papillary dermis and thickness and echogenicity of the subcutaneous adipose tissue were measured in the lower leg. Popliteal vein hemodynamics were investigated using Doppler sonography. Values were compared between late afternoon and morning. Each comparison was performed for men, women in the follicular, and women in the luteal phase.ResultsFor women in the luteal phase, papillary dermis and subcutaneous adipose tissue were thicker (median 0.20 mm, interquartile range [IQR] 0.12-0.25 mm vs median 0.33 mm, IQR 0.25-0.35 mm; p < .001 and median 5.0 mm, IQR 4.3-5.5 mm vs median 5.2 mm, IQR 4.5-6.2 mm; p = .013, respectively) and subcutaneous adipose tissue echogenicity was higher (median 66.9 IQR 64.1-70.5 vs median 71.7, IQR 65.0-76.7; p = .007) in the late afternoon than in the morning. The popliteal vein velocity-time integral (VTI) was lower in the late afternoon (median 33.0 cm, IQR 27.3-40.5 cm) than in the morning (median 26.1 cm, IQR 23.5-39.6 cm; p = .043). A significant negative correlation was observed between VTI and echogenicity (r = -0.549, p = .005). These findings were reduced in women in the follicular phase, and absent in men.ConclusionLate-afternoon lower leg edema may be associated with decreased leg blood flow in women, particularly in the luteal phase.
Collapse
Affiliation(s)
- Yoriko Sei
- Department of Medical Technology, Faculty of Health Sciences, Kyorin University, Tokyo, Japan
| | - Tomonori Kishino
- Department of Clinical Engineering, Faculty of Health Sciences, Kyorin University, Tokyo, Japan
| | - Shohei Shibasaki
- Department of Medical Technology, Faculty of Health Sciences, Kyorin University, Tokyo, Japan
| | - Keiichiro Harashima
- Department of Medical Technology, Faculty of Health Sciences, Kyorin University, Tokyo, Japan
| | - Konomi Sakata
- Department of Clinical Engineering, Faculty of Health Sciences, Kyorin University, Tokyo, Japan
| | - Hiroaki Ohnishi
- Department of Laboratory Medicine, Faculty of Medicine, Kyorin University, Tokyo, Japan
| | | |
Collapse
|
7
|
Byers CG. Crystalloids versus Colloids: Same Controversy, New Information. Vet Clin North Am Small Anim Pract 2025; 55:363-377. [PMID: 40082121 DOI: 10.1016/j.cvsm.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Intravenous fluid therapy is a regular component of care for cats and dogs with goals of restoring tissue perfusion, preventing organ dysfunction, and maintaining homeostasis. Veterinarians have a plethora of fluid choices, including hypertonic, hypotonic, and isotonic crystalloids, as well as synthetic and natural colloids. Studies published in both the human and veterinary medical literature have documented adverse events associated with synthetic and natural colloids. Reports of increased incidences of acute kidney injury, need for renal replacement therapy, and mortality in a variety of patient populations have called into question the use of synthetic colloids in humans and companion animals.
Collapse
|
8
|
Friedman J, Ghio M, Cotton-Betteridge A, Karim M, Packer J, Engelhardt D, Abdullah S, Shaheen F, Taghavi S, Duchesne J, Jackson-Weaver O. Infrarenal Aortic Occlusion Causes Endothelial Injury via Mitochondrial Reactive Oxygen Species. J Surg Res 2025; 309:139-145. [PMID: 40253934 DOI: 10.1016/j.jss.2025.03.023] [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: 10/29/2024] [Revised: 02/14/2025] [Accepted: 03/22/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION The endothelial glycocalyx is an anti-inflammatory, anti-coagulant coating of endothelial cells which participates in many physiologic processes. Damage to the glycocalyx occurs secondary to several disease states, and is especially pronounced in ischemia-reperfusion (I/R) injury. In models of trauma and hemorrhage, this damage has been shown to occur secondary to disordered succinate metabolism leading to increased mitochondrial reactive oxygen species (mitoROS). Aortic occlusion has also been associated with I/R injury to the vascular endothelium, though the precise mechanisms by which this occurs, and whether that injury includes glycocalyx shedding, are largely unknown. Consequently, very few therapeutic strategies for protection of the glycocalyx have been investigated. The aim of this study was to determine whether an experimental model of infrarenal aortic occlusion, with or without hemorrhage, causes a mitochondrial ROS-dependent glycocalyx shedding, and at what point this shedding predominantly occurs. We additionally aimed to determine whether plasma succinate was elevated following aortic occlusion. MATERIALS AND METHODS Male Sprague Dawley rats were anesthetized and laparotomy performed. A jugular catheter was placed and a baseline blood sample collected. In the hemorrhage group, 3 mL of blood was drawn from the jugular catheter. The infrarenal abdominal aorta was isolated and clamped with an atraumatic vascular clamp. Thirty minutes after clamping a second blood sample was collected, followed by unclamping. Blood samples were collected 2 and 15 min after reperfusion. Blood was centrifuged, and succinate and glycocalyx-component syndecan-1 were measured in the plasma using an ELISA. All rats in the treatment arm received identical hemorrhage, clamping, and blood sampling, but were treated with mitochondrial-targeted ROS scavenger mitoTEMPOL prior to clamping the aorta. After euthanizing the rats, calf muscle of the hemorrhage group was flash frozen, sectioned, and stained for glycocalyx with wheat germ agglutinin. RESULTS Plasma syndecan-1 levels were significantly (P < 0.05) elevated 2 mins after unclamping the aorta compared to baseline in both groups. Levels remained increased, though not statistically significantly, at 15 min after reperfusion. No significant increase in syndecan-1 was seen after 30 min of ischemia. MitoTEMPOL treatment prevented an increase in plasma syndecan-1. Correspondingly, glycocalyx staining intensity in mitoTEMPOL-treated rats was increased compared to control. Plasma succinate was significantly elevated at 15 min after reperfusion and was not affected by treatment with mitoTEMPOL. CONCLUSIONS To our knowledge this is the first study which demonstrates a successful therapeutic strategy in the treatment of endothelial glycocalyx shedding caused by aortic occlusion and hemorrhage in an animal model. Our study shows that glycocalyx shedding is attributable to mitochondrial ROS, and occurs following distal reperfusion rather than occlusion of the aorta or during hemorrhage. Consistent with findings in hemorrhagic shock, plasma succinate levels are elevated as a result of aortic occlusion and are upstream of ROS. Taken together, our findings suggest that in the setting of aortic occlusion, a therapeutic intervention could be performed prior to reperfusion which would substantially reduce injury to the vascular endothelium.
Collapse
Affiliation(s)
- Jessica Friedman
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana; Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Michael Ghio
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | | | - Mardeen Karim
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Jacob Packer
- Tulane University School of Medicine, New Orleans, Louisiana
| | - David Engelhardt
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Sarah Abdullah
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Farhana Shaheen
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Sharven Taghavi
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Juan Duchesne
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Olan Jackson-Weaver
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
9
|
Guo Y, Quirk K, Kelley DH, Thomas JH. Advection and diffusion in perivascular and extracellular spaces in the brain. J R Soc Interface 2025; 22:20250010. [PMID: 40393523 DOI: 10.1098/rsif.2025.0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/06/2025] [Accepted: 02/25/2025] [Indexed: 05/22/2025] Open
Abstract
Knowledge of the relative importance of advection and diffusion in clearing waste from the brain has been elusive, especially concerning the extracellular space (ECS). With local and global computational models of the mouse brain, we explore how the presence or absence of advection in the ECS affects solute transport. Without advection in the ECS, clearance would occur by diffusion into flowing cerebrospinal fluid in perivascular spaces (PVSs) or elsewhere, but we find this process to be severely limited by build-up of solute in the PVSs. We simulate flow in the ECS driven by a pressure drop between arteriole and venule PVSs, which enhances clearance considerably. To assess the relative importance of advection and diffusion, we introduce a local Péclet number [Formula: see text], a dimensionless scalar field. For our simulations, [Formula: see text] through much of the ECS but [Formula: see text] near PVSs near the brain surface. This local dominance of advection in the ECS establishes a clearance mechanism markedly different from that produced by diffusion alone. In network simulations that explore different parameter values and efflux routes, the pressures needed to drive the PVS flows measured in vivo are unrealistically large for most cases lacking ECS flow. Collectively, our models indicate that a flow in the ECS is necessary to explain experimental measurements and maintain homeostasis.
Collapse
Affiliation(s)
- Yisen Guo
- Mechanical Engineering, University of Rochester, Rochester, NY, USA
| | - Keelin Quirk
- Mechanical Engineering, University of Rochester, Rochester, NY, USA
| | - Douglas H Kelley
- Mechanical Engineering, University of Rochester, Rochester, NY, USA
| | - John H Thomas
- Mechanical Engineering, University of Rochester, Rochester, NY, USA
| |
Collapse
|
10
|
Yong J, Wang D, Kwok L, Mahmud SAZ, Hakobyan K, Lord MS, Mao G. Interfacial Interactions between Neural Tracing Lectin-Gold Nanoparticle Conjugate and Cell Membrane Glycoproteins. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:10161-10176. [PMID: 40237069 PMCID: PMC12044695 DOI: 10.1021/acs.langmuir.4c05034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025]
Abstract
The use of neural tracers as targeting molecules for drug delivery has been previously established as a novel and efficient method of neural drug delivery. The wheat germ agglutinin-horseradish peroxidase conjugate (WGAHRP) is a common neural tracer, which has been used extensively to decipher neural pathways in vertebrates. It has been widely reported to bind to cell surfaces and be transported in a retrograde fashion (from synapses toward the cell body) via dynein motors along microtubules within axons and transynaptically between neurons. Here we report on the differential binding between WGAHRP and gold-conjugated WGAHRP (AuNP-WGAHRP) to the glycoprotein profiles extracted from two neuronal cell lines and one skeletal muscle cell line, as well as the binding kinetics to heparin. From proteomic analysis of the extracted glycoproteins, we suggest the identity of cell surface glycoproteins involved in the retrograde transport of WGAHRP. This study illuminates the interfacial and molecular interactions of protein-gold conjugates with native ligands and opens the door for the identification of new targets for neural tracing and nervous system-related drug delivery.
Collapse
Affiliation(s)
- Joel Yong
- School
of Chemical Engineering, University of New
South Wales, Sydney 2052, Australia
| | - Dan Wang
- Graduate
School of Biomedical Engineering, University
of New South Wales, Sydney 2052, Australia
| | - Lachlan Kwok
- School
of Chemical Engineering, University of New
South Wales, Sydney 2052, Australia
| | - Sk Al Zaheri Mahmud
- School
of Chemical Engineering, University of New
South Wales, Sydney 2052, Australia
| | - Karen Hakobyan
- School
of Chemical Engineering, University of New
South Wales, Sydney 2052, Australia
| | - Megan S. Lord
- Graduate
School of Biomedical Engineering, University
of New South Wales, Sydney 2052, Australia
| | - Guangzhao Mao
- School
of Chemical Engineering, University of New
South Wales, Sydney 2052, Australia
- School
of Engineering, Institute for Materials and Processes, The University of Edinburgh, Robert Stevenson Road, Edinburgh, EH9 3FB, U.K.
| |
Collapse
|
11
|
Yap JQ, Nikouee A, Lau JE, Walsh G, Zang QS. Mitochondria at the Heart of Sepsis: Mechanisms, Metabolism, and Sex Differences. Int J Mol Sci 2025; 26:4211. [PMID: 40362448 PMCID: PMC12071423 DOI: 10.3390/ijms26094211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Sepsis is a life-threatening condition that occurs when the body is unable to effectively combat infection, leading to systemic inflammation and multi-organ failure. Interestingly, females exhibit lower sepsis incidence and improved clinical outcomes compared to males. However, the mechanisms underlying these sex-specific differences remain poorly understood. While sex hormones have been a primary focus, emerging evidence suggests that non-hormonal factors also play contributory roles. Despite sex differences in sepsis, clinical management is the same for both males and females, with treatment focused on combating infection using antibiotics and hemodynamic support through fluid therapy. However, even with these interventions, mortality remains high, highlighting the need for more effective and targeted therapeutic strategies. Sepsis-induced cardiomyopathy (SIC) is a key contributor to multi-organ failure and is characterized by left ventricular dilation and impaired cardiac contractility. In this review, we explore sex-specific differences in sepsis and SIC, with a particular focus on mitochondrial metabolism. Mitochondria generate the ATP required for cardiac function through fatty acid and glucose oxidation, and recent studies have revealed distinct metabolic profiles between males and females, which can further differ in the context of sepsis and SIC. Targeting these metabolic pathways could provide new avenues for sepsis treatment.
