1
|
Wang B, Li Y, Ouyang Q, Xu MT, Wang YY, Fu SJ, Liu WQ, Liu XT, Ling H, Zhang X, Xiu RJ, Liu MM. Strain- and sex-dependent variability in hepatic microcirculation and liver function in mice. World J Gastroenterol 2025; 31:101058. [DOI: 10.3748/wjg.v31.i15.101058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/02/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND The integrity and functionality of the hepatic microcirculation are essential for maintaining liver health, which is influenced by sex and genetic background. Understanding these variations is crucial for addressing disparities in liver disease outcomes.
AIM To investigate the sexual dimorphism and genetic heterogeneity of liver microcirculatory function in mice.
METHODS We assessed hepatic microhemodynamics in BALB/c, C57BL/6J, and KM mouse strains using laser Doppler flowmetry and wavelet analysis. We analyzed the serum levels of alanine transaminase, glutamic acid aminotransferase, total bile acid, total protein, alkaline phosphatase, and glucose. Histological and immunohistochemical staining were employed to quantify microvascular density and the expression levels of cluster of differentiation (CD) 31, and estrogen receptor α, and β. Statistical analyses, including the Mantel test and Pearson correlation, were conducted to determine the relationships among hepatic function, microcirculation, and marcocirculation between different sexes and across genetic backgrounds.
RESULTS We identified sex-based disparities in hepatic microhemodynamics across all strains, with males exhibiting higher microvascular perfusion and erythrocyte concentration, but lower blood velocity. Strain-specific differences were evident, particularly in the endothelial oscillatory characteristics of the erythrocyte concentration. No sex-dependent differences in estrogen receptor expression were observed, while significant variations in CD31 expression and microvascular density were observed. The correlations highlighted relationships between hepatic microhemodynamics and liver function indicators.
CONCLUSION Our findings indicate the influence of genetic and sex differences on hepatic microcirculation and liver function, highlighting the necessity of incorporating both genetic background and sex into hepatic physiology studies and potential liver disease management strategies.
Collapse
Affiliation(s)
- Bing Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yuan Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Qin Ouyang
- Department of Pathology, Wangjing Hospital, China Academy of Chinese Medical Science, Beijing 100102, China
| | - Meng-Ting Xu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Ying-Yu Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Sun-Jing Fu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Wei-Qi Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Xue-Ting Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Hao Ling
- Department of Radiology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410004, Hunan Province, China
| | - Xu Zhang
- Laboratory of Electron Microscopy, Ultrastructural Pathology Center, Peking University First Hospital, Beijing 100034, China
| | - Rui-Juan Xiu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Ming-Ming Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
- Diabetes Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| |
Collapse
|
2
|
Rolf-Pissarczyk M, Schussnig R, Fries TP, Fleischmann D, Elefteriades JA, Humphrey JD, Holzapfel GA. Mechanisms of aortic dissection: From pathological changes to experimental and in silico models. PROGRESS IN MATERIALS SCIENCE 2025; 150:101363. [PMID: 39830801 PMCID: PMC11737592 DOI: 10.1016/j.pmatsci.2024.101363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Aortic dissection continues to be responsible for significant morbidity and mortality, although recent advances in medical data assimilation and in experimental and in silico models have improved our understanding of the initiation and progression of the accumulation of blood within the aortic wall. Hence, there remains a pressing necessity for innovative and enhanced models to more accurately characterize the associated pathological changes. Early on, experimental models were employed to uncover mechanisms in aortic dissection, such as hemodynamic changes and alterations in wall microstructure, and to assess the efficacy of medical implants. While experimental models were once the only option available, more recently they are also being used to validate in silico models. Based on an improved understanding of the deteriorated microstructure of the aortic wall, numerous multiscale material models have been proposed in recent decades to study the state of stress in dissected aortas, including the changes associated with damage and failure. Furthermore, when integrated with accessible patient-derived medical data, in silico models prove to be an invaluable tool for identifying correlations between hemodynamics, wall stresses, or thrombus formation in the deteriorated aortic wall. They are also advantageous for model-guided design of medical implants with the aim of evaluating the deployment and migration of implants in patients. Nonetheless, the utility of in silico models depends largely on patient-derived medical data, such as chosen boundary conditions or tissue properties. In this review article, our objective is to provide a thorough summary of medical data elucidating the pathological alterations associated with this disease. Concurrently, we aim to assess experimental models, as well as multiscale material and patient data-informed in silico models, that investigate various aspects of aortic dissection. In conclusion, we present a discourse on future perspectives, encompassing aspects of disease modeling, numerical challenges, and clinical applications, with a particular focus on aortic dissection. The aspiration is to inspire future studies, deepen our comprehension of the disease, and ultimately shape clinical care and treatment decisions.
Collapse
Affiliation(s)
| | - Richard Schussnig
- High-Performance Scientific Computing, University of Augsburg, Germany
- Institute of Structural Analysis, Graz University of Technology, Austria
| | - Thomas-Peter Fries
- Institute of Structural Analysis, Graz University of Technology, Austria
| | - Dominik Fleischmann
- 3D and Quantitative Imaging Laboratory, Department of Radiology, Stanford University, USA
| | | | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, USA
| | - Gerhard A. Holzapfel
- Institute of Biomechanics, Graz University of Technology, Austria
- Department of Structural Engineering, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
3
|
Salimi-Afjani N, Rieben R, Obrist D. Pulsatile-flow culture: a novel system for assessing vascular-cell dynamics. LAB ON A CHIP 2025; 25:1755-1766. [PMID: 40019369 PMCID: PMC11869938 DOI: 10.1039/d4lc00949e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
We describe a model system for vascular-cell culture where recirculating fluid flow in standard culture plates is generated by gravity using a combination of platform tilt and rotation (nutation). Placed inside a cell-culture incubator, variable nutation speeds provide pulsatile shear stresses to vascular cells within the physiological range. The effect of these stresses on cells is demonstrated here using standard laboratory techniques such as immunofluorescent staining, immunoblot, and supernatant analyses. This gravity-driven model framework is well-suited for assessing dynamic conditions for mono- and co-cultures. In addition, the modular design and the use of off-the-shelf components make the system economical and scalable.
Collapse
Affiliation(s)
- Neda Salimi-Afjani
- Department for BioMedical Research, University of Bern, Murtenstrasse 28, 3008 Bern, Switzerland.
- Graduate School for Cellular and Biomedical Sciences, Mittelstrasse 43, 3012 Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research, University of Bern, Murtenstrasse 28, 3008 Bern, Switzerland.
| | - Dominik Obrist
- ARTORG Center for Biomedical Engineering Research, University of Bern, Freiburgstrasse 3, 3010 Bern, Switzerland
| |
Collapse
|
4
|
Speziale P, Foster TJ, Arciola CR. The endothelium at the interface between tissues and Staphylococcus aureus in the bloodstream. Clin Microbiol Rev 2025; 38:e0009824. [PMID: 39807893 PMCID: PMC11905367 DOI: 10.1128/cmr.00098-24] [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: 01/16/2025] Open
Abstract
SUMMARYStaphylococcus aureus is a major human pathogen. It can cause many types of infections, in particular bacteremia, which frequently leads to infective endocarditis, osteomyelitis, sepsis, and other debilitating diseases. The development of secondary infections is based on the bacterium's ability to associate with endothelial cells lining blood vessels. The success of endothelial colonization and infection by S. aureus relies on its ability to express a wide array of cell wall-anchored and secreted virulence factors. Establishment of endothelial infection by the pathogen is a multistep process involving adhesion, invasion, extravasation, and dissemination of the bacterium into surrounding tissues. The process is dependent on the type of endothelium in different organs (tissues) and pathogenetic potential of the individual strains. In this review, we report an update on the organization of the endothelium in the vessels, the structure and function of the virulence factors of S. aureus, and the several aspects of bacteria-endothelial cell interactions. After these sections, we will discuss recent advances in understanding the specific mechanisms of infections that develop in the heart, bone and joints, lung, and brain. Finally, we describe how neutrophils bind to endothelial cells, migrate to the site of infection to kill bacteria in the tissues, and how staphylococci counteract neutrophils' actions. Knowledge of the molecular details of S. aureus-endothelial cell interactions will promote the development of new therapeutic strategies and tools to combat this formidable pathogen.
Collapse
Affiliation(s)
- Pietro Speziale
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Pavia, Italy
| | - Timothy J Foster
- Department of Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Carla Renata Arciola
- Laboratory of Pathology of Implant Infections, Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
5
|
Ghorbanpour S, Cartland SP, Chen H, Seth S, Ecker RC, Richards C, Aksentijevic D, Padula MP, Cole L, Warkiani ME, Kavurma MM, McClements L. The FKBPL-based therapeutic peptide, AD-01, protects the endothelium from hypoxia-induced damage by stabilising hypoxia inducible factor-α and inflammation. J Transl Med 2025; 23:309. [PMID: 40069829 PMCID: PMC11895374 DOI: 10.1186/s12967-025-06118-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 01/08/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Endothelial dysfunction is a hallmark feature of cardiovascular disease (CVD), yet the underlying mechanisms are still poorly understood. This has impeded the development of effective therapies, particularly for peripheral artery disease. FK506-binding protein like (FKBPL) and its therapeutic peptide mimetic, AD-01, are crucial negative regulators of angiogenesis, however their roles in CVD are unknown. In this study, we aimed to elucidate the FKBPL-mediated mechanisms involved in regulating endothelial dysfunction induced by hypoxia or inflammation, and to determine whether AD-01 can effectively restore endothelial function under these conditions. METHODS Hindlimb ischemia was induced in mice by ligating the proximal and distal ends of the right femoral artery, and, after three days, the gastrocnemius muscle was collected for immunofluorescence staining, and RNA extraction. A 3D in vitro microfluidics model was developed to determine the endothelial cell migration and impact of FKBPL following treatments with: (i) 24 µM FKBPL targeted siRNA, (ii) 1 mM hypoxia inducible factor (HIF-1)α activator (DMOG), (iii) 50% (v/v) macrophage conditioned media (MCM), ± 100 nM AD-01. Unbiased, untargeted proteomic analysis was conducted via LC-MS/MS to identify protein targets of AD-01. RESULTS FKBPL expression is substantially downregulated in mice after hindlimb ischemia (p < 0.05, protein; p < 0.001, mRNA), correlating with increased neovascularization and altered vascular adhesion molecule expression. In our real-time advanced 3D microfluidics model, hypoxia suppressed FKBPL (p < 0.05) and VE-cadherin (p < 0.001) expression, leading to increased endothelial cell number and migration (p < 0.001), which was restored by AD-01 treatment (p < 0.01). Under inflammatory conditions, FKBPL (p < 0.01) and HIF-1α (p < 0.05) expression was elevated, correlating with increased endothelial cell migration (p < 0.05). Unlike hypoxia, AD-01 did not influence endothelial cell migration under inflammatory conditions, but normalized FKBPL (p < 0.001), HIF-1α (p < 0.05) and CD31 (P < 0.05), expression, in 3D microfluidic cell culture. Proteomic analysis revealed that AD-01 treatment in hypoxia enhanced the abundance of tissue remodelling and vascular integrity proteins including collagen alpha-1(XIX) chain and junctional cadherin associated-5 (JCAD) proteins. CONCLUSIONS FKBPL represents an important novel mechanism in hypoxia and inflammation-induced angiogenesis. The FKBPL-based therapeutic peptide, AD-01, could be a viable treatment option for CVD-related endothelial cell dysfunction.
Collapse
Affiliation(s)
- Sahar Ghorbanpour
- School of Life Sciences & Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Siân Peta Cartland
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Hao Chen
- School of Life Sciences & Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Sanchit Seth
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- TissueGnostics Australia Pty Ltd, Brisbane, Australia
- Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Rupert C Ecker
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- TissueGnostics Australia Pty Ltd, Brisbane, Australia
- Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Claire Richards
- School of Life Sciences & Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Dunja Aksentijevic
- Centre for Biochemical Pharmacology, School of Medicine and Dentistry, William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK
| | - Matthew P Padula
- School of Life Sciences & Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Louise Cole
- The Australian Institute for Microbiology and Infection (AIMI), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
| | | | - Lana McClements
- School of Life Sciences & Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
6
|
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
|
7
|
Shah W, Gong Y, Qiao X, Lu Y, Ding Y, Zhang Z, Gao Y. Exploring Endothelial Cell Dysfunction's Impact on the Brain-Retina Microenvironment Connection: Molecular Mechanisms and Implications. Mol Neurobiol 2025:10.1007/s12035-025-04714-x. [PMID: 39904964 DOI: 10.1007/s12035-025-04714-x] [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: 06/24/2024] [Accepted: 01/19/2025] [Indexed: 02/06/2025]
Abstract
The intricate linking between the health of blood vessels and the functioning of neurons has attracted growing attention in the context of disorders that affect the neurological environment. Endothelial cells, forming the blood-brain barrier and blood-retinal barrier, play a fundamental role in maintaining the integrity of the brain-retina microenvironment connection. This review explores the molecular foundations of endothelial cell dysfunction and its implications for the brain-retina interaction. A comprehensive analysis of the complex factors contributing to endothelial dysfunction is presented, including oxidative stress, inflammation, reduced nitric oxide signaling, and disrupted vascular autoregulation. The significance of endothelial dysfunction extends to neurovascular coupling, synaptic plasticity, and trophic support. To our knowledge, there is currently no existing literature review addressing endothelial microvascular dysfunction and its interplay with the brain-retina microenvironment. The review also explains bidirectional communication between the brain and retina, highlighting how compromised endothelial function can disrupt this vital crosstalk and inhibit normal physiological processes. As neurodegenerative diseases frequently exhibit vascular involvement, a deeper comprehension of the interaction between endothelial cells and neural tissue holds promise for innovative therapeutic strategies. By targeting endothelial dysfunction, we may enhance our ability to preserve the intricate dynamics of the brain-retina microenvironment connection and ameliorate the progression of neurological disorders.
