1
|
Deng H, Eichmann A, Schwartz MA. Fluid Shear Stress-Regulated Vascular Remodeling: Past, Present, and Future. Arterioscler Thromb Vasc Biol 2025; 45:882-900. [PMID: 40207366 DOI: 10.1161/atvbaha.125.322557] [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: 04/11/2025]
Abstract
The vascular system remodels throughout life to ensure adequate perfusion of tissues as they grow, regress, or change metabolic activity. Angiogenesis, the sprouting of new blood vessels to expand the capillary network, versus regression, in which endothelial cells die or migrate away to remove unneeded capillaries, controls capillary density. In addition, upstream arteries adjust their diameters to optimize blood flow to downstream vascular beds, which is controlled primarily by vascular endothelial cells sensing fluid shear stress (FSS) from blood flow. Changes in capillary density and small artery tone lead to changes in the resistance of the vascular bed, which leads to changes in flow through the arteries that feed these small vessels. The resultant decreases or increases in FSS through these vessels then stimulate their inward or outward remodeling, respectively. This review summarizes our knowledge of endothelial FSS-dependent vascular remodeling, offering insights into potential therapeutic interventions. We first provide a historical overview, then discuss the concept of set point and mechanisms of low-FSS-mediated and high-FSS-mediated inward and outward remodeling. We then cover in vivo animal models, molecular mechanisms, and clinical implications. Understanding the mechanisms underlying physiological endothelial FSS-mediated vascular remodeling and their failure due to mutations or chronic inflammatory and metabolic stresses may lead to new therapeutic strategies to prevent or treat vascular diseases.
Collapse
Affiliation(s)
- Hanqiang Deng
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
| | - Anne Eichmann
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
| | - Martin A Schwartz
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale School of Medicine, New Haven, CT (M.A.S.)
- Department of Biomedical Engineering, Yale School of Engineering, New Haven, CT (M.A.S.)
| |
Collapse
|
2
|
Hu Q, Chen H, Lan J, Chen Y, Liu Z, Xiong Y, Zhou W, Zhong Z, Ye Q. KLF10 Induced by Hypothermic Machine Perfusion Alleviates Renal Inflammation Through BIRC2 /Noncanonical NF-κB Pathway. Transplantation 2025; 109:e273-e286. [PMID: 39716345 DOI: 10.1097/tp.0000000000005314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
BACKGROUND Hypothermic machine perfusion (HMP) is becoming the main preservation method for donation after circulatory death (DCD) kidneys. It can provide continuous flow and form shear stress (SS) upon endothelial cells (ECs), thereby regulating EC injury. Krüppel-like factor 10 (KLF10) has been shown to lessen vascular damage. However, how SS and KLF10 impact HMP-regulated injury is unclear. METHODS In this study, we investigated the influences of KLF10 on HMP in animal models and human renal biopsy and explored how SS affected KLF10 expression in a parallel-plate flow chamber system. Chromatin Immunoprecipitation sequencing and luciferase assay were performed to seek the target genes of KLF10. The influences of KLF10 on HMP-regulated injury were investigated by transfecting si-KLF10 adeno-associated virus serotype 9 into rat kidneys. The molecular expression was examined using immunofluorescence staining, Western blotting, and quantitative polymerase chain reaction. RESULTS Our results show KLF10 expression was augmented in human, rabbit, and rat DCD kidneys after HMP. HMP improved ECs and tubule injury and attenuated inflammation; however, the knockdown of KLF10 reversed this effect. SS regulated KLF10 expression in ECs by affecting F-actin, and KLF10 could maintain ECs homeostasis. Chromatin Immunoprecipitation sequencing and luciferase assay revealed that baculoviral inhibitor of apoptosis protein repeat-containing 2 (BIRC2) is a target gene of KLF10. Furthermore, BIRC2 linked to nuclear factor kappa B (NF-κB)-inducing kinase, induced NF-κB)-inducing kinase ubiquitination, and resulted in inhibiting the noncanonical NF-κB pathway. CONCLUSIONS SS can mediate KLF10 expression, whereas HMP can protect against warm ischemic injury by reducing inflammation via KLF10/BIRC2/noncanonical NF-κB pathway. Therefore, KLF10 might be a novel target for improving DCD kidney quality.
Collapse
Affiliation(s)
- Qianchao Hu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Hao Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Jia'nan Lan
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Yiwen Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Zhongzhong Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Yan Xiong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Wei Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Zibiao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
- The 3rd Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, Changsha, China
| |
Collapse
|
3
|
Li J, Yu Y, Tranquillo RT. Computational construction and design optimization of a novel tri-tube heart valve. Biomech Model Mechanobiol 2025:10.1007/s10237-025-01956-5. [PMID: 40418405 DOI: 10.1007/s10237-025-01956-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/26/2025] [Indexed: 05/27/2025]
Abstract
A finite-element-based algorithm for the in silico construction of a novel tri-tube heart valve was developed to facilitate optimization of the leaflet geometry. An anisotropic hyperelastic model fitted to high-strain rate planar equibiaxial tension and compression data was used to approximate the nonlinear and anisotropic material behavior of biologically-engineered tubes and simulate valve closure under steady back pressure and steady forward flow. Four metrics were considered to evaluate valve performance in simulated closure: coaptation area, regurgitation area, pinwheel index, and prolapse area. Response surfaces revealed competing objectives between metrics for a valve of target 24 mm diameter in terms of two design parameters, tube diameter and leaflet height. A multi-objective genetic algorithm determined an intermediate tube diameter and leaflet height (16 mm and 11 mm, respectively) of the design space as optimal. Additionally, steady flow simulations were performed using two-way fluid-structure interaction with selected designs to examine washout behind leaflets with particle tracking. One design close to the optimal point for valve closure indicated washout for particles initially distributed behind leaflets. Though comprehensive valve design optimization requires flow analysis over multiple valve cycles to capture all effects associated with flow, this methodology based on diastolic state geometry optimization followed by steady washout analysis reduces the space of design variables for further optimization.
Collapse
Affiliation(s)
- Jirong Li
- Department of Biomedical Engineering, University of Minnesota, 7-114 NHH, 312 Church St SE, Minneapolis, MN, 55455, USA
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Yijiang Yu
- Department of Biomedical Engineering, University of Minnesota, 7-114 NHH, 312 Church St SE, Minneapolis, MN, 55455, USA
| | - Robert T Tranquillo
- Department of Biomedical Engineering, University of Minnesota, 7-114 NHH, 312 Church St SE, Minneapolis, MN, 55455, USA.
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
4
|
Harden JE, David Branch J, Reynolds LJ. Impacts of Physical Inactivity Models on Endothelial Function: A Systematic Review. Sports Med 2025:10.1007/s40279-025-02238-x. [PMID: 40388068 DOI: 10.1007/s40279-025-02238-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Endothelial dysfunction is associated with cardiovascular disease and cardiac events. Numerous studies demonstrate that a reduction in physical activity/exercise levels are associated with poor endothelial function. Yet, these studies use a plethora of models to mimic reduced activity levels which may have widely different results on endothelial function. It is pertinent to collectively review these articles to provide a comprehensive understanding of the impact of reduced activity on vascular health, as endothelial function is one of many factors that influences vascular tone. OBJECTIVE The purpose of this systematic review is to examine and synthesize the current literature regarding the effects of acutely (≤ ~ 2 months) reducing physical activity on endothelial function. METHODS This systematic review contains a search of two databases (PubMed, Web of Science) conducted by two reviewers. After screening and review, the search yielded 20 studies that were appraised and reviewed. RESULTS Articles were separated into four categories based on the type of inactivity intervention: reduction in daily physical activity interventions, detraining (removal of exercise) interventions, bed rest interventions, and immobilization interventions. Acute physical inactivity interventions were largely shown to reduce upper and lower limb artery flow-mediated dilation and lower limb microvascular function. CONCLUSION The results indicate that those studies with increased time spent in inactivity, as well as increased severity of inactivity, were more likely to have negative endothelial function outcomes. Future research should examine differences in the severity of physical inactivity interventions regarding endothelial function.
Collapse
Affiliation(s)
- Joel E Harden
- Integrative Cardiometabolic Research Laboratory, School of Exercise Science, Old Dominion University, 1006C Student Recreation CTR, Norfolk, VA, 23529, USA
| | - J David Branch
- School of Exercise Science, Old Dominion University, Norfolk, VA, USA
| | - Leryn J Reynolds
- Integrative Cardiometabolic Research Laboratory, School of Exercise Science, Old Dominion University, 1006C Student Recreation CTR, Norfolk, VA, 23529, USA.
| |
Collapse
|
5
|
Zhao GJ, Han SY, Li Y, Yuan D, Qin S, Li Y, Jang H, Chen LJ, Wei TYW, He M, Li YS, Bouman Chen Z, Shi L, Chien S, Shyy JYJ. METTL3 mediates atheroprone flow-induced glycolysis in endothelial cells. Proc Natl Acad Sci U S A 2025; 122:e2424796122. [PMID: 40327688 PMCID: PMC12088407 DOI: 10.1073/pnas.2424796122] [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: 12/04/2024] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Atheroprone flow-increased glycolysis in vascular endothelial cells (ECs) is pivotal in EC dysfunction and the initiation of atherosclerosis. Methyltransferase 3 (METTL3) is a major m6A methyltransferase for RNA N6-mehtyladenosine (m6A) modifications to regulate epitranscriptome and cellular functions. With the atheroprone flow upregulating METTL3 and m6A RNA hypermethylation, we investigate the role of METTL3 in atheroprone flow-induced glycolysis in ECs in vitro and in vivo. Compared to pulsatile shear stress (PS, atheroprotective flow), oscillatory shear stress (OS, atheroprone flow) increases METTL3 expression to enhance the m6A modifications of mRNAs encoding HK1, PFKFB3, and GCKR, which are rate-limiting enzymes of glycolysis. These augmented m6A modifications increase the expressions of HK1 and PFKFB3 while decreasing GCKR, resulting in elevated EC glycolysis, as revealed by seahorse analysis. Moreover, a stimulated Raman scattering (SRS) imaging study demonstrates the elevation of glucose incorporation into de novo synthesized lipids in ECs under atheroprone flow in vitro and in vivo. Empagliflozin, a sodium-glucose cotransporter-2 inhibitor (SGLT2i) drug, represses METTL3 expression, thereby mitigating OS-induced glycolysis in ECs. These data suggest mechanisms by which METTL3 links EC mechanotransduction with metabolic reprogramming under atherogenic conditions.
Collapse
Affiliation(s)
- Guo-Jun Zhao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou450052, China
- Division of Cardiology, Department of Medicine, University of California, La Jolla, CA92093
| | - So Yun Han
- Division of Cardiology, Department of Medicine, University of California, La Jolla, CA92093
| | - Yajuan Li
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
- Institute of Engineering in Medicine, University of California, La Jolla, CA92093
| | - Dongqiang Yuan
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA91010
| | - Shuo Qin
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
| | - Yuhan Li
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
| | - Hongje Jang
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
| | - Li-Jing Chen
- Institute of Engineering in Medicine, University of California, La Jolla, CA92093
| | - Tong-You Wade Wei
- Division of Cardiology, Department of Medicine, University of California, La Jolla, CA92093
| | - Ming He
- Division of Cardiology, Department of Medicine, University of California, La Jolla, CA92093
| | - Yi-Shun Li
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
- Institute of Engineering in Medicine, University of California, La Jolla, CA92093
| | - Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA91010
| | - Lingyan Shi
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
| | - Shu Chien
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
- Institute of Engineering in Medicine, University of California, La Jolla, CA92093
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, University of California, La Jolla, CA92093
- Institute of Engineering in Medicine, University of California, La Jolla, CA92093
| |
Collapse
|
6
|
Ghosh S, Mandal S, Das A, Shukla PC, Das S. Probing Shear-Induced Endothelial Activation in Vascular Conditions Using On-Chip Models. ACS Biomater Sci Eng 2025; 11:2677-2689. [PMID: 40214744 DOI: 10.1021/acsbiomaterials.4c02164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
Hemodynamic fluctuations at vessel bifurcation impact the development of atherosclerosis and aneurysms. A novel glass capillary tube-based lithography-free technique was used for fabricating vessel bifurcations with stenosis and aneurysm at the junction of bifurcation to determine the endothelial response to arterial shear rates in vitro. At variable shear rates of 1-2000 s-1, representative of conditions in the aorta, the endothelial cell responses under flow disturbances encountered in stenosed and aneurysmal vessels were modeled. Mechanical disturbances induce greater endothelial activation at stenosis, while increased VE-cadherin expression deters activation at dilations. The endothelial responses to disturbed flow were better observed at the area of bifurcation, where the increase in shear forces and reduced pressure marginally compensated for cellular activation. The comparative model was established using an image analysis approach for the assessment of endothelial responses toward disease progression at bifurcations. No significant differences in endothelial markers were observed under inflammatory stress and physiologically relevant mechanical stresses due to compensatory effects of inflammatory cytokines inducing NF-κβ activation, as seen using this frugal approach.