Collapse
Affiliation(s)
- John Q. Yap
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Azadeh Nikouee
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Jessie E. Lau
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Gabriella Walsh
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Qun Sophia Zang
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
- Cardiovascular Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| |
Collapse
|
12
|
Li HL, Liu N, Sosnowik S, Yung M, Gong H. Endothelial glycocalyx in different flow regions of the trabecular outflow pathway in bovine eyes. Front Cell Dev Biol 2025; 13:1569569. [PMID: 40352667 PMCID: PMC12061873 DOI: 10.3389/fcell.2025.1569569] [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: 02/01/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Background Glycocalyx is a hair-like structure covering the endothelium of the aqueous outflow pathway. While trabecular outflow is segmental circumferentially around the eye, regional differences in glycocalyx morphology remain largely unexplored. This study investigated glycocalyx variations in the different structures along the trabecular outflow pathway in high-flow (HF) and low-flow (LF) regions of bovine eyes. Methods Enucleated bovine eyes (n = 8) were perfused with fluorescein to identify HF and LF regions. The glycocalyx was labeled with Alcian Blue 8GX, and radial wedges from the anterior chamber angles of both HF and LF regions were processed for transmission electron microscopy. Glycocalyx thickness and coverage were quantified using ImageJ and compared between different outflow pathway locations in HF and LF regions. Glycocalyx measurements at intracellular (I-pores) and border pores (B-pores), the percentage of glycocalyx-unfilled pores, as well as the percentage of giant vacuoles (GVs) with and without I-pores with glycocalyx lining the inner membrane were evaluated. Results Glycocalyx thickness and coverage did not differ significantly between HF and LF regions. However, thickness progressively increased from the proximal (trabecular meshwork) to the distal (episcleral veins) outflow pathway. In both I-pores and B-pores, the glycocalyx was present near the basal opening, edge, and center of the pores, with thickness increasing toward the center. No significant differences in the percentage of glycocalyx-filled pores were observed between HF and LF regions. However, the percentage of GVs with I-pores exhibiting glycocalyx lining the inner cellular membrane was significantly higher (100%) than that of those without I-pores (16%). Conclusion No regional differences were found between HF and LF regions, but glycocalyx thickness progressively increased from the proximal to the distal outflow pathway, potentially reflecting varying shear stress conditions. The significantly higher percentage of GVs with I-pores containing glycocalyx lining the inner cellular membrane compared to those without I-pores suggests a relationship between aqueous outflow dynamics and glycocalyx synthesis. These findings provide a morphological basis for future research on glycocalyx alterations in glaucoma and their impact on outflow resistance.
Collapse
Affiliation(s)
- Hoi-Lam Li
- Ophthalmology, Boston University School of Medicine, Boston, MA, United States
| | - Neil Liu
- Ophthalmology, Boston University School of Medicine, Boston, MA, United States
| | - Shayna Sosnowik
- Ophthalmology, Boston University School of Medicine, Boston, MA, United States
- Department of Ophthalmology, MedVet, Columbus, Worthington, OH, United States
| | - Michelle Yung
- Ophthalmology, Boston University School of Medicine, Boston, MA, United States
| | - Haiyan Gong
- Ophthalmology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
13
|
Pavlidis G, Kountouri A, Katogiannis K, Thymis J, Nikolaou PE, Chania C, Karalis J, Kostelli G, Michalopoulou E, Katsanaki E, Parissis J, Vink H, Long R, Tsiodras S, Lambadiari V, Ikonomidis I. Effects of 4-month treatment with glycocalyx dietary supplement on endothelial glycocalyx and vascular function after COVID-19 infection. Eur J Clin Invest 2025:e70058. [PMID: 40270280 DOI: 10.1111/eci.70058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/11/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has been associated with impaired endothelial and vascular function. We investigated whether intervention with glycocalyx dietary supplement (GDS), containing glucosamine sulfate and fucoidan, improves endothelial glycocalyx and vascular function after COVID-19 infection. METHODS Fifty-seven convalescent patients 14 days after mild-to-moderate COVID-19 infection managed in an outpatient setting were randomized to receive GDS (n = 29) or placebo (n = 28) for 4 consecutive months. We measured at baseline and at 4 months: (a) perfused boundary region (PBR) of the sublingual microvessels with a diameter range of 4-25 μm, as a marker of endothelial glycocalyx integrity, (b) pulse wave velocity and augmentation index, (c) coronary flow reserve using Doppler echocardiography, and (d) malondialdehyde and protein carbonyls as oxidative stress markers. RESULTS Four months after treatment, patients who received GDS showed a greater reduction in PBR 4-25 μm (-6.8% vs. -1.3%), pulse wave velocity (-13.2% vs. -3%), augmentation index (-28.5% vs. -2.5%), malondialdehyde (-26% vs. -2.9%), protein carbonyls (-31.3% vs. -1%) and a greater increase in coronary flow reserve (12.9% vs. 1.6%) compared to placebo (p < .05). In the GDS group, the reduction in PBR 4-25 μm was associated with the corresponding decrease in pulse wave velocity (r = .31, p = .047), malondialdehyde, and protein carbonyls, as well as with the increase in coronary flow reserve (r = -.59, p = .008) at follow-up. Post-treatment, none of the patients under GDS reported post-COVID symptoms compared to 21.4% of the patients under placebo. CONCLUSION Four-month treatment with GDS may improve endothelial glycocalyx and vascular function after COVID-19 infection. CLINICAL TRIAL REGISTRATION URL: https://www. CLINICALTRIALS gov. Unique identifier: NCT05185934.
Collapse
Affiliation(s)
- George Pavlidis
- 2nd Department of Cardiology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Kountouri
- 2nd Propaedeutic Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Katogiannis
- 2nd Department of Cardiology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - John Thymis
- 2nd Department of Cardiology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Christina Chania
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - John Karalis
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Gabriella Kostelli
- 2nd Department of Cardiology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni Michalopoulou
- 2nd Department of Cardiology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni Katsanaki
- 2nd Department of Cardiology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - John Parissis
- University Department of Emergency Medicine, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Hans Vink
- GlycoCalyx Research Institute, Alpine, Utah, USA
| | - Robert Long
- GlycoCalyx Research Institute, Alpine, Utah, USA
| | - Sotirios Tsiodras
- 4th Department of Internal Medicine, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vaia Lambadiari
- 2nd Propaedeutic Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ignatios Ikonomidis
- 2nd Department of Cardiology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
14
|
Mitra R, Pentland K, Kolev S, Eden M, Levine E, Oakes JM, Ebong EE. Co-therapy with S1P and heparan sulfate derivatives to restore endothelial glycocalyx and combat pro-atherosclerotic endothelial dysfunction. Life Sci 2025; 377:123662. [PMID: 40280298 DOI: 10.1016/j.lfs.2025.123662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
AIMS Endothelial cell (EC) glycocalyx (GCX) shedding from disturbed blood flow and chemical factors leads to low-density lipoprotein infiltration, reduced nitric oxide synthesis, vascular dysfunction and atherosclerosis. This study evaluates a therapy combining sphingosine-1-phosphate (S1P) and heparin (heparan sulfate derivative). We hypothesized that heparin/S1P co-treatment repairs mechanically damaged EC GCX in disturbed flow (DF) regions and restores anti-atherosclerotic mechanotransduction to treat cardiovascular disease. MATERIALS AND METHODS We used a parallel-plate flow chamber to simulate flow conditions in vitro and a partial carotid ligation mouse model to mimic DF in vivo. Heparin and albumin-bound S1P were administered to assess their reparative effects on the endothelial GCX. Fluorescent staining, confocal microscopy, and ultrasound evaluated endothelial cell function and endothelial-dependent vascular function. Barrier functionality was assessed via macrophage uptake. Heparin/S1P mechanism-of-action insights were gained through fluid dynamics simulations and staining of GCX synthesis enzyme and S1P receptor. Statistical analyses validated the results. KEY FINDINGS The in vitro data showed that heparin/S1P therapy improves DF-conditioned ECs by restoring GCX and elevating vasodilator eNOS (endothelial-type nitric oxide synthase) expression. In vivo studies confirmed GCX degradation, vessel inflammation, hyperpermeability, and wall thickening in the mouse model's partially ligated left carotid artery. Heparin/S1P treatment restored GCX thickness and coverage, reduced inflammation and hyperpermeability, and inhibited vessel wall thickening. SIGNIFICANCE This work introduces a new approach to regenerating the EC GCX and restoring its function in ECs under DF conditions, offering a groundbreaking solution for preventing cardiovascular diseases like atherosclerosis.
Collapse
Affiliation(s)
- Ronodeep Mitra
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Kaleigh Pentland
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Svilen Kolev
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Matthew Eden
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Erel Levine
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Jessica M Oakes
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Eno E Ebong
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States; Department of Bioengineering, Northeastern University, Boston, MA, United States; Department of Neuroscience, Albert Einstein College of Medicine, NY, New York, United States.
| |
Collapse
|
15
|
Sirch M, Wörle D, Huber MG, Westerhausen C. Enhanced Adhesion of Mildly Positively Charged Vesicles to Endothelial Cells with Shed Glycocalyx. ACS OMEGA 2025; 10:14858-14865. [PMID: 40290995 PMCID: PMC12019467 DOI: 10.1021/acsomega.4c10054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 04/30/2025]
Abstract
The glycocalyx of endothelial cells is a dynamic, gel-like layer of glycoproteins, proteoglycans, and glycolipids that lines the luminal surface of blood vessels, playing a critical role in vascular permeability, mechanotransduction, and protection against shear stress. In this study, we investigated the in vitro adhesion of giant unilamellar vesicles (GUVs) composed of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC). Specifically, we examined mildly positively charged DOTAP-DMPC (20:80) GUVs, based on positively charged DOTAP and neutral DMPC but exhibiting an overall mild positive charge in physiological buffer, and neutral DMPC GUVs, which show a negative charge in physiological buffer. Adhesion to human umbilical vein endothelial cells (HUVEC) was studied under three culture conditions: dynamic (intact glycocalyx), static (underdeveloped glycocalyx), and glycocalyx-shed (degraded glycocalyx). Vesicles were produced via electroformation, stained with Texas Red dye, and perfused over endothelial cells at a controlled velocity to simulate slow blood flow. Adhesion was tracked using fluorescence microscopy combined with cell segmentation techniques. Adhesion of DOTAP-DMPC vesicles was significantly enhanced-by approximately 3.5-fold-on glycocalyx-shed cells compared to cells with an intact glycocalyx. In contrast, DMPC vesicles showed no adhesion under any condition. Analysis of vesicle size distributions revealed no significant differences between adherent and nonadherent vesicles or between DOTAP-DMPC and DMPC vesicles. These findings provide insights into the role of the endothelial glycocalyx in regulating adhesion, with potential implications for tumor cell interactions with the endothelium and mechanisms underlying DOTAP-based transfection.
Collapse
Affiliation(s)
- Manuel
M. Sirch
- Institute
of Theoretical Medicine, Physiology, University
of Augsburg, Augsburg 86159, Germany
- Institute
of Physics, University of Augsburg, Augsburg 86159, Germany
| | - David Wörle
- Institute
of Theoretical Medicine, Physiology, University
of Augsburg, Augsburg 86159, Germany
| | - Marina G. Huber
- Institute
of Theoretical Medicine, Physiology, University
of Augsburg, Augsburg 86159, Germany
| | - Christoph Westerhausen
- Institute
of Theoretical Medicine, Physiology, University
of Augsburg, Augsburg 86159, Germany
- Institute
of Physics, University of Augsburg, Augsburg 86159, Germany
- Center
for NanoScience (CeNS), Ludwig-Maximilians-Universität
Munich, Munich 80799, Germany
| |
Collapse
|
16
|
Bolger C, Pyne DB, McKune AJ. Concerns regarding interpretation in systematic review of exercise and endothelial glycocalyx. Eur J Clin Invest 2025:e70048. [PMID: 40237126 DOI: 10.1111/eci.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Affiliation(s)
- Craig Bolger
- University of Canberra Research Institute for Sport and Exercise, Bruce, Australian Capital Territory, Australia
- ACT Brumbies Rugby, Canberra, Australian Capital Territory, Australia
| | - David B Pyne
- University of Canberra Research Institute for Sport and Exercise, Bruce, Australian Capital Territory, Australia
| | - Andrew J McKune
- University of Canberra Research Institute for Sport and Exercise, Bruce, Australian Capital Territory, Australia
- School of Health Sciences, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
17
|
Piekoszewska-Ziętek P, Pańczyk-Tomaszewska M, Olczak-Kowalczyk D. Long-Term Impact of Interdisciplinary Collaboration on Oral Health in Children with Nephrotic Syndrome: A 12-Year Retrospective Study. J Clin Med 2025; 14:2696. [PMID: 40283525 PMCID: PMC12027630 DOI: 10.3390/jcm14082696] [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: 03/07/2025] [Revised: 03/27/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Objectives: Children with nephrotic syndrome (NS) are prone to oral health issues due to immunosuppression and systemic inflammation, which may exacerbate their renal condition. The objective of this study was to evaluate the impact of a 12-year interdisciplinary collaboration between pediatric dentists and nephrologists on oral health in children with NS. Methods: A retrospective analysis of 80 NS patients-40 assessed in 2012 and 40 in 2024-was conducted using caries indices (dmft/DMFT), Plaque Index, and Gingival Index. Statistical tests assessed differences between groups (p < 0.05). Results: The prevalence of active caries significantly decreased (50% vs. 78%; p = 0.011), with fewer decayed permanent teeth (0.96 ± 1.56 vs. 2.66 ± 2.51; p = 0.003) and improved oral hygiene (good hygiene in 52.5% vs. 30%; p = 0.041) in the 2024 group. Gingivitis was less severe compared to 2012. Conclusions: Long-term interdisciplinary care significantly improved oral health in children with NS. These improvements may contribute to reduced systemic inflammation and better overall disease control. Integrating dental care into NS management is recommended to support long-term outcomes.