Collapse
Affiliation(s)
- Wahid Shah
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Yuxing Gong
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Xin Qiao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Yaling Lu
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Yufei Ding
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Ziting Zhang
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Yuan Gao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China.
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
8
|
Vilahur G, Fuster V. Interplay between platelets and coagulation: from protective haemostasis to pathological arterial thrombosis. Eur Heart J 2025; 46:413-423. [PMID: 39673717 DOI: 10.1093/eurheartj/ehae776] [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: 06/25/2024] [Revised: 08/28/2024] [Accepted: 10/27/2024] [Indexed: 12/16/2024] Open
Abstract
Haemostasis refers to the physiological process aimed at repairing vessel injury and preventing bleeding. It involves four interlinked stages culminating in the formation of a platelet-fibrin haemostatic plug that is eventually dissolved once the vessel heals. In contrast, arterial thrombosis is a pathological condition resulting from atheroma exposure, triggering the formation of a platelet-rich thrombus that may obstruct blood flow, leading to the clinical manifestations of ischaemic cardiovascular disease. The following review will provide a comprehensive overview of the finely regulated endogenous antithrombotic mechanisms responsible for maintaining the haemostatic balance and preventing intravascular thrombosis. Thereafter, it will further detail the different stages and mechanisms governing the intricate interplay between the vessel, platelets, and the coagulation cascade in haemostasis, highlighting the most recent advances in platelet biology and function, to further elucidate the differential traits and players contributing to pathological arterial thrombus growth. The review will also delve into the impact of emerging cardiovascular risk factors on tilting the haemostatic balance towards a pro-thrombotic state, thereby increasing the patient's vulnerability to thrombotic events. Finally, it will underscore the importance of early screening for subclinical atherosclerosis through advanced imaging technologies capable of quantifying plaque burden and metabolic activity since they may set the stage for an increased thrombotic risk. Implementing proactive interventions to halt atherosclerosis progression or inducing its regression at early stages is crucial for preserving haemostasis and reducing the likelihood of ischaemic atherothrombotic disease.
Collapse
Affiliation(s)
- Gemma Vilahur
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Carrer Sant Quintí 77-79, Barcelona 08041, Spain
- CiberCV, Institute Carlos III, Madrid 28029, Spain
| | - Valentin Fuster
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
9
|
Groten SA, van den Eshof BL, van Alphen FPJ, Meijer AB, van den Biggelaar M, Hoogendijk AJ. Integrative phosphoproteomic analyses reveal hemostatic-endothelial signaling interplay. J Thromb Haemost 2025; 23:717-730. [PMID: 39442624 DOI: 10.1016/j.jtha.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/18/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND The vascular endothelial cell (EC) monolayer plays a crucial part in maintaining hemostasis. An extensive array of G protein-coupled receptors allows ECs to dynamically act on key hemostatic stimuli such as thrombin and histamine. The impact of these individual stimuli on EC signal transduction has been the subject of various studies, but insight into discordant and concordant EC signaling between different G protein-coupled receptors remains limited. OBJECTIVES To elucidate histamine and protease-activated receptor (PAR1-4) signaling cascades in ECs, discern overlapping and diverging regulation between these stimuli and their effect on the EC monolayer. METHODS We employed stable isotope labeling by amino acids in cell culture mass spectrometry-based phosphoproteomics on in vitro cultured blood outgrowth ECs stimulated with histamine and different PAR1 to 4 peptides. We investigated key phosphosites through immuno(fluorescence) staining and determined effects on barrier function through transendothelial resistance assays. RESULTS EC histamine activation initiated an extensive (kinase) signaling network (including MAPK3, STAT3, and CTNND1). PAR1 and PAR2 receptors induced highly similar signaling cascades, whereas PAR3 and PAR4 induced minimal phospho-regulation. Integration of all applied stimuli indicated uniquely activated proteins between both stimuli, as well as a general overlapping activation of cell junction and actin cytoskeletal proteins. CONCLUSION We provide an integrative phosphoproteomic analysis of histamine and PAR agonists in the endothelium that highlights the endothelial response programs that are at the basis of regulating hemostasis.
Collapse
Affiliation(s)
- Stijn A Groten
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Bart L van den Eshof
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | | | - Alexander B Meijer
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands; Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | - Arie J Hoogendijk
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands.
| |
Collapse
|
10
|
Gomez-Salinero JM, Redmond D, Rafii S. Microenvironmental determinants of endothelial cell heterogeneity. Nat Rev Mol Cell Biol 2025:10.1038/s41580-024-00825-w. [PMID: 39875728 DOI: 10.1038/s41580-024-00825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
During development, endothelial cells (ECs) undergo an extraordinary specialization by which generic capillary microcirculatory networks spanning from arteries to veins transform into patterned organotypic zonated blood vessels. These capillary ECs become specialized to support the cellular and metabolic demands of each specific organ, including supplying tissue-specific angiocrine factors that orchestrate organ development, maintenance of organ-specific functions and regeneration of injured adult organs. Here, we illustrate the mechanisms by which microenvironmental signals emanating from non-vascular niche cells induce generic ECs to acquire specific inter-organ and intra-organ functional attributes. We describe how perivascular, parenchymal and immune cells dictate vascular heterogeneity and capillary zonation, and how this system is maintained through tissue-specific signalling activated by vasculogenic and angiogenic factors and deposition of matrix components. We also discuss how perturbation of organotypic vascular niche cues lead to erasure of EC signatures, contributing to the pathogenesis of disease processes. We also describe approaches that use reconstitution of tissue-specific signatures of ECs to promote regeneration of damaged organs.
Collapse
Affiliation(s)
- Jesus M Gomez-Salinero
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
11
|
Jimenez-Trinidad FR, Calvo-Gomez S, Sabaté M, Brugaletta S, Campuzano V, Egea G, Dantas AP. Extracellular Vesicles as Mediators of Endothelial Dysfunction in Cardiovascular Diseases. Int J Mol Sci 2025; 26:1008. [PMID: 39940780 PMCID: PMC11816526 DOI: 10.3390/ijms26031008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
This comprehensive review aims to provide a thorough overview of the vital role that extracellular vesicles (EVs) play in endothelial dysfunction, particularly emphasizing how physiological factors-such as sex and aging-along with significant cardiovascular risk factors, influence this process. The review covers studies ranging from the first description of EVs in 1945 to contemporary insights into their biological roles in intercellular signaling and endothelial dysfunction. A comprehensive analysis of peer-reviewed articles and reviews indexed in the PubMed database was conducted to compile the information. Initially, Medical Subject Headings (MeSH) terms included keywords aimed at providing general knowledge about the role of EVs in the regulation of endothelial signaling, such as "extracellular vesicles", "endothelium", and "intercellular signaling". Subsequently, terms related to the pathophysiological implications of EV interactions with endothelial dysfunction and cardiovascular disease were added, including "cardiovascular disease", "sex", "aging", "atherosclerosis", "obesity", and "diabetes". Additionally, the potential applications of EVs in cardiovascular disease were explored using the MeSH terms "extracellular vesicles", "cardiovascular disease", "biomarker", and "therapeutic strategy". The results of this bibliographical review reveal that EVs have the capacity to induce various cellular responses within the cardiovascular system and play a significant role in the complex landscape of endothelial dysfunction and cardiovascular disease. The composition of the EV cargo is subject to modification by pathophysiological conditions such as sex, aging, and cardiovascular risk factors, which result in a complex regulatory influence on endothelial function and neighboring cells when released from a dysfunctional endothelium. Moreover, the data suggest that this field still requires further exploration, as EV biology is continuously evolving, presenting a dynamic and engaging area for research. A deeper understanding of the molecular cargo involved in EV-endothelium interactions could yield valuable biomarkers for monitoring cardiovascular disease progression and facilitate the development of innovative bioengineered therapeutic strategies to enhance patient outcomes.
Collapse
Affiliation(s)
- Francisco Rafael Jimenez-Trinidad
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (F.R.J.-T.); (V.C.); (G.E.)
- Institut Clínic Cardiovascular (ICCV), Hospital Clínic, 08036 Barcelona, Spain; (M.S.); (S.B.)
- Division of Respiratory, Cardiovascular and Renal Pathobiology and Bioengineering, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Sergi Calvo-Gomez
- Department of Biomedical Sciences, School of Medicine, Universitat Internacional de Catalunya (UIC), 08195 Barcelona, Spain;
| | - Manel Sabaté
- Institut Clínic Cardiovascular (ICCV), Hospital Clínic, 08036 Barcelona, Spain; (M.S.); (S.B.)
- Division of Respiratory, Cardiovascular and Renal Pathobiology and Bioengineering, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Salvatore Brugaletta
- Institut Clínic Cardiovascular (ICCV), Hospital Clínic, 08036 Barcelona, Spain; (M.S.); (S.B.)
- Division of Respiratory, Cardiovascular and Renal Pathobiology and Bioengineering, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Victoria Campuzano
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (F.R.J.-T.); (V.C.); (G.E.)
- Rare Diseases Biomedical Research Network Center (CIBERER), Instituto de Salud Carlos III, 28222 Madrid, Spain
| | - Gustavo Egea
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (F.R.J.-T.); (V.C.); (G.E.)
- Division of Respiratory, Cardiovascular and Renal Pathobiology and Bioengineering, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Center of Medical Genetics, University of Antwerpen, 2659 Edegem, Belgium
| | - Ana Paula Dantas
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (F.R.J.-T.); (V.C.); (G.E.)
- Institut Clínic Cardiovascular (ICCV), Hospital Clínic, 08036 Barcelona, Spain; (M.S.); (S.B.)
- Division of Respiratory, Cardiovascular and Renal Pathobiology and Bioengineering, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| |
Collapse
|
12
|
Crystal GJ, Pagel PS. Perspectives on the History of Coronary Physiology: Discovery of Major Principles and Their Clinical Correlates. J Cardiothorac Vasc Anesth 2025; 39:220-243. [PMID: 39278733 DOI: 10.1053/j.jvca.2024.08.017] [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: 06/20/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 09/18/2024]
Abstract
Coronary circulation plays an essential role in delivering oxygen and metabolic substrates to satisfy the considerable energy demand of the heart. This article reviews the history that led to the current understanding of coronary physiology, beginning with William Harvey's revolutionary discovery of systemic blood circulation in the 17th century, and extending through the 20th century when the major mechanisms regulating coronary blood flow (CBF) were elucidated: extravascular compressive forces, metabolic control, pressure-flow autoregulation, and neural pathways. Pivotal research studies providing evidence for each of these mechanisms are described, along with their clinical correlates. The authors describe the major role played by researchers in the 19th century, who formulated basic principles of hemodynamics, such as Poiseuille's law, which provided the conceptual foundation for experimental studies of CBF regulation. Targeted research studies in coronary physiology began in earnest around the turn of the 20th century. Despite reliance on crude experimental techniques, the pioneers in coronary physiology made groundbreaking discoveries upon which our current knowledge is predicated. Further advances in coronary physiology were facilitated by technological developments, including methods to measure phasic CBF and its regional distribution, and by biochemical discoveries, including endothelial vasoactive molecules and adrenergic receptor subtypes. The authors recognize the invaluable contribution made by basic scientists toward the understanding of CBF regulation, and the enormous impact that this fundamental information has had on improving clinical diagnosis, decision-making, and patient care.
Collapse
Affiliation(s)
- George J Crystal
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL.
| | - Paul S Pagel
- Anesthesia Service, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI
| |
Collapse
|
13
|
Acar Y, Managh A, Hill EJ, Roach P. Tuning Surface Chemistry Impacts on Cardiac Endothelial and Smooth Muscle Cell Development. J Biomed Mater Res A 2025; 113:e37846. [PMID: 39668801 DOI: 10.1002/jbm.a.37846] [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: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death worldwide, with approx. Twenty million deaths in 2021. Cardiovascular implants are among the most used biomaterials in the clinical world. However, poor endothelialisation and rapid thrombosis remains a challenge. Simple chemical surface modification techniques can be used to steer biological interactions without affecting the bioimplants' overall bulk characteristics such as radiopacity and flexibility. Although silanes are well studied for protein and cell interactions, the methodical investigation of cardiac endothelial cell (EC) alongside smooth muscle cell (SMC) to mimic natural arterial environments has been limited. In this study, these cells have been investigated on surfaces functionalized with methyl, amine, thiol, methacrylate, and fluorine organosilane groups. Cardiac EC and SMC growth was investigated with metabolic activity, time lapse imaging, and immunofluorescent staining techniques. The results demonstrated that the surfaces tested are able to selectively regulate the viability and growth of the cells. Aminosilane modified surfaces displayed 2-fold higher metabolic activity with HUVEC and 2-fold less metabolic activity with HCASMC cell lines, compared to tissue culture plastic controls. The amino-modification outperformed all other chemistries tested in terms of ability to promote the proliferation of ECs, while importantly reducing the activity of SMCs. This report demonstrates that aminosilane modified surfaces have the potential to be utilized in novel cardiovascular implants, which could improve biological integration in the short and possibly longer-term. The findings of this study suggest that specific chemical modifications of the surface can enhance endothelial cell activity while minimizing the proliferation of smooth muscle cells, which are often associated with thrombosis. This highlights the potential of carefully engineered surface chemistries to improve the clinical outcomes of cardiovascular implants.