Collapse
Affiliation(s)
- Swachhatoa Ghosh
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur 721302, India
| | - Shreya Mandal
- Department of Bioscience and Biotechnology, IIT Kharagpur, Kharagpur 721302, India
| | - Abhijit Das
- Department of Bioscience and Biotechnology, IIT Kharagpur, Kharagpur 721302, India
| | - Praphulla C Shukla
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur 721302, India
| | - Soumen Das
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur 721302, India
| |
Collapse
|
7
|
Bai B, Liu L, Liu Y, Yang S, Wu H, Zhang K, Shi L, Zhang Q. High Hcy regulates fluid shear stress pathway activity through histone H3K79 homocysteinylation in hyperhomocysteinemia-related child hypertension. J Transl Med 2025; 23:525. [PMID: 40346632 PMCID: PMC12065145 DOI: 10.1186/s12967-025-06483-6] [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: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND The rise of hypertension in children has been increasingly associated with hyperhomocysteinemia (HHcy), which is recognized as a major risk factor. However, the underlying mechanisms linking homocysteine and hypertension (termed HHYP) are not fully understood. METHODS This study utilized plasma samples from 27 control children and 27 children with HHYP (aged 8 ~ 16 years) for TMT6-labeled proteomic quantification, identifying significant altered proteins. Bioinformatics analysis revealed pathway alterations. Verification was carried out via parallel reaction monitoring (PRM) and western blot (WB) analyses. Additionally, a rat model of HHYP induced by high methionine diets, and umbilical vein endothelial cell models exposed to high homocysteine (hcy) levels were developed to investigate the molecular underpinnings further. Protein expression changes and epigenetic modifications were assessed using WB, immunohistochemistry (IHC), and ChIP-qPCR techniques. RESULTS Key findings indicated that 357 proteins and 69 pathways were altered in children with HHYP. Specifically, 12 proteins within the fluid shear stress and atherosclerosis (FSSA) pathway showed differential expression, including the downregulation of TRX1 and GPX1 and the upregulation of ICAM1. The same expression patterns were noted in both the HHYP rat aortic tissues and the high hcy cultured endothelial cells. Moreover, elevated H3K79hcy modification levels were observed alongside epigenetic regulation of genes related to the FSSA pathway. Importantly, folic acid (FA), a medication frequently used in the clinical treatment of HHYP, has been demonstrated to effectively reverse H3K79hcy modifications and restore the disrupted FSSA pathway in both animal models and cell cultures. CONCLUSIONS The present study suggests that HHcy may contribute to hypertension through the epigenetic dysregulation of the FSSA pathway mediated by H3K79hcy. Furthermore, the pediatric proteomics data gleaned from this study offer new clinical insights into the pathophysiology of HHYP in children.
Collapse
Affiliation(s)
- Baoling Bai
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, No. 2 Yabao Rd, Beijing, 100020, China
| | - Lingyun Liu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, No. 2 Yabao Rd, Beijing, 100020, China
| | - Yanyan Liu
- Department of Cardiology, Capital Center for Children's Health, Capital Medical University, No. 2 Yabao Rd, Beijing, 100020, China
| | - Shuangshuang Yang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, No. 2 Yabao Rd, Beijing, 100020, China
| | - Haojie Wu
- Department of Cardiology, Capital Center for Children's Health, Capital Medical University, No. 2 Yabao Rd, Beijing, 100020, China
| | - Kexin Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, No. 2 Yabao Rd, Beijing, 100020, China
| | - Lin Shi
- Department of Cardiology, Capital Center for Children's Health, Capital Medical University, No. 2 Yabao Rd, Beijing, 100020, China.
| | - Qin Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, No. 2 Yabao Rd, Beijing, 100020, China.
| |
Collapse
|
8
|
Otsuki T, Suwabe K, Yoshikawa T, Kotani K, Zempo-Miyaki A. Nailfold capillary patterns in ballgame and endurance athletes. Front Physiol 2025; 16:1568972. [PMID: 40308570 PMCID: PMC12040917 DOI: 10.3389/fphys.2025.1568972] [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: 01/31/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Purpose Nailfold capillary patterns can be observed noninvasively using light microscopy. Nailfold capillaries are straight, U-shaped, and densely looped in healthy individuals and may be altered by disease or lifestyle factors, such as diet. However, the effects of daily physical activity and exercise training on nailfold capillary patterns remain unclear. This study aimed to examine the effects of exercise training on nailfold capillary patterns by investigating these patterns in endurance athletes, ballgame athletes, and sedentary healthy men. Methods Five healthy men participated in nailfold capillary pattern measurements on three consecutive days to test the reproducibility and bilateral differences in the nailfold capillary loop density, length, and width measured using light microscopy and a commercial analysis system. The nailfold capillaries of 10 endurance athletes (endurance group; eight long-distance runners and two triathletes), 10 ballgame athletes (ballgame group; seven soccer players and three basketball players), and nine sedentary healthy men (sedentary group) were then examined using light microscopy. Results The day-to-day coefficient of variation for the nailfold capillary loop density, length, and width were 4.9% ± 1.6%, 7.5% ± 1.3%, and 4.2% ± 1.5%, respectively. No significant differences in these measurements were observed between the dominant and non-dominant hands. Nailfold capillary density was greater in the ballgame group than in the endurance and sedentary groups. Capillary loop length was shorter in the ballgame group than in the endurance and sedentary groups. No significant differences in capillary loop density and length were observed between the endurance and sedentary groups. No significant intergroup differences were observed in capillary loop width. Conclusion These results suggest that the nailfold capillary patterns of male ballgame athletes differ from those of endurance athletes and sedentary healthy men. Ballgame training may affect nailfold capillary patterns in sedentary healthy men.
Collapse
Affiliation(s)
- Takeshi Otsuki
- Faculty of Sport and Health Sciences, Ryutsu Keizai University, Ryugasaki, Japan
| | | | | | | | | |
Collapse
|
9
|
Lightfoot A, Lewis JW, Patten DA, Shetty S, Hewett PW, Mansour AA, McGettrick HM, Iqbal AJ. Differential expression of endothelial derived galectins in response to shear stress. Exp Cell Res 2025; 447:114521. [PMID: 40107440 DOI: 10.1016/j.yexcr.2025.114521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Endothelial cells function as mechanosensors, dynamically altering their functional response based on varying shear stress/flow patterns to maintain vascular homeostasis. Disturbed flow leads to endothelium dysfunction, promoting conditions such as atherosclerosis. Understanding the molecular impact of flow is crucial for the development of new therapeutic targets for vascular diseases. Galectins have been implicated in vascular diseases, specifically their role in inflammation. However, the regulation of endothelial galectins by shear stress remains unexplored. METHODS Galectin gene and protein expression were analysed from publicly available datasets or in human umbilical endothelial cells (HUVEC) and human arterial endothelial cells (HAEC) cultured under either shear stress induced by orbital shaking or static conditions by qPCR, immunofluorescence imaging and ELISA. RESULTS Laminar shear stress upregulated LGALS9 and downregulated LGALS1, while disturbed flow reversed these effects. Complex shear environments significantly increased Gal-3 and Gal-9 expression at both gene and protein levels, with distinct variations in surface expression and secretion. In vivo single-cell RNA sequencing (scRNAseq) revealed reduced Lgals9 expression in endothelial cells exposed to disturbed flow in carotid artery ligation models compared to laminar flow. SIGNIFICANCE These findings highlight that endothelial galectin expression is shear-regulated, which has significant implications for understanding galectin biology and there potential as therapeutic targets in vascular diseases influenced by shear stress.
Collapse
Affiliation(s)
- Abbey Lightfoot
- Department of Cardiovascular Sciences, College of Medicine and Health University of Birmingham, Birmingham, B15 2TT, UK
| | - Jonathan W Lewis
- Department of Inflammation and Ageing, College of Medicine and Health University of Birmingham, Birmingham, B15 2TT, UK
| | - Daniel A Patten
- Department of Immunology and Immunotherapy, College of Medicine and Health University of Birmingham, Birmingham, B15 2TT, UK
| | - Shishir Shetty
- Department of Immunology and Immunotherapy, College of Medicine and Health University of Birmingham, Birmingham, B15 2TT, UK
| | - Peter W Hewett
- Department of Cardiovascular Sciences, College of Medicine and Health University of Birmingham, Birmingham, B15 2TT, UK
| | - Adel Abo Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Helen M McGettrick
- Department of Inflammation and Ageing, College of Medicine and Health University of Birmingham, Birmingham, B15 2TT, UK.
| | - Asif J Iqbal
- Department of Cardiovascular Sciences, College of Medicine and Health University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
10
|
Yan K, Ye W, Martínez A, Geronzi L, Escrig P, Tomasi J, Rochette M, Haigron P, Bel-Brunon A. Fluid-structure-growth modeling in ascending aortic aneurysm: capability to reproduce a patient case. Biomech Model Mechanobiol 2025; 24:405-422. [PMID: 39760773 DOI: 10.1007/s10237-024-01915-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025]
Abstract
Predicting the evolution of ascending aortic aneurysm (AscAA) growth is a challenge, complicated by the intricate interplay of aortic geometry, tissue behavior, and blood flow dynamics. We investigate a flow-structural growth and remodeling (FSG) model based on the homogenized constrained mixture theory to simulate realistic AscAA growth evolution. Our approach involves initiating a finite element model with an initial elastin insult, driven by the distribution of Time-Averaged Wall Shear Stress (TAWSS) derived from computational fluid dynamics simulations. Through FSG simulation, we first calibrate the growth and remodeling material parameters to reproduce the growth observed on a patient-specific case. Then, we explore the influence of two critical parameters: the direction of the inlet jet flow, which affects the zone of significant TAWSS, and prestretch, which impacts the tissue homeostatic state. Our results show that calibrating material parameters, inlet flow direction, and prestretch allows to reproduce the observed growth, and that prestretch calibration and inlet flow direction significantly influence the simulated growth pattern. Our workflow can be applied to additional patient cases to confirm these tendencies and progress toward a predictive tool for clinical decision support.
Collapse
Affiliation(s)
- Kexin Yan
- ANSYS, 69621, Villeurbanne, France.
- CHU Rennes, Inserm, LTSI - UMR 1099, Univ Rennes, 35000, Rennes, France.
- CNRS, LaMCoS, UMR5259, INSA Lyon, 69621, Villeurbanne, France.
| | | | | | - Leonardo Geronzi
- Department of Enterprise Engineering "Mario Lucertini", University of Rome "Tor Vergata", Rome, Italy
| | - Pierre Escrig
- CHU Rennes, Inserm, LTSI - UMR 1099, Univ Rennes, 35000, Rennes, France
| | - Jacques Tomasi
- CHU Rennes, Inserm, LTSI - UMR 1099, Univ Rennes, 35000, Rennes, France
| | | | - Pascal Haigron
- CHU Rennes, Inserm, LTSI - UMR 1099, Univ Rennes, 35000, Rennes, France
| | | |
Collapse
|
11
|
Ali A, Yun S. Multifaceted Role of Notch Signaling in Vascular Health and Diseases. Biomedicines 2025; 13:837. [PMID: 40299408 PMCID: PMC12024539 DOI: 10.3390/biomedicines13040837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Notch signaling is evolutionarily conserved from Drosophila to mammals and it functions as an essential modulator of vascular growth and development by directing endothelial cell specification, proliferation, migration, arteriovenous differentiation, inflammation, and apoptosis. The interplay between Notch and other signaling pathways plays a homeostatic role by modulating multiple vascular functions, including permeability regulation, angiogenesis, and vascular remodeling. This review explores current knowledge on Notch signaling in vascular development, homeostasis, and disease. It also discusses recent developments in understanding how endothelial Notch signaling affects vascular inflammation via cytokines or aberrant shear stress in endothelial cells while addressing the reciprocal relationship between Notch signaling and inflammation.