Collapse
|
18
|
Abrashev H, Abrasheva D, Nikolov N, Ananiev J, Georgieva E. A Systematic Review of Endothelial Dysfunction in Chronic Venous Disease-Inflammation, Oxidative Stress, and Shear Stress. Int J Mol Sci 2025; 26:3660. [PMID: 40332237 PMCID: PMC12026777 DOI: 10.3390/ijms26083660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
Chronic venous disease (CVD) is among the most common diseases in industrialized countries and has a significant socioeconomic impact. The diversity of clinical symptoms and manifestations of CVD pose major challenges in routine diagnosis and treatment. Despite the high prevalence and the huge number of venous surgical interventions performed every day, a substantial proportion of the etiopathogenesis remains unclear. There are several widely advocated and generally valid theories of "peri-capillary fibrin cuffs" and "white cell trapping hypothesis", which consider the role of venous reflux/obstruction, inflammation, vascular remodeling, hemodynamic changes, genetic and social risk factors. There are several specific provoking factors for the development of venous reflux: incompetence of the valve system, inflammation of the vascular wall, and venous hypertension. Over the past few years, increasing scientific data has demonstrated the link between oxidative stress, endothelial dysfunction, and vascular inflammation. High levels of oxidants and persistent inflammation can cause cumulative changes in hemodynamics, resulting in permanent and irreversible damage to the microcirculation and endothelial cells. Production of reactive oxygen species and expression of inflammatory cytokines and adhesion molecules are involved in a vicious cycle of venous wall remodeling. The interaction of ROS, and in particular, the superoxide anion radical, with nitric oxide leads to a decrease in NO bioavailability, followed by the initiation of prolonged vasoconstriction and hypoxia and impairment of vascular tone. This review addresses the role of ED, oxidative, and hemodynamic stress in the CVD mediation. Based on predefined inclusion and exclusion criteria, we conducted a systematic review of published scientific articles using PubMed, PMC Europe, Scopus, WoS, MEDLINE, and Google Scholar databases in the interval from 24 April 2002 to 1 April 2025. The current review included studies (n = 197) scientific articles, including new reviews, updates, and grey literature, which were evaluated according to eligibility criteria. The selection process was performed using a standardized form according to PRISMA rules, the manual search of the databases, and a double-check to ensure transparent and complete reporting of reviews. Studies had to report quantitative assessments of the relationship between vascular endothelial dysfunction, inflammation, oxidative stress, and shear stress in a chronic venous disease.
Collapse
Affiliation(s)
- Hristo Abrashev
- Department of Vascular Surgery, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Despina Abrasheva
- II Department of Internal Medicine Therapy: Cardiology, Rheumatology, Hematology and Gastroenterology, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Nadelin Nikolov
- Vascular Surgery Department, National Heart Hospital, 1000 Sofia, Bulgaria;
| | - Julian Ananiev
- Department of General and Clinical Pathology, Forensic Medicine, Deontology and Dermatovenerology, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Ekaterina Georgieva
- Department of General and Clinical Pathology, Forensic Medicine, Deontology and Dermatovenerology, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria;
| |
Collapse
|
19
|
Risinger WB, Matheson PJ, Franklin ME, Lakshmanan J, Li Y, Harbrecht BG, Smith JW. Plasma resuscitation restores glomerular hyaluronic acid and mitigates hemorrhage-induced glomerular dysfunction. J Trauma Acute Care Surg 2025:01586154-990000000-00962. [PMID: 40205640 DOI: 10.1097/ta.0000000000004623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
BACKGROUND Acute renal dysfunction following hemorrhagic shock and resuscitation carries significant morbidity and mortality. While shock-induced shedding of the glycocalyx is well described within the pulmonary and splanchnic vasculature, less is known regarding early alterations to the glycocalyx of the renal microcirculation, particularly the glomerulus. We sought to evaluate the impact of hemorrhagic shock and resuscitation modalities on glomerular glycocalyx metabolism and function. We hypothesized that fresh frozen plasma resuscitation would attenuate glomerular glycocalyx shedding and reduce glomerular barrier dysfunction. METHODS Male Sprague-Dawley rats were subjected to 60 minutes of hemorrhagic shock to 40% of baseline mean arterial pressure, followed by resuscitation with shed whole blood and either lactated Ringer's or fresh frozen plasma. Experimental groups included the following: (a) baseline, (b) post-hemorrhagic shock, (c) post-lactated Ringer's resuscitation, and (d) post-plasma resuscitation. Enzyme-linked immunosorbent assays and immunohistochemistry were used to evaluate alterations of syndecan-1 and hyaluronic acid within the glomerular glycocalyx. Urine protein concentration was measured as a surrogate for glomerular function, and expression of cubilin and megalin was quantified to evaluate renal tubule protein reabsorptive capacity. RESULTS Despite evidence of systemic glycocalyx shedding, hemorrhagic shock and resuscitation did not alter glomerular synedcan-1 expression. However, shock induced shedding of hyaluronic acid from the glomerular glycocalyx. While hyaluronic acid breakdown was exacerbated by crystalloid resuscitation, plasma utilization restored levels back to baseline. Urine protein concentration drastically increased following hemorrhagic shock and resuscitation with lactated Ringer's. By contrast, plasma administration reduced urine protein levels back to normal. Renal cortex cubilin and megalin expression did not differ among the experimental groups, suggesting that alterations in urine protein were driven by changes in glomerular function. CONCLUSION Plasma-based resuscitation appears to reverse shock-induced shedding of glomerular hyaluronic acid and attenuates glomerular barrier dysfunction. Differential shedding of the glomerular glycocalyx may represent a novel pathway in acute kidney injury pathophysiology.
Collapse
Affiliation(s)
- William B Risinger
- From the Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | | | | | | | | | | | | |
Collapse
|
20
|
Larsen R, Kucharz K, Aydin S, Micael MKB, Choudhury B, Paulchakrabarti M, Lønstrup M, Lin DC, Abeln M, Münster-Kühnel A, Toledo AG, Lauritzen M, Esko JD, Daneman R. Multi-omic analysis reveals the unique glycan landscape of the blood-brain barrier glycocalyx. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.645297. [PMID: 40291667 PMCID: PMC12026896 DOI: 10.1101/2025.04.07.645297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The blood-brain barrier (BBB) glycocalyx is the dense layer of glycans and glycoconjugates that coats the luminal surface of the central nervous system (CNS) vasculature. Despite being the first point of contact between the blood and brain, not much is known about the BBB glycocalyx. Here, we performed a multi-omic investigation of the BBB glycocalyx which revealed a unique glycan landscape characterized by enrichment of sialic acid, chondroitin sulfate, and hyaluronan. We found that the BBB glycocalyx was thicker than glycocalyces in the peripheral vasculature and that hyaluronan was the major contributor to its ultrastructure. Using endothelial RNA sequencing, we found potential genetic determinants for these differences, including BBB enrichment of genes involved in sialic acid addition and peripheral enrichment of Tmem2 and Hyal2, the only known cell-surface hyaluronidases. Glycocalyx degradation and increases in vascular permeability are widely associated with inflammation. However, we found that the BBB glycocalyx remains largely unchanged in neuroinflammation during the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis and that its degradation is not sufficient to alter BBB permeability in health. Moreover, we showed that CNS endothelial sialic acid removal delays onset of EAE, indicating that BBB glycocalyx sialic acid may contribute to the progression of neuroinflammation. These findings underscore the unique and robust nature of the BBB glycocalyx and provide targets and tools for future studies into its role in health and neuroinflammation.
Collapse
|
21
|
Wang Y, Zhang Z, Qu X, Zhou G. Role of the endothelial cell glycocalyx in sepsis-induced acute kidney injury. Front Med (Lausanne) 2025; 12:1535673. [PMID: 40255592 PMCID: PMC12006053 DOI: 10.3389/fmed.2025.1535673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/25/2025] [Indexed: 04/22/2025] Open
Abstract
Sepsis-induced acute kidney injury (S-AKI) is a common complication of sepsis. It occurs at high incidence and is associated with a high level of mortality in the intensive care unit (ICU). The pathophysiologic mechanisms underlying S-AKI are complex, and include renal vascular endothelial cell dysfunction. The endothelial glycocalyx (EG) is a polysaccharide/protein complex located on the cell membrane at the luminal surface of vascular endothelial cells that has anti-inflammatory, anti-thrombotic, and endothelial protective effects. Recent studies have shown that glycocalyx damage plays a causal role in S-AKI progression. In this review, we first describe the structure, location, and basic function of the EG. Second, we analyze the underlying mechanisms of EG degradation in sepsis and S-AKI. Finally, we provide a summary of the potential therapeutic strategies that target the EG.
Collapse
Affiliation(s)
- Yixun Wang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Critical Care Medicine, Yichang Central People's Hospital, Yichang, China
- Yichang Sepsis Clinical Research Center, Yichang, Hubei, China
| | - Zhaohui Zhang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Critical Care Medicine, Yichang Central People's Hospital, Yichang, China
- Yichang Sepsis Clinical Research Center, Yichang, Hubei, China
| | - Xingguang Qu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Critical Care Medicine, Yichang Central People's Hospital, Yichang, China
- Yichang Sepsis Clinical Research Center, Yichang, Hubei, China
| | - Gaosheng Zhou
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Critical Care Medicine, Yichang Central People's Hospital, Yichang, China
- Yichang Sepsis Clinical Research Center, Yichang, Hubei, China
| |
Collapse
|
22
|
Ohmura K, Tomita H, Okada H, Nakayama N, Ohe N, Izumo T, Hara A. Visualizing the endothelial glycocalyx in human glioma vasculature. Brain Tumor Pathol 2025; 42:33-42. [PMID: 40035915 PMCID: PMC11993481 DOI: 10.1007/s10014-025-00498-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: 11/20/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025]
Abstract
Gliomas are the most common primary brain tumors in adults. However, glioblastoma is especially difficult to treat despite advancements in treatment. Therefore, new and more effective treatments are needed. The endothelial glycocalyx covers the luminal surface of the endothelium and plays an important role in vascular homeostasis. Tumor blood vessels normally have increased permeability, but some of them mimic normal cerebral blood vessels constituting the blood-brain barrier and retain drug-barrier function. Therefore, brain tumor vessels are considered to constitute the blood-tumor barrier. There are few reports on the endothelial glycocalyx in human brain tumor vessels. We aimed to visualize the endothelial glycocalyx in human brain tumor vessels and evaluate its microstructural differences in glioma vessels and normal capillaries. Surgical specimens from patients with glioma who underwent tumor resection at our institution were evaluated. We visualized the microstructures of the brain tumor vessels in human glioma specimens using electron microscopy with lanthanum nitrate. The endothelial glycocalyx was identified in the human glioma vasculature and its microstructure varied between the tumor margin and core. These variations may influence tumor angiogenesis and vascular remodeling, contributing to advancements in targeted therapies and diagnostics for human gliomas.
Collapse
Affiliation(s)
- Kazufumi Ohmura
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan.
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu, Japan.
| | - Hideshi Okada
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu, Japan.
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan.
| | - Noriyuki Nakayama
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Naoyuki Ohe
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tsuyoshi Izumo
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| |
Collapse
|
23
|
Iliakis CS, Crotta S, Wack A. The Interplay Between Innate Immunity and Nonimmune Cells in Lung Damage, Inflammation, and Repair. Annu Rev Immunol 2025; 43:395-422. [PMID: 40036704 DOI: 10.1146/annurev-immunol-082323-031852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
As the site of gas exchange, the lung is critical for organismal survival. It is also subject to continual environmental insults inflicted by pathogens, particles, and toxins. Sometimes, these insults result in structural damage and the initiation of an innate immune response. Operating in parallel, the immune response aims to eliminate the threat, while the repair process ensures continual physiological function of the lung. The inflammatory response and repair processes are thus inextricably linked in time and space but are often studied in isolation. Here, we review the interplay of innate immune cells and nonimmune cells during lung insult and repair. We highlight how cellular cross talk can fine-tune the circuitry of the immune response, how innate immune cells can facilitate or antagonize proper organ repair, and the prolonged changes to lung immunity and physiology that can result from acute immune responses and repair processes.