Collapse
Affiliation(s)
- Yasemin Acar
- School of Science, Department of Chemistry, Loughborough University, UK
| | - Amy Managh
- School of Science, Department of Chemistry, Loughborough University, UK
| | - Eric James Hill
- School of Science, Department of Chemistry, Loughborough University, UK
| | - Paul Roach
- School of Science, Department of Chemistry, Loughborough University, UK
| |
Collapse
|
14
|
Hostler AC, Hahn WW, Hu MS, Rennert R, Fischer KS, Barrera JA, Duscher D, Januszyk M, Henn D, Sivaraj D, Yasmeh JP, Kussie HC, Granoski MB, Padmanabhan J, Vial IN, Riegler J, Wu JC, Longaker MT, Chen K, Maan ZN, Gurtner GC. Endothelial-specific CXCL12 regulates neovascularization during tissue repair and tumor progression. FASEB J 2024; 38:e70210. [PMID: 39698751 DOI: 10.1096/fj.202401307r] [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: 07/02/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024]
Abstract
C-X-C motif chemokine ligand 12 (CXCL12; Stromal Cell-Derived Factor 1 [SDF-1]), most notably known for its role in embryogenesis and hematopoiesis, has been implicated in tumor pathophysiology and neovascularization. However, its cell-specific role and mechanism of action have not been well characterized. Previous work by our group has demonstrated that hypoxia-inducible factor (HIF)-1 modulates downstream CXCL12 expression following ischemic tissue injury. By utilizing a conditional CXCL12 knockout murine model, we demonstrate that endothelial-specific deletion of CXCL12 (eKO) modulates ischemic tissue survival, altering tissue repair and tumor progression without affecting embryogenesis and morphogenesis. Loss of endothelial-specific CXCL12 disrupts critical endothelial-fibroblast crosstalk necessary for stromal growth and vascularization. Using murine parabiosis with novel transcriptomic technologies, we demonstrate that endothelial-specific CXCL12 signaling results in downstream recruitment of non-inflammatory circulating cells, defined by neovascularization modulating genes. These findings indicate an essential role for endothelial-specific CXCL12 expression during the neovascular response in tissue injury and tumor progression.
Collapse
Affiliation(s)
- Andrew C Hostler
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - William W Hahn
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Michael S Hu
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Robert Rennert
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Katharina S Fischer
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Janos A Barrera
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Dominik Duscher
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Michael Januszyk
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Dominic Henn
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Dharshan Sivaraj
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Jonathan P Yasmeh
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Hudson C Kussie
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Maia B Granoski
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Jagannath Padmanabhan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Ivan N Vial
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Johannes Riegler
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Kellen Chen
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Zeshaan N Maan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Geoffrey C Gurtner
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
15
|
Dalimunthe A, Carensia Gunawan M, Dhiya Utari Z, Dinata MR, Halim P, Estherina S. Pakpahan N, Sitohang AI, Sukarno MA, Yuandani, Harahap Y, Setyowati EP, Park MN, Yusoff SD, Zainalabidin S, Prananda AT, Mahadi MK, Kim B, Harahap U, Syahputra RA. In-depth analysis of lupeol: delving into the diverse pharmacological profile. Front Pharmacol 2024; 15:1461478. [PMID: 39605919 PMCID: PMC11598436 DOI: 10.3389/fphar.2024.1461478] [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: 07/08/2024] [Accepted: 09/30/2024] [Indexed: 11/29/2024] Open
Abstract
Lupeol, a naturally occurring lupane-type pentacyclic triterpenoid, is widely distributed in various edible vegetables, fruits, and medicinal plants. Notably, it is found in high concentrations in plants like Tamarindus indica, Allanblackia monticola, and Emblica officinalis, among others. Quantitative studies have highlighted its presence in Elm bark, Olive fruit, Aloe leaf, Ginseng oil, Mango pulp, and Japanese Pear bark. This compound is synthesized from squalene through the mevalonate pathway and can also be synthetically produced in the lab, addressing challenges in natural product synthesis. Over the past four decades, extensive research has demonstrated lupeol's multifaceted pharmacological properties, including anti-inflammatory, antioxidant, anticancer, and antibacterial effects. Despite its significant therapeutic potential, clinical applications of lupeol have been limited by its poor water solubility and bioavailability. Recent advancements have focused on nano-based delivery systems to enhance its bioavailability, and the development of various lupeol derivatives has further amplified its bioactivity. This review provides a comprehensive overview of the latest advancements in understanding the pharmacological benefits of lupeol. It also discusses innovative strategies to improve its bioavailability, thereby enhancing its clinical efficacy. The aim is to consolidate current knowledge and stimulate further research into the therapeutic potential of lupeol and its derivatives.
Collapse
Affiliation(s)
- Aminah Dalimunthe
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Mega Carensia Gunawan
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Zahirah Dhiya Utari
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Muhammad Riza Dinata
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | | | - Alex Insandus Sitohang
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - M. Andriansyah Sukarno
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Yuandani
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | | | | | - Moon Nyeo Park
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Syaratul Dalina Yusoff
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Satirah Zainalabidin
- Biomedical Science, Centre of Toxicology and Health Risk Study, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Arya Tjipta Prananda
- Faculty of Medicine, Universitas Sumatera Utara, Medan, Sumatera Utara, Indonesia
| | - Mohd Kaisan Mahadi
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Bonglee Kim
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Urip Harahap
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
16
|
Patterson PD, Hilditch CJ, Martin SE, Roach DGL, Weaver MD, Okerman TS, Hostler D, Weiss LS, Reis SE. Comparison of 45-min nap versus no-nap during simulated night shift work on endothelial function: a randomized crossover feasibility trial. Pilot Feasibility Stud 2024; 10:137. [PMID: 39533414 PMCID: PMC11555913 DOI: 10.1186/s40814-024-01569-2] [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: 05/09/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Night shift workers face increased risk of cardiovascular disease (CVD) compared to non-shift workers. Evidence supports on-shift napping and regular non-invasive monitoring of endothelial function for risk mitigation, yet neither strategy is widely used. METHODS We evaluated the feasibility of non-invasive assessment of peripheral arterial tone (PAT) to assess the effect of napping during simulated night shift work on endothelial function. We used a single-site, randomized crossover trial of simulated night shift work with a 45-min nap condition versus a control, no-nap condition (ClinicalTrials.gov NCT05436951). RESULTS The primary outcome was the number of participants with ≥ 70% of endothelial function assessments. Secondary outcomes included mean reactive hyperemia index (RHI), BP, and cognitive performance with the brief psychomotor vigilance task (PVT-B). Of the 10 consented, 9 completed both conditions. All participants exceeded feasibility benchmarks. Mean RHI did not differ by nap condition, and the delta from pre- to post measure did not differ (difference in delta = - 0.26, 95% CI - 1.09, 0.58). Hourly PVT-B assessments from 19:00 to 07:00 h did not differ by nap condition. Compared to pre-nap measures, cognitive performance on the PVT-B was poorest at + 0 min post-nap. CONCLUSION Our findings can inform larger studies evaluating the effects of night shift work and napping on endothelial function. TRIAL REGISTRATIONS ClinicalTrials.gov (NCT05436951, registered on June 23, 2022).
Collapse
Affiliation(s)
- P Daniel Patterson
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA.
- Emergency Medicine Program, Department of Community Health Services and Rehabilitation Science, School of Health and Rehabilitation Sciences, University of Pittsburgh, PA, Pittsburgh, 15261, USA.
| | - Cassie J Hilditch
- Fatigue Countermeasures Laboratory, San José State University, CA, San José, 95192, USA
| | - Sarah E Martin
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA
| | - David G L Roach
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA
| | - Matthew D Weaver
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, MA, Boston, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, MA, Boston, 02115, USA
| | - Tiffany S Okerman
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA
| | - David Hostler
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, NY, Buffalo, 14214, USA
| | - Leonard S Weiss
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA
| | - Steven E Reis
- Department of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA
| |
Collapse
|
17
|
Aksu U, Yavuz-Aksu B, Goswami N. Microcirculation: Current Perspective in Diagnostics, Imaging, and Clinical Applications. J Clin Med 2024; 13:6762. [PMID: 39597906 PMCID: PMC11595220 DOI: 10.3390/jcm13226762] [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: 10/04/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
This review discusses the pivotal role of microcirculation in maintaining tissue oxygenation and waste removal and highlights its significance in various pathological conditions. It delves into the cellular mechanisms underlying hemodynamic coherence, elucidating the roles of the endothelium, glycocalyx, and erythrocytes in sustaining microcirculatory integrity. Furthermore, the review gives comprehensive information about microcirculatory changes observed in cardiac surgery, sepsis, shock, and COVID-19 disease. Through comprehensive exploration, the review underscores the intricate relationship between microcirculation, disease states, and clinical outcomes, emphasizing the importance of understanding and monitoring microvascular dynamics in critical care settings.
Collapse
Affiliation(s)
- Ugur Aksu
- Biology Department, Science Faculty, Istanbul University, Istanbul 34459, Turkey
| | - Berna Yavuz-Aksu
- Duzen Laboratory Group, Biochemistry Section, Istanbul 34394, Turkey;
| | - Nandu Goswami
- Gravitational Physiology and Medicine Research Unit, Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, 3810 Graz, Austria
- Center for Space and Aviation Health, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates
| |
Collapse
|
18
|
Ma J, Yang L, Wu J, Huang Z, Zhang J, Liu M, Li M, Luo J, Wang H. Unraveling the Molecular Mechanisms of SIRT7 in Angiogenesis: Insights from Substrate Clues. Int J Mol Sci 2024; 25:11578. [PMID: 39519130 PMCID: PMC11546391 DOI: 10.3390/ijms252111578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Angiogenesis, a vital physiological or pathological process regulated by complex molecular networks, is widely implicated in organismal development and the pathogenesis of various diseases. SIRT7, a member of the Sirtuin family of nicotinamide adenine dinucleotide + (NAD+) dependent deacetylases, plays crucial roles in cellular processes such as transcriptional regulation, cell metabolism, cell proliferation, and genome stability maintenance. Characterized by its enzymatic activities, SIRT7 targets an array of substrates, several of which exert regulatory effects on angiogenesis. Experimental evidence from in vitro and in vivo studies consistently demonstrates the effects of SIRT7 in modulating angiogenesis, mediated through various molecular mechanisms. Consequently, understanding the regulatory role of SIRT7 in angiogenesis holds significant promise, offering novel avenues for therapeutic interventions targeting either SIRT7 or angiogenesis. This review delineates the putative molecular mechanisms by which SIRT7 regulates angiogenesis, taking its substrates as a clue, endeavoring to elucidate experimental observations by integrating knowledge of SIRT7 substrates and established angiogenenic mechanisms.