Collapse
Affiliation(s)
| | - Sanguk Yun
- Department of Biotechnology, Inje University, Gimhae 50834, Republic of Korea;
| |
Collapse
|
12
|
Cheng CK, Wang N, Wang L, Huang Y. Biophysical and Biochemical Roles of Shear Stress on Endothelium: A Revisit and New Insights. Circ Res 2025; 136:752-772. [PMID: 40146803 PMCID: PMC11949231 DOI: 10.1161/circresaha.124.325685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Hemodynamic shear stress, the frictional force exerted by blood flow on the endothelium, mediates vascular homeostasis. This review examines the biophysical nature and biochemical effects of shear stress on endothelial cells, with a particular focus on its impact on cardiovascular pathophysiology. Atherosclerosis develops preferentially at arterial branches and curvatures, where disturbed flow patterns are most prevalent. The review also highlights the range of shear stress across diverse human arteries and its temporal variations, including aging-related alterations. This review presents a summary of the critical mechanosensors and flow-sensitive effectors that respond to shear stress, along with the downstream cellular events that they regulate. The review evaluates experimental models for studying shear stress in vitro and in vivo, as well as their potential limitations. The review discusses strategies targeting shear stress, including pharmacological approaches, physiological means, surgical interventions, and gene therapies. Furthermore, the review addresses emerging perspectives in hemodynamic research, including single-cell sequencing, spatial omics, metabolomics, and multiomics technologies. By integrating the biophysical and biochemical aspects of shear stress, this review offers insights into the complex interplay between hemodynamics and endothelial homeostasis at the preclinical and clinical levels.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, China (C.K.C., L.W., Y.H.)
| | - Nanping Wang
- Laboratory for Molecular Vascular Biology and Bioengineering, and Wuhu Hospital, Health Science Center, East China Normal University, Shanghai (N.W.)
| | - Li Wang
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, China (C.K.C., L.W., Y.H.)
| | - Yu Huang
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, China (C.K.C., L.W., Y.H.)
| |
Collapse
|
13
|
Akter MZ, Tufail F, Ahmad A, Oh YW, Kim JM, Kim S, Hasan MM, Li L, Lee DW, Kim YS, Lee SJ, Kim HS, Ahn Y, Choi YJ, Yi HG. Harnessing native blueprints for designing bioinks to bioprint functional cardiac tissue. iScience 2025; 28:111882. [PMID: 40177403 PMCID: PMC11964760 DOI: 10.1016/j.isci.2025.111882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Cardiac tissue lacks regenerative capacity, making heart transplantation the primary treatment for end-stage heart failure. Engineered cardiac tissues developed through three-dimensional bioprinting (3DBP) offer a promising alternative. However, reproducing the native structure, cellular diversity, and functionality of cardiac tissue requires advanced cardiac bioinks. Major obstacles in CTE (cardiac tissue engineering) include accurately characterizing bioink properties, replicating the cardiac microenvironment, and achieving precise spatial organization. Optimizing bioink properties to closely mimic the extracellular matrix (ECM) is essential, as deviations may result in pathological effects. This review encompasses the rheological and electromechanical properties of bioinks and the function of the cardiac microenvironment in the design of functional cardiac constructs. Furthermore, it focuses on improving the rheological characteristics, printability, and functionality of bioinks, offering valuable perspectives for developing new bioinks especially designed for CTE.
Collapse
Affiliation(s)
- Mst Zobaida Akter
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Fatima Tufail
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ashfaq Ahmad
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yoon Wha Oh
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jung Min Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seoyeon Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Md Mehedee Hasan
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Longlong Li
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Weon Lee
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yong Sook Kim
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Su-jin Lee
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Youngkeun Ahn
- Division of Cardiology, Department of Internal Medicine, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Yeong-Jin Choi
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science (KIMS), Changwon 51508, Republic of Korea
- Advanced Materials Engineering, Korea National University of Science and Technology (UST), Changwon, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
14
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [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: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
15
|
Wu X, Xiao H, Ma L. The application of computational fluid dynamics in hepatic portal vein haemodynamics research: a narrative review. Quant Imaging Med Surg 2025; 15:2605-2620. [PMID: 40160636 PMCID: PMC11948386 DOI: 10.21037/qims-24-1593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 01/15/2025] [Indexed: 04/02/2025]
Abstract
Background and Objective The diagnosis and treatment of many liver diseases are related to the assessment of the hepatic portal vein (PV). Noninvasive methods (medical imaging) and invasive methods (hepatic vein catheterization) are commonly used to analyse the haemodynamic information of the PV. In recent years, computational fluid dynamics (CFD) has emerged as a transformative tool in haemodynamics research, revolutionizing the understanding of blood flow behaviour, especially in various artery systems. The purpose of this review is the following: (I) introduce clinicians to CFD as a novel tool and describe its role in PV assessment; and (II) for clinicians and researchers who already use CFD, outline the progress in the application of CFD to the PV. Methods The English-language literature published from 1987 (when the first study supporting the study's aim appeared) to 2024 was selected for inclusion in a narrative review. Key Content and Findings This narrative review commences with an overview of principles of CFD and methods in PV studies, which involve model establishment, grid partitioning, boundary condition formulation, and error analysis. The focus then shifts to CFD's impact on the examination of the PV under different conditions such as portal hypertension in liver cirrhosis, PV thrombosis, post-transjugular intrahepatic portosystemic shunt (TIPS) procedure, and evaluation of the PV after liver transplantation. Finally, challenges and future directions about the CFD application in PV are outlined. Conclusions CFD has potential application value in PV haemodynamics, but of the few studies available, most involve only small samples. Therefore, more research is needed to clarify the feasibility and reliability of this new tool.
Collapse
Affiliation(s)
- Xian Wu
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, China
- Department of Ultrasound, West China Tianfu Hospital of Sichuan University, Chengdu, China
| | - Hong Xiao
- State Key Laboratory of Hydraulics and Mountain River Engineering, Sichuan University, Chengdu, China
| | - Lin Ma
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, China
- Department of Ultrasound, West China Tianfu Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Deuter D, Haj A, Brawanski A, Krenkel L, Schmidt NO, Doenitz C. Fast simulation of hemodynamics in intracranial aneurysms for clinical use. Acta Neurochir (Wien) 2025; 167:56. [PMID: 40029490 PMCID: PMC11876267 DOI: 10.1007/s00701-025-06469-9] [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: 07/21/2024] [Accepted: 02/14/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND A widely accepted tool to assess hemodynamics, one of the most important factors in aneurysm pathophysiology, is Computational Fluid Dynamics (CFD). As current workflows are still time consuming and difficult to operate, CFD is not yet a standard tool in the clinical setting. There it could provide valuable information on aneurysm treatment, especially regarding local risks of rupture, which might help to optimize the individualized strategy of neurosurgical dissection during microsurgical aneurysm clipping. METHOD We established and validated a semi-automated workflow using 3D rotational angiographies of 24 intracranial aneurysms from patients having received aneurysm treatment at our centre. Reconstruction of vessel geometry and generation of volume meshes was performed using AMIRA 6.2.0 and ICEM 17.1. For solving ANSYS CFX was used. For validational checks, tests regarding the volumetric impact of smoothing operations, the impact of mesh sizes on the results (grid convergence), geometric mesh quality and time tests for the time needed to perform the workflow were conducted in subgroups. RESULTS Most of the steps of the workflow were performed directly on the 3D images requiring no programming experience. The workflow led to final CFD results in a mean time of 22 min 51.4 s (95%-CI 20 min 51.562 s-24 min 51.238 s, n = 5). Volume of the geometries after pre-processing was in mean 4.46% higher than before in the analysed subgroup (95%-CI 3.43-5.50%). Regarding mesh sizes, mean relative aberrations of 2.30% (95%-CI 1.51-3.09%) were found for surface meshes and between 1.40% (95%-CI 1.07-1.72%) and 2.61% (95%-CI 1.93-3.29%) for volume meshes. Acceptable geometric mesh quality of volume meshes was found. CONCLUSIONS We developed a semi-automated workflow for aneurysm CFD to benefit from hemodynamic data in the clinical setting. The ease of handling opens the workflow to clinicians untrained in programming. As previous studies have found that the distribution of hemodynamic parameters correlates with thin-walled aneurysm areas susceptible to rupture, these data might be beneficial for the operating neurosurgeon during aneurysm surgery, even in acute cases.
Collapse
Affiliation(s)
- Daniel Deuter
- Klinik und Poliklinik für Neurochirurgie, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany.
| | - Amer Haj
- Klinik und Poliklinik für Neurochirurgie, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Alexander Brawanski
- Klinik und Poliklinik für Neurochirurgie, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Lars Krenkel
- Regensburg Center of Biomedical Engineering (RCBE), OTH Regensburg and University of Regensburg, 93053, Regensburg, Germany
| | - Nils-Ole Schmidt
- Klinik und Poliklinik für Neurochirurgie, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Christian Doenitz
- Klinik und Poliklinik für Neurochirurgie, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| |
Collapse
|
17
|
Ruisch J, Bakker JMKD, Helvert MV, Jebbink EG, Holewijn S, Reijnen MMPJ, Korte CLD, Saris AECM. Quantification of Blood Flow in the Carotid Bifurcation of Healthy Subjects. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2025; 72:309-320. [PMID: 40031281 DOI: 10.1109/tuffc.2025.3529285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Locally disturbed blood flow patterns are known to create an atherogenic environment, particularly in the presence of other cardiovascular risk factors. Given the geometry of a healthy carotid artery, complex flow patterns are expected to be present. This study aims to characterize (complex) blood flow patterns and estimate flow-derived parameters in the carotid bifurcation of healthy subjects. Ultrasound-based velocity vector imaging (US-VVI) was acquired in the carotid bifurcation of 20 healthy subjects. Hemodynamic parameters, including temporal velocity profile, vector complexity (VC), vortex presence, and wall shear stress (WSS), were derived and compared between two age groups (20-30 and 65-75 years). Lower velocities and higher VC values were observed in the older age group for all timepoints. The highest presence of vortices was observed during the systolic deceleration, which was more exposed in younger subjects (5 out of 10) compared to older subjects (3 out of 9). A quick build-up and consequent resolving of the vortices was reflected by the relatively short vortex duration, with a vortex presence of 11.4% (7.9-15.6) and 13.1% (5.9-18.6) as a percentage of the cardiac cycle in younger and older subjects, respectively. Larger WSS estimates, represented as median along the complete vessel wall, were found in the younger subjects at all timestamps, except at systolic deceleration. In conclusion, the presence of complex flow patterns was confirmed in healthy subjects and multiple flow-derived hemodynamic parameters were evaluated in two age groups, providing an insight into age-related differences in hemodynamics. Aging seemed to result in higher vector complexities, whereas the presence of recirculating flow is less in older subjects.
Collapse
|
18
|
Aye SSS, Fang Z, Wu MCL, Lim KS, Ju LA. Integrating microfluidics, hydrogels, and 3D bioprinting for personalized vessel-on-a-chip platforms. Biomater Sci 2025; 13:1131-1160. [PMID: 39834160 DOI: 10.1039/d4bm01354a] [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: 01/22/2025]
Abstract
Thrombosis, a major cause of morbidity and mortality worldwide, presents a complex challenge in cardiovascular medicine due to the intricacy of clotting mechanisms in living organisms. Traditional research approaches, including clinical studies and animal models, often yield conflicting results due to the inability to control variables in these complex systems, highlighting the need for more precise investigative tools. This review explores the evolution of in vitro thrombosis models, from conventional polydimethylsiloxane (PDMS)-based microfluidic devices to advanced hydrogel-based systems and cutting-edge 3D bioprinted vascular constructs. We discuss how these emerging technologies, particularly vessel-on-a-chip platforms, are enabling researchers to control previously unmanageable factors, thereby offering unprecedented opportunities to pinpoint specific clotting mechanisms. While PDMS-based devices offer optical transparency and fabrication ease, their inherent limitations, including non-physiological rigidity and surface properties, have driven the development of hydrogel-based systems that better mimic the extracellular matrix of blood vessels. The integration of microfluidics with biomimetic materials and tissue engineering approaches has led to the development of sophisticated models capable of simulating patient-specific vascular geometries, flow dynamics, and cellular interactions under highly controlled conditions. The advent of 3D bioprinting further enables the creation of complex, multi-layered vascular structures with precise spatial control over geometry and cellular composition. Despite significant progress, challenges remain in achieving long-term stability, incorporating immune components, and translating these models to clinical applications. By providing a comprehensive overview of current advancements and future prospects, this review aims to stimulate further innovation in thrombosis research and accelerate the development of more effective, personalized approaches to thrombosis prevention and treatment.
Collapse
Affiliation(s)
- San Seint Seint Aye
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
| | - Zhongqi Fang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
| | - Mike C L Wu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
| | - Khoon S Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
- School of Medical Sciences, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2006, Australia
- Heart Research Institute, Newtown, NSW 2042, Australia
| |
Collapse
|
19
|
Beresova L, Vitecek J, Protivánková I, Dudka M, Chroma K, Skrott Z, Buchtova T, Poláková K, Novotny J, Novakova L, Bartek J, Mistrik M. Uncovering pre-cytokinetic block in cancer cells under shear stress using a disturbed flow-generating device. Sci Rep 2025; 15:6457. [PMID: 39987149 PMCID: PMC11846833 DOI: 10.1038/s41598-024-83058-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 12/11/2024] [Indexed: 02/24/2025] Open
Abstract
During metastasis, cancer cells navigate through harsh conditions, including various mechanical forces in the bloodstream, highlighting the need to understand the impact of mechanical and shear stresses on cancer cells. To overcome the current methodological limitations of such research, here we present a new device that replicates similar conditions by applying shear stress on cultured cells. The device provides a less complex, easily accessible alternative to traditional fluidics while generating fluid shear stress values comparable to those in human veins and capillaries. The device allows analyses of large cell numbers in standard cell culture flasks and incubators. Using this device to explore the shear stress-induced responses of various human cell lines, we discovered a previously unknown, reversible pre-cytokinetic block occurring in cells that lose anchorage during mitosis and are kept under constant shear stress. Notably, some cancer cell lines appear to bypass this unorthodox cell-cycle block, suggesting its role as a safety checkpoint to restrict the proliferation of cancer cells in the bloodstream and their overall spreading potential. These findings provide new insights into the diverse responses of normal and cancer cells to shear stress and highlight the potential of our technology for research on circulating tumor cells and metastatic spread.