Collapse
Affiliation(s)
- Chrysante S Iliakis
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| | - Stefania Crotta
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| |
Collapse
|
24
|
Lu CH, Lee CE, Nakamoto ML, Cui B. Cellular Signaling at the Nano-Bio Interface: Spotlighting Membrane Curvature. Annu Rev Phys Chem 2025; 76:251-277. [PMID: 40258240 PMCID: PMC12043246 DOI: 10.1146/annurev-physchem-090722-021151] [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] [Indexed: 04/23/2025]
Abstract
No longer viewed as a passive consequence of cellular activities, membrane curvature-the physical shape of the cell membrane-is now recognized as an active constituent of biological processes. Nanoscale topographies on extracellular matrices or substrate surfaces impart well-defined membrane curvatures on the plasma membrane. This review examines biological events occurring at the nano-bio interface, the physical interface between the cell membrane and surface nanotopography, which activates intracellular signaling by recruiting curvature-sensing proteins. We encompass a wide range of biological processes at the nano-bio interface, including cell adhesion, endocytosis, glycocalyx redistribution, regulation of mechanosensitive ion channels, cell migration, and differentiation. Despite the diversity of processes, we call attention to the critical role of membrane curvature in each process. We particularly highlight studies that elucidate molecular mechanisms involving curvature-sensing proteins with the hope of providing comprehensive insights into this rapidly advancing area of research.
Collapse
Affiliation(s)
- Chih-Hao Lu
- Department of Chemistry, Stanford University, Stanford, California, USA;
- Wu-Tsai Neuroscience Institute and Sarafan ChEM-H Institute, Stanford University, Stanford, California, USA
| | - Christina E Lee
- Wu-Tsai Neuroscience Institute and Sarafan ChEM-H Institute, Stanford University, Stanford, California, USA
- Biophysics Program, Stanford University School of Medicine, Stanford, California, USA
| | - Melissa L Nakamoto
- Department of Chemistry, Stanford University, Stanford, California, USA;
- Wu-Tsai Neuroscience Institute and Sarafan ChEM-H Institute, Stanford University, Stanford, California, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, California, USA;
- Wu-Tsai Neuroscience Institute and Sarafan ChEM-H Institute, Stanford University, Stanford, California, USA
| |
Collapse
|
25
|
Çakir MU, Karduz G, Aksu U. Experimental and clinical perspectives on glycocalyx integrity and its relation to acute respiratory distress syndrome. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167745. [PMID: 39987847 DOI: 10.1016/j.bbadis.2025.167745] [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: 10/13/2024] [Revised: 02/02/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
The development of microcirculation imaging devices has significantly advanced our comprehension of the capillary environment's dynamics. Early research suggested that erythrocytes did not contact the vessel's inner surface due to the Fåhraeus effect, implying the presence of a covering on the endothelial cell surface. Subsequent electron microscopy studies revealed this layer to be a complex part of the vessel wall, now known as the endothelial glycocalyx (EG). The EG is a network of proteoglycans and glycoproteins bound to the endothelial membrane, incorporating soluble molecules from the endothelium and plasma. Over time, studies have elucidated the structure, function, and therapeutic targets of the glycocalyx, underscoring its pivotal role in vascular biology. The presence of cellular extensions of lung tissue cells in both vascular and nonvascular areas demonstrates the pivotal role of the glycocalyx in pulmonary vascular leak, surfactant dysfunction, impaired lung compliance and gas exchange abnormalities, which are hallmarks of acute respiratory distress syndrome (ARDS). It is of the utmost importance to elucidate the mechanisms underlying alveolocapillary glycocalyx degradation to develop efficacious treatments for ARDS, which has a mortality rate of 35 %. An understanding of the glycocalyx's role in vascular integrity provides a foundation for exploring new therapeutic avenues to mitigate lung injury and improve clinical outcomes in ARDS patients.
Collapse
Affiliation(s)
- Muzaffer Utku Çakir
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Türkiye
| | - Gülsüm Karduz
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Türkiye
| | - Ugur Aksu
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Türkiye.
| |
Collapse
|
26
|
Muendlein A, Leiherer A, Drexel H. Evaluation of circulating glypican 4 as a novel biomarker in disease - A comprehensive review. J Mol Med (Berl) 2025; 103:355-364. [PMID: 39961831 DOI: 10.1007/s00109-025-02520-5] [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: 10/03/2024] [Revised: 11/18/2024] [Accepted: 01/18/2025] [Indexed: 04/17/2025]
Abstract
Glypican 4 (GPC4), a member of the cell surface heparan sulfate proteoglycan family, plays a crucial role in regulating various cell signaling and developmental processes. Its ability to be released from the cell surface into the bloodstream through shedding makes it a promising blood-based biomarker in health and disease. In this context, circulating GPC4 has been initially proposed as an insulin-sensitizing adipokine being linked with various conditions of insulin resistance. In addition, serum levels of GPC4 can indicate glycocalyx shedding and associated pathophysiological states, such as systemic inflammation. Particularly in a morbid and elderly population, increased GPC4 concentrations may reflect general organ dysfunction and an advanced state of multimorbidity, showing a strong association with the prognosis of severe conditions such as heart failure or advanced cancer. This comprehensive review is the first to summarize the existing scientific knowledge on the role of circulating GPC4 as a novel diagnostic and prognostic biomarker across different pathologic conditions. We also discuss in detail the putative underlying pathophysiological mechanisms behind these findings.
Collapse
Affiliation(s)
- A Muendlein
- Vorarlberg Institute for Vascular Investigation & Treatment, (VIVIT), Feldkirch, Austria.
| | - A Leiherer
- Vorarlberg Institute for Vascular Investigation & Treatment, (VIVIT), Feldkirch, Austria
- Medical Central Laboratories, Feldkirch, Austria
- Private University in the Principality of Liechtenstein (UFL), Triesen, Principality of Liechtenstein
| | - H Drexel
- Vorarlberg Institute for Vascular Investigation & Treatment, (VIVIT), Feldkirch, Austria
- Private University in the Principality of Liechtenstein (UFL), Triesen, Principality of Liechtenstein
- Academic Teaching Hospital Feldkirch, Feldkirch, Austria
- Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
27
|
Ninham BW, Bunkin N, Battye M. The endothelial surface layer-glycocalyx - Universal nano-infrastructure is fundamental to physiology, cell traffic and a complementary neural network. Adv Colloid Interface Sci 2025; 338:103401. [PMID: 39862802 DOI: 10.1016/j.cis.2025.103401] [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: 07/18/2024] [Revised: 01/13/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
The glycocalyx and its associated endothelial surface layer which lines all cell membranes and most tissues, dwarfs the phospholipid membrane of cells in extent. Its major components are sulphated polymers like heparan and chondroitin sulphates and hyaluronic acid. These form a fuzzy layer of unknown structure and function. It has become increasingly clear that the ESL-GC complex must play many roles. We postulate it has a self-organised infrastructure that directs cell traffic, acts in defence against pathogens and other cells, and in diseases like diabetes, and heart disease, besides being a playground for a host of biochemical activity. Based on an analogous sulphated polymeric system Nafion, the fuel cell polymer, we suggest a model for the structure of the ESL-GC complex and how it functions. Taken together with parallel developments in physical chemistry, in nanobubbles, their stability in physiological media, and reactivity, we believe the model may throw light on a variety of phenomena, diabetes and some other diseases.
Collapse
Affiliation(s)
- Barry W Ninham
- Materials Physics (formerly Department of Applied Mathematics), Research School of Physics, Australian National University, Canberra, ACT 2600, Australia.
| | - Nikolai Bunkin
- Department of Fundamental Sciences, Bauman Moscow State Technical University, 2nd Baumanskaya Str. 5, 105005 Moscow, Russia
| | | |
Collapse
|
28
|
Wattchow NE, Pullen BJ, Indraratna AD, Nankivell V, Everest-Dass A, Psaltis PJ, Kolarich D, Nicholls SJ, Packer NH, Bursill CA. The emerging role of glycans and the importance of sialylation in cardiovascular disease. Atherosclerosis 2025; 403:119172. [PMID: 40138819 DOI: 10.1016/j.atherosclerosis.2025.119172] [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: 11/21/2024] [Revised: 02/13/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025]
Abstract
Glycosylation is the process by which glycans (i.e. 'sugars') are enzymatically attached to proteins or lipids to form glycoconjugates. Growing evidence points to glycosylation playing a central role in atherosclerosis. Glycosylation occurs in all human cells and post-translationally modifies many signalling molecules that regulate cardiovascular disease, affecting their binding and function. Glycoconjugates are present in abundance on the vascular endothelium and on circulating lipoproteins, both of which have well-established roles in atherosclerotic plaque development. Sialic acid is a major regulator of glycan function and therefore the process of sialylation, in which sialic acid is added to glycans, is likely to be entwined in any regulation of atherosclerosis. Glycans and sialylation regulators have the potential to present as new biomarkers that predict atherosclerotic disease or as targets for pharmacological intervention, as well as providing insights into novel cardiovascular mechanisms. Moreover, the asialoglycoprotein receptor 1 (ASGR1), a glycan receptor, is emerging as an exciting new regulator of lipid metabolism and coronary artery disease. This review summarises the latest advances in the growing body of evidence that supports an important role for glycosylation and sialylation in the regulation of atherosclerosis.
Collapse
Affiliation(s)
- Naomi E Wattchow
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, 5000, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5000, Australia
| | - Benjamin J Pullen
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, 5000, Australia; Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP), Australia
| | - Anuk D Indraratna
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland, 4222, Australia
| | - Victoria Nankivell
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, 5000, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5000, Australia; Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP), Australia
| | - Arun Everest-Dass
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP), Australia; Institute for Biomedicine and Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland, 4222, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, 5000, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5000, Australia; Department of Cardiology, Royal Adelaide Hospital, Adelaide, South Australia, 5000, Australia
| | - Daniel Kolarich
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP), Australia; Institute for Biomedicine and Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland, 4222, Australia
| | - Stephen J Nicholls
- Victorian Heart Institute, Monash University, Clayton, Victoria, 3168, Australia
| | - Nicolle H Packer
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5000, Australia; Institute for Biomedicine and Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland, 4222, Australia; School of Natural Sciences, Macquarie University, Macquarie Park, New South Wales, 2109, Australia; Australian Research Council (ARC) Centre of Excellence for Synthetic Biology, Australia
| | - Christina A Bursill
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, 5000, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5000, Australia; Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP), Australia.
| |
Collapse
|
29
|
Bridges CA, Fu L, Yeow J, Huang X, Jackson M, Kuchel RP, Sterling JD, Baker SM, Lord MS. The interplay between endothelial glycocalyx maturity and both the toxicity and intracellular uptake of charged nanoparticles. Acta Biomater 2025; 196:293-306. [PMID: 40058617 DOI: 10.1016/j.actbio.2025.03.012] [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: 10/29/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 04/05/2025]
Abstract
Nanoparticles are widely studied for delivering treatments to target tissues, but few have reached clinical use. Most nanoparticles encounter blood vessels on their way to target tissues. The inner surface of these vessels is lined with endothelial cells covered by a glycocalyx, an extracellular matrix rich in anionic glycans. The role of the glycocalyx in nanoparticle interactions is not well understood. Here, we demonstrate that endothelial cells need extended culture times to synthesize a mature glycocalyx. Our research shows that branched polyethyleneimine functionalized gold nanoparticles bind to endothelial cells expressing either a developing or mature glycocalyx, with the interaction involving hyaluronan and heparan sulfate. These nanoparticles are subsequently internalized. Similar results were seen with poly(L-arginine). A mature glycocalyx protects cells by reducing the toxicity of these cationic nanoparticles. In contrast, lipoic acid-functionalized gold nanoparticles are internalized by cells with a developing glycocalyx, but not a mature one. Poly(L-glutamic acid) only interacts with cells when major glycans in the glycocalyx are degraded. These findings highlight the complex relationship between nanoparticle charge and structure, and their effects on toxicity, binding, and uptake by endothelial cells. This offers important insights for improving nanoparticle interactions with blood vessels in health and disease. STATEMENT OF SIGNIFICANCE: Endothelial cells lining blood vessels form a barrier through which nanoparticles must cross to reach target tissues. These cells are covered with a layer called the glycocalyx, which is rich in anionic glycans. However, the role of the glycocalyx in how nanoparticles interact with cells remains underexplored. Our research revealed that cells with a mature glycocalyx internalize cationic nanoparticles and experience reduced cytotoxicity. Conversely, a mature glycocalyx prevents anionic nanoparticles from entering cells. These results suggest that the structure of both the nanoparticles and the glycocalyx should be considered in future studies to improve the use of nanoparticles for medical applications.
