1
|
Grego A, Fernandes C, Fonseca I, Dias-Neto M, Costa R, Leite-Moreira A, Oliveira SM, Trindade F, Nogueira-Ferreira R. Endothelial dysfunction in cardiovascular diseases: mechanisms and in vitro models. Mol Cell Biochem 2025:10.1007/s11010-025-05289-w. [PMID: 40259179 DOI: 10.1007/s11010-025-05289-w] [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: 02/09/2025] [Accepted: 04/08/2025] [Indexed: 04/23/2025]
Abstract
Endothelial cells (ECs) are arranged side-by-side to create a semi-permeable monolayer, forming the inner lining of every blood vessel (micro and macrocirculation). Serving as the first barrier for circulating molecules and cells, ECs represent the main regulators of vascular homeostasis being able to respond to environmental changes, either physical or chemical signals, by producing several factors that regulate vascular tone and cellular adhesion. Healthy endothelium has anticoagulant properties that prevent the adhesion of leukocytes and platelets to the vessel walls, contributing to resistance to thrombus formation, and regulating inflammation, and vascular smooth muscle cell proliferation. Many risk factors of cardiovascular diseases (CVDs) promote the endothelial expression of chemokines, cytokines, and adhesion molecules. The resultant endothelial activation can lead to endothelial cell dysfunction (ECD). In vitro models of ECD allow the study of cellular and molecular mechanisms of disease and provide a research platform for screening potential therapeutic agents. Even though alternative models are available, such as animal models or ex vivo models, in vitro models offer higher experimental flexibility and reproducibility, making them a valuable tool for the understanding of pathophysiological mechanisms of several diseases, such as CVDs. Therefore, this review aims to synthesize the currently available in vitro models regarding ECD, emphasizing CVDs. This work will focus on 2D cell culture models (endothelial cell lines and primary ECs), 3D cell culture systems (scaffold-free and scaffold-based), and 3D cell culture models (such as organ-on-a-chip). We will dissect the role of external stimuli-chemical and mechanical-in triggering ECD.
Collapse
Affiliation(s)
- Ana Grego
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Cristiana Fernandes
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Ivo Fonseca
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Marina Dias-Neto
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Raquel Costa
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - Adelino Leite-Moreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Cardiothoracic Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Sandra Marisa Oliveira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Fábio Trindade
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Rita Nogueira-Ferreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
2
|
Taylor MS, Francis M, Choi CS. Flow-Dependent Modulation of Endothelial Ca 2+ Dynamics by Small Conductance Ca 2+-Activated K + Channels in Mouse Carotid Arteries. Biomedicines 2024; 12:2900. [PMID: 39767806 PMCID: PMC11727411 DOI: 10.3390/biomedicines12122900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Small conductance Ca2+ activated K+ channels (KCa2.3) are important regulators of vascular function. They provide Ca2+-dependent hyperpolarization of the endothelial membrane potential, promoting agonist-induced vasodilation. Another important mechanism of influence may occur through positive feedback regulation of endothelial Ca2+ signals, likely via amplification of influx through membrane cation channels. KCa2.3 channels have recently been implicated in flow-mediated dilation of the arterial vasculature and may contribute to the crucial homeostatic role of shear stress in preventing vascular wall remodeling and progressive vascular disease (i.e., atherosclerosis). The impact of KCa2.3 channels on endothelial Ca2+ signaling under physiologically relevant shear stress conditions remains unknown. METHODS In the current study, we employ mice expressing an endothelium-specific Ca2+ fluorophore (cdh5-GCaMP8) to characterize the KCa2.3 channel influence on the dynamic Ca2+ signaling profile along the arterial endothelium in the presence and absence of shear-stress. RESULTS Our data indicate KCa2.3 channels have a minimal influence on basal Ca2+ signaling in the carotid artery endothelium in the absence of flow, but they contribute substantially to amplification of Ca2+ dynamics in the presence of flow and their influence can be augmented through exogenous positive modulation. CONCLUSIONS The findings suggest a pivotal role for KCa2.3 channels in adjusting the profile of homeostatic dynamic Ca2+ signals along the arterial intima under flow.
Collapse
Affiliation(s)
- Mark S. Taylor
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA; (M.F.); (C.-S.C.)
| | | | | |
Collapse
|
3
|
Baek IH, Helms V, Kim Y. Genetically Engineered Filamentous Bacteriophages Displaying TGF-β1 Promote Angiogenesis in 3D Microenvironments. J Funct Biomater 2024; 15:314. [PMID: 39590518 PMCID: PMC11594957 DOI: 10.3390/jfb15110314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Combined 3D cell culture in vitro assays with microenvironment-mimicking systems are effective for cell-based screening tests of drug and chemical toxicity. Filamentous bacteriophages have diverse applications in material science, drug delivery, tissue engineering, energy, and biosensor development. Specifically, genetically modified bacteriophages have the potential to deliver therapeutic molecules or genes to targeted tumor tissues. The engineered bacteriophages in this study significantly enhanced endothelial cell migration and tube formation within the extracellular matrix (ECM). Compared to TGF-β1 alone and non-modified phages, the presence of TGF-β1 on the bacteriophages demonstrated superior performance as a continuous stimulant in the microenvironment, effectively promoting these angiogenic processes. Assays, including RT-qPCR, ELISA, and fluorescence microscopy, confirmed the expression of angiogenic markers such as CD31, validating the formation of 3D angiogenic structures. Our findings indicate that the TGF-β1 displayed by bacteriophages likely acted as a chemotactic factor, promoting the migration, proliferation, and tube formation of endothelial cells (ECs) within the ECM. Although direct contact between ECs and bacteriophages was not explicitly confirmed, the observed effects strongly suggest that TGF-β1-RGD bacteriophages contributed to the stimulation of angiogenic processes. The formation of angiogenic structures by ECs in the ECM was confirmed as three-dimensional and regulated by the surface treatment of microfluidic channels. These results suggest that biocompatible TGF-β1-displaying bacteriophages could continuously stimulate the microenvironment in vitro for angiogenesis models. Furthermore, we demonstrated that these functionalized bacteriophages have the potential to be utilized as versatile biomaterials in the field of biomedical engineering. Similar strategies could be applied to develop angiogenic matrices for tissue engineering in in vitro assays.
Collapse
Affiliation(s)
- In-Hyuk Baek
- Environmental Safety Group, Korea Institute of Science & Technology Europe GmbH, Campus E71, 66123 Saarbrücken, Germany;
- Center for Bioinformatics, Saarland University, 66123 Saarbrücken, Germany;
| | - Volkhard Helms
- Center for Bioinformatics, Saarland University, 66123 Saarbrücken, Germany;
| | - Youngjun Kim
- Environmental Safety Group, Korea Institute of Science & Technology Europe GmbH, Campus E71, 66123 Saarbrücken, Germany;
| |
Collapse
|
4
|
Kim J, Won C, Ham S, Han H, Shin S, Jang J, Lee S, Kwon C, Cho S, Park H, Lee D, Lee WJ, Lee T, Lee JH. Increased Susceptibility to Mechanical Stretch Drives the Persistence of Keloid Fibroblasts: An Investigation Using a Stretchable PDMS Platform. Biomedicines 2024; 12:2169. [PMID: 39457482 PMCID: PMC11504861 DOI: 10.3390/biomedicines12102169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Keloids are a common fibrotic disease of the skin, with the pathological hallmark of excessive extracellular matrix synthesis due to abnormal fibroblast activity. Since keloids clinically arise in areas of high mechanical tension, the mechanotransductory pathway may be attributed to its pathogenesis. We aimed to establish a preclinical platform to elucidate the underlying mechanism of keloid development and its clinical persistence. METHODS We fabricated a mechanically stretchable polydimethylsiloxane cell culture platform; with its mimicry of the in vivo cyclic stretch of skeletal muscles, cells showed higher proliferation compared with conventional modalities. RESULTS In response to mechanical strain, TGF-β and type 1 collagen showed significant increases, suggesting possible TGF-β/Smad pathway activation via mechanical stimulation. Protein candidates selected by proteomic analysis were evaluated, indicating that key molecules involved in cell signaling and oxidative stress were significantly altered. Additionally, the cytoskeletal network of keloid fibroblasts showed increased expression of its components after periodic mechanical stimulation. CONCLUSIONS Herein, we demonstrated and validated the existing body of knowledge regarding profibrotic mechanotransduction signaling pathways in keloid fibroblasts. Cyclic stretch, as a driving force, could help to decipher the tension-mediated biomechanical processes, leading to the development of optimized therapeutic targets.
Collapse
Affiliation(s)
- Jihee Kim
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.K.); (S.H.); (S.S.); (J.J.)
| | - Chihyeong Won
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, Republic of Korea; (C.W.); (H.H.); (S.L.); (C.K.); (S.C.); (H.P.)