Collapse
Affiliation(s)
- Lucie Beresova
- Laboratory of Genome Integrity, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University, Olomouc, Czech Republic
| | - Jan Vitecek
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Iva Protivánková
- Laboratory of Genome Integrity, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University, Olomouc, Czech Republic
| | - Michal Dudka
- Department of Optics, Faculty of Science, Palacky University, Olomouc, Czech Republic
| | - Katarina Chroma
- Laboratory of Genome Integrity, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University, Olomouc, Czech Republic
| | - Zdenek Skrott
- Laboratory of Genome Integrity, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University, Olomouc, Czech Republic
| | - Tereza Buchtova
- Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
| | - Kamila Poláková
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic
| | - Jan Novotny
- Faculty of Mechanical Engineering, Jan Evangelista Purkyně University, Ústí Nad Labem, Czech Republic
| | - Ludmila Novakova
- Faculty of Mechanical Engineering, Jan Evangelista Purkyně University, Ústí Nad Labem, Czech Republic
| | - Jiri Bartek
- Laboratory of Genome Integrity, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University, Olomouc, Czech Republic
- Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Martin Mistrik
- Laboratory of Genome Integrity, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University, Olomouc, Czech Republic.
| |
Collapse
|
20
|
Ma X, Huang T, Chen X, Li Q, Liao M, Fu L, Huang J, Yuan K, Wang Z, Zeng Y. Molecular mechanisms in liver repair and regeneration: from physiology to therapeutics. Signal Transduct Target Ther 2025; 10:63. [PMID: 39920130 PMCID: PMC11806117 DOI: 10.1038/s41392-024-02104-8] [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: 11/08/2023] [Revised: 09/02/2024] [Accepted: 12/12/2024] [Indexed: 02/09/2025] Open
Abstract
Liver repair and regeneration are crucial physiological responses to hepatic injury and are orchestrated through intricate cellular and molecular networks. This review systematically delineates advancements in the field, emphasizing the essential roles played by diverse liver cell types. Their coordinated actions, supported by complex crosstalk within the liver microenvironment, are pivotal to enhancing regenerative outcomes. Recent molecular investigations have elucidated key signaling pathways involved in liver injury and regeneration. Viewed through the lens of metabolic reprogramming, these pathways highlight how shifts in glucose, lipid, and amino acid metabolism support the cellular functions essential for liver repair and regeneration. An analysis of regenerative variability across pathological states reveals how disease conditions influence these dynamics, guiding the development of novel therapeutic strategies and advanced techniques to enhance liver repair and regeneration. Bridging laboratory findings with practical applications, recent clinical trials highlight the potential of optimizing liver regeneration strategies. These trials offer valuable insights into the effectiveness of novel therapies and underscore significant progress in translational research. In conclusion, this review intricately links molecular insights to therapeutic frontiers, systematically charting the trajectory from fundamental physiological mechanisms to innovative clinical applications in liver repair and regeneration.
Collapse
Affiliation(s)
- Xiao Ma
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiangzheng Chen
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qian Li
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mingheng Liao
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Fu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiwei Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yong Zeng
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
21
|
Ma R, Gong L, Dong C, Utsumi T, Qi J, Zhuang ZW, Zhang X, Yang Y, McConnell MJ, Huang HC, Iwakiri Y. Hepatic Arterial Flow-Induced Portal Tract Fibrosis in Portal Hypertension: The Role of VCAM-1 and Osteopontin-Expressing Macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.634947. [PMID: 39975181 PMCID: PMC11838461 DOI: 10.1101/2025.01.31.634947] [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
Background The liver undergoes significant hemodynamic changes during surgery, transplantation, or cirrhosis with portal hypertension(PH). The hepatic artery buffer response(HABR), which compensates for reduced portal venous flow by increasing hepatic artery(HA) flow, is hypothesized to induce pathological portal tract remodeling. This study investigates the molecular mechanisms underlying this process. Methods PH was induced in Sprague-Dawley rats via partial portal vein ligation(PPVL). Structural evaluation(microCT), immune cell profiling, hemodynamic measurements, and transcriptomic analysis in macrophages(Mϕ) from sham or PPVL rats were conducted. Results MicroCT revealed decreased portal vein flow and increased HA flow correlated with portal pressure(r=0.799, p<0.01). A 2-fold increase in portal tract fibrosis(p<0.001) was observed with increased α-SMA+ myofibroblasts in PPVL rats. CD68+ Mϕ peaked at 10 days post-PPVL, and their depletion significantly reduced fibrosis(p<0.001), indicating critical roles of Mϕ in portal tract remodeling. VCAM-1 was elevated in HA endothelium and portal fibroblasts (PFs); VCAM-1 neutralization reduced collagen accumulation(p<0.05), CD68+ Mϕ(46.3%, p<0.01), and CD3+ T cells(18%, p<0.05). Mϕ-conditioned medium increased VCAM-1 in PFs(8-fold, p<0.001) and enhanced PF migration, while VCAM-1 knockdown reduced this effect (p<0.01). Single-cell RNA sequencing data(GSE171904) and RNA-FISH revealed increased interactions between osteopontin (Spp1)+ Mϕ and PFs, with Spp1+ Mϕ driving fibrosis. Spp1 knockdown in Mϕ co-culture reduced PF fibrogenic markers, while recombinant Spp1 upregulated Col1a1, Fn1, and Acta2 expression in PFs. Conclusion Increased VCAM-1 in arterial endothelial cells and PFs facilitates the recruitment of Spp1+ Mϕ, which drive HA flow-mediated vascular remodeling and portal tract fibrosis. These findings highlight arterial flow-induced fibrosis as a key mechanism in PH, potentially contributing to disease progression and decompensation. Synopsis Liver hemodynamic changes in portal hypertension drive extracellular matrix accumulation and portal tract remodeling via Spp1+ macrophages. This study highlights how altered blood flow induces fibrosis, and its potential role in decompensation, and identifies therapeutic targets for advanced liver disease.
Collapse
|
22
|
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
|
23
|
Liu MY, Wang M, Liu J, Sun AQ, He CS, Cong X, Kong W, Li W. Hemodynamic disturbance and mTORC1 activation: Unveiling the biomechanical pathogenesis of thoracic aortic aneurysms in Marfan syndrome. J Pharm Anal 2025; 15:101120. [PMID: 39989903 PMCID: PMC11847113 DOI: 10.1016/j.jpha.2024.101120] [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: 03/27/2024] [Revised: 09/19/2024] [Accepted: 10/10/2024] [Indexed: 02/25/2025] Open
Abstract
Thoracic aortic aneurysm (TAA) significantly endangers the lives of individuals with Marfan syndrome (MFS), yet the intricacies of their biomechanical origins remain elusive. Our investigation delves into the pivotal role of hemodynamic disturbance in the pathogenesis of TAA, with a particular emphasis on the mechanistic contributions of the mammalian target of rapamycin (mTOR) signaling cascade. We uncovered that activation of the mTOR complex 1 (mTORC1) within smooth muscle cells, instigated by the oscillatory wall shear stress (OSS) that stems from disturbed flow (DF), is a catalyst for TAA progression. This revelation was corroborated through both an MFS mouse model (Fbn1 +/C1039G) and clinical MFS specimens. Crucially, our research demonstrates a direct linkage between the activation of the mTORC1 pathway and the intensity in OSS. Therapeutic administration of rapamycin suppresses mTORC1 activity, leading to the attenuation of aberrant SMC behavior, reduced inflammatory infiltration, and restoration of extracellular matrix integrity-collectively decelerating TAA advancement in our mouse model. These insights posit the mTORC1 axis as a strategic target for intervention, offering a novel approach to manage TAAs in MFS and potentially pave insights for current treatment paradigms.
Collapse
Affiliation(s)
- Ming-Yuan Liu
- Department of Vascular Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing Center of Vascular Surgery, Beijing, 100050, China
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Meili Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Junjun Liu
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - An-Qiang Sun
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Chang-Shun He
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Xin Cong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- The Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- The Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
| | - Wei Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, 100044, China
- The Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
| |
Collapse
|
24
|
Arduini A, Fleming SJ, Xiao L, Hall AW, Akkad AD, Chaffin MD, Bendinelli KJ, Tucker NR, Papangeli I, Mantineo H, Flores-Bringas P, Babadi M, Stegmann CM, García-Cardeña G, Lindsay ME, Klattenhoff C, Ellinor PT. Transcriptional profile of the rat cardiovascular system at single-cell resolution. Cell Rep 2025; 44:115091. [PMID: 39709602 PMCID: PMC11781962 DOI: 10.1016/j.celrep.2024.115091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/24/2024] [Accepted: 11/28/2024] [Indexed: 12/24/2024] Open
Abstract
We sought to characterize cellular composition across the cardiovascular system of the healthy Wistar rat, an important model in preclinical cardiovascular research. We performed single-nucleus RNA sequencing (snRNA-seq) in 78 samples in 10 distinct regions, including the four chambers of the heart, ventricular septum, sinoatrial node, atrioventricular node, aorta, pulmonary artery, and pulmonary veins, which produced 505,835 nuclei. We identified 26 distinct cell types and additional subtypes, with different cellular composition across cardiac regions and tissue-specific transcription for each cell type. Several cell subtypes were region specific, including a subtype of vascular smooth muscle cells enriched in the large vasculature. We observed tissue-enriched cellular communication networks, including heightened Nppa-Npr1/2/3 signaling in the sinoatrial node. The existence of tissue-restricted cell types suggests regional regulation of cardiovascular physiology. Our detailed transcriptional characterization of each cell type offers the potential to identify novel therapeutic targets and improve preclinical models of cardiovascular disease.
Collapse
Affiliation(s)
- Alessandro Arduini
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA
| | - Stephen J Fleming
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ling Xiao
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amelia W Hall
- Gene Regulation Observatory, The Broad Institute, Cambridge, MA 02142, USA
| | - Amer-Denis Akkad
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge, MA 02142, USA
| | - Mark D Chaffin
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA
| | - Kayla J Bendinelli
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA
| | | | - Irinna Papangeli
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge, MA 02142, USA
| | - Helene Mantineo
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Mehrtash Babadi
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Guillermo García-Cardeña
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Mark E Lindsay
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Carla Klattenhoff
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge, MA 02142, USA
| | - Patrick T Ellinor
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiology Division, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
25
|
Schröder K. Sodium-Glucose-Cotransporter-2 Inhibitor Therapy and Intermitted Fasting in Cardiorenal Syndrome: The Role of Glucose-Mediated Oxidative Stress. J Clin Med 2025; 14:746. [PMID: 39941418 PMCID: PMC11818847 DOI: 10.3390/jcm14030746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Cardiorenal syndrome (CRS) is a complex clinical disorder characterized by the interplay between heart and kidney dysfunction. This condition is exacerbated by comorbidities such as diabetes mellitus, which contribute to glucose-mediated oxidative stress, further complicating the management of CRS. The management of CRS has evolved with the discovery of sodium-glucose-cotransporter-2 (SGLT2) inhibitors, which have been established as effective agents in reducing hyperglycemia and demonstrated cardiorenal protective effects. Concurrently, intermittent fasting has gained attention as an intervention without pharmacological treatment for its metabolic benefits, including improved glucose metabolism and insulin regulation and sensitivity, both with a potential reduction in oxidative stress. This review provides a summary of current findings on the roles of SGLT2 inhibitors and intermittent fasting in managing CRS, with a particular focus on glucose-mediated oxidative stress. We evaluate the mechanisms by which these interventions exert their effects, identify gaps in current research, and offer recommendations for future studies. While both SGLT2 inhibitors and intermittent fasting demonstrate potential in managing CRS, more research is needed to elucidate their long-term efficacy, safety, and potential synergistic effects.