Collapse
Affiliation(s)
- Claire A Bridges
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Lu Fu
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jonathan Yeow
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Xiaojing Huang
- Molecular Surface Interaction Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Miriam Jackson
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Rhiannon P Kuchel
- Electron Microscope Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - James D Sterling
- College of Innovation, Entrepreneurship, and Economic Development, Missouri University of Science and Technology, Rolla, MO 65409, USA
| | | | - Megan S Lord
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
30
|
Yang L, Guan X, Cheng J, Ni L, Yao H, Gao Y, Zhu K, Shi X, Li B, Lin Y. VAMP8 as a biomarker and potential therapeutic target for endothelial cell dysfunction in atherosclerosis. Gene 2025; 942:149231. [PMID: 39800194 DOI: 10.1016/j.gene.2025.149231] [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: 05/30/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Endothelial cell dysfunction has a critical role in the pathophysiology of atherosclerosis. This study aims to uncover pivotal genes and pathways linked to endothelial cell dysfunction in atherosclerosis, as well as to ascertain the assumed causal effects and potential mechanisms. METHODS Datasets relevant to endothelial cell dysfunction in atherosclerosis were collected and divided into training and validation sets. Following differential analysis, we constructed a protein-protein interaction (PPI) network and a molecular interaction map of common-differentially expressed genes (co-DEGs) with proteins known to be involved in atherosclerotic endothelial cell dysfunction. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genome (KEGG), and Gene Set Enrichment Analysis (GSEA) were also conducted. Moreover, human umbilical vein endothelial cells (HUVECs) were cultured in circumstances characterized by elevated glucose levels to establish a cellular injury model simulating atherosclerotic conditions, and quantitative Polymerase Chain Reaction (qPCR) experiments were conducted to validate the differences of co-DEGs. Subsequently, the Summary-data-based Mendelian Randomization (SMR) method was employed. Additionally, we employed the Western Blot (WB) technique to validate the differential expression of VAMP8. Finally, we identified the differential expression of VAMP8 in the validation set and further validated its differential expression by collecting fresh blood samples from 20 patients with atherosclerosis and 20 healthy individuals. RESULTS 14 co-DEGs (FABP5, GULP1, COL4A5, VAMP8, FABP4, PFN2, ANGPT2, TFPI2, NUPR1, SULF1, FGF13, BASP1, EPB41L3, and PBK) were identified. SMR analysis confirmed 10 potential causal effect genes: PSRC1, VAMP8, FES, HNRNPUL1, CFDP1, SAP130, MDN1, OPRL1, UTP11, and HOXC4. The qPCR and WB experiments demonstrated that VAMP8 was significantly upregulated in the injured HUVECs group (p < 0.0001). Compared to the control group, VAMP8 was markedly increased in the blood samples of patients with atherosclerosis (p < 0.0001). CONCLUSIONS VAMP8 may potentially serve as a pathogenic gene in the process of endothelial dysfunction in atherosclerosis.
Collapse
Affiliation(s)
- Luqun Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xin Guan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Jiangwei Cheng
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Lin Ni
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Huijing Yao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yuping Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Kaiyi Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xiushan Shi
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Bingjie Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
31
|
Jakka SR, Mugesh G. Emerging Role of Noncovalent Interactions and Disulfide Bond Formation in the Cellular Uptake of Small Molecules and Proteins. Chem Asian J 2025; 20:e202401734. [PMID: 39831847 DOI: 10.1002/asia.202401734] [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: 11/30/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 01/22/2025]
Abstract
Intracellular delivery of proteins and small molecules is an important barrier in the development of strategies to deliver functional proteins and therapeutics into the cells to realize their full potential in biotechnology, biomedicine, cell-based therapies, and gene editing protein systems. Most of the intracellular protein delivery strategies involve the conjugation of cell penetrating peptides to enable the permeability of plasma membrane of mammalian cells to allow proteins to enter cytosol. The conjugations of small molecules such as (p-methylphenyl) glycine, pyrenebutyrate and cysteines are used for the same purpose. Molecular level interactions are governed mostly by ionic (cationic/anionic), covalent and noncovalent interactions with various molecular entities of glycocalyx matrix on plasma membrane lipid bilayer. Although the role of noncovalent interactions in cellular uptake is not fully understood, several recent advances have focused on the noncovalent interaction-based strategies of intracellular delivery of small molecules and proteins into mammalian cells. These are achieved by simple modification of protein surfaces with chemical moieties which can form noncovalent interactions other than hydrogen bonding. In this review, we describe the recent advances and the mechanistic aspects of intracellular delivery and role of noncovalent interactions in the cellular uptake of proteins and small molecules.
Collapse
Affiliation(s)
- Surendar R Jakka
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560 012, India
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560 012, India
| |
Collapse
|
32
|
Xu XX, Tian Y, Pu Y, Che B, Luo H, Liu Y, Liu YJ, Jing G. Bacterial Swimming and Accumulation on Endothelial Cell Surfaces. J Phys Chem B 2025; 129:2647-2655. [PMID: 39983743 DOI: 10.1021/acs.jpcb.4c08666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Abstract
Flagellar-driven locomotion plays a critical role in the bacterial attachment and colonization of surfaces, contributing to the risks of contamination and infection. Extensive efforts to uncover the underlying principles governing bacterial motility near surfaces have relied on idealized assumptions about surrounding artificial surfaces. However, in the context of living systems, the role of cells from tissues and organs becomes increasingly critical, particularly in bacterial swimming and adhesion, yet it remains poorly understood. Here, we propose using biological surfaces composed of vascular endothelial cells to experimentally investigate bacterial motion and interaction behaviors. Our results reveal that bacterial trapping observed on inorganic surfaces is counteractively manifested with reduced radii of circular motion on cellular surfaces. Additionally, two distinct modes of bacterial adhesion were identified: tight and loose adhesion. Interestingly, the presence of living cells enhances bacterial surface enrichment, and imposed flow intensifies this accumulation via a bias-swimming effect. These results surprisingly indicate that physical effects remain the dominant factor regulating bacterial motility and accumulation at the single-cell-layer level in vitro, bridging the gap between simplified hydrodynamic mechanisms and complex biological surfaces with relevance to biofilm formation and bacterial contamination.
Collapse
Affiliation(s)
- Xin-Xin Xu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yangguang Tian
- School of Physics, Northwest University, Xi'an 710127, China
| | - Yuhe Pu
- School of Physics, Northwest University, Xi'an 710127, China
| | - Bingchen Che
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Hao Luo
- School of Physics, Northwest University, Xi'an 710127, China
| | - Yanan Liu
- School of Physics, Northwest University, Xi'an 710127, China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Guangyin Jing
- School of Physics, Northwest University, Xi'an 710127, China
| |
Collapse
|
33
|
Ahn SJ, Goya B, Bertomo C, Sciortino R, Racchumi G, Bonilla LG, Anrather J, Iadecola C, Faraco G. Neutrophil stalling does not mediate the increase in tau phosphorylation and the cognitive impairment associated with high salt diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640593. [PMID: 40093148 PMCID: PMC11908121 DOI: 10.1101/2025.02.27.640593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
High dietary salt intake has powerful effects on cerebral blood vessels and has emerged as a risk factor for stroke and cognitive impairment. In mice, high salt diet (HSD) leads to reduced cerebral blood flow (CBF), tau hyperphosphorylation and cognitive dysfunction. However, it is still unclear whether the reduced CBF is responsible for the effects of HSD on tau and cognition. Capillary stalling has emerged as a cause of CBF reduction and cognitive impairment in models of Alzheimer's disease and diabetes. Therefore, we tested the hypothesis that capillary stalling also contributes to the CBF reduction and cognitive impairment in HSD. Using two-photon imaging, we found that HSD increased stalling of neutrophils in brain capillaries and decreased CBF. Neutrophil depletion reduced the number of stalled capillaries and restored resting CBF but did not prevent tau phosphorylation or cognitive impairment. These novel findings show that, capillary stalling contribute to CBF reduction in HSD, but not to tau phosphorylation and cognitive deficits. Therefore, the hypoperfusion caused by capillary stalling is not the main driver of the tau phosphorylation and cognitive impairment.
Collapse
|
34
|
Soliman Y, Al-Khodor J, Yildirim Köken G, Mustafaoglu N. A guide for blood-brain barrier models. FEBS Lett 2025; 599:599-644. [PMID: 39533665 DOI: 10.1002/1873-3468.15053] [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: 05/13/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Understanding the intricate mechanisms underlying brain-related diseases hinges on unraveling the pivotal role of the blood-brain barrier (BBB), an essential dynamic interface crucial for maintaining brain equilibrium. This review offers a comprehensive analysis of BBB physiology, delving into its cellular and molecular components while exploring a wide range of in vivo and in vitro BBB models. Notably, recent advancements in 3D cell culture techniques are explicitly discussed, as they have significantly improved the fidelity of BBB modeling by enabling the replication of physiologically relevant environments under flow conditions. Special attention is given to the cellular aspects of in vitro BBB models, alongside discussions on advances in stem cell technologies, providing valuable insights into generating robust cellular systems for BBB modeling. The diverse array of cell types used in BBB modeling, depending on their sources, is meticulously examined in this comprehensive review, scrutinizing their respective derivation protocols and implications. By synthesizing diverse approaches, this review sheds light on the improvements of BBB models to capture physiological conditions, aiding in understanding BBB interactions in health and disease conditions to foster clinical developments.
Collapse
Affiliation(s)
- Yomna Soliman
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Faculty of Pharmacy, Mansoura University, Egypt
| | - Jana Al-Khodor
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
| | | | - Nur Mustafaoglu
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Sabancı University Nanotechnology Research and Application Center, Istanbul, Turkey
| |
Collapse
|
35
|
Shi SM, Suh RJ, Shon DJ, Garcia FJ, Buff JK, Atkins M, Li L, Lu N, Sun B, Luo J, To NS, Cheung TH, McNerney MW, Heiman M, Bertozzi CR, Wyss-Coray T. Glycocalyx dysregulation impairs blood-brain barrier in ageing and disease. Nature 2025; 639:985-994. [PMID: 40011765 PMCID: PMC11946907 DOI: 10.1038/s41586-025-08589-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/03/2025] [Indexed: 02/28/2025]
Abstract
The blood-brain barrier (BBB) is highly specialized to protect the brain from harmful circulating factors in the blood and maintain brain homeostasis1,2. The brain endothelial glycocalyx layer, a carbohydrate-rich meshwork composed primarily of proteoglycans, glycoproteins and glycolipids that coats the BBB lumen, is a key structural component of the BBB3,4. This layer forms the first interface between the blood and brain vasculature, yet little is known about its composition and roles in supporting BBB function in homeostatic and diseased states. Here we find that the brain endothelial glycocalyx is highly dysregulated during ageing and neurodegenerative disease. We identify significant perturbation in an underexplored class of densely O-glycosylated proteins known as mucin-domain glycoproteins. We demonstrate that ageing- and disease-associated aberrations in brain endothelial mucin-domain glycoproteins lead to dysregulated BBB function and, in severe cases, brain haemorrhaging in mice. Finally, we demonstrate that we can improve BBB function and reduce neuroinflammation and cognitive deficits in aged mice by restoring core 1 mucin-type O-glycans to the brain endothelium using adeno-associated viruses. Cumulatively, our findings provide a detailed compositional and structural mapping of the ageing brain endothelial glycocalyx layer and reveal important consequences of ageing- and disease-associated glycocalyx dysregulation on BBB integrity and brain health.
Collapse
Affiliation(s)
- Sophia M Shi
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ryan J Suh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - D Judy Shon
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Francisco J Garcia
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Josephine K Buff
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Micaiah Atkins
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
| | - Lulin Li
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Nannan Lu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Bryan Sun
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Ning-Sum To
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Tom H Cheung
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Daniel and Mayce Yu Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - M Windy McNerney
- Department of Psychiatry, Stanford University School of Medicine, Stanford, CA, USA
- MIRECC, Department of Veterans Affairs, Palo Alto, CA, USA
| | - Myriam Heiman
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| | - Tony Wyss-Coray
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA.