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA 91125, USA
| | - Seoyoon Ham
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.K.); (S.H.); (S.S.); (J.J.)
| | - Heetak Han
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, Republic of Korea; (C.W.); (H.H.); (S.L.); (C.K.); (S.C.); (H.P.)
| | - Sungsik Shin
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.K.); (S.H.); (S.S.); (J.J.)
| | - Jieun Jang
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.K.); (S.H.); (S.S.); (J.J.)
| | - Sanghyeon Lee
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, Republic of Korea; (C.W.); (H.H.); (S.L.); (C.K.); (S.C.); (H.P.)
| | - Chaebeen Kwon
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, Republic of Korea; (C.W.); (H.H.); (S.L.); (C.K.); (S.C.); (H.P.)
| | - Sungjoon Cho
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, Republic of Korea; (C.W.); (H.H.); (S.L.); (C.K.); (S.C.); (H.P.)
| | - Hyeonjoo Park
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, Republic of Korea; (C.W.); (H.H.); (S.L.); (C.K.); (S.C.); (H.P.)
| | - Dongwon Lee
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (D.L.); (W.J.L.)
| | - Won Jai Lee
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (D.L.); (W.J.L.)
| | - Taeyoon Lee
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, Republic of Korea; (C.W.); (H.H.); (S.L.); (C.K.); (S.C.); (H.P.)
| | - Ju Hee Lee
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (J.K.); (S.H.); (S.S.); (J.J.)
| |
Collapse
|
5
|
Katsuta H, Sokabe M, Hirata H. From stress fiber to focal adhesion: a role of actin crosslinkers in force transmission. Front Cell Dev Biol 2024; 12:1444827. [PMID: 39193363 PMCID: PMC11347286 DOI: 10.3389/fcell.2024.1444827] [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: 06/06/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
The contractile apparatus, stress fiber (SF), is connected to the cell adhesion machinery, focal adhesion (FA), at the termini of SF. The SF-FA complex is essential for various mechanical activities of cells, including cell adhesion to the extracellular matrix (ECM), ECM rigidity sensing, and cell migration. This mini-review highlights the importance of SF mechanics in these cellular activities. Actin-crosslinking proteins solidify SFs by attenuating myosin-driven flows of actin and myosin filaments within the SF. In the solidified SFs, viscous slippage between actin filaments in SFs and between the filaments and the surrounding cytosol is reduced, leading to efficient transmission of myosin-generated contractile force along the SFs. Hence, SF solidification via actin crosslinking ensures exertion of a large force to FAs, enabling FA maturation, ECM rigidity sensing and cell migration. We further discuss intracellular mechanisms for tuning crosslinker-modulated SF mechanics and the potential relationship between the aberrance of SF mechanics and pathology including cancer.
Collapse
Affiliation(s)
- Hiroki Katsuta
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masahiro Sokabe
- Human Information Systems Laboratories, Kanazawa Institute of Technology, Hakusan, Japan
| | - Hiroaki Hirata
- Department of Applied Bioscience, Kanazawa Institute of Technology, Hakusan, Japan
| |
Collapse
|
6
|
Nwokoye PN, Abilez OJ. Blood vessels in a dish: the evolution, challenges, and potential of vascularized tissues and organoids. Front Cardiovasc Med 2024; 11:1336910. [PMID: 38938652 PMCID: PMC11210405 DOI: 10.3389/fcvm.2024.1336910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/19/2024] [Indexed: 06/29/2024] Open
Abstract
Vascular pathologies are prevalent in a broad spectrum of diseases, necessitating a deeper understanding of vascular biology, particularly in overcoming the oxygen and nutrient diffusion limit in tissue constructs. The evolution of vascularized tissues signifies a convergence of multiple scientific disciplines, encompassing the differentiation of human pluripotent stem cells (hPSCs) into vascular cells, the development of advanced three-dimensional (3D) bioprinting techniques, and the refinement of bioinks. These technologies are instrumental in creating intricate vascular networks essential for tissue viability, especially in thick, complex constructs. This review provides broad perspectives on the past, current state, and advancements in key areas, including the differentiation of hPSCs into specific vascular lineages, the potential and challenges of 3D bioprinting methods, and the role of innovative bioinks mimicking the native extracellular matrix. We also explore the integration of biophysical cues in vascularized tissues in vitro, highlighting their importance in stimulating vessel maturation and functionality. In this review, we aim to synthesize these diverse yet interconnected domains, offering a broad, multidisciplinary perspective on tissue vascularization. Advancements in this field will help address the global organ shortage and transform patient care.
Collapse
Affiliation(s)
- Peter N. Nwokoye
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Oscar J. Abilez
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
- Division of Pediatric CT Surgery, Stanford University, Stanford, CA, United States
- Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, United States
- Bio-X Program, Stanford University, Stanford, CA, United States
| |
Collapse
|
7
|
Baek KI, Ryu K. Role of Flow-Sensitive Endothelial Genes in Atherosclerosis and Antiatherogenic Therapeutics Development. J Cardiovasc Transl Res 2024; 17:609-623. [PMID: 38010480 DOI: 10.1007/s12265-023-10463-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease that is the underlying cause of cardiovascular disease which initiates from endothelial dysfunction from genetic and environmental risk factors, including biomechanical forces: blood flow. Endothelial cells (ECs) lining the inner arterial wall regions exposed to disturbed flow are prone to atherosclerosis development, whereas the straight regions exposed to stable flow are spared from the disease. These flow patterns induce genome- and epigenome-wide changes in gene expression in ECs. Through the sweeping changes in gene expression, disturbed flow reprograms ECs from athero-protected cell types under the stable flow condition to pro-atherogenic cell conditions. The pro-atherogenic changes induced by disturbed flow, in combination with additional risk factors such as hypercholesterolemia, lead to the progression of atherosclerosis. The flow-sensitive genes and proteins are critical in understanding the mechanisms and serve as novel targets for antiatherogenic therapeutics.
Collapse
Affiliation(s)
- Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Kitae Ryu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Department of Biotechnology, The University of Suwon, 17, Wauan-Gil, Bongdam-Eup, Hwaseong-Si, Gyeonggi-Do, 18323, Republic of Korea.
| |
Collapse
|
8
|
Hauger PC, Hordijk PL. Shear Stress-Induced AMP-Activated Protein Kinase Modulation in Endothelial Cells: Its Role in Metabolic Adaptions and Cardiovascular Disease. Int J Mol Sci 2024; 25:6047. [PMID: 38892235 PMCID: PMC11173107 DOI: 10.3390/ijms25116047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Endothelial cells (ECs) line the inner surface of all blood vessels and form a barrier that facilitates the controlled transfer of nutrients and oxygen from the circulatory system to surrounding tissues. Exposed to both laminar and turbulent blood flow, ECs are continuously subject to differential mechanical stimulation. It has been well established that the shear stress associated with laminar flow (LF) is atheroprotective, while shear stress in areas with turbulent flow (TF) correlates with EC dysfunction. Moreover, ECs show metabolic adaptions to physiological changes, such as metabolic shifts from quiescence to a proliferative state during angiogenesis. The AMP-activated protein kinase (AMPK) is at the center of these phenomena. AMPK has a central role as a metabolic sensor in several cell types. Moreover, in ECs, AMPK is mechanosensitive, linking mechanosensation with metabolic adaptions. Finally, recent studies indicate that AMPK dysregulation is at the center of cardiovascular disease (CVD) and that pharmacological targeting of AMPK is a promising and novel strategy to treat CVDs such as atherosclerosis or ischemic injury. In this review, we summarize the current knowledge relevant to this topic, with a focus on shear stress-induced AMPK modulation and its consequences for vascular health and disease.
Collapse
Affiliation(s)
| | - Peter L. Hordijk
- Department of Physiology, Amsterdam UMC, Amsterdam Cardiovascular Sciences, Microcirculation, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| |
Collapse
|
9
|
Zambrano BA, Wilson SI, Zook S, Vekaria B, Moreno MR, Kassi M. Computational investigation of outflow graft variation impact on hemocompatibility profile in LVADs. Artif Organs 2024; 48:375-385. [PMID: 37962282 DOI: 10.1111/aor.14679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 10/17/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Hemocompatibility-related adverse events (HRAE) occur commonly in patients with left ventricular assist devices (LVADs) and add to morbidity and mortality. It is unclear whether the outflow graft orientation can impact flow conditions leading to HRAE. This study presents a simulation-based approach using exact patient anatomy from medical images to investigate the influence of outflow cannula orientation in modulating flow conditions leading to HRAEs. METHODS A 3D model of a proximal aorta and outflow graft was reconstructed from a computed tomography (CT) scan of an LVAD patient and virtually modified to model multiple cannula orientations (n = 10) by varying polar (cranio-caudal) (n = 5) and off-set (anterior-posterior) (n = 2) angles. Time-dependent computational flow simulations were then performed for each anatomical orientation. Qualitative and quantitative hemodynamics metrics of thrombogenicity including time-averaged wall shear stress (TAWSS), oscillatory shear index (OSI), endothelial cell platelet activation potential (ECAP), particle residence time (PRT), and platelet activation potential (PLAP) were analyzed. RESULTS Within the simulations performed, endothelial cell activation potential (ECAP) and particle residence time (PRT) were found to be lowest with a polar angle of 85°, regardless of offset angle. However, polar angles that produced parameters at levels least associated with thrombosis varied when the offset angle was changed from 0° to 12°. For offset angles of 0° and 12° respectively, flow shear was lowest at 65° and 75°, time averaged wall shear stress (TAWSS) was highest at 85° and 35°, and platelet activation potential (PLAP) was lowest at 65° and 45°. CONCLUSION This study suggests that computational fluid dynamic modeling based on patient-specific anatomy can be a powerful analytical tool when identifying optimal positioning of an LVAD. Contrary to previous work, our findings suggest that there may be an "ideal" outflow cannula for each individual patient based on a CFD-based hemocompatibility profile.