Collapse
Affiliation(s)
- Katrin Schröder
- Institute of Cardiovascular Physiology, Medical Faculty, Goethe University, 60590 Frankfurt, Germany; ; Tel.: +49-(0)69-6301-83660; Fax: +49-(0)69-6301-7668
- German Center of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| |
Collapse
|
26
|
Miao X, Chen T, Lang Z, Wu Y, Wu X, Zhu Z, Xu RX. Design, fabrication, and application of bioengineering vascular networks based on microfluidic strategies. J Mater Chem B 2025; 13:1252-1269. [PMID: 39691980 DOI: 10.1039/d4tb02047b] [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: 12/19/2024]
Abstract
Vascularization is a critical component of tissue engineering research and is essential for enhancing the success rate of tissue construction and function. Over the past decade, researchers have explored various methods to construct in vitro vascular networks, including 3D printing, cell sphere technology, and microfluidics. Microfluidic technology has garnered significant attention due to its notable advantages in precision, controllability, flexibility, and applicability. It can be primarily classified into two modes: (i) the pre-designed mode, which involves creating vascular networks by pre-designing vascular channels and seeding endothelial cells, encompassing microfluidic chips and microfluidic spinning technologies; and (ii) the self-assembly mode, where cell spheres are fabricated using microfluidic technology and subsequently self-assemble into vascular networks. In this review, we first provide a brief overview of the normal physiological and pathological characteristics of vascular networks, followed by a discussion of the factors to be considered in designing in vitro vascular networks, and conclude with an examination of the classification of technologies for the preparation of microfluidic vascular networks and recent advancements. It is anticipated that in vitro vascular network models will soon be successfully applied in regenerative medicine and drug development.
Collapse
Affiliation(s)
- Xiaoping Miao
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
| | - Tianao Chen
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
| | - Zhongliang Lang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
- Department of Plastic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China.
| | - Yongqi Wu
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
| | - Xizhi Wu
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
| | - Zhiqiang Zhu
- Department of Plastic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China.
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Ronald X Xu
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| |
Collapse
|
27
|
Simões-Faria R, Daems M, Peacock HM, Declercq M, Willems A, Jones EAV, Ghesquière B. Wall shear stress modulates metabolic pathways in endothelial cells. Metabolomics 2025; 21:16. [PMID: 39832080 PMCID: PMC11753319 DOI: 10.1007/s11306-024-02214-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Hemodynamic forces play a crucial role in modulating endothelial cell (EC) behavior, significantly influencing blood vessel responses. While traditional in vitro studies often explore ECs under static conditions, ECs are exposed to various hemodynamic forces in vivo. This study investigates how wall shear stress (WSS) influences EC metabolism, focusing on the interplay between WSS and key metabolic pathways. OBJECTIVES The aim of this study is to examine the effects of WSS on EC metabolism, specifically evaluating its impact on central carbon metabolism and glycolysis using transcriptomics and tracer metabolomics approaches. METHODS ECs were exposed to WSS, and transcriptomic analysis was performed to assess gene expression changes related to metabolic pathways. Tracer metabolomics was used to track metabolic fluxes, focusing on glutamine and glycolytic metabolism. Additionally, chemical inhibition of glutamate dehydrogenase was conducted to evaluate its role in EC fitness under WSS. RESULTS Transcriptomic data revealed upregulation of glutamine and glutamate pathways, alongside downregulation of glycolytic activity in ECs exposed to WSS. Tracer metabolomics confirmed that WSS promotes glutamine anaplerosis into the Krebs cycle, while decreasing glycolytic metabolism. Suppression of glutamate dehydrogenase impaired EC fitness under WSS conditions. CONCLUSION Our findings illuminate that ECs subjected to WSS exhibit a preference for glutamine as a key nutrient source for central carbon metabolism pathways, indicating diminished reliance on glycolysis. This study elucidates the nutritional predilections and regulatory mechanisms governing EC metabolism under WSS in vitro, underscoring the pivotal role of physical stimuli in shaping EC metabolic responses.
Collapse
Affiliation(s)
- Rita Simões-Faria
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Margo Daems
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Hanna M Peacock
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Mathias Declercq
- Department of Development and Regeneration, CF Centre, Woman and Child, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Anton Willems
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Elizabeth A V Jones
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Bart Ghesquière
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
- Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium.
| |
Collapse
|
28
|
Nakagami T, Kato Y, Watanabe K, Kimura R, Sato T, Hayashi T, Suda S. Repeated Ischemic Stroke due to in situ Thrombus at the Middle Cerebral Artery in a Patient with Essential Thrombocythemia. Intern Med 2025; 64:281-285. [PMID: 38811226 PMCID: PMC11802207 DOI: 10.2169/internalmedicine.3590-24] [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: 02/02/2024] [Accepted: 04/05/2024] [Indexed: 05/31/2024] Open
Abstract
Essential thrombocythemia (ET) is a myeloproliferative neoplasm that is a rare cause of ischemic stroke. We herein report a 70-year-old man with JAK2 V617F mutation-positive ET who experienced ischemic stroke twice in 1 month due to transient stenosis. In both events, transient stenosis formed at the same curvature of the right middle cerebral artery, and the thrombus disappeared with the initiation of antiplatelet agents. The formation of in situ thrombus at the curvature of the intracranial vessels may be a unique characteristic of JAK2 V617F mutation-positive ET patients.
Collapse
Affiliation(s)
- Toru Nakagami
- Department of Neurology and Cerebrovascular Medicine, Saitama Medical University International Medical Center, Japan
| | - Yuji Kato
- Department of Neurology and Cerebrovascular Medicine, Saitama Medical University International Medical Center, Japan
| | - Kaito Watanabe
- Department of Neurology and Cerebrovascular Medicine, Saitama Medical University International Medical Center, Japan
| | - Ryutaro Kimura
- Department of Neurology and Cerebrovascular Medicine, Saitama Medical University International Medical Center, Japan
| | - Tsugumi Sato
- Department of Pathology, Saitama Medical University International Medical Center, Japan
| | - Takeshi Hayashi
- Department of Neurology and Cerebrovascular Medicine, Saitama Medical University International Medical Center, Japan
| | - Satoshi Suda
- Department of Neurology and Cerebrovascular Medicine, Saitama Medical University International Medical Center, Japan
| |
Collapse
|
29
|
Wang HS, Hsu BG, Wang JH, Yang CF. Increased serum galectin-3 level is associated with endothelial dysfunction and cardiovascular events in patients with hypertension. Heliyon 2025; 11:e41111. [PMID: 39758383 PMCID: PMC11699377 DOI: 10.1016/j.heliyon.2024.e41111] [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: 03/28/2024] [Revised: 09/15/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Background Endothelial dysfunction can lead to various harmful cardiovascular complications. The importance of galectin-3 (Gal-3) has been proposed in some cardiac diseases related to chronic inflammation. However, its role in hypertension-induced endothelial dysfunction remains unclear. Methods We enrolled 120 patients with hypertension, assessed their baseline characteristics, and monitored their 7-year cardiovascular outcomes. We performed an enzyme-linked immunosorbent assay to measure serum Gal-3 levels. The vascular reactivity index (VRI) was examined by digital thermal monitoring. Patients with VRI <1.0, 1.0 to <2.0, and ≥2.0 were defined as having poor, intermediate, and good vascular reactivity, respectively. Results Among the recruited patients, 12 had poor vascular reactivity, 57 had intermediate vascular reactivity, and 51 had good vascular reactivity. Older age, higher total cholesterol levels, higher low-density lipoprotein cholesterol levels, lower estimated glomerular filtration rate, and higher Gal-3 levels were associated with poor endothelial dysfunction. Multivariate linear regression analysis showed that age and Gal-3 levels were correlated with VRI. During the 7-year follow-up period, patients with higher Gal-3 levels had more cardiovascular events. Conclusions Higher Gal-3 levels are associated with endothelial dysfunction and unfavorable cardiovascular outcomes in patients with hypertension, suggesting its potential role in the hypertension-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Hui-Sheng Wang
- Division of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Bang-Gee Hsu
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ji-Hung Wang
- Division of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Cardiovascular Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chiu-Fen Yang
- Division of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Cardiovascular Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| |
Collapse
|
30
|
Huang J, Fan Y, Wang Y, Liu J. The effects of enhanced external counter-pulsation on post-acute sequelae of COVID-19: A narrative review. Open Med (Wars) 2025; 20:20241067. [PMID: 39802655 PMCID: PMC11716443 DOI: 10.1515/med-2024-1067] [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: 05/17/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 01/16/2025] Open
Abstract
Some of the millions of patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have developed new sequelae after recovering from the initial disease, termed post-acute sequelae of coronavirus disease 2019 (PASC). One symptom is anxiety, which is likely due to three etiologies: brain structural changes, neuroendocrine disruption, and neurotransmitter alterations. This review provides an overview of the current literature on the pathophysiological pathways linking coronavirus disease 2019 to anxiety, as well as the possible mechanisms of action in which an increasingly scrutinized treatment method, enhanced external counter-pulsation (EECP), is able to alleviate anxiety. SARS-CoV-2 triggers increased inflammatory cytokine production, as well as oxidative stress; these processes contribute to the aforementioned three etiologies. The potential treatment approach of EECP, involving sequenced inflation and deflation of specifically-placed airbags, has become of increasing interest, as it has been found to alleviate PASC-associated anxiety by improving patient cardiovascular function. These functional improvements were achieved by EECP stimulating anti-inflammatory and pro-angiogenic processes, as well as improving endothelial cell function and coronary blood flow, partially via counteracting against the negative effects of SARS-CoV-2 infection on the renin-angiotensin-aldosterone system. Therefore, EECP could promote both psychosomatic and cardiac rehabilitation. Further research, though, is still needed to fully determine its benefits and mechanism of action.
Collapse
Affiliation(s)
- Jiecheng Huang
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
| | - Yuxuan Fan
- Department of Rehabilitation, Tongji Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai, China
| | - Yongshun Wang
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
- Department of Cardiology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Jingjin Liu
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
- Department of Cardiology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| |
Collapse
|
31
|
Lim J, Truong HD, Song TY, Giam WJH, Koh EL, Tan JKS. The interdependent hemodynamic influence between abdominal aortic aneurysm and renal artery stenosis. Sci Rep 2024; 14:31986. [PMID: 39738423 PMCID: PMC11685789 DOI: 10.1038/s41598-024-83622-x] [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: 07/23/2024] [Accepted: 12/15/2024] [Indexed: 01/02/2025] Open
Abstract
Cardiovascular diseases remain a leading cause of morbidity and mortality worldwide with abdominal aortic aneurysm (AAA) and renal artery stenosis (RAS) standing out as significant contributors to the vascular pathology spectrum. While these conditions have traditionally been approached as distinct entities, emerging evidence suggests a compelling interdependent relationship between AAA and RAS, challenging the conventional siloed understanding. The confluence of AAA and RAS represents a complex interplay within the cardiovascular system, one that is often overlooked in clinical practice and research. Here, we reveal a bidirectional consequential impact between these two diseases. The location of the AAA sac was investigated for its specific influence on the risk of RAS development. Although studies have shown a higher coincidence between the suprarenal AAA and RAS, our findings demonstrated that the presence of a suprarenal AAA correlated with the lowest risk of RAS development among the three investigated AAA locations. Notably, we also highlighted that the pre-existence of stenosis in the renal artery poses an elevated risk for the formation of suprarenal AAA, assessed by an increased wall shear stress gradient on the aortic wall. Our findings prompt a paradigm shift in the understanding and treatment of AAA and RAS in clinical practice.
Collapse
Affiliation(s)
- Jiaqi Lim
- Department of Biomedical Engineering, National University of Singapore, Block E7 #06-02, 15 Kent Ridge Cres, Singapore, 119276, Singapore
- NUS Graduate School - Integrative Sciences and Engineering Programme, National University of Singapore, 21 Lower Kent Ridge Road, Singapore, 119077, Singapore
- The N.1 Institute for Health, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore
| | - Hung Dong Truong
- Department of Biomedical Engineering, National University of Singapore, Block E7 #06-02, 15 Kent Ridge Cres, Singapore, 119276, Singapore
| | - Tae Yoon Song
- Department of Biomedical Engineering, National University of Singapore, Block E7 #06-02, 15 Kent Ridge Cres, Singapore, 119276, Singapore
| | - Wilkin Jing Han Giam
- Department of Biomedical Engineering, National University of Singapore, Block E7 #06-02, 15 Kent Ridge Cres, Singapore, 119276, Singapore
| | - Evelyn Linyi Koh
- Department of Biomedical Engineering, National University of Singapore, Block E7 #06-02, 15 Kent Ridge Cres, Singapore, 119276, Singapore
| | - Justin Kok Soon Tan
- Department of Biomedical Engineering, National University of Singapore, Block E7 #06-02, 15 Kent Ridge Cres, Singapore, 119276, Singapore.