Collapse
Affiliation(s)
- Junjie Ma
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| | - Liqian Yang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| | - Jiaxing Wu
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| | - Zhihong Huang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| | - Jiaqi Zhang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| | - Minghui Liu
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing 100191, China; (M.L.); (M.L.)
| | - Meiting Li
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing 100191, China; (M.L.); (M.L.)
| | - Jianyuan Luo
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing 100191, China; (M.L.); (M.L.)
| | - Haiying Wang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| |
Collapse
|
19
|
Oliveira-Paula GH, Martins AC, Ferrer B, Tinkov AA, Skalny AV, Aschner M. The impact of manganese on vascular endothelium. Toxicol Res 2024; 40:501-517. [PMID: 39345740 PMCID: PMC11436708 DOI: 10.1007/s43188-024-00260-1] [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: 05/04/2024] [Revised: 07/10/2024] [Accepted: 07/26/2024] [Indexed: 10/01/2024] Open
Abstract
Manganese (Mn) is an essential trace element involved in various physiological processes, but excessive exposure may lead to toxicity. The vascular endothelium, a monolayer of endothelial cells within blood vessels, is a primary target of Mn toxicity. This review provides a comprehensive overview of the impact of Mn on vascular endothelium, focusing on both peripheral and brain endothelial cells. In vitro studies have demonstrated that high concentrations of Mn can induce endothelial cell cytotoxicity, increase permeability, and disrupt cell-cell junctions through mechanisms involving oxidative stress, mitochondrial damage, and activation of signaling pathways, such as Smad2/3-Snail. Conversely, low concentrations of Mn may protect endothelial cells from the deleterious effects of high glucose and advanced glycation end-products. In the central nervous system, Mn can cross the blood-brain barrier (BBB) and accumulate in the brain parenchyma, leading to neurotoxicity. Several transport mechanisms, including ZIP8, ZIP14, and SPCA1, have been identified for Mn uptake by brain endothelial cells. Mn exposure can impair BBB integrity by disrupting tight junctions and increasing permeability. In vivo studies have corroborated these findings, highlighting the importance of endothelial barriers in mediating Mn toxicity in the brain and kidneys. Maintaining optimal Mn homeostasis is crucial for preserving endothelial function, and further research is needed to develop targeted therapeutic strategies to prevent or mitigate the adverse effects of Mn overexposure. Graphical Abstract
Collapse
Affiliation(s)
| | - Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - Alexey A. Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003 Russia
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435 Russia
| | - Anatoly V. Skalny
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003 Russia
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435 Russia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| |
Collapse
|
20
|
Matsumoto T, Nagano T, Taguchi K, Kobayashi T, Tanaka-Totoribe N. Toll-like receptor 3 involvement in vascular function. Eur J Pharmacol 2024; 979:176842. [PMID: 39033837 DOI: 10.1016/j.ejphar.2024.176842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/24/2024] [Accepted: 07/19/2024] [Indexed: 07/23/2024]
Abstract
Maintaining endothelial cell (EC) and vascular smooth muscle cell (VSMC) integrity is an important component of human health and disease because both EC and VSMC regulate various functions, including vascular tone control, cellular adhesion, homeostasis and thrombosis regulation, proliferation, and vascular inflammation. Diverse stressors affect functions in both ECs and VSMCs and abnormalities of functions in these cells play a crucial role in cardiovascular disease initiation and progression. Toll-like receptors (TLRs) are important detectors of pathogen-associated molecular patterns derived from various microbes and viruses as well as damage-associated molecular patterns derived from damaged cells and perform innate immune responses. Among TLRs, several studies reveal that TLR3 plays a key role in initiation, development and/or protection of diseases, and an emerging body of evidence indicates that TLR3 presents components of the vasculature, including ECs and VSMCs, and plays a functional role. An agonist of TLR3, polyinosinic-polycytidylic acid [poly (I:C)], affects ECs, including cell death, inflammation, chemoattractant, adhesion, permeability, and hemostasis. Poly (I:C) also affects VSMCs including inflammation, proliferation, and modulation of vascular tone. Moreover, alterations of vascular function induced by certain molecules and/or interventions are exerted through TLR3 signaling. Hence, we present the association between TLR3 and vascular function according to the latest studies.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Second Department of Pharmacology, School of Pharmaceutical Sciences, Kyushu University of Medical Science, Nobeoka, Miyazaki, 882-8508, Japan.
| | - Takayuki Nagano
- Second Department of Pharmacology, School of Pharmaceutical Sciences, Kyushu University of Medical Science, Nobeoka, Miyazaki, 882-8508, Japan
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Naoko Tanaka-Totoribe
- First Department of Pharmacology, School of Pharmaceutical Sciences, Kyushu University of Medical Science, Nobeoka, Miyazaki, 882-8508, Japan
| |
Collapse
|
21
|
Zhou L, Li J, Wang J, Niu X, Li J, Zhang K. Pathogenic role of PFKFB3 in endothelial inflammatory diseases. Front Mol Biosci 2024; 11:1454456. [PMID: 39318551 PMCID: PMC11419998 DOI: 10.3389/fmolb.2024.1454456] [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: 06/25/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
The differentiation of vascular endothelial cells and the formation of new blood vessels are inseparable from the energy supply and regulation of metabolism. The budding of blood vessels is a starting point of glycolysis pathway in angiogenesis. Phosphofructokinase-2/fructose 2,6-biophosphatase 3 (PFKFB3), a key rate-limiting enzyme in glycolysis, exhibits strong kinase activity. Inhibition of PFKFB3 can reduce the rate of glycolysis, thereby inhibiting the budding of blood vessels, resulting in inhibition of pathological angiogenesis. In this review, the role of PFKFB3 in the angiogenesis of inflammatory diseases was summarized, and the endothelial inflammatory diseases associated with PFKFB3 were reviewed.
Collapse
Affiliation(s)
- Ling Zhou
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Taiyuan Central Hospital, Taiyuan, China
| | - Juan Li
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Taiyuan Central Hospital, Taiyuan, China
| | - Juanjuan Wang
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Taiyuan Central Hospital, Taiyuan, China
| | - Xuping Niu
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Taiyuan Central Hospital, Taiyuan, China
| | - Junqin Li
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Taiyuan Central Hospital, Taiyuan, China
| | - Kaiming Zhang
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Taiyuan Central Hospital, Taiyuan, China
| |
Collapse
|
22
|
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
|
23
|
Groten SA, Smit ER, van den Biggelaar M, Hoogendijk AJ. The proteomic landscape of in vitro cultured endothelial cells across vascular beds. Commun Biol 2024; 7:989. [PMID: 39143368 PMCID: PMC11324761 DOI: 10.1038/s42003-024-06649-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 07/29/2024] [Indexed: 08/16/2024] Open
Abstract
Blood vessel endothelial cells (EC) display heterogeneity across vascular beds, which is anticipated to drive site-specific vascular pathology. This heterogeneity is assessed using transcriptomics in vivo, and functional assays in vitro, but how proteomes compare across human in vitro cultured ECs remains incompletely characterized. We generated an in-depth human EC proteomic landscape (>8000 proteins) across six organs and two in vitro models in steady-state and upon IFNγ-induced inflammation. EC proteomes displayed a high similarity and organ-specific proteins were limited. Variation between ECs was mainly based on proliferation and differentiation processes in which Blood outgrowth endothelial cells (BOEC) and Human umbilical vein cells (HUVEC) represented the extremes of proteomic phenotypes. The IFNγ response was highly conserved across all samples. Harnessing dynamics in protein abundances we delineated VWF and VE-Cadherin correlation networks. This EC landscape provides an extensive proteomic addition in studying EC biology and heterogeneity from an in vitro perspective.
Collapse
Affiliation(s)
- Stijn A Groten
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Eva R Smit
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | | | - Arie J Hoogendijk
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands.
| |
Collapse
|
24
|
Jin T, Fu Z, Zhou L, Chen L, Wang J, Wang L, Yan S, Li T, Jin P. GelMA loaded with platelet lysate promotes skin regeneration and angiogenesis in pressure ulcers by activating STAT3. Sci Rep 2024; 14:18345. [PMID: 39112598 PMCID: PMC11306777 DOI: 10.1038/s41598-024-67304-2] [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: 01/02/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
Pressure ulcers (PU) are caused by persistent long-term pressure, which compromises the integrity of the epidermis, dermis, and subcutaneous adipose tissue layer by layer, making it difficult to heal. Platelet products such as platelet lysate (PL) can promote tissue regeneration by secreting numerous growth factors based on clinical studies on skin wound healing. However, the components of PL are difficult to retain in wounds. Gelatin methacrylate (GelMA) is a photopolymerizable hydrogel that has lately emerged as a promising material for tissue engineering and regenerative medicine. The PL liquid was extracted, flow cytometrically detected for CD41a markers, and evenly dispersed in the GelMA hydrogel to produce a surplus growth factor hydrogel system (PL@GM). The microstructure of the hydrogel system was observed under a scanning electron microscope, and its sustained release efficiency and biological safety were tested in vitro. Cell viability and migration of human dermal fibroblasts, and tube formation assays of human umbilical vein endothelial cells were applied to evaluate the ability of PL to promote wound healing and regeneration in vitro. Real-time polymerase chain reaction (PCR) and western blot analyses were performed to elucidate the skin regeneration mechanism of PL. We verified PL's therapeutic effectiveness and histological analysis on the PU model. PL promoted cell viability, migration, wound healing and angiogenesis in vitro. Real-time PCR and western blot indicated PL suppressed inflammation and promoted collagen I synthesis by activating STAT3. PL@GM hydrogel system demonstrated optimal biocompatibility and favorable effects on essential cells for wound healing. PL@GM also significantly stimulated PU healing, skin regeneration, and the formation of subcutaneous collagen and blood vessels. PL@GM could accelerate PU healing by promoting fibroblasts to migrate and secrete collagen and endothelial cells to vascularize. PL@GM promises to be an effective and convenient treatment modality for PU, like chronic wound treatment.
Collapse
Affiliation(s)
- Tingting Jin
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Zexin Fu
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liuyi Zhou
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Lulu Chen
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ji Wang
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Lu Wang
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Sheng Yan
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Ting Li
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.
| | - Peihong Jin
- Center for Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.
| |
Collapse
|
25
|
Rather MA, Khan A, Jahan S, Siddiqui AJ, Wang L. Influence of Tau on Neurotoxicity and Cerebral Vasculature Impairment Associated with Alzheimer's Disease. Neuroscience 2024; 552:1-13. [PMID: 38871021 DOI: 10.1016/j.neuroscience.2024.05.042] [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: 12/10/2023] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
Alzheimer's disease is a fatal chronic neurodegenerative condition marked by a gradual decline in cognitive abilities and impaired vascular function within the central nervous system. This affliction initiates its insidious progression with the accumulation of two aberrant protein entities including Aβ plaques and neurofibrillary tangles. These chronic elements target distinct brain regions, steadily erasing the functionality of the hippocampus and triggering the erosion of memory and neuronal integrity. Several assumptions are anticipated for AD as genetic alterations, the occurrence of Aβ plaques, altered processing of amyloid precursor protein, mitochondrial damage, and discrepancy of neurotropic factors. In addition to Aβ oligomers, the deposition of tau hyper-phosphorylates also plays an indispensable part in AD etiology. The brain comprises a complex network of capillaries that is crucial for maintaining proper function. Tau is expressed in cerebral blood vessels, where it helps to regulate blood flow and sustain the blood-brain barrier's integrity. In AD, tau pathology can disrupt cerebral blood supply and deteriorate the BBB, leading to neuronal neurodegeneration. Neuroinflammation, deficits in the microvasculature and endothelial functions, and Aβ deposition are characteristically detected in the initial phases of AD. These variations trigger neuronal malfunction and cognitive impairment. Intracellular tau accumulation in microglia and astrocytes triggers deleterious effects on the integrity of endothelium and cerebral blood supply resulting in further advancement of the ailment and cerebral instability. In this review, we will discuss the impact of tau on neurovascular impairment, mitochondrial dysfunction, oxidative stress, and the role of hyperphosphorylated tau in neuron excitotoxicity and inflammation.
Collapse
Affiliation(s)
- Mashoque Ahmad Rather
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, United States.
| | - Andleeb Khan
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, 226026, India
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail City, Saudi Arabia
| | - Lianchun Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, United States
| |
Collapse
|
26
|
Prajapat SK, Maharana KC, Singh S. Mitochondrial dysfunction in the pathogenesis of endothelial dysfunction. Mol Cell Biochem 2024; 479:1999-2016. [PMID: 37642880 DOI: 10.1007/s11010-023-04835-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
Cardiovascular diseases (CVDs) are a matter of concern worldwide, and mitochondrial dysfunction is one of the major contributing factors. Vascular endothelial dysfunction has a major role in the development of atherosclerosis because of the abnormal chemokine secretion, inflammatory mediators, enhancement of LDL oxidation, cytokine elevation, and smooth muscle cell proliferation. Endothelial cells transfer oxygen from the pulmonary circulatory system to the tissue surrounding the blood vessels, and a majority of oxygen is transferred to the myocardium by endothelial cells, which utilise a small amount of oxygen to generate ATP. Free radicals of oxide are produced by mitochondria, which are responsible for cellular oxygen uptake. Increased mitochondrial ROS generation and reduction in agonist-stimulated eNOS activation and nitric oxide bioavailability were directly linked to the observed change in mitochondrial dynamics, resulting in various CVDs and endothelial dysfunction. Presently, the manuscript mainly focuses on endothelial dysfunction, providing a deep understanding of the various features of mitochondrial mechanisms that are used to modulate endothelial dysfunction. We talk about recent findings and approaches that may make it possible to detect mitochondrial dysfunction as a potential biomarker for risk assessment and diagnosis of endothelial dysfunction. In the end, we cover several targets that may reduce mitochondrial dysfunction through both direct and indirect processes and assess the impact of several different classes of drugs in the context of endothelial dysfunction.