Collapse
Affiliation(s)
- Byron A Zambrano
- J. Mike Walker '66 Department of Mechanical Engineering, Texas A&M University, College Station, Texas, USA
| | - Shannon I Wilson
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Salma Zook
- Houston Methodist, Department of Cardiology, Houston Methodist Research Hospital, Houston, Texas, USA
| | - Bansi Vekaria
- Houston Methodist, Department of Cardiology, Houston Methodist Research Hospital, Houston, Texas, USA
| | - Michael R Moreno
- J. Mike Walker '66 Department of Mechanical Engineering, Texas A&M University, College Station, Texas, USA
| | - Mahwash Kassi
- Houston Methodist, Department of Cardiology, Houston Methodist Research Hospital, Houston, Texas, USA
| |
Collapse
|
10
|
Hernández-Espinosa LC, Hernández-Muñoz R. Blood flow-bearing physical forces, endothelial glycocalyx, and liver enzyme mobilization: A hypothesis. J Gen Physiol 2024; 156:e202313462. [PMID: 38231124 PMCID: PMC10794122 DOI: 10.1085/jgp.202313462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/13/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024] Open
Abstract
Numerous elements involved in shear stress-induced signaling have been identified, recognizing their functions as mechanotransducing ion channels situated at cellular membranes. This form of mechanical signaling relies on transmembrane proteins and cytoplasmic proteins that restructure the cytoskeleton, contributing to mechanotransduction cascades. Notably, blood flow generates mechanical forces that significantly impact the structure and remodeling of blood vessels. The primary regulation of blood vessel responses occurs through hemodynamic forces acting on the endothelium. These mechanical events intricately govern endothelial biophysical, biochemical, and genetic responses. Endothelial cells, positioned on the intimal surface of blood vessels, have the capability to express components of the glycocalyx. This endothelial structure emerges as a pivotal factor in mechanotransduction and the regulation of vascular tone. The endothelial glycocalyx assumes diverse roles in both health and disease. Our findings propose a connection between the release of specific enzymes from the rat liver and variations in the hepatic blood flow/mass ratio. Importantly, this phenomenon is not correlated with liver necrosis. Consequently, this review serves as an exploration of the potential involvement of membrane proteins in a hypothetical mechanotransducing phenomenon capable of controlling the release of liver enzymes.
Collapse
Affiliation(s)
- Lorena Carmina Hernández-Espinosa
- Department of Cell Biology and Development, Institute of Cellular Physiology, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Rolando Hernández-Muñoz
- Department of Cell Biology and Development, Institute of Cellular Physiology, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
11
|
Coste B, Delmas P. PIEZO Ion Channels in Cardiovascular Functions and Diseases. Circ Res 2024; 134:572-591. [PMID: 38422173 DOI: 10.1161/circresaha.123.322798] [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] [Indexed: 03/02/2024]
Abstract
The cardiovascular system provides blood supply throughout the body and as such is perpetually applying mechanical forces to cells and tissues. Thus, this system is primed with mechanosensory structures that respond and adapt to changes in mechanical stimuli. Since their discovery in 2010, PIEZO ion channels have dominated the field of mechanobiology. These have been proposed as the long-sought-after mechanosensitive excitatory channels involved in touch and proprioception in mammals. However, more and more pieces of evidence point to the importance of PIEZO channels in cardiovascular activities and disease development. PIEZO channel-related cardiac functions include transducing hemodynamic forces in endothelial and vascular cells, red blood cell homeostasis, platelet aggregation, and arterial blood pressure regulation, among others. PIEZO channels contribute to pathological conditions including cardiac hypertrophy and pulmonary hypertension and congenital syndromes such as generalized lymphatic dysplasia and xerocytosis. In this review, we highlight recent advances in understanding the role of PIEZO channels in cardiovascular functions and diseases. Achievements in this quickly expanding field should open a new road for efficient control of PIEZO-related diseases in cardiovascular functions.
Collapse
Affiliation(s)
- Bertrand Coste
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| | - Patrick Delmas
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| |
Collapse
|
12
|
Wendong Y, Jiali J, Qiaomei F, Yayun W, Xianze X, Zheng S, Wei H. Biomechanical forces and force-triggered drug delivery in tumor neovascularization. Biomed Pharmacother 2024; 171:116117. [PMID: 38171243 DOI: 10.1016/j.biopha.2023.116117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024] Open
Abstract
Tumor angiogenesis is one of the typical hallmarks of tumor occurrence and development, and tumor neovascularization also exhibits distinct characteristics from normal blood vessels. As the number of cells and matrix inside the tumor increases, the biomechanical force is enhanced, specifically manifested as solid stress, fluid stress, stiffness, and topology. This mechanical microenvironment also provides shelter for tumors and intensifies angiogenesis, providing oxygen and nutritional support for tumor progression. During tumor development, the biomechanical microenvironment also emerges, which in turn feeds back to regulate the tumor progression, including tumor angiogenesis, and biochemical and biomechanical signals can regulate tumor angiogenesis. Blood vessels possess inherent sensitivity to mechanical stimuli, but compared to the extensive research on biochemical signal regulation, the study of the regulation of tumor neovascularization by biomechanical signals remains relatively scarce. Biomechanical forces can affect the phenotypic characteristics and mechanical signaling pathways of tumor blood vessels, directly regulating angiogenesis. Meanwhile, they can indirectly regulate tumor angiogenesis by causing an imbalance in angiogenesis signals and affecting stromal cell function. Understanding the regulatory mechanism of biomechanical forces in tumor angiogenesis is beneficial for better identifying and even taming the mechanical forces involved in angiogenesis, providing new therapeutic targets for tumor vascular normalization. Therefore, we summarized the composition of biomechanical forces and their direct or indirect regulation of tumor neovascularization. In addition, this review discussed the use of biomechanical forces in combination with anti-angiogenic therapies for the treatment of tumors, and biomechanical forces triggered delivery systems.
Collapse
Affiliation(s)
- Yao Wendong
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Jiang Jiali
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Fan Qiaomei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Weng Yayun
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Xie Xianze
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Shi Zheng
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| | - Huang Wei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| |
Collapse
|
13
|
Engin A. Endothelial Dysfunction in Obesity and Therapeutic Targets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:489-538. [PMID: 39287863 DOI: 10.1007/978-3-031-63657-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Parallel to the increasing prevalence of obesity in the world, the mortality from cardiovascular disease has also increased. Low-grade chronic inflammation in obesity disrupts vascular homeostasis, and the dysregulation of adipocyte-derived endocrine and paracrine effects contributes to endothelial dysfunction. Besides the adipose tissue inflammation, decreased nitric oxide (NO)-bioavailability, insulin resistance (IR), and oxidized low-density lipoproteins (oxLDLs) are the main factors contributing to endothelial dysfunction in obesity and the development of cardiorenal metabolic syndrome. While normal healthy perivascular adipose tissue (PVAT) ensures the dilation of blood vessels, obesity-associated PVAT leads to a change in the profile of the released adipo-cytokines, resulting in a decreased vasorelaxing effect. Higher stiffness parameter β, increased oxidative stress, upregulation of pro-inflammatory cytokines, and nicotinamide adenine dinucleotide phosphate (NADP) oxidase in PVAT turn the macrophages into pro-atherogenic phenotypes by oxLDL-induced adipocyte-derived exosome-macrophage crosstalk and contribute to the endothelial dysfunction. In clinical practice, carotid ultrasound, higher leptin levels correlate with irisin over-secretion by human visceral and subcutaneous adipose tissues, and remnant cholesterol (RC) levels predict atherosclerotic disease in obesity. As a novel therapeutic strategy for cardiovascular protection, liraglutide improves vascular dysfunction by modulating a cyclic adenosine monophosphate (cAMP)-independent protein kinase A (PKA)-AMP-activated protein kinase (AMPK) pathway in PVAT in obese individuals. Because the renin-angiotensin-aldosterone system (RAAS) activity, hyperinsulinemia, and the resultant IR play key roles in the progression of cardiovascular disease in obesity, RAAS-targeted therapies contribute to improving endothelial dysfunction. By contrast, arginase reciprocally inhibits NO formation and promotes oxidative stress. Thus, targeting arginase activity as a key mediator in endothelial dysfunction has therapeutic potential in obesity-related vascular comorbidities. Obesity-related endothelial dysfunction plays a pivotal role in the progression of type 2 diabetes (T2D). The peroxisome proliferator-activated receptor gamma (PPARγ) agonist, rosiglitazone (thiazolidinedione), is a popular drug for treating diabetes; however, it leads to increased cardiovascular risk. Selective sodium-glucose co-transporter-2 (SGLT-2) inhibitor empagliflozin (EMPA) significantly improves endothelial dysfunction and mortality occurring through redox-dependent mechanisms. Although endothelial dysfunction and oxidative stress are alleviated by either metformin or EMPA, currently used drugs to treat obesity-related diabetes neither possess the same anti-inflammatory potential nor simultaneously target endothelial cell dysfunction and obesity equally. While therapeutic interventions with glucagon-like peptide-1 (GLP-1) receptor agonist liraglutide or bariatric surgery reverse regenerative cell exhaustion, support vascular repair mechanisms, and improve cardiometabolic risk in individuals with T2D and obesity, the GLP-1 analog exendin-4 attenuates endothelial endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
14
|
Zhou R, Li J, Chen Z, Wang R, Shen Y, Zhang R, Zhou F, Zhang Y. Pathological hemodynamic changes and leukocyte transmigration disrupt the blood-spinal cord barrier after spinal cord injury. J Neuroinflammation 2023; 20:118. [PMID: 37210532 DOI: 10.1186/s12974-023-02787-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 04/21/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Blood-spinal cord barrier (BSCB) disruption is a key event after spinal cord injury (SCI), which permits unfavorable blood-derived substances to enter the neural tissue and exacerbates secondary injury. However, limited mechanical impact is usually followed by a large-scale BSCB disruption in SCI. How the BSCB disruption is propagated along the spinal cord in the acute period of SCI remains unclear. Thus, strategies for appropriate clinical treatment are lacking. METHODS A SCI contusion mouse model was established in wild-type and LysM-YFP transgenic mice. In vivo two-photon imaging and complementary studies, including immunostaining, capillary western blotting, and whole-tissue clearing, were performed to monitor BSCB disruption and verify relevant injury mechanisms. Clinically applied target temperature management (TTM) to reduce the core body temperature was tested for the efficacy of attenuating BSCB disruption. RESULTS Barrier leakage was detected in the contusion epicenter within several minutes and then gradually spread to more distant regions. Membrane expression of the main tight junction proteins remained unaltered at four hours post-injury. Many junctional gaps emerged in paracellular tight junctions at the small vessels from multiple spinal cord segments at 15 min post-injury. A previously unnoticed pathological hemodynamic change was observed in the venous system, which likely facilitated gap formation and barrier leakage by exerting abnormal physical force on the BSCB. Leukocytes were quickly initiated to transverse through the BSCB within 30 min post-SCI, actively facilitating gap formation and barrier leakage. Inducing leukocyte transmigration generated gap formation and barrier leakage. Furthermore, pharmacological alleviation of pathological hemodynamic changes or leukocyte transmigration reduced gap formation and barrier leakage. TTM had very little protective effects on the BSCB in the early period of SCI other than partially alleviating leukocyte infiltration. CONCLUSIONS Our data show that BSCB disruption in the early period of SCI is a secondary change, which is indicated by widespread gap formation in tight junctions. Pathological hemodynamic changes and leukocyte transmigration contribute to gap formation, which could advance our understanding of BSCB disruption and provide new clues for potential treatment strategies. Ultimately, TTM is inadequate to protect the BSCB in early SCI.