- The N.1 Institute for Health, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore.
| |
Collapse
|
32
|
Taylor MS, Francis M, Choi CS. Flow-Dependent Modulation of Endothelial Ca 2+ Dynamics by Small Conductance Ca 2+-Activated K + Channels in Mouse Carotid Arteries. Biomedicines 2024; 12:2900. [PMID: 39767806 PMCID: PMC11727411 DOI: 10.3390/biomedicines12122900] [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: 11/19/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Small conductance Ca2+ activated K+ channels (KCa2.3) are important regulators of vascular function. They provide Ca2+-dependent hyperpolarization of the endothelial membrane potential, promoting agonist-induced vasodilation. Another important mechanism of influence may occur through positive feedback regulation of endothelial Ca2+ signals, likely via amplification of influx through membrane cation channels. KCa2.3 channels have recently been implicated in flow-mediated dilation of the arterial vasculature and may contribute to the crucial homeostatic role of shear stress in preventing vascular wall remodeling and progressive vascular disease (i.e., atherosclerosis). The impact of KCa2.3 channels on endothelial Ca2+ signaling under physiologically relevant shear stress conditions remains unknown. METHODS In the current study, we employ mice expressing an endothelium-specific Ca2+ fluorophore (cdh5-GCaMP8) to characterize the KCa2.3 channel influence on the dynamic Ca2+ signaling profile along the arterial endothelium in the presence and absence of shear-stress. RESULTS Our data indicate KCa2.3 channels have a minimal influence on basal Ca2+ signaling in the carotid artery endothelium in the absence of flow, but they contribute substantially to amplification of Ca2+ dynamics in the presence of flow and their influence can be augmented through exogenous positive modulation. CONCLUSIONS The findings suggest a pivotal role for KCa2.3 channels in adjusting the profile of homeostatic dynamic Ca2+ signals along the arterial intima under flow.
Collapse
Affiliation(s)
- Mark S. Taylor
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA; (M.F.); (C.-S.C.)
| | | | | |
Collapse
|
33
|
Yu Y, Yuan H, Han Q, Shi J, Liu X, Xue Y, Li Y. SMOC2, OGN, FCN3, and SERPINA3 could be biomarkers for the evaluation of acute decompensated heart failure caused by venous congestion. Front Cardiovasc Med 2024; 11:1406662. [PMID: 39717447 PMCID: PMC11663912 DOI: 10.3389/fcvm.2024.1406662] [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: 03/25/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024] Open
Abstract
Background Venous congestion (VC) sets in weeks before visible clinical decompensation, progressively increasing cardiac strain and leading to acute heart failure (HF) decompensation. Currently, the field lacks a universally acknowledged gold standard and early detection methods for VC. Methods Using data from the GEO database, we identified VC's impact on HF through key genes using Limma and STRING databases. The potential mechanisms of HF exacerbation were explored via GO and KEGG enrichment analyses. Diagnostic genes for acute decompensated HF were discovered using LASSO, RF, and SVM-REF machine learning algorithms, complemented by single-gene GSEA analysis. A nomogram tool was developed for the diagnostic model's evaluation and application, with validation conducted on external datasets. Results Our findings reveal that VC influences 37 genes impacting HF via 8 genes, primarily affecting oxygen transport, binding, and extracellular matrix stability. Four diagnostic genes for HF's pre-decompensation phase were identified: SMOC2, OGN, FCN3, and SERPINA3. These genes showed high diagnostic potential, with AUCs for each gene exceeding 0.9 and a genomic AUC of 0.942. Conclusions Our study identifies four critical diagnostic genes for HF's pre-decompensated phase using bioinformatics and machine learning, shedding light on the molecular mechanisms through which VC worsens HF. It offers a novel approach for clinical evaluation of acute decompensated HF patient congestion status, presenting fresh insights into its pathogenesis, diagnosis, and treatment.
Collapse
Affiliation(s)
- Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huajing Yuan
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Quancheng Han
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingle Shi
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiujuan Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
34
|
Zhao Y, Cao H, Wei Y, Zheng T. The impact of different degrees of stenosis on platelet deposition in the left anterior descending branch of the coronary artery. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108445. [PMID: 39369586 DOI: 10.1016/j.cmpb.2024.108445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/15/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND AND OBJECTIVE This study aimed to investigate the impact of different stenotic degrees on platelet deposition in the left anterior descending branch of the coronary artery. METHODS The idealized model of coronary artery stenosis of 30 %, 40 %, 50 %, 60 %, 70 % and four patient-specific models of 22.17 %, 34.88 %, 51.23 % and 62.96 % were established. A discrete phase model was used to calculate the deposition of platelet particles in blood. RESULTS (1) As the stenotic degree increased from 30 % to 70 %, the maximum deposition rates were 4.23e-2 kg/(m2 ·s), 3.47e-2 kg/(m2 ·s), 0.14 kg/(m2 ·s), 0.15 kg/(m2 ·s), and 0.38 kg/(m2 ·s), respectively. (2) The greater the stenotic degree, the more points of platelet deposition. (3) Platelets were mainly deposited at the proximal segment of mild stenosis. When the stenotic degree exceeded 50 %, the deposition position moved to the distal segment of the stenosis. (4) The results in the real coronary artery models were similar to those in the idealized model. CONCLUSION The study suggests that the location and number of platelet deposition are related to the degree of stenosis. Moderate to severe stenosis is more likely to spread downstream.
Collapse
Affiliation(s)
- Yiming Zhao
- Sichuan-tibet Railway Co. LTD, Chengdu 610036, China
| | - Haoyao Cao
- Department of Mechanics & Engineering, College Architecture & Environment, Sichuan University, Chengdu 610065, China; Sichuan University Yibin Park / Yibin Institute of Industrial Technology, Yibin 644000, China
| | - Yongtao Wei
- Department of Mechanics & Engineering, College Architecture & Environment, Sichuan University, Chengdu 610065, China.
| | - Tinghui Zheng
- Department of Mechanics & Engineering, College Architecture & Environment, Sichuan University, Chengdu 610065, China; Med-X Center for Informatics, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
35
|
Wang Y, Chatterjee E, Li G, Xu J, Xiao J. Force-sensing protein expression in response to cardiovascular mechanotransduction. EBioMedicine 2024; 110:105412. [PMID: 39481337 PMCID: PMC11554632 DOI: 10.1016/j.ebiom.2024.105412] [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: 07/12/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Force-sensing biophysical cues in microenvironment, including extracellular matrix performances, stretch-mediated mechanics, shear stress and flow-induced hemodynamics, have a significant influence in regulating vascular morphogenesis and cardiac remodeling by mechanotransduction. Once cells perceive these extracellular mechanical stimuli, Piezo activation promotes calcium influx by forming integrin-adhesion-coupling receptors. This induces robust contractility of cytoskeleton structures to further transmit biomechanical alternations into nuclei by regulating Hippo-Yes associated protein (YAP) signaling pathway between cytoplasmic and nuclear translocation. Although biomechanical stimuli are widely studied in cardiovascular diseases, the expression of force-sensing proteins in response to cardiovascular mechanotransduction has not been systematically concluded. Therefore, this review will summarize the force-sensing Piezo, cytoskeleton and YAP proteins to mediate extracellular mechanics, and also give the prominent emphasis on intrinsic connection of these mechanical proteins and cardiovascular mechanotransduction. Extensive insights into cardiovascular mechanics may provide some new strategies for cardiovascular clinical therapy.
Collapse
Affiliation(s)
- Yongtao Wang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China
| | - Emeli Chatterjee
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jiahong Xu
- Department of Cardiology, Shanghai Gongli Hospital, Shanghai 200135, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
36
|
Ma L, Ge P, Zeng C, Liu C, Yin Z, Ya X, Zhai Y, He Q, Li J, Ye X, Zhang Q, Wang R, Zhang D, Zhang Y, Zhao J. Prognostic value of morphology and hemodynamics in moyamoya disease for long-term outcomes and disease progression. Sci Rep 2024; 14:28182. [PMID: 39548256 PMCID: PMC11568140 DOI: 10.1038/s41598-024-79608-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024] Open
Abstract
To explore the relationship between morphological and hemodynamic parameters, baseline characteristics, and long-term outcomes in patients with moyamoya disease (MMD) using a computational fluid dynamics model. We retrospectively reviewed 129 patients at Beijing Tiantan hospital between July 2020 and December 2021. Perioperative clinical variables and Suzuki stage were recorded. Logistic regression analysis was employed to identify the risk factors for unfavorable long-term outcomes. The association between morphological, CT perfusion parameters, hemodynamic parameters and the Suzuki stage, clinical variables of MMD was also analyzed. Patients with high relative Wall Shear Stress (rWSS) were older and had more cases with higher Suzuki stage and worse follow-up mRS scores (p < 0.05). High rWSS at the terminal ICA and diabetes mellitus were identified as independent predictors of unfavorable long-term outcomes [OR = 3.039(1.191-7.754), p = 0.020; OR = 3.164(1.141-8.723), p = 0.027, respectively]. ROC analysis demonstrated that predictive models incorporating rWSS improved AUC values, with the highest AUC in Model 2 (AUC = 0.889). High rWSS was significantly associated with future TIA and stroke events (p = 0.032). We speculated that high rWSS and diabetes mellitus were independent risk factors for unfavorable long-term outcomes in patients with MMD. rWSS and morphological parameters are crucial for predicting MMD progression and understanding its pathogenesis.
Collapse
Affiliation(s)
- Long Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Peicong Ge
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| | - Chaofan Zeng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zihan Yin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiaolong Ya
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yuanren Zhai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Junsheng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xun Ye
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Dong Zhang
- Department of Neurosurgery, Beijing Hospital, Beijing, China
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
37
|
Li M, Wu J, Yang T, Zhao Y, Ren P, Chang L, Shi P, Yang J, Liu Y, Li X, Wang P, Cao Y. Engineered Biomimetic Nanoparticles-Mediated Targeting Delivery of Allicin Against Myocardial Ischemia-Reperfusion Injury by Inhibiting Ferroptosis. Int J Nanomedicine 2024; 19:11275-11292. [PMID: 39524923 PMCID: PMC11550785 DOI: 10.2147/ijn.s478276] [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: 05/14/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Background Cardiac microvascular damage is substantially related with the onset of myocardial ischaemia-reperfusion (IR) injury. Reportedly, allicin (AL) effectively protects the cardiac microvascular system from IR injury. However, the unsatisfactory therapeutic efficacy of current drugs and insufficient drug delivery to the damaged heart are major concerns. Here, inspired by the natural interaction between neutrophils and inflamed cardiac microvascular endothelial cells (CMECs), a neutrophil membrane-camouflaged nanoparticle for non-invasive active-targeting therapy for IR injury by improving drug delivery to the injured heart is constructed. Methods In this study, we engineered mesoporous silica nanoparticles (MSNs) coated with a neutrophil membrane to act as a drug delivery system, encapsulating AL. The potential of the nanoparticles (named AL@MSNs@NM) for specific targeting of infarcted myocardium was assessed using small animal vivo imaging system. The cardiac function of AL@MSNs@NM after treatment was evaluated by Animal Ultrasound Imaging system, HE staining, and Laser Speckle Imaging System. The therapeutic mechanism was analyzed by ELISA kits, immunofluorescence, and PCR. Results We discovered that AL@MSNs@NM significantly improves cardiac function index, reduced infarct size and fibrosis, increased vascular perfusion in ischemic areas, and also promoted the function of CMECs, including migration, tube formation, shear stress adaptation, and nitric oxide production. Further research revealed that AL@MSNs@NM have cardio-protective functions in IR rats by inhibiting CMEC ferroptosis and increasing platelet endothelial cell adhesion molecule-1 (PECAM-1) expression. Conclusion Our results indicated that AL@MSNs@NM significantly reversed CMEC ferroptosis and increased PECAM-1 expression, enhanced cardiac function, and reduced myocardial infarction size. Therefore, this strategy demonstrates that engineered biomimetic nanotechnology effectively delivers AL for targeted therapy of myocardial infarction.
Collapse
Affiliation(s)
- Minghui Li
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Jiabi Wu
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Tao Yang
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Yuhang Zhao
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Ping Ren
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Lingling Chang
- Department of Physiology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Pilong Shi
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Jing Yang
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Yuhang Liu
- Department of Physiology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Xiaolei Li
- Department of Pathology, Jiangsu College of Nursing, Jiangsu, 223003, People’s Republic of China
| | - Peng Wang
- Department of Physiology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Yonggang Cao
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| |
Collapse
|
38
|
Alateeq K, Walsh EI, Cherbuin N. High Blood Pressure and Impaired Brain Health: Investigating the Neuroprotective Potential of Magnesium. Int J Mol Sci 2024; 25:11859. [PMID: 39595928 PMCID: PMC11594239 DOI: 10.3390/ijms252211859] [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: 09/07/2024] [Revised: 10/27/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
High blood pressure (BP) is a significant contributor to the disease burden globally and is emerging as an important cause of morbidity and mortality in the young as well as the old. The well-established impact of high BP on neurodegeneration, cognitive impairment, and dementia is widely acknowledged. However, the influence of BP across its full range remains unclear. This review aims to explore in more detail the effects of BP levels on neurodegeneration, cognitive function, and dementia. Moreover, given the pressing need to identify strategies to reduce BP levels, particular attention is placed on reviewing the role of magnesium (Mg) in ageing and its capacity to lower BP levels, and therefore potentially promote brain health. Overall, the review aims to provide a comprehensive synthesis of the evidence linking BP, Mg and brain health. It is hoped that these insights will inform the development of cost-effective and scalable interventions to protect brain health in the ageing population.