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA.
| |
Collapse
|
36
|
Lai SK, Lee ZQ, Tan TI, Tan BH, Sugrue RJ. Evidence that the cell glycocalyx envelops respiratory syncytial virus (RSV) particles that form on the surface of RSV-infected human airway cells. Virology 2025; 604:110415. [PMID: 40044247 DOI: 10.1016/j.virol.2025.110415] [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: 10/03/2024] [Revised: 12/29/2024] [Accepted: 01/16/2025] [Indexed: 05/11/2025]
Abstract
We examined how respiratory syncytial virus (RSV) particles circumvent the overlying glycocalyx on virus-infected A549 cells. The glycocalyx was detected using the lectin WGA-AL488 probe, and the antibodies anti-HS and anti-syndecan-4 that detect heparin sulphate (HS) and the syndecan-4 protein (SYND4) respectively. Imaging of RSV-infected cells provided evidence that the glycocalyx envelopes the virus filaments as they form, and that components of the glycocalyx such as HS moieties and SYND4 are displayed on the surface of the mature virus filaments. Recombinant expression of the G protein in these cells suggested that the G protein was trafficked into pre-existing filamentous cellular structures with a well-defined glycocalyx, further suggesting that the glycocalyx is maintained at the site of virus particle assembly. These data provide evidence that during RSV particle assembly the virus filaments become enveloped by the glycocalyx, and that the glycocalyx should be considered as a structural component of virus filaments.
Collapse
Affiliation(s)
- Soak Kuan Lai
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Zhi Qi Lee
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Trina Isabel Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Boon Huan Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Republic of Singapore
| | - Richard J Sugrue
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore.
| |
Collapse
|
37
|
Andrijauskas A, Andrijauskas P, Jovaišienė I, Valaika A, Jovaisa T, Urbonas K, Činčikas D, Svediene S, Scupakova N, Puodziukaite L, Budra M, Kalinauskas G, Stankevičius E. Ask a Doctor a Question: A Clinician's Message to the Industry. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:368. [PMID: 40142179 PMCID: PMC11944065 DOI: 10.3390/medicina61030368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 03/28/2025]
Abstract
The medical industry is an integral part of the delivery of healthcare. Collaboration between academic institutions, healthcare providers, and the industry are necessary but not devoid of flaws. This expert opinion article calls for closer attention to be paid by the medical industry to "what a frontline clinician needs" rather than relying solely on experts' opinions and stake holders' requests in planning future products and features. The need for the monitoring of tissue fluid accumulation is discussed from the point of view of practicing anaesthesiology and intensive care specialists in the context of the potential missed opportunity to have it be already available.
Collapse
Affiliation(s)
- Audrius Andrijauskas
- Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (A.A.); (I.J.); (T.J.); (D.Č.); (S.S.); (L.P.)
| | - Povilas Andrijauskas
- II Department of Anaesthesiology and Intensive Care, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania; (K.U.); (N.S.)
| | - Ieva Jovaišienė
- Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (A.A.); (I.J.); (T.J.); (D.Č.); (S.S.); (L.P.)
| | - Arūnas Valaika
- Clinic of Cardiac and Vascular Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (A.V.); (M.B.); (G.K.)
| | - Tomas Jovaisa
- Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (A.A.); (I.J.); (T.J.); (D.Č.); (S.S.); (L.P.)
| | - Karolis Urbonas
- II Department of Anaesthesiology and Intensive Care, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania; (K.U.); (N.S.)
| | - Darius Činčikas
- Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (A.A.); (I.J.); (T.J.); (D.Č.); (S.S.); (L.P.)
| | - Saule Svediene
- Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (A.A.); (I.J.); (T.J.); (D.Č.); (S.S.); (L.P.)
| | - Nadezda Scupakova
- II Department of Anaesthesiology and Intensive Care, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania; (K.U.); (N.S.)
| | - Lina Puodziukaite
- Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (A.A.); (I.J.); (T.J.); (D.Č.); (S.S.); (L.P.)
| | - Mindaugas Budra
- Clinic of Cardiac and Vascular Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (A.V.); (M.B.); (G.K.)
| | - Gintaras Kalinauskas
- Clinic of Cardiac and Vascular Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania; (A.V.); (M.B.); (G.K.)
| | - Edgaras Stankevičius
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania;
| |
Collapse
|
38
|
Famá EAB, Pinhal MAS. Extracellular matrix components in preeclampsia. Clin Chim Acta 2025; 568:120132. [PMID: 39798685 DOI: 10.1016/j.cca.2025.120132] [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: 11/11/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Preeclampsia (PE) is a gestational complication affecting 5% to 10% of all pregnancies. PE is characterized by hypertension and endothelial dysfunction, whose etiology involves, among other factors, alterations in the extracellular matrix (ECM) that can compromise vascular remodeling and trophoblast invasion, ie, processes essential for placental development. Endothelial dysfunction is caused by release of antiangiogenic factors, mainly a soluble fms-like tyrosine kinase-1 (sFlt-1), which antagonizes two endothelial angiogenic factors, the vascular endothelial growth factor (VEGF) and placental growth factor (PLGF). This angiogenic imbalance contributes to clinical symptoms including hypertension and multisystem dysfunction. This review aims to summarize recent advances in understanding PE, particularly with altered ECM components such as heparan sulfate proteoglycans, the glycosidase heparanase, fibronectin, collagen XVIII (endostatin), and metalloproteases. This comprehensive narrative review was conducted on PubMed from 1994 to 2024, focusing on articles on the pathophysiology of PE, particularly endothelial dysfunction caused by ECM modifications. The data shows a reduced expression of matrix metalloproteinases, increased collagen fragment XVIII, and significant changes in fibronectin associated with PE. Furthermore, endothelial dysfunction was associated with increased degradation of heparan sulfate chains from proteoglycans and increased sFlt-1. Understanding these ECM modifications is crucial for developing potential new therapeutic interventions that improve maternal and fetal outcome in PE.
Collapse
Affiliation(s)
- Eduardo Augusto Brosco Famá
- Obstetrics/Gynecology Department, Centro Universitário Faculdade de Medicina ABC (FMABC), Santo André, São Paulo, Brazil.
| | | |
Collapse
|
39
|
Hsieh LTH, Hall BS, Newcombe J, Mendum TA, Varela SS, Umrania Y, Deery MJ, Shi WQ, Diaz-Delgado J, Salguero FJ, Simmonds RE. The Mycobacterium ulcerans toxin mycolactone causes destructive Sec61-dependent loss of the endothelial glycocalyx and vessel basement membrane to drive skin necrosis. eLife 2025; 12:RP86931. [PMID: 39913180 PMCID: PMC11801798 DOI: 10.7554/elife.86931] [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] [Indexed: 02/07/2025] Open
Abstract
The drivers of tissue necrosis in Mycobacterium ulcerans infection (Buruli ulcer disease) have historically been ascribed solely to the directly cytotoxic action of the diffusible exotoxin, mycolactone. However, its role in the clinically evident vascular component of disease aetiology remains poorly explained. We have now dissected mycolactone's effects on human primary vascular endothelial cells in vitro. We show that mycolactone-induced changes in endothelial morphology, adhesion, migration, and permeability are dependent on its action at the Sec61 translocon. Unbiased quantitative proteomics identified a profound effect on proteoglycans, driven by rapid loss of type II transmembrane proteins of the Golgi, including enzymes required for glycosaminoglycan (GAG) synthesis, combined with a reduction in the core proteins themselves. Loss of the glycocalyx is likely to be of particular mechanistic importance, since knockdown of galactosyltransferase II (beta-1,3-galactotransferase 6; B3GALT6), the GAG linker-building enzyme, phenocopied the permeability and phenotypic changes induced by mycolactone. Additionally, mycolactone depleted many secreted basement membrane components and microvascular basement membranes were disrupted in vivo during M. ulcerans infection in the mouse model. Remarkably, exogenous addition of laminin-511 reduced endothelial cell rounding, restored cell attachment and reversed the defective migration caused by mycolactone. Hence supplementing mycolactone-depleted extracellular matrix may be a future therapeutic avenue, to improve wound healing rates.
Collapse
Affiliation(s)
- Louise Tzung-Harn Hsieh
- Discipline of Microbes, Infection & Immunity, School of Biosciences, Faculty of Health and Medical Sciences, University of SurreyGuildfordUnited Kingdom
| | - Belinda S Hall
- Discipline of Microbes, Infection & Immunity, School of Biosciences, Faculty of Health and Medical Sciences, University of SurreyGuildfordUnited Kingdom
| | - Jane Newcombe
- Discipline of Microbes, Infection & Immunity, School of Biosciences, Faculty of Health and Medical Sciences, University of SurreyGuildfordUnited Kingdom
| | - Tom A Mendum
- Discipline of Microbes, Infection & Immunity, School of Biosciences, Faculty of Health and Medical Sciences, University of SurreyGuildfordUnited Kingdom
| | - Sonia Santana Varela
- Discipline of Microbes, Infection & Immunity, School of Biosciences, Faculty of Health and Medical Sciences, University of SurreyGuildfordUnited Kingdom
| | - Yagnesh Umrania
- Cambridge Centre for Proteomics, University of CambridgeCambridgeUnited Kingdom
| | - Michael J Deery
- Cambridge Centre for Proteomics, University of CambridgeCambridgeUnited Kingdom
| | - Wei Q Shi
- Department of Chemistry, Ball State UniversityMuncieUnited States
| | - Josué Diaz-Delgado
- Texas A&M Veterinary Medical Diagnostic LaboratoryCollege StationUnited States
| | | | - Rachel E Simmonds
- Discipline of Microbes, Infection & Immunity, School of Biosciences, Faculty of Health and Medical Sciences, University of SurreyGuildfordUnited Kingdom
| |
Collapse
|
40
|
Erning K, Wilson KL, Smith CS, Nguyen L, Joesph NI, Irengo R, Cao LY, Cumaran M, Shi Y, Lyu S, Riley L, Dunn TW, Carmichael ST, Segura T. Clustered VEGF Nanoparticles in Microporous Annealed Particle (MAP) Hydrogel Accelerates Functional Recovery and Brain Tissue Repair after Stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635733. [PMID: 39974959 PMCID: PMC11838428 DOI: 10.1101/2025.01.30.635733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Ischemic stroke, a blockage in the vasculature of the brain that results in insufficient blood flow, is one of the world's leading causes of disability. The cascade of inflammation and cell death that occurs immediately following stroke drives vascular and functional loss that does not fully recover over time, and no FDA-approved therapies exist that stimulate regeneration post-stroke. We have previously developed a hydrogel scaffold that delivered heparin nanoparticles with and without VEGF bound to their surface to promote angiogenesis and reduce inflammation, respectively. However, the inclusion of the naked heparin nanoparticles warranted concern over the development of bleeding complications. Here, we explore how microporous annealed particle (MAP) scaffolds functionalized with VEGF coated heparin nanoparticles can both reduce inflammation and promote angiogenesis - without the inclusion of free heparin nanoparticles. We show that our updated design not only successfully promotes de novo tissue formation, including the development of mature vessels and neurite sprouting, but it also leads to functional improvement in a photothrombotic stroke model. In addition, we find increased astrocyte infiltration into the infarct site correlated with mature vessel formation. This work demonstrates how our biomaterial design can enhance endogenous regeneration post-stroke while eliminating the need for excess heparin.
Collapse
|
41
|
Ferreira G, Cardozo R, Chavarria L, Santander A, Sobrevia L, Chang W, Gundersen G, Nicolson GL. The LINC complex in blood vessels: from physiology to pathological implications in arterioles. J Physiol 2025. [PMID: 39898417 DOI: 10.1113/jp285906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
The LINC (linker of nucleoskeleton and cytoskeleton) complex is a critical component of the cellular architecture that bridges the nucleoskeleton and cytoskeleton and mediates mechanotransduction to and from the nucleus. Though it plays important roles in all blood vessels, it is in arterioles that this complex plays a pivotal role in maintaining endothelial cell integrity, regulating vascular tone, forming new microvessels and modulating responses to mechanical and biochemical stimuli. It is also important in vascular smooth muscle cells and fibroblasts, where it possibly plays a role in the contractile to secretory phenotypic transformation during atherosclerosis and vascular ageing, and in fibroblasts' migration and inflammatory responses in the adventitia. Physiologically, the LINC complex contributes to the stability of arteriolar structure, adaptations to changes in blood flow and injury repair mechanisms. Pathologically, dysregulation or mutations in LINC complex components can lead to compromised endothelial function, vascular remodelling and exacerbation of cardiovascular diseases such as atherosclerosis (arteriolosclerosis). This review summarizes our current understanding of the roles of the LINC complex in cells from arterioles, highlighting its most important physiological functions, exploring its implications for vascular pathology and emphasizing some of its functional characteristics in endothelial cells. By elucidating the LINC complex's role in health and disease, we aim to provide insights that could improve future therapeutic strategies targeting LINC complex-related vascular disorders.