Collapse
Affiliation(s)
- Suresh Kumar Prajapat
- National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, Hajipur, Bihar, India
| | - Krushna Ch Maharana
- National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, Hajipur, Bihar, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotions Industrial Park (EPIP), Industrial Area, Dist: Vaishali, Hajipur, Bihar, 844102, India.
| |
Collapse
|
27
|
Hong SG, Kennelly JP, Williams KJ, Bensinger SJ, Mack JJ. Flow-mediated modulation of the endothelial cell lipidome. Front Physiol 2024; 15:1431847. [PMID: 39119214 PMCID: PMC11307263 DOI: 10.3389/fphys.2024.1431847] [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: 05/13/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
The luminal surface of the endothelium is exposed to dynamic blood flow patterns that are known to affect endothelial cell phenotype. While many studies have documented the phenotypic changes by gene or protein expression, less is known about the role of blood flow pattern on the endothelial cell (EC) lipidome. In this study, shotgun lipidomics was conducted on human aortic ECs (HAECs) exposed to unidirectional laminar flow (UF), disturbed flow (DF), or static conditions for 48 h. A total of 520 individual lipid species from 17 lipid subclasses were detected. Total lipid abundance was significantly increased for HAECs exposed to DF compared to UF conditions. Despite the increase in the total lipid abundance, HAECs maintained equivalent composition of each lipid subclass (% of total lipid) under DF and UF. However, by lipid composition (% of total subclass), 28 lipid species were significantly altered between DF and UF. Complimentary RNA sequencing of HAECs exposed to UF or DF revealed changes in transcripts involved in lipid metabolism. Shotgun lipidomics was also performed on HAECs exposed to pro-inflammatory agonists lipopolysaccharide (LPS) or Pam3CSK4 (Pam3) for 48 h. Exposure to LPS or Pam3 reshaped the EC lipidome in both unique and overlapping ways. In conclusion, exposure to flow alters the EC lipidome and ECs undergo stimulus-specific lipid reprogramming in response to pro-inflammatory agonist exposure. Ultimately, this work provides a resource to profile the transcriptional and lipidomic changes that occur in response to applied flow that can be accessed by the vascular biology community to further dissect and extend our understanding of endothelial lipid biology.
Collapse
Affiliation(s)
- Soon-Gook Hong
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - John P. Kennelly
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Kevin J. Williams
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Lipidomics Lab, University of California, Los Angeles, Los Angeles, CA, United States
| | - Steven J. Bensinger
- UCLA Lipidomics Lab, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Julia J. Mack
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
28
|
Sénémaud J, Skarbek C, Hernandez B, Song R, Lefevre I, Bianchi E, Castier Y, Nicoletti A, Bureau C, Caligiuri G. Camouflaging endovascular stents with an endothelial coat using CD31 domain 1 and 2 mimetic peptides. JVS Vasc Sci 2024; 5:100213. [PMID: 39257386 PMCID: PMC11386311 DOI: 10.1016/j.jvssci.2024.100213] [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: 02/27/2024] [Accepted: 07/17/2024] [Indexed: 09/12/2024] Open
Abstract
Objective Implantation of an endovascular device disrupts the homeostatic CD31:CD31 interactions among quiescent endothelial cells (ECs), platelets, and circulating leukocytes. The aim of this study was to design an endothelial-mimetic coating of nitinol and cobalt-chromium (CoCr) surfaces and stents using synthetic CD31 peptides, to promote device endothelialization and pacific integration within the arterial wall. Methods Peptides mimicking the domains 1 (D1) and 2 (D2) of CD31 were synthetized and immobilized onto experimental nitinol and CoCr surfaces using a three-step, dip-coating, mussel-inspired protocol using copper-free click chemistry. Human aortic EC phenotype and endothelialization assessment using parallel scratch tests were carried out using five synthetic CD31 peptides coated on 4.8-mm nitinol and CoCr flat disks and were compared with control disks. The CD31 peptide exhibiting the best results in vitro was then immobilized on clinical-grade 3 × 40-mm self-expanding nitinol and 2.5 × 20.0-mm balloon-expandable CoCr stents. Such devices were implanted in native arteries of White New Zealand rabbits, and compared with control uncoated bare metal stents (BMS) and drug-eluting stents 7 and 30 days after implantation using resin cross-sections and scanning electron microscopy (n = 2-3 per group at each time point). Results Membrane-distal CD31 D1 and D2 peptides exhibited a distinct capability to foster a healthy endothelial phenotype and to promote endothelialization in vitro. By day 7 after implantation, CD31 nitinol and CoCr stents were evenly covered by wholesome ECs, devoid of thromboinflammatory signs, in contrast with both BMS and drug-eluting stents. Such results were consistent until day 30. Conclusions Membrane-distal CD31 biomimetic peptides seem to camouflage the device surface effectively, preventing local reactions and promoting rapid and seamless endovascular integration.
Collapse
Affiliation(s)
- Jean Sénémaud
- Department of Vascular Surgery, Bichat University Hospital, Paris, France
- Université Paris Cité, Paris, France
- Laboratory for Vascular Translational Science, INSERM U1148, Paris, France
| | - Charles Skarbek
- Université Paris Cité, Paris, France
- Laboratory for Vascular Translational Science, INSERM U1148, Paris, France
| | - Belen Hernandez
- Laboratory for Vascular Translational Science, INSERM U1148, Paris, France
- Université Sorbonne Paris Nord, Paris, France
| | - Ran Song
- Sino Medical Sciences Technology Inc., Tianjin, China
| | | | - Elisabetta Bianchi
- Peptide Chemistry Unit, Peptides & Small Molecules R&D Department, IRBM SpA, Pomezia, Roma, Italy
| | - Yves Castier
- Department of Vascular Surgery, Bichat University Hospital, Paris, France
- Université Paris Cité, Paris, France
| | - Antonino Nicoletti
- Université Paris Cité, Paris, France
- Laboratory for Vascular Translational Science, INSERM U1148, Paris, France
| | - Christophe Bureau
- Sino Medical Sciences Technology Inc., Tianjin, China
- AlchiMedics S.A.S., Paris, France
| | - Giuseppina Caligiuri
- Université Paris Cité, Paris, France
- Laboratory for Vascular Translational Science, INSERM U1148, Paris, France
- Department of Cardiology, Bichat University Hospital, Paris, France
| |
Collapse
|
29
|
Wang S, Fang R, Huang L, Zhou L, Liu H, Cai M, Sha’aban A, Yu C, Akkaif MA. Acupuncture in Traditional Chinese Medicine: A Complementary Approach for Cardiovascular Health. J Multidiscip Healthc 2024; 17:3459-3473. [PMID: 39050695 PMCID: PMC11268752 DOI: 10.2147/jmdh.s476319] [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: 05/23/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
Cardiovascular diseases (CVDs) are increasingly prevalent in clinical settings. With the continuous improvement of people's living standards, the gradual acceleration of the pace of life, and the deterioration of the living environment in recent years, the incidence of CVDs is increasing annually. The prevalence of CVDs among individuals aged 50 and above is notably elevated, posing a significant risk to patients' well-being and lives. At this juncture, numerous clinical treatment choices are available for managing CVDs, with traditional Chinese medicine (TCM) therapy standing out as a practical, safe, and reliable option. Over the recent years, there has been growing acknowledgement among both medical professionals and patients. With the expanding integration of TCM in the treatment of various clinical conditions, the use of TCM in managing CVDs has gained significant attention within the medical community, potentially emerging as an efficacious approach for addressing cardiovascular diseases. This article conducts a comprehensive review of the TCM approach, particularly acupuncture, as a supplementary treatment for CVDs, highlighting its ability to effectively lower blood pressure, decrease coronary artery events, mitigate arrhythmias, and enhance cardiac function when used alongside conventional medication. The review underscores the promise of acupuncture in enhancing cardiovascular health, although variations in research methodologies necessitate standardized applications.
Collapse
Affiliation(s)
- Shengfeng Wang
- Rehabilitation Department of Chinese Medicine, The Fourth People’s Hospital of Kunshan, Kunshan City, Jiangsu Province, People’s Republic of China
| | - Ruxue Fang
- Rehabilitation Department of Chinese Medicine, The Fourth People’s Hospital of Kunshan, Kunshan City, Jiangsu Province, People’s Republic of China
| | - Lei Huang
- Rehabilitation Department of Chinese Medicine, The Fourth People’s Hospital of Kunshan, Kunshan City, Jiangsu Province, People’s Republic of China
| | - Liping Zhou
- Rehabilitation Department of Chinese Medicine, The Fourth People’s Hospital of Kunshan, Kunshan City, Jiangsu Province, People’s Republic of China
- Department of Community Health, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, Pulau Pinang, Malaysia
| | - Haibo Liu
- Department of Cardiology, QingPu Branch of Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Meiling Cai
- Department of Obstetrics and Gynecology, Qingpu Branch, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Abubakar Sha’aban
- Health and Care Research Wales Evidence Centre, Cardiff University, Heath Park, Cardiff, UK
| | - Chunxiang Yu
- Rehabilitation Department of Chinese Medicine, The Fourth People’s Hospital of Kunshan, Kunshan City, Jiangsu Province, People’s Republic of China
| | - Mohammed Ahmed Akkaif
- Department of Cardiology, QingPu Branch of Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
30
|
Thijssen VLJL. Vascular galectins in tumor angiogenesis and cancer immunity. Semin Immunopathol 2024; 46:3. [PMID: 38990363 PMCID: PMC11239785 DOI: 10.1007/s00281-024-01014-9] [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: 01/02/2024] [Accepted: 03/13/2024] [Indexed: 07/12/2024]
Abstract
Sustained tumor angiogenesis, i.e., the induction and maintenance of blood vessel growth by tumor cells, is one of the hallmarks of cancer. The vascularization of malignant tissues not only facilitates tumor growth and metastasis, but also contributes to immune evasion. Important players in all these processes are the endothelial cells which line the luminal side of blood vessel. In the tumor vasculature, these cells are actively involved in angiogenesis as well in the hampered recruitment of immune cells. This is the result of the abnormal tumor microenvironment which triggers both angiostimulatory and immune inhibitory gene expression profiles in endothelial cells. In recent years, it has become evident that galectins constitute a protein family that is expressed in the tumor endothelium. Moreover, several members of this glycan-binding protein family have been found to facilitate tumor angiogenesis and stimulate immune suppression. All this has identified galectins as potential therapeutic targets to simultaneously hamper tumor angiogenesis and alleviate immune suppression. The current review provides a brief introduction in the human galectin protein family. The current knowledge regarding the expression and regulation of galectins in endothelial cells is summarized. Furthermore, an overview of the role that endothelial galectins play in tumor angiogenesis and tumor immunomodulation is provided. Finally, some outstanding questions are discussed that should be addressed by future research efforts. This will help to fully understand the contribution of endothelial galectins to tumor progression and to exploit endothelial galectins for cancer therapy.
Collapse
Affiliation(s)
- Victor L J L Thijssen
- Radiation Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands.
- Center for Experimental and Molecular Medicine, Laboratory for Experimental Oncology and Radiobiology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
- Cancer Center Amsterdam, Cancer Biology & Immunology, Amsterdam, The Netherlands.
| |
Collapse
|
31
|
Hong SG, Kennelly JP, Williams KJ, Bensinger SJ, Mack JJ. Flow-Mediated Modulation of the Endothelial Cell Lipidome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598934. [PMID: 38915541 PMCID: PMC11195170 DOI: 10.1101/2024.06.13.598934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The luminal surface of the endothelium is exposed to dynamic blood flow patterns that are known to affect endothelial cell phenotype. While many studies have documented the phenotypic changes by gene or protein expression, less is known about the role of blood flow pattern on the endothelial cell (EC) lipidome. In this study, shotgun lipidomics was conducted on human aortic ECs (HAECs) exposed to unidirectional laminar flow (UF), disturbed flow (DF), or static conditions for 48 hrs. A total of 520 individual lipid species from 17 lipid subclasses were detected. Total lipid abundance was significantly increased for HAECs exposed to DF compared to UF conditions. Despite the increase in the total lipid abundance, HAECs maintained equivalent composition of each lipid subclass (% of total lipid) under both DF and UF. However, by lipid composition (% of total subclass), 28 lipid species were significantly altered between DF and UF. Complimentary RNA sequencing of HAECs exposed to UF or DF revealed changes in transcripts involved in lipid metabolism. Shotgun lipidomics was also performed on HAECs exposed to pro-inflammatory agonists lipopolysaccharide (LPS) or Pam3CSK4 (Pam3) for 48 hrs. Exposure to LPS or Pam3 reshaped the EC lipidome in both unique and overlapping ways. In conclusion, exposure to flow alters the EC lipidome and ECs undergo stimulus-specific lipid reprogramming in response to pro-inflammatory agonist exposure. Ultimately, this work provides a resource to profile the transcriptional and lipidomic changes that occur in response to applied flow that can be accessed by the vascular biology community to further dissect and extend our understanding of endothelial lipid biology.