Collapse
Affiliation(s)
- Khawlah Alateeq
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT 2601, Australia; (K.A.); (E.I.W.)
- Radiological Science, College of Applied Medical Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Erin I. Walsh
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT 2601, Australia; (K.A.); (E.I.W.)
| | - Nicolas Cherbuin
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT 2601, Australia; (K.A.); (E.I.W.)
| |
Collapse
|
39
|
Xu Z, Rasteh AM, Dong A, Wang P, Liu H. Identification of molecular targets of Hypericum perforatum in blood for major depressive disorder: a machine-learning pharmacological study. Chin Med 2024; 19:141. [PMID: 39385284 PMCID: PMC11465934 DOI: 10.1186/s13020-024-01018-5] [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: 06/05/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is one of the most common psychiatric disorders worldwide. Hypericum perforatum (HP) is a traditional herb that has been shown to have antidepressant effects, but its mechanism is unclear. This study aims to identify the molecular targets of HP for the treatment of MDD. METHODS We performed differential analysis and weighted gene co-expression network analysis (WGCNA) with blood mRNA expression cohort of MDD and healthy control to identify DEGs and significant module genes (gene list 1). Three databases, CTD, DisGeNET, and GeneCards, were used to retrieve MDD-related gene intersections to obtain MDD-predicted targets (gene list 2). The validated targets were retrieved from the TCMSP database (gene list 3). Based on these three gene lists, 13 key pathways were identified. The PPI network was constructed by extracting the intersection of genes and HP-validated targets on all key pathways. Key therapeutic targets were obtained using MCODE and machine learning (LASSO, SVM-RFE). Clinical diagnostic assessments (Nomogram, Correlation, Intergroup expression), and gene set enrichment analysis (GSEA) were performed for the key targets. In addition, immune cell analysis was performed on the blood mRNA expression cohort of MDD to explore the association between the key targets and immune cells. Finally, molecular docking prediction was performed for the targets of HP active ingredients on MDD. RESULTS Differential expression analysis and WGCNA module analysis yielded 933 potential targets for MDD. Three disease databases were intersected with 982 MDD-predicted targets. The TCMSP retrieved 275 valid targets for HP. Separate enrichment analysis intersected 13 key pathways. Five key targets (AKT1, MAPK1, MYC, EGF, HSP90AA1) were finally screened based on all enriched genes and HP valid targets. Combined with the signaling pathway and immune cell analysis suggested the effect of peripheral immunity on MDD and the important role of neutrophils in immune inflammation. Finally, the binding of HP active ingredients (quercetin, kaempferol, and luteolin) and all 5 key targets were predicted based on molecular docking. CONCLUSIONS The active constituents of Hypericum perforatum can act on MDD and key targets and pathways of this action were identified.
Collapse
Affiliation(s)
- Zewen Xu
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | | | | | - Panpan Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Hengrui Liu
- Cancer Research Institute, Jinan University, Guangzhou, China.
- Tianjin Yinuo Biomedical Co., Ltd, Tianjin, China.
| |
Collapse
|
40
|
Mote N, Kubik S, Polacheck WJ, Baker BM, Trappmann B. A nanoporous hydrogel-based model to study chemokine gradient-driven angiogenesis under luminal flow. LAB ON A CHIP 2024; 24:4892-4906. [PMID: 39308400 DOI: 10.1039/d4lc00460d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
The growth of new blood vessels through angiogenesis is a highly coordinated process, which is initiated by chemokine gradients that activate endothelial cells within a perfused parent vessel to sprout into the surrounding 3D tissue matrix. While both biochemical signals from pro-angiogenic factors, as well as mechanical cues originating from luminal fluid flow that exerts shear stress on the vessel wall, have individually been identified as major regulators of endothelial cell sprouting, it remains unclear whether and how both types of cues synergize. To fill this knowledge gap, here, we created a 3D biomimetic model of chemokine gradient-driven angiogenic sprouting, in which a micromolded tube inside a hydrogel matrix is seeded with endothelial cells and connected to a perfusion system to control fluid flow rates and resulting shear forces on the vessel wall. To allow for the formation of chemokine gradients despite the presence of luminal flow, a nanoporous synthetic hydrogel that supports angiogenesis but limits the interstitial flow proved crucial. Using this system, we find that luminal flow and resulting shear stress is a major regulator of the speed and morphogenesis of angiogenic sprouting, whose action is mediated through changes in vascular permeability.
Collapse
Affiliation(s)
- Nidhi Mote
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Sarah Kubik
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, 27514 USA
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, 27514 USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, 2174 Lurie BME Building, 1101 Beal Avenue, Ann Arbor, MI, 48109 USA
| | - Britta Trappmann
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 6, 44227 Dortmund, Germany.
| |
Collapse
|
41
|
Ya X, Ma L, Li H, Ge P, Zheng Z, Mou S, Liu C, Zhang Y, Wang R, Zhang Q, Ye X, Zhang D, Zhao J. Exploring the relationship between hemodynamics and the immune microenvironment in carotid atherosclerosis: Insights from CFD and CyTOF technologies. J Cereb Blood Flow Metab 2024; 44:1733-1744. [PMID: 38833561 PMCID: PMC11494853 DOI: 10.1177/0271678x241251976] [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: 10/13/2023] [Revised: 02/24/2024] [Accepted: 03/28/2024] [Indexed: 06/06/2024]
Abstract
Carotid atherosclerosis is a major cause of stroke. Hemodynamic forces, such as shear stress and oscillatory shear, play an important role in the initiation and progression of atherosclerosis. The alteration of the immune microenvironment is the fundamental pathological mechanism by which diverse external environmental factors impact the formation and progression of plaques. However, Current research on the relationship between hemodynamics and immunity in atherosclerosis still lack of comprehensive understanding. In this study, we combined computational fluid dynamics (CFD) and Mass cytometry (CyTOF) technologies to explore the changes in the immune microenvironment within plaques under different hemodynamic conditions. Our results indicated that neutrophils were enriched in adverse flow environments. M2-like CD163+CD86+ macrophages were predominantly enriched in high WSS and low OSI environments, while CD163-CD14+ macrophages were enriched in low WSS and high OSI environments. Functional analysis further revealed T cell pro-inflammatory activation and dysregulation in modulation, along with an imbalance in M1-like/M2-like macrophages, suggesting their potential involvement in the progression of atherosclerotic lesions mediated by adverse flow patterns. Our study elucidated the potential mechanisms by which hemodynamics regulated the immune microenvironment within plaques, providing intervention targets for future precision therapies.
Collapse
Affiliation(s)
- Xiaolong Ya
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Long Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Hao Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Peicong Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhiyao Zheng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Siqi Mou
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xun Ye
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Dong Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
42
|
Limbu S, McCloskey KE. An Endothelial Cell Is Not Simply an Endothelial Cell. Stem Cells Dev 2024; 33:517-527. [PMID: 39030822 PMCID: PMC11564855 DOI: 10.1089/scd.2024.0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/18/2024] [Indexed: 07/22/2024] Open
Abstract
Endothelial cells (ECs) are a multifaceted component of the vascular system with roles in immunity, maintaining tissue fluid balance, and vascular tone. Dysregulation or dysfunction of ECs can have far-reaching implications, leading pathologies ranging from cardiovascular diseases, such as hypertension and atherosclerosis, ischemia, chronic kidney disease, blood-brain barrier integrity, dementia, and tumor metastasis. Recent advancements in regenerative medicine have highlighted the potential of stem cell-derived ECs, particularly from induced pluripotent stem cells, to treat ischemic tissues, as well as models of vascular integrity. This review summarizes what is known in the generation of ECs with an emphasis on tissue-specific ECs and EC subphenotypes important in the development of targeted cell-based therapies for patient treatment.
Collapse
Affiliation(s)
- Shiwani Limbu
- Quantitative and System Biology Graduate Program, University of California, Merced, USA
| | - Kara E. McCloskey
- Quantitative and System Biology Graduate Program, University of California, Merced, USA
- Materials Science and Engineering Department, University of California, Merced, USA
| |
Collapse
|
43
|
Wang Y, Liu M, Zhang W, Liu H, Jin F, Mao S, Han C, Wang X. Mechanical strategies to promote vascularization for tissue engineering and regenerative medicine. BURNS & TRAUMA 2024; 12:tkae039. [PMID: 39350780 PMCID: PMC11441985 DOI: 10.1093/burnst/tkae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 10/04/2024]
Abstract
Vascularization is a major challenge in the field of tissue engineering and regenerative medicine. Mechanical factors have been demonstrated to play a fundamental role in vasculogenesis and angiogenesis and can affect the architecture of the generated vascular network. Through the regulation of mechanical factors in engineered tissues, various mechanical strategies can be used to optimize the preformed vascular network and promote its rapid integration with host vessels. Optimization of the mechanical properties of scaffolds, including controlling scaffold stiffness, increasing surface roughness and anisotropic structure, and designing interconnected, hierarchical pore structures, is beneficial for the in vitro formation of vascular networks and the ingrowth of host blood vessels. The incorporation of hollow channels into scaffolds promotes the formation of patterned vascular networks. Dynamic stretching and perfusion can facilitate the formation and maturation of preformed vascular networks in vitro. Several indirect mechanical strategies provide sustained mechanical stimulation to engineered tissues in vivo, which further promotes the vascularization of implants within the body. Additionally, stiffness gradients, anisotropic substrates and hollow channels in scaffolds, as well as external cyclic stretch, boundary constraints and dynamic flow culture, can effectively regulate the alignment of vascular networks, thereby promoting better integration of prevascularized engineered tissues with host blood vessels. This review summarizes the influence and contribution of both scaffold-based and external stimulus-based mechanical strategies for vascularization in tissue engineering and elucidates the underlying mechanisms involved.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Meixuan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Wei Zhang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Huan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Fang Jin
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Shulei Mao
- Department of Burns and Plastic Surgery, Quhua Hospital of Zhejiang, 62 Wenchang Road, Quhua, Quzhou 324004, China
| | - Chunmao Han
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Xingang Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| |
Collapse
|
44
|
Chen L, Qu H, Liu B, Chen BC, Yang Z, Shi DZ, Zhang Y. Low or oscillatory shear stress and endothelial permeability in atherosclerosis. Front Physiol 2024; 15:1432719. [PMID: 39314624 PMCID: PMC11417040 DOI: 10.3389/fphys.2024.1432719] [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: 05/14/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Endothelial shear stress is a tangential stress derived from the friction of the flowing blood on the endothelial surface of the arterial wall and is expressed in units of force/unit area (dyne/cm2). Branches and bends of arteries are exposed to complex blood flow patterns that generate low or oscillatory endothelial shear stress, which impairs glycocalyx integrity, cytoskeleton arrangement and endothelial junctions (adherens junctions, tight junctions, gap junctions), thus increasing endothelial permeability. The lipoproteins and inflammatory cells penetrating intima due to the increased endothelial permeability characterizes the pathological changes in early stage of atherosclerosis. Endothelial cells are critical sensors of shear stress, however, the mechanisms by which the complex shear stress regulate endothelial permeability in atherosclerosis remain unclear. In this review, we focus on the molecular mechanisms of the endothelial permeability induced by low or oscillatory shear stress, which will shed a novel sight in early stage of atherosclerosis.
Collapse
Affiliation(s)
- Li Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Hua Qu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine, Beijing, China
| | - Bin Liu
- The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Bing-Chang Chen
- Graduate school, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Zhen Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Da-Zhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Ying Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| |
Collapse
|
45
|
Okamoto T, Hashimoto Y, Iemitsu M, Ogoh S. Acute effects of static stretching exercise-induced decrease in arterial stiffness on maximal aerobic capacity. J Sports Med Phys Fitness 2024; 64:849-856. [PMID: 38842370 DOI: 10.23736/s0022-4707.24.15758-1] [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: 06/07/2024]
Abstract
BACKGROUND We recently have reported that individual day-to-day arterial stiffness variations are associated with maximal aerobic capacity. However, the evidence of this phenomenon was not provided sufficiently. The present study aimed to examine whether a decrease in arterial stiffness through static stretching exercise could enhance maximal aerobic capacity. METHODS Twelve healthy young men (age 22±2 years, mean and standard deviation) participated in this study and underwent two separate sessions in a randomized controlled crossover design: a single session of a whole-body static stretching exercise protocol that involved the trunk, upper limb, and lower limb (stretch condition), and sedentary control where they rested in the exercise room. Brachial-ankle pulse wave velocity (baPWV) was measured as an index of systemic arterial stiffness before, immediately after and at 30 min after both conditions. Maximal oxygen uptake (V̇O2max) was assessed using a graded power test on an electronically braked cycle ergometer after these measurements. RESULTS As we expected, there was a significant decrease in the baPWV at 30 min after the stretch trial compared to baseline values (P=0.01). The baPWV in the stretch condition was lower than that of the control condition, while V̇O2max in the stretch condition was higher than that of the control condition (P=0.03). CONCLUSIONS Based on these findings, it can be inferred that an acute reduction in arterial stiffness may contribute to change in maximal aerobic capacity.