Collapse
Affiliation(s)
- Gonzalo Ferreira
- Department of Biophysics, Faculty of Medicine, Universidad de La República, Montevideo, Uruguay
| | - Romina Cardozo
- Department of Biophysics, Faculty of Medicine, Universidad de La República, Montevideo, Uruguay
| | - Luisina Chavarria
- Department of Biophysics, Faculty of Medicine, Universidad de La República, Montevideo, Uruguay
| | - Axel Santander
- Department of Biophysics, Faculty of Medicine, Universidad de La República, Montevideo, Uruguay
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville, Spain
- Medical School (Faculty of Medicine), Sao Paulo State University (UNESP), Sao Paulo, Brazil
- Faculty of Medicine and Biomedical Sciences, University of Queensland Centre for Clinical Research (UQCCR), University of Queensland, QLD, Herston, Queensland, Australia
- Tecnologico de Monterrey, Eutra, The Institute for Obesity Research (IOR), School of Medicine and Health Sciences, Monterrey, Nuevo León, Mexico
| | - Wakam Chang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Gregg Gundersen
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Garth L Nicolson
- Department of Molecular Pathology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| |
Collapse
|
42
|
Du Y, Han B, Biere K, Abdelmalek N, Shu X, Song C, Chen G, Li N, Tuschen M, Wu H, Sun S, Choukér A, Long M, Moser D. Lunar and Martian gravity alter immune cell interactions with endothelia in parabolic flight. NPJ Microgravity 2025; 11:4. [PMID: 39900929 PMCID: PMC11791073 DOI: 10.1038/s41526-024-00456-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 12/28/2024] [Indexed: 02/05/2025] Open
Abstract
Returning to the moon and traveling to Mars represent the main targets of human space exploration missions within the upcoming decades. Comparable to microgravity, partial gravity in these destinations is assumed to dysregulate immune functions, thereby threatening astronauts´ health. To investigate the impact of partial gravity on immune cell attachment to vessel endothelia, THP-1 cells and HUVEC cell layers were monitored in a flow chamber system during parabolic flight in lunar (0.16 g) or Martian (0.38 g) gravity. Focus was set on floating speed, cell adhesion, surface molecule expression and cytoskeletal reorganization under basal and TNF-induced inflammatory environment. Floating speed of THP-1 cells was increased in partial gravity, which was accompanied by a successively lower adhesion to the endothelial HUVEC cells. Expression levels of the adhesion markers Mac-1 on THP-1 cells as well as ICAM-1 on HUVECs were found elevated in lunar and Martian gravity, which was aggravated by TNF. Analysis of cytoskeletal organization in HUVECs revealed reduced intracellular F-actin microfilament networks and a stronger cell directionality with stress fiber alignment at cell borders in partial gravity, which was intensified by TNF. In summary, altered immune cell - endothelium interactions as quantified in partial gravity conditions show similarities to cellular behavior in microgravity. However, the different magnitudes of effects in dependence of gravitational level still need to be assessed in further investigations.
Collapse
Affiliation(s)
- Yu Du
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, 100190, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Bing Han
- Laboratory of Translational Research 'Stress and Immunity', Department of Anesthesiology, LMU Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Katharina Biere
- Laboratory of Translational Research 'Stress and Immunity', Department of Anesthesiology, LMU Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Nathalie Abdelmalek
- Laboratory of Translational Research 'Stress and Immunity', Department of Anesthesiology, LMU Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Xinyu Shu
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, 100190, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Chaoyang Song
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, 100190, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Guangyao Chen
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, 100190, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Ning Li
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, 100190, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Marina Tuschen
- Laboratory of Translational Research 'Stress and Immunity', Department of Anesthesiology, LMU Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Huan Wu
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, 100190, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Shujin Sun
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, 100190, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Alexander Choukér
- Laboratory of Translational Research 'Stress and Immunity', Department of Anesthesiology, LMU Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.
| | - Mian Long
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, 100190, Beijing, China.
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China.
| | - Dominique Moser
- Laboratory of Translational Research 'Stress and Immunity', Department of Anesthesiology, LMU Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
43
|
Lu Z, Fang P, Li S, Xia D, Zhang J, Wu X, Pan J, Cai H, Fu L, Sun G, You Q. Lactylation of Histone H3k18 and Egr1 Promotes Endothelial Glycocalyx Degradation in Sepsis-Induced Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407064. [PMID: 39721014 PMCID: PMC11831459 DOI: 10.1002/advs.202407064] [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: 06/24/2024] [Revised: 12/12/2024] [Indexed: 12/28/2024]
Abstract
Circulating lactate is a critical biomarker for sepsis-induced acute lung injury (S-ALI) and is strongly associated with poor prognosis. However, whether elevated lactate directly promotes S-ALI and the specific mechanism involved remain unclear. Here, this work shows that lactate causes pulmonary endothelial glycocalyx degradation and worsens ALI during sepsis. Mechanistically, lactate increases the lactylation of K18 of histone H3, which is enriched at the promoter of EGR1 and promotes its transcription, leading to upregulation of heparanase in pulmonary microvascular endothelial cells. In addition, multiple lactylation sites are identified in EGR1, and lactylation is confirmed to occur mainly at K364. K364 lactylation of EGR1 facilitates its interaction with importin-α, in turn promoting its nuclear localization. Importantly, this work identifies KAT2B as a novel lactyltransferase whose GNAT domain directly mediates the lactylation of EGR1 during S-ALI. In vivo, suppression of lactate production or genetic knockout of EGR1 mitigated glycocalyx degradation and ALI and improved survival outcomes in mice with polymicrobial sepsis. Therefore, this study reveals that the crosstalk between metabolic reprogramming in endothelial cells and epigenetic modifications plays a critical role in the pathological processes of S-ALI.
Collapse
Affiliation(s)
- Zongqing Lu
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Pu Fang
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Shuai Li
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Dunling Xia
- Department of Emergency MedicineFirst Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Jingjing Zhang
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Xianghui Wu
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Jingjing Pan
- Department of Respiratory Intensive Care UnitAnhui Chest HospitalHefei230022China
| | - Haijian Cai
- Center for Scientific ResearchAnhui Medical UniversityHefei230032China
| | - Lin Fu
- Department of Respiratory and Critical Care MedicineSecond Affiliated Hospital of Anhui Medical UniversityHefei230601China
| | - Gengyun Sun
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Qinghai You
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| |
Collapse
|
44
|
Wang C, Lv J, Yang M, Fu Y, Wang W, Li X, Yang Z, Lu J. Recent advances in surface functionalization of cardiovascular stents. Bioact Mater 2025; 44:389-410. [PMID: 39539518 PMCID: PMC11558551 DOI: 10.1016/j.bioactmat.2024.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Cardiovascular diseases (CVD) are the leading global threat to human health. The clinical application of vascular stents improved the survival rates and quality of life for patients with cardiovascular diseases. However, despite the benefits stents bring to patients, there are still notable complications such as thrombosis and in-stent restenosis (ISR). Surface modification techniques represent an effective strategy to enhance the clinical efficacy of vascular stents and reduce complications. This paper reviews the development strategies of vascular stents based on surface functional coating technologies aimed at addressing the limitations in clinical application, including the inhibition of intimal hyperplasia, promotion of re-endothelialization. These strategies have improved endothelial repair and inhibited vascular remodeling, thereby promoting vascular healing post-stent implantation. However, the pathological microenvironment of target vessels and the lipid plaques are key pathological factors in the development of atherosclerosis (AS) and impaired vascular repair after percutaneous coronary intervention (PCI). Therefore, restoring normal physiological environment and removing the plaques are also treatment focuses after PCI for promoting vascular repair. Unfortunately, research in this area is limited. This paper reviews the advancements in vascular stents based on surface engineering technologies over the past decade, providing guidance for the development of stents.
Collapse
Affiliation(s)
- Chuanzhe Wang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, 523059, Dongguan, Guangdong, China
| | - Jie Lv
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, Chengdu, Sichuan, China
| | - Mengyi Yang
- School of Materials Science and Engineering, Key Lab of Advanced Technology for Materials of Education Ministry, Southwest Jiaotong University, 610031, Chengdu, China
| | - Yan Fu
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, 523059, Dongguan, Guangdong, China
| | - Wenxuan Wang
- School of Materials Science and Engineering, Key Lab of Advanced Technology for Materials of Education Ministry, Southwest Jiaotong University, 610031, Chengdu, China
| | - Xin Li
- Department of Cardiology, Third People's Hospital of Chengdu Affiliated to Southwest Jiaotong University, 610072, Chengdu, Sichuan, China
| | - Zhilu Yang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, 523059, Dongguan, Guangdong, China
| | - Jing Lu
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, Chengdu, Sichuan, China
| |
Collapse
|
45
|
Garcia FJ, Heiman M. Molecular and cellular characteristics of cerebrovascular cell types and their contribution to neurodegenerative diseases. Mol Neurodegener 2025; 20:13. [PMID: 39881338 PMCID: PMC11780804 DOI: 10.1186/s13024-025-00799-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: 11/10/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Many diseases and disorders of the nervous system suffer from a lack of adequate therapeutics to halt or slow disease progression, and to this day, no cure exists for any of the fatal neurodegenerative diseases. In part this is due to the incredible diversity of cell types that comprise the brain, knowledge gaps in understanding basic mechanisms of disease, as well as a lack of reliable strategies for delivering new therapeutic modalities to affected areas. With the advent of single cell genomics, it is now possible to interrogate the molecular characteristics of diverse cell populations and their alterations in diseased states. More recently, much attention has been devoted to cell populations that have historically been difficult to profile with bulk single cell technologies. In particular, cell types that comprise the cerebrovasculature have become increasingly better characterized in normal and neurodegenerative disease contexts. In this review, we describe the current understanding of cerebrovasculature structure, function, and cell type diversity and its role in the mechanisms underlying various neurodegenerative diseases. We focus on human and mouse cerebrovasculature studies and discuss both origins and consequences of cerebrovascular dysfunction, emphasizing known cell type-specific vulnerabilities in neuronal and cerebrovascular cell populations. Lastly, we highlight how novel insights into cerebrovascular biology have impacted the development of modern therapeutic approaches and discuss outstanding questions in the field.
Collapse
Affiliation(s)
- Francisco J Garcia
- The Picower Institute for Learning and Memory, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Myriam Heiman
- The Picower Institute for Learning and Memory, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
| |
Collapse
|
46
|
Batty L, Park J, Qin L, Riaz M, Lin Y, Xu Z, Gao X, Li X, Lopez C, Zhang W, Hoareau M, Fallon ME, Huang Y, Luo H, Luo J, Ménoret S, Li P, Jiang Z, Smith P, Sachs DH, Tellides G, Ignacio Anegon, Pober JS, Liu P, Qyang Y. Vascular endothelial cells derived from transgene-free pig induced pluripotent stem cells for vascular tissue engineering. Acta Biomater 2025; 193:171-184. [PMID: 39681154 DOI: 10.1016/j.actbio.2024.12.033] [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: 07/08/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/18/2024]
Abstract
Induced pluripotent stem cells (iPSCs) hold great promise for the treatment of cardiovascular diseases through cell-based therapies, but these therapies require extensive preclinical testing that is best done in species-in-species experiments. Pigs are a good large animal model for these tests due to the similarity of their cardiovascular system to humans. However, a lack of adequate pig iPSCs (piPSCs) that are analogous to human iPSCs has greatly limited the potential usefulness of this model system. Herein, transgene-free piPSCs with true pluripotency were generated by using reprogramming factors in an optimized pig pluripotency medium. Using an effective differentiation protocol, piPSCs were used to derive endothelial cells (ECs) which displayed EC markers and functionality comparable to native pig ECs. Further, piPSC-ECs demonstrated suitability for vascular tissue engineering, producing a tissue engineered vascular conduit (TEVC) that displayed the upregulation of flow responding markers. In an in vivo functional study, these piPSC-EC-TEVCs maintained the expression of endothelial markers and prevented thrombosis as interposition inferior vena cava grafts in immunodeficient rats. The piPSCs described in this study open up the possibility of unique preclinical species-in-species large animal modeling for the furtherance of modeling of cell-based cardiovascular tissue engineering therapies. STATEMENT OF SIGNIFICANCE: While there has been significant progress in the development of cellularized cardiovascular tissue engineered therapeutics using stem cells, few of them have moved into clinical trials. This is due to the lack of a robust preclinical large animal model to address the high safety and efficacy standards for transplanted therapeutics. In this study, pig stem cells that are analagous to human's were created to address this bottleneck. They demonstrated the ability to differentiate into functional endothelial cells and were able to create a tissue engineered therapeutic that is analogous to a human therapy. With these cells, future experiments testing the safety and efficacy of tissue engineered constructs are possible, bringing these crucial therapeutics closer to the patients that need them.