Collapse
|
32
|
Nwokocha C, Palacios J, Ojukwu VE, Nna VU, Owu DU, Nwokocha M, McGrowder D, Orie NN. Oxidant-induced disruption of vascular K + channel function: implications for diabetic vasculopathy. Arch Physiol Biochem 2024; 130:361-372. [PMID: 35757993 DOI: 10.1080/13813455.2022.2090578] [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: 05/27/2022] [Accepted: 06/07/2022] [Indexed: 11/02/2022]
Abstract
Diabetes in humans a chronic metabolic disorder characterised by hyperglycaemia, it is associated with an increased risk of cardiovascular disease, disruptions to metabolism and vascular functions. It is also linked to oxidative stress and its complications. Its role in vascular dysfunctions is generally reported without detailed impact on the molecular mechanisms. Potassium ion channel (K+ channels) are key regulators of vascular tone, and as membrane proteins, are modifiable by oxidant stress associated with diabetes. This review manuscript examined the impact of oxidant stress on vascular K+ channel functions in diabetes, its implication in vascular complications and metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
| | - Javier Palacios
- Department of Pharmacy, Faculty of Health Sciences, Arturo Prat University, Iquique, Chile
| | - Victoria E Ojukwu
- Basic Medical Sciences, University of the West Indies, Mona, Kingston, Jamaica
| | - Victor Udo Nna
- Department of Physiology, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Daniel Udofia Owu
- Department of Physiology, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Magdalene Nwokocha
- Department of Pathology, Faculty of Medical Sciences, University of the West Indies, Mona, Kingston, Jamaica
| | - Donovan McGrowder
- Department of Pathology, Faculty of Medical Sciences, University of the West Indies, Mona, Kingston, Jamaica
| | - Nelson N Orie
- Centre of Metabolism and Inflammation, University College London, London, UK
| |
Collapse
|
33
|
Madaudo C, Coppola G, Parlati ALM, Corrado E. Discovering Inflammation in Atherosclerosis: Insights from Pathogenic Pathways to Clinical Practice. Int J Mol Sci 2024; 25:6016. [PMID: 38892201 PMCID: PMC11173271 DOI: 10.3390/ijms25116016] [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: 04/28/2024] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
This comprehensive review explores the various scenarios of atherosclerosis, a systemic and chronic arterial disease that underlies most cardiovascular disorders. Starting from an overview of its insidious development, often asymptomatic until it reaches advanced stages, the review delves into the pathophysiological evolution of atherosclerotic lesions, highlighting the central role of inflammation. Insights into clinical manifestations, including heart attacks and strokes, highlight the disease's significant burden on global health. Emphasis is placed on carotid atherosclerosis, clarifying its epidemiology, clinical implications, and association with cognitive decline. Prevention strategies, lifestyle modifications, risk factor management, and nuanced antithrombotic treatment considerations are critical to managing cardiovascular complications, thus addressing a crucial aspect of cardiovascular health.
Collapse
Affiliation(s)
- Cristina Madaudo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| | - Giuseppe Coppola
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| | | | - Egle Corrado
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| |
Collapse
|
34
|
Akhigbe R, Odetayo A, Akhigbe T, Hamed M, Ashonibare P. Pathophysiology and management of testicular ischemia/reperfusion injury: Lessons from animal models. Heliyon 2024; 10:e27760. [PMID: 38694115 PMCID: PMC11058307 DOI: 10.1016/j.heliyon.2024.e27760] [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: 11/10/2023] [Revised: 02/24/2024] [Accepted: 03/06/2024] [Indexed: 05/03/2024] Open
Abstract
Testicular torsion is a urological emergency that involves the twisting of the spermatic cord along its course. Compelling pieces of evidence have implicated oxidative stress-sensitive signaling in pathogenesis of testicular I/R injury. Although, surgical detorsion is the mainstay management; blockade of the pathways involved in the pathogenesis may improve the surgical outcome. Experimental studies using various testicular I/R models have been reported in a bid to explore the mechanisms associated with testicular I/R and evaluate the benefits of potential therapeutic measures; however, most are limited by their shortcomings. Thus, this review was intended to describe the details of the available testicular I/R models as well as their merits and drawbacks, the pathophysiological basis and consequences of testicular I/R, and the pharmacological agents that have being proposed to confer testicular benefits against testicular I/R. This provides an understanding of the pathophysiological events and available models used in studying testicular I/R. In addition, this research provides evidence-based molecules with therapeutic potentials as well as their mechanisms of action in testicular I/R.
Collapse
Affiliation(s)
- R.E. Akhigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | - A.F. Odetayo
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
- Department of Physiology, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - T.M. Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
- Breeding and Plant Genetics Unit, Department of Agronomy, Osun State University, Osun State, Nigeria
| | - M.A. Hamed
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
- Department of Medical Laboratory Science, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
- The Brainwill Laboratory, Osogbo, Osun State, Nigeria
| | - P.J. Ashonibare
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| |
Collapse
|
35
|
Kaczara P, Czyzynska-Cichon I, Kus E, Kurpinska A, Olkowicz M, Wojnar-Lason K, Pacia MZ, Lytvynenko O, Baes M, Chlopicki S. Liver sinusoidal endothelial cells rely on oxidative phosphorylation but avoid processing long-chain fatty acids in their mitochondria. Cell Mol Biol Lett 2024; 29:67. [PMID: 38724891 PMCID: PMC11084093 DOI: 10.1186/s11658-024-00584-8] [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: 12/20/2023] [Accepted: 04/25/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND It is generally accepted that endothelial cells (ECs), primarily rely on glycolysis for ATP production, despite having functional mitochondria. However, it is also known that ECs are heterogeneous, and their phenotypic features depend on the vascular bed. Emerging evidence suggests that liver sinusoidal ECs (LSECs), located in the metabolically rich environment of the liver, show high metabolic plasticity. However, the substrate preference for energy metabolism in LSECs remains unclear. METHODS Investigations were conducted in primary murine LSECs in vitro using the Seahorse XF technique for functional bioenergetic assays, untargeted mass spectrometry-based proteomics to analyse the LSEC proteome involved in energy metabolism pathways, liquid chromatography-tandem mass spectrometry-based analysis of acyl-carnitine species and Raman spectroscopy imaging to track intracellular palmitic acid. RESULTS This study comprehensively characterized the energy metabolism of LSECs, which were found to depend on oxidative phosphorylation, efficiently fuelled by glucose-derived pyruvate, short- and medium-chain fatty acids and glutamine. Furthermore, despite its high availability, palmitic acid was not directly oxidized in LSEC mitochondria, as evidenced by the acylcarnitine profile and etomoxir's lack of effect on oxygen consumption. However, together with L-carnitine, palmitic acid supported mitochondrial respiration, which is compatible with the chain-shortening role of peroxisomal β-oxidation of long-chain fatty acids before further degradation and energy generation in mitochondria. CONCLUSIONS LSECs show a unique bioenergetic profile of highly metabolically plastic ECs adapted to the liver environment. The functional reliance of LSECs on oxidative phosphorylation, which is not a typical feature of ECs, remains to be determined.
Collapse
Affiliation(s)
- Patrycja Kaczara
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland.
| | - Izabela Czyzynska-Cichon
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Edyta Kus
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Anna Kurpinska
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Mariola Olkowicz
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Kamila Wojnar-Lason
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
- Jagiellonian University Medical College, Department of Pharmacology, Grzegorzecka 16, 31-531, Krakow, Poland
| | - Marta Z Pacia
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Olena Lytvynenko
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Myriam Baes
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Laboratory of Cell Metabolism, 3000, Leuven, Belgium
| | - Stefan Chlopicki
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
- Jagiellonian University Medical College, Department of Pharmacology, Grzegorzecka 16, 31-531, Krakow, Poland
| |
Collapse
|
36
|
Matheson C, Simovic T, Heefner A, Colon M, Tunon E, Cobb K, Thode C, Breland A, Cobb CO, Nana-Sinkam P, Garten R, Rodriguez-Miguelez P. Evidence of premature vascular dysfunction in young adults who regularly use e-cigarettes and the impact of usage length. Angiogenesis 2024; 27:229-243. [PMID: 38345700 PMCID: PMC11021332 DOI: 10.1007/s10456-023-09903-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/27/2023] [Indexed: 04/17/2024]
Abstract
BACKGROUND Electronic (e-) cigarettes are increasingly popular tobacco products on the US market. Traditional tobacco products are known to cause vascular dysfunction, one of the earliest indicators of cardiovascular disease (CVD) development. However, little is known about the effect of regular e-cigarette use on vascular function. The purpose of this study was to investigate the impact of regular e-cigarette use on vascular function and cardiovascular health in young, healthy adults. METHODS Twenty-one regular users of e-cigarettes (ECU) and twenty-one demographically matched non-users (NU) completed this study. Vascular health was assessed in the cutaneous microcirculation through different reactivity tests to evaluate overall functionality, endothelium-dependent vasodilation (EDD), and endothelium-independent vasodilation (EID). Macrovascular function was assessed using flow-mediated dilation (FMD). RESULTS Our results suggest that regular users of e-cigarettes present with premature microvascular impairment when compared to non-users. Specifically, they exhibit lower hyperemic (p = 0.003), thermal (p = 0.010), and EDD (p = 0.004) responses. No differences in EID between the groups were identified. We also identified that individuals who use e-cigarettes for longer than 3 years also present with systemic manifestations, as observed by significantly reduced macrovascular (p = 0.002) and microvascular (p ≤ 0.044) function. CONCLUSIONS Our novel data suggests that young, apparently healthy, regular users of e-cigarettes present with premature vascular dysfunction in the microcirculation when compared to non-users. We have also identified systemic vascular dysfunction affecting both the micro and macrovasculature in those young individuals who used e-cigarettes for longer than 3 years. Taken together, these findings associate regular e-cigarette use with premature vascular dysfunctions and adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Chloe Matheson
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, 817 West Franklin Street, Richmond, VA, 23284, USA
| | - Tijana Simovic
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, 817 West Franklin Street, Richmond, VA, 23284, USA
| | - Allison Heefner
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, 817 West Franklin Street, Richmond, VA, 23284, USA
- School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Marisa Colon
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, 817 West Franklin Street, Richmond, VA, 23284, USA
| | - Enrique Tunon
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, 817 West Franklin Street, Richmond, VA, 23284, USA
| | - Kolton Cobb
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, 817 West Franklin Street, Richmond, VA, 23284, USA
| | - Christopher Thode
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, 817 West Franklin Street, Richmond, VA, 23284, USA
| | - Alison Breland
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Caroline O Cobb
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Patrick Nana-Sinkam
- Division of Pulmonary and Critical Care, Virginia Commonwealth University, Richmond, VA, USA
| | - Ryan Garten
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, 817 West Franklin Street, Richmond, VA, 23284, USA
| | - Paula Rodriguez-Miguelez
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, 817 West Franklin Street, Richmond, VA, 23284, USA.
- Division of Pulmonary and Critical Care, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
37
|
Park S, Cho SW. Bioengineering toolkits for potentiating organoid therapeutics. Adv Drug Deliv Rev 2024; 208:115238. [PMID: 38447933 DOI: 10.1016/j.addr.2024.115238] [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: 09/26/2023] [Revised: 01/28/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Organoids are three-dimensional, multicellular constructs that recapitulate the structural and functional features of specific organs. Because of these characteristics, organoids have been widely applied in biomedical research in recent decades. Remarkable advancements in organoid technology have positioned them as promising candidates for regenerative medicine. However, current organoids still have limitations, such as the absence of internal vasculature, limited functionality, and a small size that is not commensurate with that of actual organs. These limitations hinder their survival and regenerative effects after transplantation. Another significant concern is the reliance on mouse tumor-derived matrix in organoid culture, which is unsuitable for clinical translation due to its tumor origin and safety issues. Therefore, our aim is to describe engineering strategies and alternative biocompatible materials that can facilitate the practical applications of organoids in regenerative medicine. Furthermore, we highlight meaningful progress in organoid transplantation, with a particular emphasis on the functional restoration of various organs.
Collapse
Affiliation(s)
- Sewon Park
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea; Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
38
|
Krajcsir B, Pócsi M, Fejes Z, Nagy B, Kappelmayer J, Beke Debreceni I. Ponatinib Induces a Procoagulant Phenotype in Human Coronary Endothelial Cells via Inducing Apoptosis. Pharmaceutics 2024; 16:559. [PMID: 38675220 PMCID: PMC11055157 DOI: 10.3390/pharmaceutics16040559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
BCR-ABL tyrosine kinase inhibitors (TKIs) are effective drugs in the treatment of patients with chronic myeloid leukemia. However, based on clinical studies, ponatinib was associated with the development of thrombotic complications. Since endothelial cells (ECs) regulate blood coagulation, their abnormal phenotype may play a role in the development of thrombotic events. We here aimed to investigate the effect of ponatinib on the procoagulant activity of cultured endothelial cells in vitro. Human coronary artery endothelial cells (HCAECs) were incubated with 50, 150, and 1000 nM of ponatinib. Subsequently, phosphatidylserine (PS) exposure and endothelial microvesicles (EMVs) were measured by flow cytometry. In addition, EC- and EMV-dependent thrombin generation was analyzed. To investigate pro-apoptotic effects of ponatinib, the level of Bax and Bcl-xL proteins were studied using Western blot and F3, THBD, and VCAM1 mRNAs were quantified by qPCR. Therapeutic concentrations of ponatinib significantly increased PS expression on ECs and the amount of EMVs which significantly shortened the time parameters of thrombin generation. In addition, these changes were associated with an increased ratio of Bax and Bcl-xL proteins in the presence of the decreased THBD mRNA level. Overall, ponatinib enhances the procoagulant activity of ECs via inducing apoptosis, which may contribute to thrombotic events.