Collapse
Affiliation(s)
- Rubing Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100191, People's Republic of China
- Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of P.R. China, Beijing, 100191, People's Republic of China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, People's Republic of China
| | - Junzhao Li
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100191, People's Republic of China
- Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of P.R. China, Beijing, 100191, People's Republic of China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, People's Republic of China
| | - Zhengyang Chen
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Hubei, Wuhan, 430060, People's Republic of China
| | - Rong Zhang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100191, People's Republic of China
- Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of P.R. China, Beijing, 100191, People's Republic of China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, People's Republic of China
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, People's Republic of China.
| | - Yong Zhang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100191, People's Republic of China.
- Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of P.R. China, Beijing, 100191, People's Republic of China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, People's Republic of China.
| |
Collapse
|
15
|
Ciavarella C, Motta I, Vasuri F, Palumbo T, Lisi AP, Costa A, Astolfi A, Valente S, Versura P, Fornasiero EF, Mauro R, Gargiulo M, Pasquinelli G. The PPAR-γ Agonist Pioglitazone Modulates Proliferation and Migration in HUVEC, HAOSMC and Human Arteriovenous Fistula-Derived Cells. Int J Mol Sci 2023; 24:4424. [PMID: 36901853 PMCID: PMC10003103 DOI: 10.3390/ijms24054424] [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: 12/29/2022] [Revised: 02/12/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
The failure of arteriovenous fistulas (AVFs) following intimal hyperplasia (IH) increases morbidity and mortality rates in patients undergoing hemodialysis for chronic kidney disease. The peroxisome-proliferator associated receptor (PPAR-γ) may be a therapeutic target in IH regulation. In the present study, we investigated PPAR-γ expression and tested the effect of pioglitazone, a PPAR-γ agonist, in different cell types involved in IH. As cell models, we used Human Endothelial Umbilical Vein Cells (HUVEC), Human Aortic Smooth Muscle Cells (HAOSMC), and AVF cells (AVFCs) isolated from (i) normal veins collected at the first AVF establishment (T0), and (ii) failed AVF with IH (T1). PPAR-γ was downregulated in AVF T1 tissues and cells, in comparison to T0 group. HUVEC, HAOSMC, and AVFC (T0 and T1) proliferation and migration were analyzed after pioglitazone administration, alone or in combination with the PPAR-γ inhibitor, GW9662. Pioglitazone negatively regulated HUVEC and HAOSMC proliferation and migration. The effect was antagonized by GW9662. These data were confirmed in AVFCs T1, where pioglitazone induced PPAR-γ expression and downregulated the invasive genes SLUG, MMP-9, and VIMENTIN. In summary, PPAR-γ modulation may represent a promising strategy to reduce the AVF failure risk by modulating cell proliferation and migration.
Collapse
Affiliation(s)
- Carmen Ciavarella
- DIMEC—Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Center for Applied Biomedical Research (CRBA), University of Bologna, 40138 Bologna, Italy
| | - Ilenia Motta
- DIMEC—Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Center for Applied Biomedical Research (CRBA), University of Bologna, 40138 Bologna, Italy
| | - Francesco Vasuri
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Teresa Palumbo
- Alma Mater Institute on Healthy Planet, University of Bologna, 40138 Bologna, Italy
| | - Anthony Paul Lisi
- DIMEC—Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA 19102, USA
| | - Alice Costa
- DIMEC—Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Annalisa Astolfi
- DIMEC—Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Sabrina Valente
- DIMEC—Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Center for Applied Biomedical Research (CRBA), University of Bologna, 40138 Bologna, Italy
| | - Piera Versura
- DIMEC—Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Center for Applied Biomedical Research (CRBA), University of Bologna, 40138 Bologna, Italy
- Ophtalmology Unit, IRCSS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Eugenio F. Fornasiero
- Department of Neuro-Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Raffaella Mauro
- Vascular Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Mauro Gargiulo
- DIMEC—Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Vascular Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Gianandrea Pasquinelli
- DIMEC—Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
16
|
Wang C, Xu H, Liao X, Wang W, Wu W, Li W, Niu L, Li Z, Li A, Sun Y, Huang W, Song F. Hypertension Promotes the Proliferation and Migration of ccRCC Cells by Downregulation of TIMP3 in Tumor Endothelial Cells through the miR-21-5p/TGFBR2/P38/EGR1 Axis. Mol Cancer Res 2023; 21:62-75. [PMID: 36125433 DOI: 10.1158/1541-7786.mcr-22-0089] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 07/15/2022] [Accepted: 09/16/2022] [Indexed: 02/03/2023]
Abstract
Recent studies have demonstrated that hypertension correlates with tumorigenesis and prognosis of clear-cell renal cell carcinoma (ccRCC); however, the underlying molecular mechanisms remain unclear. By analyzing bulk and single-cell RNA sequencing data and experimental examining of surgical excised ccRCC samples, we found that tissue inhibitors of metalloproteinases 3 (TIMP3), a pivotal paracrine factor in suppressing tumor progression, was significantly reduced in the tumor endothelial cells of patients with hypertensive ccRCC. Besides, in tumor xenograft of NCG mouse model, compared with saline normotensive group the expression of TIMP3 was significantly decreased in the angiotensin II-induced hypertension group. Treating human umbilical vein endothelial cells (HUVEC) with the plasma of patients with hypertensive ccRCC and miR-21-5p, elevated in the plasma of patients with hypertensive ccRCC, reduced the expression of TIMP3 compared with normotensive and control littermates. We also found that the inhibition of TIMP3 expression by miR-21-5p was not through directly targeting at 3'UTR of TIMP3 but through suppressing the expression of TGFβ receptor 2 (TGFBR2). In addition, the knockout of TGFBR2 reduced TIMP3 expression in HUVECs through P38/EGR1 (early growth response protein 1) signaling axis. Moreover, via coculture of ccRCC cell lines with HUVECs and mouse tumor xenograft model, we discovered that the TIMP3 could suppress the proliferation and migration of ccRCC. IMPLICATIONS Overall, our findings shed new light on the role of hypertension in promoting the progression of ccRCC and provide a potential therapeutic target for patients with ccRCC with hypertension.
Collapse
Affiliation(s)
- Chenguang Wang
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Haibo Xu
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.,International Cancer Center, Shenzhen University School of Medicine, Shenzhen, Guangdong, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, Guangdong, China
| | - Xinhui Liao
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Weiming Wang
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.,International Cancer Center, Shenzhen University School of Medicine, Shenzhen, Guangdong, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, Guangdong, China
| | - Wanjun Wu
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.,International Cancer Center, Shenzhen University School of Medicine, Shenzhen, Guangdong, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, Guangdong, China
| | - Wujiao Li
- Clinical laboratory, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Liman Niu
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.,International Cancer Center, Shenzhen University School of Medicine, Shenzhen, Guangdong, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, Guangdong, China
| | - Zhichao Li
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.,International Cancer Center, Shenzhen University School of Medicine, Shenzhen, Guangdong, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, Guangdong, China
| | - Aolin Li
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.,International Cancer Center, Shenzhen University School of Medicine, Shenzhen, Guangdong, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, Guangdong, China
| | - Yangyang Sun
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Weiren Huang
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.,International Cancer Center, Shenzhen University School of Medicine, Shenzhen, Guangdong, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, Guangdong, China
| | - Fei Song
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.,International Cancer Center, Shenzhen University School of Medicine, Shenzhen, Guangdong, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, Guangdong, China
| |
Collapse
|
17
|
Ebrahimi Z, Farsinejad A, Mohammadi MH, Ahmadizad S. Comparable effects of circuit and traditional resistance exercise on platelet α2bβ3 receptor and platelet activation and function. Clin Hemorheol Microcirc 2022; 83:293-303. [PMID: 36565105 DOI: 10.3233/ch-221603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Resistance exercise induces thrombocytosis and increases platelet activation and function. These changes might be related to exercise variables including exercise intensity and type. OBJECTIVE We compared the effects of traditional resistance exercise (TRE) and circuit resistance exercise (CRE) on cellular markers of platelet activation and function. METHODS In this crossover study ten healthy male (mean±SD: age, 25.6±2.4 years) subjects performed TRE encompassed 3 sets of 10 repetitions at 100% of 10-RM (10 repetition maximum) for 6 exercises, and CRE protocols included 3 sets of 10 repetitions at 100% of 10-RM for all 6 exercises consecutively, in two separate weeks. To measure platelet indices, PAC1, CD41a, CD42b and CD62P three blood samples were taken before, immediately after exercise, and after 30 min recovery. RESULTS Lactate concentration, blood pressure, platelet count (PLT), and mean platelet volume (MPV) were significantly (p < 0.05) increased following both resistance exercise trials. Significant increases in PAC1, and CD62P; and significant reductions for CD42b and CD41a were detected following both REs (p < 0.05). However, changes in PAC1 and CD62P were significantly different between the two protocols (p < 0.05), with higher increases detected following CRE. CONCLUSIONS Acute RE increases platelet indices and platelet activation; and that CRE results in higher platelet activation than TRE, probably due to exercise-induced increases in shear stress.