Collapse
Affiliation(s)
- Takanobu Okamoto
- Faculty of Sport Science, Nippon Sport Science University, Tokyo, Japan -
- Research Institute of Sports Science, Nippon Sport Science University, Tokyo, Japan -
| | - Yuto Hashimoto
- Research Institute of Sports Science, Nippon Sport Science University, Tokyo, Japan
| | - Motoyuki Iemitsu
- Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| | - Shigehiko Ogoh
- Department of Biomedical Engineering, Toyo University, Saitama, Japan
| |
Collapse
|
46
|
Allen MF, Park SY, Kwak YS. Oxidative stress and vascular dysfunction: Potential therapeutic targets and therapies in peripheral artery disease. Microvasc Res 2024; 155:104713. [PMID: 38914307 DOI: 10.1016/j.mvr.2024.104713] [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: 11/13/2023] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
Peripheral artery disease (PAD) is the manifestation of atherosclerosis characterized by the accumulation of plaques in the arteries of the lower limbs. Interestingly, growing evidence suggests that the pathology of PAD is multifaceted and encompasses both vascular and skeletal muscle dysfunctions, which contributes to blunted physical capabilities and diminished quality of life. Importantly, it has been suggested that many of these pathological impairments may stem from blunted reduction-oxidation (redox) handling. Of note, in those with PAD, excessive production of reactive oxygen species (ROS) outweighs antioxidant capabilities resulting in oxidative damage, which may have systemic consequences. It has been suggested that antioxidant supplementation may be able to assist in handling ROS. However, the activation of various ROS production sites makes it difficult to determine the efficacy of these antioxidant supplements. Therefore, this review focuses on the common cellular mechanisms that facilitate ROS production and discusses how excessive ROS may impair vascular and skeletal muscle function in PAD. Furthermore, we provide insight for current and potential antioxidant therapies, specifically highlighting activation of the Kelch-like ECH-associated protein 1 (Keap1) - Nuclear Factor Erythroid 2-related factor 2 (Nrf2) pathway as a potential pharmacological therapy to combat ROS accumulation and aid in vascular function, and physical performance in patients with PAD. Altogether, this review provides a better understanding of excessive ROS in the pathophysiology of PAD and enhances our perception of potential therapeutic targets that may improve vascular function, skeletal muscle function, walking capacity, and quality of life in patients with PAD.
Collapse
Affiliation(s)
- Michael F Allen
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE, United States of America
| | - Song-Young Park
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE, United States of America; Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Yi-Sub Kwak
- Department of Physical Education, College of Arts, Design, and Sports Science, Dong-Eui University, Busan, Republic of Korea.
| |
Collapse
|
47
|
Pandian K, Huang L, Junaid A, Harms A, van Zonneveld AJ, Hankemeier T. Tracer-based metabolomics for profiling nitric oxide metabolites in a 3D microvessels-on-chip model. FASEB J 2024; 38:e70005. [PMID: 39171967 DOI: 10.1096/fj.202400553r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/19/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Endothelial dysfunction, prevalent in cardiovascular diseases (CVDs) and linked to conditions like diabetes, hypertension, obesity, renal failure, or hypercholesterolemia, is characterized by diminished nitric oxide (NO) bioavailability-a key signaling molecule for vascular homeostasis. Current two-dimensional (2D) in vitro studies on NO synthesis by endothelial cells (ECs) lack the crucial laminar shear stress, a vital factor in modulating the NO-generating enzyme, endothelial nitric oxide synthase (eNOS), under physiological conditions. Here we developed a tracer-based metabolomics approach to measure NO-specific metabolites with mass spectrometry (MS) and show the impact of fluid flow on metabolic parameters associated with NO synthesis using 2D and 3D platforms. Specifically, we tracked the conversion of stable-isotope labeled NO substrate L-Arginine to L-Citrulline and L-Ornithine to determine eNOS activity. We demonstrated clear responses in human coronary artery endothelial cells (HCAECs) cultured with 13C6, 15N4-L-Arginine, and treated with eNOS stimulator, eNOS inhibitor, and arginase inhibitor. Analysis of downstream metabolites, 13C6, 15N3 L-Citrulline and 13C5, 15N2 L-Ornithine, revealed distinct outcomes. Additionally, we evaluated the NO metabolic status in static 2D culture and 3D microvessel models with bidirectional and unidirectional fluid flow. Our 3D model exhibited significant effects, particularly in microvessels exposed to the eNOS stimulator, as indicated by the 13C6, 15N3 L-Citrulline/13C5, 15N2 L-Ornithine ratio, compared to the 2D culture. The obtained results indicate that the 2D static culture mimics an endothelial dysfunction status, while the 3D model with a unidirectional fluid flow provides a more representative physiological environment that provides a better model to study endothelial dysfunction.
Collapse
Affiliation(s)
- Kanchana Pandian
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Luojiao Huang
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Abidemi Junaid
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Amy Harms
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Thomas Hankemeier
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| |
Collapse
|
48
|
Stanic B, Kokai D, Markovic Filipovic J, Tomanic T, Vukcevic J, Stojkov V, Andric N. Vascular endothelial effects of dibutyl phthalate: In vitro and in vivo evidence. Chem Biol Interact 2024; 399:111120. [PMID: 38944327 DOI: 10.1016/j.cbi.2024.111120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/24/2024] [Accepted: 06/27/2024] [Indexed: 07/01/2024]
Abstract
Dibutyl phthalate (DBP) is widely used in many consumer and personal care products. Here, we report vascular endothelial response to DBP in three different exposure scenarios: after short-term exposure (24 h) of human endothelial cells (ECs) EA.hy926 to 10-6, 10-5, and 10-4 M DBP, long-term exposure (12 weeks) of EA.hy926 cells to 10-9, 10-8, and 10-7 M DBP, and exposure of rats (28 and 90 days) to 100, 500, and 5000 mg DBP/kg food. We examined different vascular functions such as migration of ECs, adhesion of ECs to the extracellular matrix, tube formation, the morphology of rat aorta, as well as several signaling pathways involved in controlling endothelial function. Short-term in vitro exposure to DBP increased migration of ECs through G protein-coupled estrogen receptor, extracellular signal-regulated kinase 1/2, and nitric oxide (NO) signaling and decreased adhesion to gelatin. Long-term in vitro exposure to DBP transiently increased EC migration and had a bidirectional effect on EC adhesion to gelatin and tube formation. These effects were accompanied by a sustained increase in NO production and endothelial NO synthase (eNOS) and Akt activity. In vivo, exposure to DBP for 90 days decreased the aortic wall-to-lumen ratio and increased eNOS and Akt phosphorylation in ECs of rat aorta. This comparative investigation has shown that exposure to DBP may affect vascular function by altering EC migration, adhesion to gelatin, and tube formation after short- and long-term in vitro exposure and by decreasing the aortic wall-to-lumen ratio in vivo. The eNOS-NO and Akt signaling could be important in mediating the effects of DBP in long-term exposure scenarios.
Collapse
Affiliation(s)
- Bojana Stanic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia.
| | - Dunja Kokai
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | | | - Tamara Tomanic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | - Jelena Vukcevic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | - Viktor Stojkov
- University of Novi Sad, Institute of Food Technology, Serbia
| | - Nebojsa Andric
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| |
Collapse
|
49
|
Yu J, Du Q, Li X, Wei W, Fan Y, Zhang J, Chen J. Potential role of endothelial progenitor cells in the pathogenesis and treatment of cerebral aneurysm. Front Cell Neurosci 2024; 18:1456775. [PMID: 39193428 PMCID: PMC11348393 DOI: 10.3389/fncel.2024.1456775] [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: 06/29/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Cerebral aneurysm (CA) is a significant health concern that results from pathological dilations of blood vessels in the brain and can lead to severe and potentially life-threatening conditions. While the pathogenesis of CA is complex, emerging studies suggest that endothelial progenitor cells (EPCs) play a crucial role. In this paper, we conducted a comprehensive literature review to investigate the potential role of EPCs in the pathogenesis and treatment of CA. Current research indicates that a decreased count and dysfunction of EPCs disrupt the balance between endothelial dysfunction and repair, thus increasing the risk of CA formation. Reversing these EPCs abnormalities may reduce the progression of vascular degeneration after aneurysm induction, indicating EPCs as a promising target for developing new therapeutic strategies to facilitate CA repair. This has motivated researchers to develop novel treatment options, including drug applications, endovascular-combined and tissue engineering therapies. Although preclinical studies have shown promising results, there is still a considerable way to go before clinical translation and eventual benefits for patients. Nonetheless, these findings offer hope for improving the treatment and management of this condition.
Collapse
Affiliation(s)
- Jin Yu
- Department of Neurosurgery, Wuhan Asia General Hospital, Wuhan, Hubei, China
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qian Du
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Wei
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuncun Fan
- Department of Respiratory and Critical Care Medicine, Laifeng County People’s Hospital, Enshi, Hubei, China
| | - Jianjian Zhang
- Department of Neurosurgery, Wuhan Asia General Hospital, Wuhan, Hubei, China
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jincao Chen
- Department of Neurosurgery, Wuhan Asia General Hospital, Wuhan, Hubei, China
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
50
|
Ishiko S, Koller A, Deng W, Huang A, Sun D. Liposomal nanocarriers of preassembled glycocalyx restore normal venular permeability and shear stress sensitivity in sepsis: assessed quantitatively with a novel microchamber system. Am J Physiol Heart Circ Physiol 2024; 327:H390-H398. [PMID: 38874615 PMCID: PMC11427114 DOI: 10.1152/ajpheart.00138.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
The endothelial glycocalyx (EG), covering the luminal side of endothelial cells, regulates vascular permeability and senses wall shear stress. In sepsis, EG undergoes degradation leading to increased permeability and edema formation. We hypothesized that restoring EG integrity using liposomal nanocarriers of preassembled glycocalyx (LNPG) will restore normal venular permeability in lipopolysaccharide (LPS)-induced sepsis model of mice. To test this hypothesis, we designed a unique perfusion microchamber in which the permeability of isolated venules could be assessed by measuring the concentration of Evans blue dye (EBD) in microliter samples of extravascular solution (ES). Histamine-induced time- and dose-dependent increases in EBD in the ES could be measured, confirming the sensitivity of the microchamber system. Notably, the histamine-induced increase in permeability was significantly attenuated by histamine receptor (H1) antagonist, triprolidine hydrochloride. Subsequently, mice were treated with LPS or LPS + LNPG. When compared with control mice, venules from LPS-treated mice showed a significant increased permeability, which was significantly reduced by LNPG administration. Moreover, in the presence of wall shear stress, intraluminal administration of LNPG significantly reduced the permeability in isolated venules from LPS-treated mice. We have found no sex differences. In conclusion, our newly developed microchamber system allows us to quantitatively measure the permeability of isolated venules. LPS-induced sepsis increases permeability of mesenteric venules that is attenuated by in vivo LNPG administration, which also reestablished endothelial responses to shear stress. Thus, LNPG presents a promising therapeutic potential for restoring EG function and thereby mitigating vasogenic edema due to increased permeability in sepsis.NEW & NOTEWORTHY In sepsis, the degradation of the endothelial glycocalyx leads to increased venular permeability. In this study, we developed a potentially new therapeutic approach by in vivo administration of liposomal nanocarriers of preassembled glycocalyx to mice, which restored venular sensitivity to wall shear stress and permeability in lipopolysaccharide-induced sepsis, likely by restoring the integrity of the endothelial glycocalyx. Using a new microchamber system, the permeability of Evans blue dye could be quantitatively determined.
Collapse
Affiliation(s)
- Shinya Ishiko
- Department of Medicine, New York Medical College, Valhalla, New York, United States
| | - Akos Koller
- Department of Physiology, New York Medical College, Valhalla, New York, United States
- Institute of Translational Medicine, HUN-RES-SE, Cerebrovascular and Neurocognitive Disorders Research Group, Semmelweis University, Budapest, Hungary
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
- Research Center for Sports Physiology, Hungarian University of Sports Science, Budapest, Hungary
| | - Wensheng Deng
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - An Huang
- Department of Physiology, New York Medical College, Valhalla, New York, United States
| | - Dong Sun
- Department of Physiology, New York Medical College, Valhalla, New York, United States
| |
Collapse
|