Collapse
Affiliation(s)
- Bálint Krajcsir
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
- Laki Kálmán Doctoral School, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Marianna Pócsi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
| | - Zsolt Fejes
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
| | - János Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
| | - Ildikó Beke Debreceni
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.K.); (M.P.); (Z.F.); (J.K.)
| |
Collapse
|
39
|
Ta HQ, Kuppusamy M, Sonkusare SK, Roeser ME, Laubach VE. The endothelium: gatekeeper to lung ischemia-reperfusion injury. Respir Res 2024; 25:172. [PMID: 38637760 PMCID: PMC11027545 DOI: 10.1186/s12931-024-02776-4] [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: 02/09/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
The success of lung transplantation is limited by the high rate of primary graft dysfunction due to ischemia-reperfusion injury (IRI). Lung IRI is characterized by a robust inflammatory response, lung dysfunction, endothelial barrier disruption, oxidative stress, vascular permeability, edema, and neutrophil infiltration. These events are dependent on the health of the endothelium, which is a primary target of IRI that results in pulmonary endothelial barrier dysfunction. Over the past 10 years, research has focused more on the endothelium, which is beginning to unravel the multi-factorial pathogenesis and immunologic mechanisms underlying IRI. Many important proteins, receptors, and signaling pathways that are involved in the pathogenesis of endothelial dysfunction after IR are starting to be identified and targeted as prospective therapies for lung IRI. In this review, we highlight the more significant mediators of IRI-induced endothelial dysfunction discovered over the past decade including the extracellular glycocalyx, endothelial ion channels, purinergic receptors, kinases, and integrins. While there are no definitive clinical therapies currently available to prevent lung IRI, we will discuss potential clinical strategies for targeting the endothelium for the treatment or prevention of IRI. The accruing evidence on the essential role the endothelium plays in lung IRI suggests that promising endothelial-directed treatments may be approaching the clinic soon. The application of therapies targeting the pulmonary endothelium may help to halt this rapid and potentially fatal injury.
Collapse
Affiliation(s)
- Huy Q Ta
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA
| | - Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Swapnil K Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Mark E Roeser
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA
| | - Victor E Laubach
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA.
| |
Collapse
|
40
|
Alvarado-Ojeda ZA, Trejo-Moreno C, Ferat-Osorio E, Méndez-Martínez M, Fragoso G, Rosas-Salgado G. Role of Angiotensin II in Non-Alcoholic Steatosis Development. Arch Med Res 2024; 55:102986. [PMID: 38492325 DOI: 10.1016/j.arcmed.2024.102986] [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: 08/30/2023] [Revised: 02/23/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Fatty liver is a multifactorial disease characterized by excessive accumulation of lipids in hepatocytes (steatosis), insulin resistance, oxidative stress, and inflammation. This disease has a major public health impact because it is the first stage of a chronic and degenerative process in the liver that can lead to steatohepatitis, cirrhosis, and liver cancer. Although this disease is mainly diagnosed in patients with obesity, type 2 diabetes mellitus, and dyslipidemia, recent evidence indicates that vasoactive hormones such as angiotensin II (ANGII) not only promote endothelial dysfunction (ED) and hypertension, but also cause fatty liver, increase adipose tissue, and develop a pro-steatotic environment characterized by a low-grade systemic pro-inflammatory and pro-oxidant state, with elevated blood lipid levels. The role of ANGII in lipid accumulation has been little studied, so this review aims to summarize existing reports on the possible mechanism of action of ANGII in inducing lipid accumulation in hepatocytes.
Collapse
Affiliation(s)
| | - Celeste Trejo-Moreno
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca Morelos, Mexico
| | - Eduardo Ferat-Osorio
- División de Investigación en Salud, Unidad de Investigación en Epidemiología Clínica, Hospital de Especialidades, Dr. Bernardo Sepúlveda Gutiérrez, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Marisol Méndez-Martínez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| | - Gladis Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gabriela Rosas-Salgado
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca Morelos, Mexico.
| |
Collapse
|
41
|
Shekhtman O, Sioutas GS, Piavchenko G, Bhalla S, Cooke DL, Winkler E, Burkhardt JK, Srinivasan VM. Endovascular biopsy in neurointerventional surgery: A systematic review. Interv Neuroradiol 2024:15910199241240508. [PMID: 38515364 PMCID: PMC11569772 DOI: 10.1177/15910199241240508] [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: 12/13/2023] [Accepted: 03/04/2024] [Indexed: 03/23/2024] Open
Abstract
INTRODUCTION Endothelial cells (ECs) continuously line the cerebrovasculature. Molecular aberrations in the ECs are hallmarks and contributory factors to the development of cerebrovascular diseases, including intracranial aneurysms and arteriovenous malformations (AVMs). Endovascular biopsy has been introduced as a method to harvest ECs and obtain relevant biologic information. We aimed to summarize the literature on endovascular biopsy in neurointerventional surgery. METHODS We conducted a comprehensive literature search in multiple databases, identifying eligible studies focusing on neurosurgical applications of endovascular biopsy. The systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. The relevant information was collected, including study characteristics, biopsy techniques, and key findings. RESULTS Nine studies met the inclusion criteria and were included. The studies involved the collection of ECs using various endovascular devices including coils, guide wires, different stents, and forceps. Endothelial-enrichment techniques, such fluorescence-activated cell sorting (FACS), collected ECs and facilitated downstream applications of bulk or single-cell RNA sequencing (scRNAseq). The studies provided insights into gene expression profiles and identified potential biomarkers associated with intracranial aneurysms. However, challenges were observed in obtaining an adequate number of ECs and identifying consistent biomarkers. CONCLUSION Endovascular biopsy of endothelial cells (ECs) in cerebrovascular pathologies shows promise for gene expression profiling. However, many studies have been limited in sample size and underpowered to identify "signature genes" for aneurysm growth or rupture. Advancements in minimally invasive biopsy methods have potential to facilitate applications of precision medicine in the treatment of cerebrovascular disorders.
Collapse
Affiliation(s)
- Oleg Shekhtman
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Georgios S Sioutas
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Gennadii Piavchenko
- Department of Human Anatomy and Histology, Sechenov University, Moscow, Russia
| | - Shubhang Bhalla
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Daniel L Cooke
- Department of Interventional Radiology, University of California San Francisco, San Francisco, CA, USA
| | - Ethan Winkler
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jan-Karl Burkhardt
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Visish M Srinivasan
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
42
|
Sirca TB, Mureșan ME, Pallag A, Marian E, Jurca T, Vicaș LG, Tunduc IP, Manole F, Ștefan L. The Role of Polyphenols in Modulating PON1 Activity Regarding Endothelial Dysfunction and Atherosclerosis. Int J Mol Sci 2024; 25:2962. [PMID: 38474211 DOI: 10.3390/ijms25052962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/24/2024] [Accepted: 03/02/2024] [Indexed: 03/14/2024] Open
Abstract
The incidence and prevalence of cardiovascular diseases are still rising. The principal mechanism that drives them is atherosclerosis, an affection given by dyslipidemia and a pro-inflammatory state. Paraoxonase enzymes have a protective role due to their ability to contribute to antioxidant and anti-inflammatory pathways, especially paraoxonase 1 (PON1). PON1 binds with HDL (high-density lipoprotein), and high serum levels lead to a protective state against dyslipidemia, cardiovascular diseases, diabetes, stroke, nonalcoholic fatty liver disease, and many others. Modulating PON1 expression might be a treatment objective with significant results in limiting the prevalence of atherosclerosis. Lifestyle including diet and exercise can raise its levels, and some beneficial plants have been found to influence PON1 levels; therefore, more studies on herbal components are needed. Our purpose is to highlight the principal roles of Praoxonase 1, its implications in dyslipidemia, cardiovascular diseases, stroke, and other diseases, and to emphasize plants that can modulate PON1 expression, targeting the potential of some flavonoids that could be introduced as supplements in our diet and to validate the hypothesis that flavonoids have any effects regarding PON1 function.
Collapse
Affiliation(s)
- Teodora Bianca Sirca
- Doctoral School of Biomedical Sciences, University of Oradea, No. 1 University Street, 410087 Oradea, Romania
| | - Mariana Eugenia Mureșan
- Doctoral School of Biomedical Sciences, University of Oradea, No. 1 University Street, 410087 Oradea, Romania
| | - Annamaria Pallag
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania
| | - Eleonora Marian
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania
| | - Tunde Jurca
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania
| | - Laura Grațiela Vicaș
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania
| | - Ioana Paula Tunduc
- Department of Cardiology, Clinical County Emergency Hospital of Bihor, Gheorghe Doja Street 65-67, 410169 Oradea, Romania
| | - Felicia Manole
- Department of Surgery, Faculty of Medicine and Pharmacy, University of Oradea, 1st December Square 10, 410073 Oradea, Romania
| | - Liana Ștefan
- Department of Surgery, Faculty of Medicine and Pharmacy, University of Oradea, 1st December Square 10, 410073 Oradea, Romania
| |
Collapse
|
43
|
Yu X, Zhu L. Nanoparticles for the Treatment of Bone Metastasis in Breast Cancer: Recent Advances and Challenges. Int J Nanomedicine 2024; 19:1867-1886. [PMID: 38414525 PMCID: PMC10898486 DOI: 10.2147/ijn.s442768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/15/2024] [Indexed: 02/29/2024] Open
Abstract
Although the frequency of bone metastases from breast cancer has increased, effective treatment is lacking, prompting the development of nanomedicine, which involves the use of nanotechnology for disease diagnosis and treatment. Nanocarrier drug delivery systems offer several advantages over traditional drug delivery methods, such as higher reliability and biological activity, improved penetration and retention, and precise targeting and delivery. Various nanoparticles that can selectively target tumor cells without causing harm to healthy cells or organs have been synthesized. Recent advances in nanotechnology have enabled the diagnosis and prevention of metastatic diseases as well as the ability to deliver complex molecular "cargo" particles to metastatic regions. Nanoparticles can modulate systemic biodistribution and enable the targeted accumulation of therapeutic agents. Several delivery strategies are used to treat bone metastases, including untargeted delivery, bone-targeted delivery, and cancer cell-targeted delivery. Combining targeted agents with nanoparticles enhances the selective delivery of payloads to breast cancer bone metastatic lesions, providing multiple delivery advantages for treatment. In this review, we describe recent advances in nanoparticle development for treating breast cancer bone metastases.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan Province, People's Republic of China
| | - Lingling Zhu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| |
Collapse
|
44
|
Yu JE, Yeo IJ, Han SB, Yun J, Kim B, Yong YJ, Lim YS, Kim TH, Son DJ, Hong JT. Significance of chitinase-3-like protein 1 in the pathogenesis of inflammatory diseases and cancer. Exp Mol Med 2024; 56:1-18. [PMID: 38177294 PMCID: PMC10834487 DOI: 10.1038/s12276-023-01131-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/06/2023] [Accepted: 08/28/2023] [Indexed: 01/06/2024] Open
Abstract
Chitinase-3-like protein 1 (CHI3L1) is a secreted glycoprotein that mediates inflammation, macrophage polarization, apoptosis, and carcinogenesis. The expression of CHI3L1 is strongly upregulated by various inflammatory and immunological diseases, including several cancers, Alzheimer's disease, and atherosclerosis. Several studies have shown that CHI3L1 can be considered as a marker of disease diagnosis, prognosis, disease activity, and severity. In addition, the proinflammatory action of CHI3L1 may be mediated via responses to various proinflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, interleukin-6, and interferon-γ. Therefore, CHI3L1 may contribute to a vast array of inflammatory diseases. However, its pathophysiological and pharmacological roles in the development of inflammatory diseases remain unclear. In this article, we review recent findings regarding the roles of CHI3L1 in the development of inflammatory diseases and suggest therapeutic approaches that target CHI3L1.
Collapse
Affiliation(s)
- Ji Eun Yu
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
- College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Bongcheol Kim
- Senelix Co. Ltd., 25, Beobwon-ro 11-gil, Songpa-gu, Seoul, 05836, Republic of Korea
| | - Yoon Ji Yong
- PRESTI GEBIOLOGICS Co. Ltd., Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28161, Republic of Korea
| | - Young-Soo Lim
- PRESTI GEBIOLOGICS Co. Ltd., Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28161, Republic of Korea
| | - Tae Hun Kim
- Autotelic Bio Inc., Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea.
| |
Collapse
|
45
|
Wongsawat M, Glaharn S, Srisook C, Dechkhajorn W, Chaisri U, Punsawad C, Techarang T, Chotivanich K, Krudsood S, Viriyavejakul P. Immunofluorescence study of cytoskeleton in endothelial cells induced with malaria sera. Malar J 2024; 23:10. [PMID: 38183117 PMCID: PMC10770940 DOI: 10.1186/s12936-023-04833-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Endothelial cells (ECs) play a major role in malaria pathogenesis, as a point of direct contact of parasitized red blood cells to the blood vessel wall. The study of cytoskeleton structures of ECs, whose main functions are to maintain shape and provide strength to the EC membrane is important in determining the severe sequelae of Plasmodium falciparum malaria. The work investigated the cytoskeletal changes (microfilaments-actin, microtubules-tubulin and intermediate filaments-vimentin) in ECs induced by malaria sera (Plasmodium vivax, uncomplicated P. falciparum and complicated P. falciparum), in relation to the levels of pro-inflammatory cytokines. METHODS Morphology and fluorescence intensity of EC cytoskeleton stimulated with malaria sera were evaluated using immunofluorescence technique. Levels of tumour necrosis factor (TNF) and interferon (IFN)-gamma (γ) were determined using enzyme-linked immunosorbent assay (ELISA). Control experimental groups included ECs incubated with media alone and non-malaria patient sera. Experimental groups consisted of ECs incubated with malaria sera from P. vivax, uncomplicated P. falciparum and complicated P. falciparum. Morphological scores of cytoskeletal alterations and fluorescence intensity were compared across each experiment group, and correlated with TNF and IFN-γ. RESULTS The four morphological changes of cytoskeleton included (1) shrinkage of cytoskeleton and ECs with cortical condensation, (2) appearance of eccentric nuclei, (3) presence of "spiking pattern" of cytoskeleton and EC membrane, and (4) fragmentation and discontinuity of cytoskeleton and ECs. Significant damages were noted in actin filaments compared to tubulin and vimentin filaments in ECs stimulated with sera from complicated P. falciparum malaria. Morphological damages to cytoskeleton was positively correlated with fluorescence intensity and the levels of TNF and IFN-γ. CONCLUSIONS ECs stimulated with sera from complicated P. falciparum malaria showed cytoskeletal alterations and increased in fluorescence intensity, which was associated with high levels of TNF and IFN-γ. Cytoskeletal changes of ECs incubated with complicated P. falciparum malaria sera can lead to EC junctional alteration and permeability changes, which is mediated through apoptotic pathway. The findings can serve as a basis to explore measures to strengthen EC cytoskeleton and alleviate severe malaria complications such as pulmonary oedema and cerebral malaria. In addition, immunofluorescence intensity of cytoskeleton could be investigated as potential prognostic indicator for malaria severity.