Collapse
Affiliation(s)
- Zahra Ebrahimi
- Department of Biological Sciences in Sport, Faculty of Sport Sciences and Health, Shahid Beheshti University, Tehran, Iran
| | - Alireza Farsinejad
- Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Hossein Mohammadi
- HSCT Research Center, Laboratory Hematology and Blood Banking Department, School of Allied Medical Sciences, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Sajad Ahmadizad
- Department of Biological Sciences in Sport, Faculty of Sport Sciences and Health, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
18
|
Kotlyarov S, Kotlyarova A. The Importance of the Plasma Membrane in Atherogenesis. MEMBRANES 2022; 12:1036. [PMID: 36363591 PMCID: PMC9698587 DOI: 10.3390/membranes12111036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
Atherosclerotic cardiovascular diseases are an important medical problem due to their high prevalence, impact on quality of life and prognosis. The pathogenesis of atherosclerosis is an urgent medical and social problem, the solution of which may improve the quality of diagnosis and treatment of patients. Atherosclerosis is a complex chain of events, which proceeds over many years and in which many cells in the bloodstream and the vascular wall are involved. A growing body of evidence suggests that there are complex, closely linked molecular mechanisms that occur in the plasma membranes of cells involved in atherogenesis. Lipid transport, innate immune system receptor function, and hemodynamic regulation are linked to plasma membranes and their biophysical properties. A better understanding of these interrelationships will improve diagnostic quality and treatment efficacy.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacy Management and Economics, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
19
|
Vetiska S, Wälchli T, Radovanovic I, Berhouma M. Molecular and genetic mechanisms in brain arteriovenous malformations: new insights and future perspectives. Neurosurg Rev 2022; 45:3573-3593. [PMID: 36219361 DOI: 10.1007/s10143-022-01883-4] [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: 05/27/2022] [Revised: 07/30/2022] [Accepted: 10/05/2022] [Indexed: 10/17/2022]
Abstract
Brain arteriovenous malformations (bAVMs) are rare vascular lesions made of shunts between cerebral arteries and veins without the interposition of a capillary bed. The majority of bAVMs are asymptomatic, but some may be revealed by seizures and potentially life-threatening brain hemorrhage. The management of unruptured bAVMs remains a matter of debate. Significant progress in the understanding of their pathogenesis has been made during the last decade, particularly using genome sequencing and biomolecular analysis. Herein, we comprehensively review the recent molecular and genetic advances in the study of bAVMs that not only allow a better understanding of the genesis and growth of bAVMs, but also open new insights in medical treatment perspectives.
Collapse
Affiliation(s)
- Sandra Vetiska
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - Thomas Wälchli
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada.,Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada.,Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, and Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Ivan Radovanovic
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada.,Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Moncef Berhouma
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France. .,CREATIS Lab, CNRS UMR 5220, INSERM U1294, Lyon 1, University, Lyon, France.
| |
Collapse
|
20
|
Demos C, Johnson J, Andueza A, Park C, Kim Y, Villa-Roel N, Kang DW, Kumar S, Jo H. Sox13 is a novel flow-sensitive transcription factor that prevents inflammation by repressing chemokine expression in endothelial cells. Front Cardiovasc Med 2022; 9:979745. [PMID: 36247423 PMCID: PMC9561411 DOI: 10.3389/fcvm.2022.979745] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease and occurs preferentially in arterial regions exposed to disturbed blood flow (d-flow) while the stable flow (s-flow) regions are spared. D-flow induces endothelial inflammation and atherosclerosis by regulating endothelial gene expression partly through the flow-sensitive transcription factors (FSTFs). Most FSTFs, including the well-known Kruppel-like factors KLF2 and KLF4, have been identified from in vitro studies using cultured endothelial cells (ECs). Since many flow-sensitive genes and pathways are lost or dysregulated in ECs during culture, we hypothesized that many important FSTFs in ECs in vivo have not been identified. We tested the hypothesis by analyzing our recent gene array and single-cell RNA sequencing (scRNAseq) and chromatin accessibility sequencing (scATACseq) datasets generated using the mouse partial carotid ligation model. From the analyses, we identified 30 FSTFs, including the expected KLF2/4 and novel FSTFs. They were further validated in mouse arteries in vivo and cultured human aortic ECs (HAECs). These results revealed 8 FSTFs, SOX4, SOX13, SIX2, ZBTB46, CEBPβ, NFIL3, KLF2, and KLF4, that are conserved in mice and humans in vivo and in vitro. We selected SOX13 for further studies because of its robust flow-sensitive regulation, preferential expression in ECs, and unknown flow-dependent function. We found that siRNA-mediated knockdown of SOX13 increased endothelial inflammatory responses even under the unidirectional laminar shear stress (ULS, mimicking s-flow) condition. To understand the underlying mechanisms, we conducted an RNAseq study in HAECs treated with SOX13 siRNA under shear conditions (ULS vs. oscillatory shear mimicking d-flow). We found 94 downregulated and 40 upregulated genes that changed in a shear- and SOX13-dependent manner. Several cytokines, including CXCL10 and CCL5, were the most strongly upregulated genes in HAECs treated with SOX13 siRNA. The robust induction of CXCL10 and CCL5 was further validated by qPCR and ELISA in HAECs. Moreover, the treatment of HAECs with Met-CCL5, a specific CCL5 receptor antagonist, prevented the endothelial inflammation responses induced by siSOX13. In addition, SOX13 overexpression prevented the endothelial inflammation responses. In summary, SOX13 is a novel conserved FSTF, which represses the expression of pro-inflammatory chemokines in ECs under s-flow. Reduction of endothelial SOX13 triggers chemokine expression and inflammatory responses, a major proatherogenic pathway.
Collapse
Affiliation(s)
- Catherine Demos
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Janie Johnson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Aitor Andueza
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Yerin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Nicolas Villa-Roel
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Dong-Won Kang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
21
|
Hotta K, Muller-Delp J. Microvascular Adaptations to Muscle Stretch: Findings From Animals and the Elderly. Front Physiol 2022; 13:939459. [PMID: 35860661 PMCID: PMC9289226 DOI: 10.3389/fphys.2022.939459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
Microcirculation in skeletal muscle is disturbed with advancing aging, causing limited capillary blood flow and exercise incapacity. Muscle stretch has been widely performed in physical therapy, sports medicine, and health promotion. However, the effect of stretch on microvascular reactivity and muscle blood flow remains unknown. This review focuses on stretch-induced microvascular adaptations based on evidence from cultured cells, small animals, and human studies. Vascular endothelium senses and responds to mechanical stimuli including stretch. This endothelial mechanotransduction potentially plays a vital role in the stretch-induced microvascular adaptation alongside hypoxia. Aging impairs microvascular endothelial function, but muscle stretch has the potential to restore it. Muscle stretch may be an alternative to improve vascular function and enhance exercising blood flow, especially for those who have difficulties in participating in exercise due to medical, functional, or psychological reasons.
Collapse
Affiliation(s)
- Kazuki Hotta
- Department of Rehabilitation Sciences, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
- Department of Rehabilitation, Kitasato University School of Allied Health Sciences, Sagamihara, Japan
- *Correspondence: Kazuki Hotta,
| | - Judy Muller-Delp
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| |
Collapse
|
22
|
Boron M, Hauzer-Martin T, Keil J, Sun XL. Circulating Thrombomodulin: Release Mechanisms, Measurements, and Levels in Diseases and Medical Procedures. TH OPEN 2022; 6:e194-e212. [PMID: 36046203 PMCID: PMC9273331 DOI: 10.1055/a-1801-2055] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/11/2022] [Indexed: 12/02/2022] Open
Abstract
Thrombomodulin (TM) is a type-I transmembrane protein that is mainly expressed on endothelial cells and plays important roles in many biological processes. Circulating TM of different forms are also present in biofluids, such as blood and urine. Soluble TM (sTM), comprised of several domains of TM, is the major circulating TM which is generated by either enzymatic or chemical cleavage of the intact protein under different conditions. Under normal conditions, sTM is present in low concentrations (<10 ng/mL) in the blood but is elevated in several pathological conditions associated with endothelial dysfunction such as cardiovascular, inflammatory, infection, and metabolic diseases. Therefore, sTM level has been examined for monitoring disease development, such as disseminated intravascular coagulation (DIC), sepsis and multiple organ dysfunction syndrome in patients with novel coronavirus disease 2019 (COVID-19) recently. In addition, microvesicles (MVs) that contain membrane TM (MV-TM) have been found to be released from activated cells which also contribute to levels of circulating TM in certain diseases. Several release mechanisms of sTM and MV-TM have been reported, including enzymatic, chemical, and TM mutation mechanisms. Measurements of sTM and MV-TM have been developed and explored as biomarkers in many diseases. In this review, we summarize all these advances in three categories as follows: (1) release mechanisms of circulating TM, (2) methods for measuring circulating TM in biological samples, and (3) correlation of circulating TM with diseases. Altogether, it provides a whole picture of recent advances on circulating TM in health and disease.