Collapse
Affiliation(s)
- Luke Batty
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA; Department of Pathology, Yale University, New Haven, CT 06510, USA
| | - Jinkyu Park
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA; Department of Physiology, College of Medicine, Hallym University, Hallymdaehak-gil, Chuncheon-si, Gangwon-Do, 24252, South Korea
| | - Lingfeng Qin
- Department of Surgery, Yale University, New Haven, CT 06520, USA
| | - Muhammad Riaz
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Yuyao Lin
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA; Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhen Xu
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Xuefei Gao
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xin Li
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Colleen Lopez
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Wei Zhang
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Marie Hoareau
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Meghan E Fallon
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Yan Huang
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Hangqi Luo
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Jiesi Luo
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Séverine Ménoret
- Center for Research in Transplantation and Translational Immunology UMR1064, INSERM, Nantes Université, Nantes, France; Nantes Université, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
| | - Peining Li
- Department of Genetics, Yale University, New Haven, CT 06519, USA
| | - Zhenting Jiang
- Department of Earth & Planetary Sciences, Yale University, New Haven, CT 06511, USA
| | - Peter Smith
- Department of Comparative Medicine, Yale University, New Haven, CT 06520, USA
| | - David H Sachs
- Department of Surgery, Columbia Center for Translational Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - George Tellides
- Department of Surgery, Yale University, New Haven, CT 06520, USA
| | - Ignacio Anegon
- Center for Research in Transplantation and Translational Immunology UMR1064, INSERM, Nantes Université, Nantes, France; Nantes Université, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
| | - Jordan S Pober
- Department of Pathology, Yale University, New Haven, CT 06510, USA; Department of Immunobiology, Yale University, New Haven, CT 06520, USA
| | - Pentao Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Stem Cell and Regenerative Medicine Consortium, Pokfulam, Hong Kong, China
| | - Yibing Qyang
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA; Department of Pathology, Yale University, New Haven, CT 06510, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA.
| |
Collapse
|
47
|
Wang J, Ma L, Fang Y, Ye T, Li H, Lan P. Factors influencing glycocalyx degradation: a narrative review. Front Immunol 2025; 15:1490395. [PMID: 39885987 PMCID: PMC11779607 DOI: 10.3389/fimmu.2024.1490395] [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: 09/03/2024] [Accepted: 12/30/2024] [Indexed: 02/01/2025] Open
Abstract
The glycocalyx is a layer of villus-like structure covering the luminal surface of vascular endothelial cells. Damage to the glycocalyx has been proven linked to the development of many diseases. However, the factors that promote damage to the glycocalyx are not fully elaborated. This review summarizes factors leading to the reduction of the glycocalyx in detail, including inflammatory factors, ischemia-reperfusion, oxidative stress, lipids, glucose, high sodium, female sex hormones and others. Additionally, the mechanisms underlying its degradation are discussed. To better prevent and treat related diseases induced by glycocalyx degradation, it is a meaningful measure to avoid these factors.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cardiology, Nanning Hospital of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Lan Ma
- Department of Neurology, Wenzhou Traditional Chinese Medicine (TCM) Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| | - Yu Fang
- Department of Cardiology, Nanning Hospital of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Tengteng Ye
- Department of Cardiology, Nanning Hospital of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Hongbo Li
- Department of Cardiology, Nanning Hospital of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Peng Lan
- Department of Cardiology, Nanning Hospital of Traditional Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
48
|
Flores J, Nugent K. Sodium, the Vascular Endothelium, and Hypertension: A Narrative Review of Literature. Cardiol Rev 2025:00045415-990000000-00402. [PMID: 39807866 DOI: 10.1097/crd.0000000000000854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The vascular endothelium and its endothelial glycocalyx contribute to the protection of the endothelial cells from exposure to high levels of sodium and help these structures maintain normal function by regulating vascular permeability due to its buffering effect. The endothelial glycocalyx has negative surface charges that bind sodium and limit sodium entry into cells and the interstitial space. High sodium levels can disrupt this barrier and allow the movement of sodium into cells and extravascular fluid. This can generate reactive oxygen species that inhibit nitric oxide production. This leads to vasospasm and increases intravascular pressures. Overtime vascular remodeling occurs, and this changes the anatomy of blood vessels, their intrinsic stiffness, and their response to vasodilators and results in hypertension. Patients with increased salt sensitivity are potentially at more risk for this sequence of events. Studies on the degradation of the glycocalyx provide insight into the pathogenesis of clinical disorders with vascular involvement, but there is limited information available in the context of higher concentrations of sodium. Data on higher intake of sodium and the imbalance between nitric oxide and reactive oxygen species have been obtained in experimental studies and provide insights into possible outcomes in humans. The current western diet with sodium intake above recommended levels has led to the assessment of sodium sensitivity, which has been used in different populations and could become a practical tool to evaluate patients. This would potentially allow more focused recommendations regarding salt intake. This review will consider the structure of the vascular endothelium, its components, the effect of sodium on it, and the use of the salt blood test mini.
Collapse
Affiliation(s)
- Jackeline Flores
- From the Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | | |
Collapse
|
49
|
Fonseca DLM, Jäpel M, Gyamfi MA, Filgueiras IS, Baiochi GC, Ostrinski Y, Halpert G, Lavi YB, Vojdani E, Silva-Sousa T, Usuda JN, E Silva JCS, Freire PP, Nóbile AL, Adri AS, Barcelos PM, Corrêa YLG, do Vale FYN, Lopes LO, Schmidt SL, Wang X, Vahldieck C, Fels B, Schimke LF, Cabral-Miranda G, Hirata MH, AKhan TA, Yu YRA, Dalmolin RJ, Amital H, Vojdani A, Dias HD, Nakaya H, Ochs HD, Silverberg JI, Zimmerman J, Zyskind I, Rosenberg AZ, Schulze-Forster K, Heidecke H, Catar R, Moll G, Hackel A, Kusche-Vihrog K, Shoenfeld Y, Riemekasten G, Akbarzadeh R, Marques AHC, Cabral-Marques O. Dysregulated autoantibodies targeting AGTR1 are associated with the accumulation of COVID-19 symptoms. NPJ Syst Biol Appl 2025; 11:7. [PMID: 39805853 PMCID: PMC11730328 DOI: 10.1038/s41540-025-00488-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: 05/09/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19) presents a wide spectrum of symptoms, the causes of which remain poorly understood. This study explored the associations between autoantibodies (AABs), particularly those targeting G protein-coupled receptors (GPCRs) and renin‒angiotensin system (RAS) molecules, and the clinical manifestations of COVID-19. Using a cross-sectional analysis of 244 individuals, we applied multivariate analysis of variance, principal component analysis, and multinomial regression to examine the relationships between AAB levels and key symptoms. Significant correlations were identified between specific AABs and symptoms such as fever, muscle aches, anosmia, and dysgeusia. Notably, anti-AGTR1 antibodies, which contribute to endothelial glycocalyx (eGC) degradation, a process reversed by losartan, have emerged as strong predictors of core symptoms. AAB levels increased with symptom accumulation, peaking in patients exhibiting all four key symptoms. These findings highlight the role of AABs, particularly anti-AGTR1 antibodies, in determining symptom severity and suggest their involvement in the pathophysiology of COVID-19, including vascular complications.
Collapse
Affiliation(s)
- Dennyson Leandro M Fonseca
- BIH Center for Regenerative Therapies (BCRT), Julius Wolff Institute (JWI), and Berlin Institute of Health (BIH); all Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 10117, Berlin, Germany.
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, SP, Brazil.
| | - Maj Jäpel
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Michael Adu Gyamfi
- Departament of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Igor Salerno Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Gabriela Crispim Baiochi
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Yuri Ostrinski
- Institute of Microbiology and Virology, Riga Stradins University, Riga, Latvia
- The Dina Recanati School of Medicine, Reichman University, Herzliya, Israel
| | - Gilad Halpert
- The Dina Recanati School of Medicine, Reichman University, Herzliya, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Israel
| | - Yael Bublil Lavi
- Scakler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elroy Vojdani
- Regenera Medical 11860 Wilshire Blvd., Ste. 301, Los Angeles, CA, 90025, USA
| | - Thayna Silva-Sousa
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, São Paulo, Brazil
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, Fehrbelliner Str. 38, 16816, Neuruppin, Germany
| | - Júlia Nakanishi Usuda
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, Fehrbelliner Str. 38, 16816, Neuruppin, Germany
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Juan Carlo Santos E Silva
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Paula P Freire
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Adriel Leal Nóbile
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Anny Silva Adri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Pedro Marçal Barcelos
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Yohan Lucas Gonçalves Corrêa
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Fernando Yuri Nery do Vale
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Letícia Oliveira Lopes
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Solveig Lea Schmidt
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Xiaoqing Wang
- Priority Area Chronic Lung Diseases, Research Center Borstel, Borstel, Germany
| | - Carl Vahldieck
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Benedikt Fels
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Gustavo Cabral-Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Taj Ali AKhan
- Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan
- Emerging Pathogens Institute, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Yen-Rei A Yu
- University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Rodrigo Js Dalmolin
- Bioinformatics Multidisciplinary Environment, Federal University of Rio Grande do Norte, Natal, Brazil
- Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Howard Amital
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Israel
| | - Aristo Vojdani
- Immunosciences Laboratory, Inc, Los Angeles, CA, 90035, USA
| | - Haroldo Dutra Dias
- Department of Neuroscience, Institute of Biomedical Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Helder Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
- Instituto Israelita de Ensino e Pesquisa Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine, and Seattle Children's Research Institute, Seattle, WA, USA
| | - Jonathan I Silverberg
- School of Medicine and Health Sciences, George Washington University, Washington, D.C., USA
| | | | - Israel Zyskind
- Maimonides Medical Center, Brooklyn, NY, USA
- Department of Pediatrics, NYU Langone Medical Center, New York, NY, USA
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Kai Schulze-Forster
- CellTrend Gesellschaft mit beschränkter Haftung (GmbH), Luckenwalde, Germany
| | - Harald Heidecke
- CellTrend Gesellschaft mit beschränkter Haftung (GmbH), Luckenwalde, Germany
| | - Rusan Catar
- Departament of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Julius Wolff Institute (JWI), and Berlin Institute of Health (BIH); all Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 10117, Berlin, Germany
- Departament of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Alexander Hackel
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Kristina Kusche-Vihrog
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Yehuda Shoenfeld
- The Dina Recanati School of Medicine, Reichman University, Herzliya, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Israel
- Reichman University, Herzeliya, Israel
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Reza Akbarzadeh
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany.
| | - Alexandre H C Marques
- Bioinformatics Multidisciplinary Environment, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Otavio Cabral-Marques
- BIH Center for Regenerative Therapies (BCRT), Julius Wolff Institute (JWI), and Berlin Institute of Health (BIH); all Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 10117, Berlin, Germany.
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany.
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Israel.
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29, University of São Paulo (USP) School of Medicine, Sao Paulo, Brazil.
- Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Sao Paulo, Brazil.
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil.
| |
Collapse
|
50
|
Blawitzki LC, Monzel C, Schmidt S, Hartmann L. Selective Glycan Presentation in Liquid-Ordered or -Disordered Membrane Phases and its Effect on Lectin Binding. Angew Chem Int Ed Engl 2025; 64:e202414847. [PMID: 39412184 DOI: 10.1002/anie.202414847] [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: 08/05/2024] [Indexed: 11/14/2024]
Abstract
Glycan-protein interactions play a key role in various biological processes from fertilization to infections. Many of these interactions take place at the glycocalyx-a heavily glycosylated layer at the cell surface. Despite its significance, studying the glycocalyx remains challenging due to its complex, dynamic, and heterogeneous nature. This study introduces a glycocalyx model allowing for the first time to control spatial organization and heterogeneity of the glycan moieties. Glycan-mimetics with lipid-moieties that partition into either liquid-ordered (Lo, lipid rafts) or liquid-disordered (Ld) phases of giant unilamellar vesicles (GUVs), which serve as simplified cell membrane models mimicking lipid rafts, are developed. This phase-specific allocation allows controlled placement of glycan motifs in distinct membrane environments, creating heteromultivalent systems that replicate the natural glycocalyx's complexity. We show that phase localization of glycan mimetics significantly influences recruitment of protein receptors to the membrane. Glycan-conjugates in the ordered phase demonstrate enhanced lectin binding, supporting the idea that raft-like domains facilitate stronger receptor interactions. This study provides a platform for systematically investigating spatial and dynamic presentation of glycans in biological systems and presents the first experimental evidence that glycan accumulation in lipid rafts enhances receptor binding affinity, offering deeper insights into the glycocalyx's functional mechanisms.
Collapse
Affiliation(s)
- Luca-Cesare Blawitzki
- Department for Macromolecular Chemistry, University of Freiburg, Stefan-Meier-Straße 31, 79104, Freiburg i.Br., Germany
- Department for Organic and Macromolecular Chemistry, Heinrich Heine University Duesseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Cornelia Monzel
- Department for Experimental Medical Physics, Heinrich Heine University Duesseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Stephan Schmidt
- Department for Macromolecular Chemistry, University of Freiburg, Stefan-Meier-Straße 31, 79104, Freiburg i.Br., Germany
| | - Laura Hartmann
- Department for Macromolecular Chemistry, University of Freiburg, Stefan-Meier-Straße 31, 79104, Freiburg i.Br., Germany
| |
Collapse
|