Collapse
Affiliation(s)
- Mathusorn Wongsawat
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Supattra Glaharn
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Charit Srisook
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Wilanee Dechkhajorn
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Urai Chaisri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Chuchard Punsawad
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Tachpon Techarang
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Kesinee Chotivanich
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Srivicha Krudsood
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Parnpen Viriyavejakul
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand.
| |
Collapse
|
46
|
Borek-Dorosz A, Pieczara A, Orleanska J, Brzozowski K, Tipping W, Graham D, Bik E, Kubrak A, Baranska M, Majzner K. Raman microscopy reveals how cell inflammation activates glucose and lipid metabolism. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119575. [PMID: 37689141 DOI: 10.1016/j.bbamcr.2023.119575] [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: 03/31/2023] [Revised: 08/11/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023]
Abstract
Metabolism of endothelial cells (ECs) depends on the availability of the energy substrates. Since the endothelium is the first line of defence against inflammation in the cardiovascular system and its dysfunction can lead to the development of cardiovascular diseases, it is important to understand how glucose metabolism changes during inflammation. In this work, glucose uptake was studied in human microvascular endothelial cells (HMEC-1) in high glucose (HG), and additionally in an inflammatory state, using Raman imaging. HG state was induced by incubation of ECs with a deuterated glucose analogue, while the EC inflammation was caused by TNF-α pre-treatment. Spontaneous and stimulated Raman scattering spectroscopy provided comprehensive information on biochemical changes, including lipids and the extent of unsaturation induced by excess glucose in ECs., induced by excess glucose in ECs. In this work, we indicated spectroscopic markers of metabolic changes in ECs as a strong increase in the ratio of the intensity of lipids / (proteins + lipids) bands and an increase in the level of lipid unsaturation and mitochondrial changes. Inflamed ECs treated with HG, revealed enhanced glucose uptake, and intensified lipid production i.a. of unsaturated lipids. Additionally, increased cytochrome c signal in the mitochondrial region indicated higher mitochondrial activity and biogenesis. Raman spectroscopy is a powerful method for determining the metabolic markers of ED which will better inform understanding of disease onset, development, and treatment.
Collapse
Affiliation(s)
- Aleksandra Borek-Dorosz
- Jagiellonian University in Kraków, Faculty of Chemistry, Department of Chemical Physics, 2 Gronostajowa Str., Krakow, Poland
| | - Anna Pieczara
- Jagiellonian University in Kraków, Jagiellonian Centre for Experimental Therapeutics (JCET), 14 Bobrzynskiego Str., Krakow, Poland; Jagiellonian University in Kraków, Doctoral School of Exact and Natural Sciences, 11 Lojasiewicza St., Krakow, Poland
| | - Jagoda Orleanska
- Jagiellonian University in Kraków, Faculty of Chemistry, Department of Chemical Physics, 2 Gronostajowa Str., Krakow, Poland; Jagiellonian University in Kraków, Doctoral School of Exact and Natural Sciences, 11 Lojasiewicza St., Krakow, Poland
| | - Krzysztof Brzozowski
- Jagiellonian University in Kraków, Faculty of Chemistry, Department of Chemical Physics, 2 Gronostajowa Str., Krakow, Poland
| | - William Tipping
- Centre for Molecular Nanometrology, WestCHEM, Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, Glasgow G1 1RD, United Kingdom
| | - Duncan Graham
- Centre for Molecular Nanometrology, WestCHEM, Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, Glasgow G1 1RD, United Kingdom
| | - Ewelina Bik
- Jagiellonian University in Kraków, Jagiellonian Centre for Experimental Therapeutics (JCET), 14 Bobrzynskiego Str., Krakow, Poland; Academic Centre for Materials and Nanotechnology, AGH University of Science and Technology, 30 Mickiewicza Str., Krakow, Poland
| | - Adam Kubrak
- Jagiellonian University in Kraków, Faculty of Chemistry, Department of Chemical Physics, 2 Gronostajowa Str., Krakow, Poland
| | - Malgorzata Baranska
- Jagiellonian University in Kraków, Faculty of Chemistry, Department of Chemical Physics, 2 Gronostajowa Str., Krakow, Poland; Jagiellonian University in Kraków, Jagiellonian Centre for Experimental Therapeutics (JCET), 14 Bobrzynskiego Str., Krakow, Poland
| | - Katarzyna Majzner
- Jagiellonian University in Kraków, Faculty of Chemistry, Department of Chemical Physics, 2 Gronostajowa Str., Krakow, Poland.
| |
Collapse
|
47
|
Bronson R, Lyu J, Xiong J. Transcriptome analysis reveals molecular signature and cell-type difference of Homo sapiens endothelial-to-mesenchymal transition. G3 (BETHESDA, MD.) 2023; 13:jkad243. [PMID: 37857450 PMCID: PMC10700110 DOI: 10.1093/g3journal/jkad243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/01/2023] [Accepted: 10/14/2023] [Indexed: 10/21/2023]
Abstract
Endothelial-to-mesenchymal transition (EndoMT), a specific form of epithelial-to-mesenchymal transition, drives a growing number of human (Homo sapiens) pathological conditions. This emerging knowledge opens a path to discovering novel therapeutic targets for many EndoMT-associated disorders. Here, we constructed an atlas of the endothelial-cell transcriptome and demonstrated EndoMT-induced global changes in transcriptional gene expression. Our gene ontology analyses showed that EndoMT could be a specific checkpoint for leukocyte chemotaxis, adhesion, and transendothelial migration. We also identified distinct gene expression signatures underlying EndoMT across arterial, venous, and microvascular endothelial cells. We performed protein-protein interaction network analyses, identifying a class of highly connected hub genes in endothelial cells from different vascular beds. Moreover, we found that the short-chain fatty acid acetate strongly inhibits the transcriptional program of EndoMT in endothelial cells from different vascular beds across tissues. Our results reveal the molecular signature and cell-type difference of EndoMT across distinct tissue- and vascular-bed-specific endothelial cells, providing a powerful discovery tool and resource value. These results suggest that therapeutically manipulating the endothelial transcriptome could treat an increasing number of EndoMT-associated pathological conditions.
Collapse
Affiliation(s)
- Ronald Bronson
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, St.Petersburg, FL 33701, USA
- Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St.Petersburg, FL 33701, USA
| | - Junfang Lyu
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, St.Petersburg, FL 33701, USA
- Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St.Petersburg, FL 33701, USA
| | - Jianhua Xiong
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, St.Petersburg, FL 33701, USA
- Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St.Petersburg, FL 33701, USA
| |
Collapse
|
48
|
Cheng DCY, Climie RE, Shu M, Grieve SM, Kozor R, Figtree GA. Vascular aging and cardiovascular disease: pathophysiology and measurement in the coronary arteries. Front Cardiovasc Med 2023; 10:1206156. [PMID: 38089775 PMCID: PMC10715672 DOI: 10.3389/fcvm.2023.1206156] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2024] Open
Abstract
Age is a key risk factor for cardiovascular disease, including atherosclerosis. However, pathophysiological disease processes in the arteries are not an inevitable feature of aging. Large cohort studies with arterial phenotyping along with clinical and demographic data are essential to better understand factors related to the susceptibility or resilience to age-related vascular pathophysiology in humans. This review explores the mechanisms by which vascular structure and function alters with age, and how these changes relate to cardiovascular pathophysiology and disease. Features of vascular aging in the coronary arteries have historically been difficult to quantify pre-mortem due to their size and location. However, non-invasive imaging modalities including CT Coronary Angiogram are now being used to assess coronary vascular age, and further advances in imaging analysis such as the CT Fat Attenuation Index will help provide further measurement of features associated with coronary vascular aging. Currently, markers of vascular aging are not used as therapeutic targets in routine clinical practice, but non-pharmacological interventions including aerobic exercise and low salt diet, as well as anti-hypertensives have been demonstrated to reduce arterial stiffness. Advances in imaging technology, both in acquisition and advanced analysis, as well as harmonisation of measurements for researchers across the globe will be invaluable in understanding what constitutes healthy vascular aging and in identifying features of vascular aging that are associated with coronary artery disease and its adverse outcomes. Assessing such images in large cohorts can facilitate improved definitions of resilient and susceptible phenotypes to vascular aging in the coronary arteries. This is a critical step in identifying further risk factors and biomarkers within these groups and driving forward the development of novel therapies aimed at slowing or stopping age-related vascular changes in the coronary arteries.
Collapse
Affiliation(s)
- Daniel C. Y. Cheng
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Rachel E. Climie
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Matthew Shu
- Northern Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Stuart M. Grieve
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Rebecca Kozor
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Gemma A. Figtree
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Sydney, NSW, Australia
| |
Collapse
|
49
|
Stierschneider A, Wiesner C. Shedding light on the molecular and regulatory mechanisms of TLR4 signaling in endothelial cells under physiological and inflamed conditions. Front Immunol 2023; 14:1264889. [PMID: 38077393 PMCID: PMC10704247 DOI: 10.3389/fimmu.2023.1264889] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Toll-like receptor 4 (TLR4) are part of the innate immune system. They are capable of recognizing pathogen-associated molecular patterns (PAMPS) of microbes, and damage-associated molecular patterns (DAMPs) of damaged tissues. Activation of TLR4 initiates downstream signaling pathways that trigger the secretion of cytokines, type I interferons, and other pro-inflammatory mediators that are necessary for an immediate immune response. However, the systemic release of pro-inflammatory proteins is a powerful driver of acute and chronic inflammatory responses. Over the past decades, immense progress has been made in clarifying the molecular and regulatory mechanisms of TLR4 signaling in inflammation. However, the most common strategies used to study TLR4 signaling rely on genetic manipulation of the TLR4 or the treatment with agonists such as lipopolysaccharide (LPS) derived from the outer membrane of Gram-negative bacteria, which are often associated with the generation of irreversible phenotypes in the target cells or unintended cytotoxicity and signaling crosstalk due to off-target or pleiotropic effects. Here, optogenetics offers an alternative strategy to control and monitor cellular signaling in an unprecedented spatiotemporally precise, dose-dependent, and non-invasive manner. This review provides an overview of the structure, function and signaling pathways of the TLR4 and its fundamental role in endothelial cells under physiological and inflammatory conditions, as well as the advances in TLR4 modulation strategies.
Collapse
Affiliation(s)
| | - Christoph Wiesner
- Department Science & Technology, Institute Biotechnology, IMC Krems University of Applied Sciences, Krems, Austria
| |
Collapse
|
50
|
Davodabadi F, Sajjadi SF, Sarhadi M, Mirghasemi S, Nadali Hezaveh M, Khosravi S, Kamali Andani M, Cordani M, Basiri M, Ghavami S. Cancer chemotherapy resistance: Mechanisms and recent breakthrough in targeted drug delivery. Eur J Pharmacol 2023; 958:176013. [PMID: 37633322 DOI: 10.1016/j.ejphar.2023.176013] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Conventional chemotherapy, one of the most widely used cancer treatment methods, has serious side effects, and usually results in cancer treatment failure. Drug resistance is one of the primary reasons for this failure. The most significant drawbacks of systemic chemotherapy are rapid clearance from the circulation, the drug's low concentration in the tumor site, and considerable adverse effects outside the tumor. Several ways have been developed to boost neoplasm treatment efficacy and overcome medication resistance. In recent years, targeted drug delivery has become an essential therapeutic application. As more mechanisms of tumor treatment resistance are discovered, nanoparticles (NPs) are designed to target these pathways. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation. Nano-drugs have been increasingly employed in medicine, incorporating therapeutic applications for more precise and effective tumor diagnosis, therapy, and targeting. Many benefits of NP-based drug delivery systems in cancer treatment have been proven, including good pharmacokinetics, tumor cell-specific targeting, decreased side effects, and lessened drug resistance. As more mechanisms of tumor treatment resistance are discovered, NPs are designed to target these pathways. At the moment, this innovative technology has the potential to bring fresh insights into cancer therapy. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shaghayegh Mirghasemi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Nadali Hezaveh
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Samin Khosravi
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Kamali Andani
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Saeid Ghavami
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555. Katowice, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada.
| |
Collapse
|