Collapse
Affiliation(s)
- Mallorie Boron
- Department of Chemistry and Chemical and Biomedical Engineering and Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, Cleveland, Ohio, United States
| | - Tiffany Hauzer-Martin
- Department of Chemistry and Chemical and Biomedical Engineering and Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, Cleveland, Ohio, United States
| | - Joseph Keil
- Department of Chemistry and Chemical and Biomedical Engineering and Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, Cleveland, Ohio, United States
| | - Xue-Long Sun
- Department of Chemistry and Chemical and Biomedical Engineering and Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, Cleveland, Ohio, United States
| |
Collapse
|
23
|
Shar JA, Keswani SG, Grande-Allen KJ, Sucosky P. Significance of aortoseptal angle anomalies to left ventricular hemodynamics and subaortic stenosis: A numerical study. Comput Biol Med 2022; 146:105613. [PMID: 35751200 PMCID: PMC10570849 DOI: 10.1016/j.compbiomed.2022.105613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Discrete subaortic stenosis (DSS) is an obstructive cardiac disease caused by a membranous lesion in the left ventricular (LV) outflow tract (LVOT). Although its etiology is unknown, the higher prevalence of DSS in LVOT anatomies featuring a steep aortoseptal angle (AoSA) suggests a potential role for hemodynamics. Therefore, the objective of this study was to quantify the impact of AoSA steepening on the LV three-dimensional (3D) hemodynamic stress environment. METHODS A 3D LV model reconstructed from cardiac cine-magnetic resonance imaging was connected to four LVOT geometrical variations spanning the clinical AoSA range (115°-160°). LV hemodynamic stresses were characterized in terms of cycle-averaged pressure, temporal shear magnitude (TSM), and oscillatory shear index. The wall shear stress (WSS) topological skeleton was further analyzed by computing the scaled divergence of the WSS vector field. RESULTS AoSA steepening caused an increasingly perturbed subaortic flow marked by LVOT flow skewness and complex 3D secondary flow patterns. These disturbances generated WSS overloads (>45% increase in TSM vs. 160° model) on the inferior LVOT wall, and increased WSS contraction (>66% decrease in WSS divergence vs. 160° model) in regions prone to DSS membrane formation. CONCLUSIONS AoSA steepening generated substantial hemodynamic stress abnormalities in LVOT regions prone to DSS formation. Further studies are needed to assess the possible impact of such mechanical abnormalities on the tissue and cellular responses.
Collapse
Affiliation(s)
- Jason A Shar
- Department of Mechanical Engineering, Kennesaw State University, 840 Polytechnic Lane, Marietta, GA, 30060, USA.
| | - Sundeep G Keswani
- Division of Pediatric Surgery, Texas Children's Hospital, Department of Surgery, Baylor College of Medicine, USA.
| | | | - Philippe Sucosky
- Department of Mechanical Engineering, Kennesaw State University, 840 Polytechnic Lane, Marietta, GA, 30060, USA.
| |
Collapse
|
24
|
A facile cell culture device for studying nuclear and mitochondrial response of endothelial cells to hydrostatic pressure. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.04.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
NiONP-Induced Oxidative Stress and Mitochondrial Impairment in an In Vitro Pulmonary Vascular Cell Model Mimicking Endothelial Dysfunction. Antioxidants (Basel) 2022; 11:antiox11050847. [PMID: 35624710 PMCID: PMC9137840 DOI: 10.3390/antiox11050847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/20/2022] [Accepted: 04/24/2022] [Indexed: 01/27/2023] Open
Abstract
The development and use of nanomaterials, especially of nickel oxide nanoparticles (NiONPs), is expected to provide many benefits but also has raised concerns about the potential human health risks. Inhaled NPs are known to exert deleterious cardiovascular side effects, including pulmonary hypertension. Consequently, patients with pulmonary hypertension (PH) could be at increased risk for morbidity. The objective of this study was to compare the toxic effects of NiONPs on human pulmonary artery endothelial cells (HPAEC) under physiological and pathological conditions. The study was conducted with an in vitro model mimicking the endothelial dysfunction observed in PH. HPAEC were cultured under physiological (static and normoxic) or pathological (20% cycle stretch and hypoxia) conditions and exposed to NiONPs (0.5–5 μg/cm2) for 4 or 24 h. The following endpoints were studied: (i) ROS production using CM-H2DCF-DA and MitoSOX probes, (ii) nitrite production by the Griess reaction, (iii) IL-6 secretion by ELISA, (iv) calcium signaling with a Fluo-4 AM probe, and (v) mitochondrial dysfunction with TMRM and MitoTracker probes. Our results evidenced that under pathological conditions, ROS and nitrite production, IL-6 secretions, calcium signaling, and mitochondria alterations increased compared to physiological conditions. Human exposure to NiONPs may be associated with adverse effects in vulnerable populations with cardiovascular risks.
Collapse
|
26
|
Intimal Hyperplasia of Arteriovenous Fistula. Ann Vasc Surg 2022; 85:444-453. [PMID: 35472499 DOI: 10.1016/j.avsg.2022.04.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022]
Abstract
Intimal hyperplasia (IH), a crucial histopathological injury, forms the basis of vascular stenosis and thrombogenesis. In addition, it is common in maladies such as stenosis at the anastomosis of arteriovenous fistula and restenosis after angioplasty. Various cellular and noncellular components play critical parts in the advancement of IH. This article reviews the distinctive components of IH, such as endothelial dysfunction, multiplication, and movement of vascular smooth muscle cells. Finally, in addition to synthesis of large amounts of extracellular matrix and inflammatory responses, which have frequently been studied in recent years, we offer a premise for clinical treatment with vascular smooth muscle cells.
Collapse
|
27
|
Oxidative Stress, Vascular Endothelium, and the Pathology of Neurodegeneration in Retina. Antioxidants (Basel) 2022; 11:antiox11030543. [PMID: 35326193 PMCID: PMC8944517 DOI: 10.3390/antiox11030543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress (OS) is an imbalance between free radicals/ROS and antioxidants, which evokes a biological response and is an important risk factor for diseases, in both the cardiovascular system and central nervous system (CNS). The underlying mechanisms driving pathophysiological complications that arise from OS remain largely unclear. The vascular endothelium is emerging as a primary target of excessive glucocorticoid and catecholamine action. Endothelial dysfunction (ED) has been implicated to play a crucial role in the development of neurodegeneration in the CNS. The retina is known as an extension of the CNS. Stress and endothelium dysfunction are suspected to be interlinked and associated with neurodegenerative diseases in the retina as well. In this narrative review, we explore the role of OS-led ED in the retina by focusing on mechanistic links between OS and ED, ED in the pathophysiology of different retinal neurodegenerative conditions, and how a better understanding of the role of endothelial function could lead to new therapeutic approaches for neurodegenerative diseases in the retina.
Collapse
|
28
|
Tsai CL, Huang CY, Lu YC, Pai LM, Horák D, Ma YH. Cyclic Strain Mitigates Nanoparticle Internalization by Vascular Smooth Muscle Cells. Int J Nanomedicine 2022; 17:969-981. [PMID: 35280334 PMCID: PMC8909538 DOI: 10.2147/ijn.s337942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/27/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Chia-Liang Tsai
- Department of Physiology and Pharmacology, Chang Gung University, Taoyuan, 33302, Taiwan, Republic of China
| | - Ching-Yun Huang
- Institute of Biomedical Sciences, Chang Gung University, Taoyuan, 33302, Taiwan, Republic of China
| | - Yi-Ching Lu
- Department of Physiology and Pharmacology, Chang Gung University, Taoyuan, 33302, Taiwan, Republic of China
| | - Li-Mei Pai
- Department of Biochemistry & Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan, Republic of China
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan, Republic of China
| | - Daniel Horák
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Prague 6, 162 06, Czech Republic
| | - Yunn-Hwa Ma
- Department of Physiology and Pharmacology, Chang Gung University, Taoyuan, 33302, Taiwan, Republic of China
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan, Republic of China
- Correspondence: Yunn-Hwa Ma, Department of Physiology and Pharmacology, Chang Gung University, Guishan, Taoyuan, 33302, Taiwan, Republic of China, Email
| |
Collapse
|
29
|
Ando J, Yamamoto K. Hemodynamic Forces, Endothelial Mechanotransduction, and Vascular Diseases. Magn Reson Med Sci 2022; 21:258-266. [PMID: 34024868 PMCID: PMC9680547 DOI: 10.2463/mrms.rev.2021-0018] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/23/2021] [Indexed: 11/09/2022] Open
Abstract
Cells in the tissues and organs of a living body are subjected to mechanical forces, such as pressure, friction, and tension from their surrounding environment. Cells are equipped with a mechanotransduction mechanism by which they perceive mechanical forces and transmit information into the cell interior, thereby causing physiological or pathogenetic mechano-responses. Endothelial cells (ECs) lining the inner surface of blood vessels are constantly exposed to shear stress caused by blood flow and a cyclic strain caused by intravascular pressure. A number of studies have shown that ECs are sensitive to changes in these hemodynamic forces and alter their morphology and function, sometimes by modifying gene expression. The mechanism of endothelial mechanotransduction has been elucidated, and the plasma membrane has recently been shown to act as a mechanosensor. The lipid order and cholesterol content of plasma membranes change immediately upon the exposure of ECs to hemodynamic forces, resulting in a change in membrane fluidity. These changes in a plasma membrane's physical properties affect the conformation and function of various ion channels, receptors, and microdomains (such as caveolae and primary cilia), thereby activating a wide variety of downstream signaling pathways. Such endothelial mechanotransduction works to maintain circulatory homeostasis; however, errors in endothelial mechanotransduction can cause abnormalities in vascular physiological function, leading to the initiation and progression of various vascular diseases, such as hypertension, thrombosis, aneurysms, and atherosclerosis. Recent advances in detailed imaging technology and computational fluid dynamics analysis have enabled us to evaluate the hemodynamic forces acting on vascular tissue accurately, contributing greatly to our understanding of vascular mechanotransduction and the pathogenesis of vascular diseases, as well as the development of new therapies for vascular diseases.
Collapse
Affiliation(s)
- Joji Ando
- Laboratory of Biomedical Engineering, School of Medicine, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Kimiko Yamamoto
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
30
|
Walter R, Hunter K, Stenmark K, Kheyfets VO. Hemodynamically Unloading the Distal Pulmonary Circulation in Pulmonary Hypertension: A Modeling Study. J Biomech Eng 2022; 144:024503. [PMID: 34251418 PMCID: PMC8547017 DOI: 10.1115/1.4051719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 07/05/2021] [Indexed: 02/03/2023]
Abstract
Pulmonary hypertension (PH) is a progressive disease that is characterized by a gradual increase in both resistive and reactive pulmonary arterial (PA) impedance. Previous studies in a rodent model of PH have shown that reducing the hemodynamic load in the left lung (by banding the left PA) reverses this remodeling phenomenon. However, banding a single side of the pulmonary circulation is not a viable clinical option, so-using in silico modeling-we evaluated if the banding effect can be recreated by replacing the proximal vasculature with a compliant synthetic PA. We developed a computational model of the pulmonary circulation by combining a one-dimensional model of the proximal vasculature with a zero-dimensional line transmission model to the 12th generation. Using this model, we performed four simulations: (1) Control; (2) PH; (3) PH with a stenosis in the left PA; and (4) PH with proximal vessel compliance returned to Control levels. Simulations revealed that vascular changes associated with PH result in an increase in pulse pressure (PP), maximum pressure (Pmax), maximum wall shear stress (WSS), and maximum circumferential stress (σθθ) relative to controls, in the distal circulation. Banding the left PA reduced these measurements of hemodynamic stress in the left lung, but increases them in the right lung. Furthermore, left PA banding increased reactive PA impedance. However, returning the proximal PA compliance to Control levels simultaneously decreased all measures of hemodynamic stress in both lungs, and returned reactive PA impedance to normal levels. In conclusion, if future in vivo studies support the idea of hemodynamic unloading as an effective therapy for PH, this can be surgically achieved by replacing the proximal PA with a compliant prosthesis, and it will have the added benefit of reducing reactive right ventricular afterload.
Collapse
Affiliation(s)
- Rachelle Walter
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kendall Hunter
- Department of Bioengineering and Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO 80045
| | - Kurt Stenmark
- Division of Pediatric Critical Care Medicine and Cardiovascular Pulmonary Research, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO 80045
| | - Vitaly O. Kheyfets
- Department of Bioengineering and Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO 80045
| |
Collapse
|
31
|
Chen Y, Gao Q, Li J, Mao F, Tang R, Jiang H. Activation of Topological Defects Induces a Brittle-to-Ductile Transition in Epithelial Monolayers. PHYSICAL REVIEW LETTERS 2022; 128:018101. [PMID: 35061486 DOI: 10.1103/physrevlett.128.018101] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 12/08/2021] [Indexed: 06/14/2023]
Abstract
Epithelial monolayers are subjected to various mechanical forces, such as stretching, shearing, and compression. Thus, its mechanical response to external loadings is essential for its biological functions. However, the mechanism of the fracture failure of the epithelial monolayer remains poorly understood. Here, by introducing a new type of topological transition, i.e., detach transition or T4 transition, we develop a modified cellular vertex model to investigate the rupture of the cell monolayer. Interestingly, we find a brittle-to-ductile transition in epithelial monolayers, which is controlled by the mechanical properties of single cells and cell-cell contacts. We reveal that the external loadings can activate cell rearrangement in ductile cell monolayers. The plastic deformation results from the nucleation and propagation of "pentagon-heptagon defects" in analogy with the topological defects commonly seen in 2D materials. By using a simplified four-cell model, we further demonstrate that the brittle-to-ductile transition is induced by the competition between cell rearrangement and cell detachment. Our work provides a new theoretical framework to study the rupture of living tissues and may have important implications for many other biological processes, such as wound healing and tissue morphogenesis.
Collapse
Affiliation(s)
- Yixia Chen
- Hefei National Laboratory for Physical Science at the Microscale, CAS Key Laboratory of Mechanical Behavior and Design of Materials, CAS Center for Excellence in Complex System Mechanics, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Qigan Gao
- Hefei National Laboratory for Physical Science at the Microscale, CAS Key Laboratory of Mechanical Behavior and Design of Materials, CAS Center for Excellence in Complex System Mechanics, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jingchen Li
- Hefei National Laboratory for Physical Science at the Microscale, CAS Key Laboratory of Mechanical Behavior and Design of Materials, CAS Center for Excellence in Complex System Mechanics, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Fangtao Mao
- Hefei National Laboratory for Physical Science at the Microscale, CAS Key Laboratory of Mechanical Behavior and Design of Materials, CAS Center for Excellence in Complex System Mechanics, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Ruowen Tang
- Hefei National Laboratory for Physical Science at the Microscale, CAS Key Laboratory of Mechanical Behavior and Design of Materials, CAS Center for Excellence in Complex System Mechanics, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Hongyuan Jiang
- Hefei National Laboratory for Physical Science at the Microscale, CAS Key Laboratory of Mechanical Behavior and Design of Materials, CAS Center for Excellence in Complex System Mechanics, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
32
|
Amar K, Wei F, Chen J, Wang N. Effects of forces on chromatin. APL Bioeng 2021; 5:041503. [PMID: 34661040 PMCID: PMC8516479 DOI: 10.1063/5.0065302] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/27/2021] [Indexed: 12/29/2022] Open
Abstract
Chromatin is a unique structure of DNA and histone proteins in the cell nucleus and the site of dynamic regulation of gene expression. Soluble factors are known to affect the chromatin structure and function via activating or inhibiting specific transcription factors. Forces on chromatin come from exogenous stresses on the cell surface and/or endogenous stresses, which are regulated by substrate mechanics, geometry, and topology. Forces on chromatin involve direct (via adhesion molecules, cytoskeleton, and the linker of nucleoskeleton and cytoskeleton complexes) and indirect (via diffusion and/or translocation processes) signaling pathways to modulate levels of chromatin folding and deformation to regulate transcription, which is controlled by histone modifications and depends on magnitude, direction, rate/frequency, duration, and modes of stresses. The rapid force transmission pathway activates multiple genes simultaneously, and the force may act like a "supertranscription factor." The indirect mechanotransduction pathways and the rapid force transmission pathway together exert sustained impacts on the chromatin, the nucleus, and cell functions.
Collapse
Affiliation(s)
- Kshitij Amar
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Fuxiang Wei
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Junwei Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Ning Wang
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
33
|
Kotlyarov S. Diversity of Lipid Function in Atherogenesis: A Focus on Endothelial Mechanobiology. Int J Mol Sci 2021; 22:11545. [PMID: 34768974 PMCID: PMC8584259 DOI: 10.3390/ijms222111545] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is one of the most important problems in modern medicine. Its high prevalence and social significance determine the need for a better understanding of the mechanisms of the disease's development and progression. Lipid metabolism and its disorders are one of the key links in the pathogenesis of atherosclerosis. Lipids are involved in many processes, including those related to the mechanoreception of endothelial cells. The multifaceted role of lipids in endothelial mechanobiology and mechanisms of atherogenesis are discussed in this review. Endothelium is involved in ensuring adequate vascular hemodynamics, and changes in blood flow characteristics are detected by endothelial cells and affect their structure and function.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
34
|
Deweirdt J, Ducret T, Quignard JF, Freund-Michel V, Lacomme S, Gontier E, Muller B, Marthan R, Guibert C, Baudrimont I. Effects of FW2 Nanoparticles Toxicity in a New In Vitro Pulmonary Vascular Cells Model Mimicking Endothelial Dysfunction. Cardiovasc Toxicol 2021; 22:14-28. [PMID: 34524626 DOI: 10.1007/s12012-021-09679-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/22/2021] [Indexed: 11/29/2022]
Abstract
Several epidemiological studies have revealed the involvement of nanoparticles (NPs) in respiratory and cardiovascular mortality. In this work, the focus will be on the effect of manufactured carbon black NPs for risk assessment of consumers and workers, as human exposure is likely to increase. Since the pulmonary circulation could be one of the primary targets of inhaled NPs, patients suffering from pulmonary hypertension (PH) could be a population at risk. To compare the toxic effect of carbon black NPs in the pulmonary circulation under physiologic and pathological conditions, we developed a new in vitro model mimicking the endothelial dysfunction and vascular dynamics observed in vascular pathology such as PH. Human pulmonary artery endothelial cells were cultured under physiological conditions (static and normoxia 21% O2) or under pathological conditions (20% cycle stretch and hypoxia 1% O2). Then, cells were treated for 4 or 6 h with carbon black FW2 NPs from 5 to 10 µg/cm2. Different endpoints were studied: (i) NPs internalization by transmission electronic microscopy; (ii) oxidative stress by CM-H2DCFDA probe and electron paramagnetic resonance; (iii) NO (nitrites and nitrates) production by Griess reaction; (iv) inflammation by ELISA assay; and (v) calcium signaling by confocal microscopy. The present study characterizes the in vitro model mimicking endothelial dysfunction in PH and indicates that, under such pathological conditions, oxidative stress and inflammation are increased along with calcium signaling alterations, as compared to the physiological conditions. Human exposure to carbon black NPs could produce greater deleterious effects in vulnerable patients suffering from cardiovascular diseases.
Collapse
Affiliation(s)
- J Deweirdt
- University of Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U 1045, 33000, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, 33604, Pessac, France
| | - T Ducret
- University of Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U 1045, 33000, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, 33604, Pessac, France
| | - J-F Quignard
- University of Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U 1045, 33000, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, 33604, Pessac, France
| | - V Freund-Michel
- University of Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U 1045, 33000, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, 33604, Pessac, France
| | - S Lacomme
- CNRS, Bordeaux Imaging Center UMS 3420 CNRS - US4 INSERM, 33000, Bordeaux, France
| | - E Gontier
- CNRS, Bordeaux Imaging Center UMS 3420 CNRS - US4 INSERM, 33000, Bordeaux, France
| | - B Muller
- University of Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U 1045, 33000, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, 33604, Pessac, France
| | - R Marthan
- University of Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U 1045, 33000, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, 33604, Pessac, France.,CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, 33000, Bordeaux, France
| | - C Guibert
- University of Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U 1045, 33000, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, 33604, Pessac, France
| | - I Baudrimont
- University of Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U 1045, 33000, Bordeaux, France. .,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, 33604, Pessac, France.
| |
Collapse
|
35
|
Fini EM, Salimian M, Ahmadizad S. Responses of platelet CD markers and indices to resistance exercise with and without blood flow restriction in patients with type 2 diabetes. Clin Hemorheol Microcirc 2021; 80:281-289. [PMID: 34511492 DOI: 10.3233/ch-211229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Diabetes mellitus is a common disorder with the risk of vascular injury. OBJECTIVE The aim of this study was to compare the effects of low-intensity resistance exercise with blood flow restriction versus high-intensity resistance exercise on platelet CD markers and indices in patients with type 2 diabetes. METHODS Fifteen female patients with type 2 diabetes (Mean±SD; age, 47.6±7.2 yrs) randomly completed two resistance exercise at an intensity corresponding to 20% and 80% of one-repetition maximum (1-RM), with and without blood flow restriction (REBFR and RE), respectively. We measured markers of platelet activation (P-selectin, GpIIb/IIIa, and CD42) and platelet indices before and immediately after exercise, and after 30 min recovery. RESULTS Platelet count (PLT) and plateletcrit (PCT) increased in response to REBFR more than the RE (p < 0.05), though, no significant differences in PDW and MPV were observed (p < 0.05). Although P-selectin (CD62P), CD61, CD41, and CD42 were reduced following resistance exercise in both trials, these reductions were non-significant (p < 0.05). Besides, no significant between-group differences were found for platelet CD markers (p < 0.05). CONCLUSIONS It is concluded that REBFR induces thrombocytosis, but responses of platelet CD markers in patients with type 2 diabetes are similar following low-intensity REBFR and high-intensity RE.
Collapse
Affiliation(s)
- Elahe Malekyian Fini
- Department of Biological Sciences in Sport, Faculty of Sport Sciences and Health, Shahid Beheshti University, Tehran, Iran
| | - Morteza Salimian
- Platelet Research Laboratory, Kashan University of Medical Sciences, Kashan, Iran
| | - Sajad Ahmadizad
- Department of Biological Sciences in Sport, Faculty of Sport Sciences and Health, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
36
|
Association between Physical Activity and Seasonal Variations in Metabolic and Vascular Function in Adults. ENDOCRINES 2021. [DOI: 10.3390/endocrines2020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
This article highlights the association between physical activity (PA) and seasonal variations in metabolic and vascular function in adults. Increasing PA is an important method for preventing cardiovascular disease (CVD) and all-cause mortality by improving blood glucose, blood pressure, blood lipid profiles, body fat, insulin resistance, and vascular function, such as endothelial function. Conversely, various factors, such as seasonal climate conditions, may affect the amount of PA that individuals undertake. Changes in PA often induce seasonal variations in metabolic and vascular function; the deterioration of such functions in winter is the most prominent, and there is clear evidence of an increased risk of CVD in this season. Understanding the influence of PA on seasonal variations observed in metabolic and vascular function is necessary for the management of these physiological functions. In this article summary, few studies have proven that maintaining PA can suppress the variations, and it remains unclear what types, intensities, and durations of regular PA are effective for circumventing seasonal impact. In addition to further studies, there is a need to educate individuals about the strategies to manage PA and other aspects of their lifestyles throughout the year, particularly in winter.
Collapse
|
37
|
Dey K, Roca E, Ramorino G, Sartore L. Progress in the mechanical modulation of cell functions in tissue engineering. Biomater Sci 2021; 8:7033-7081. [PMID: 33150878 DOI: 10.1039/d0bm01255f] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In mammals, mechanics at multiple stages-nucleus to cell to ECM-underlie multiple physiological and pathological functions from its development to reproduction to death. Under this inspiration, substantial research has established the role of multiple aspects of mechanics in regulating fundamental cellular processes, including spreading, migration, growth, proliferation, and differentiation. However, our understanding of how these mechanical mechanisms are orchestrated or tuned at different stages to maintain or restore the healthy environment at the tissue or organ level remains largely a mystery. Over the past few decades, research in the mechanical manipulation of the surrounding environment-known as substrate or matrix or scaffold on which, or within which, cells are seeded-has been exceptionally enriched in the field of tissue engineering and regenerative medicine. To do so, traditional tissue engineering aims at recapitulating key mechanical milestones of native ECM into a substrate for guiding the cell fate and functions towards specific tissue regeneration. Despite tremendous progress, a big puzzle that remains is how the cells compute a host of mechanical cues, such as stiffness (elasticity), viscoelasticity, plasticity, non-linear elasticity, anisotropy, mechanical forces, and mechanical memory, into many biological functions in a cooperative, controlled, and safe manner. High throughput understanding of key cellular decisions as well as associated mechanosensitive downstream signaling pathway(s) for executing these decisions in response to mechanical cues, solo or combined, is essential to address this issue. While many reports have been made towards the progress and understanding of mechanical cues-particularly, substrate bulk stiffness and viscoelasticity-in regulating the cellular responses, a complete picture of mechanical cues is lacking. This review highlights a comprehensive view on the mechanical cues that are linked to modulate many cellular functions and consequent tissue functionality. For a very basic understanding, a brief discussion of the key mechanical players of ECM and the principle of mechanotransduction process is outlined. In addition, this review gathers together the most important data on the stiffness of various cells and ECM components as well as various tissues/organs and proposes an associated link from the mechanical perspective that is not yet reported. Finally, beyond addressing the challenges involved in tuning the interplaying mechanical cues in an independent manner, emerging advances in designing biomaterials for tissue engineering are also explored.
Collapse
Affiliation(s)
- Kamol Dey
- Department of Applied Chemistry and Chemical Engineering, Faculty of Science, University of Chittagong, Bangladesh
| | | | | | | |
Collapse
|
38
|
Cameron T, Bennet T, Rowe EM, Anwer M, Wellington CL, Cheung KC. Review of Design Considerations for Brain-on-a-Chip Models. MICROMACHINES 2021; 12:441. [PMID: 33921018 PMCID: PMC8071412 DOI: 10.3390/mi12040441] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Abstract
In recent years, the need for sophisticated human in vitro models for integrative biology has motivated the development of organ-on-a-chip platforms. Organ-on-a-chip devices are engineered to mimic the mechanical, biochemical and physiological properties of human organs; however, there are many important considerations when selecting or designing an appropriate device for investigating a specific scientific question. Building microfluidic Brain-on-a-Chip (BoC) models from the ground-up will allow for research questions to be answered more thoroughly in the brain research field, but the design of these devices requires several choices to be made throughout the design development phase. These considerations include the cell types, extracellular matrix (ECM) material(s), and perfusion/flow considerations. Choices made early in the design cycle will dictate the limitations of the device and influence the end-point results such as the permeability of the endothelial cell monolayer, and the expression of cell type-specific markers. To better understand why the engineering aspects of a microfluidic BoC need to be influenced by the desired biological environment, recent progress in microfluidic BoC technology is compared. This review focuses on perfusable blood-brain barrier (BBB) and neurovascular unit (NVU) models with discussions about the chip architecture, the ECM used, and how they relate to the in vivo human brain. With increased knowledge on how to make informed choices when selecting or designing BoC models, the scientific community will benefit from shorter development phases and platforms curated for their application.
Collapse
Affiliation(s)
- Tiffany Cameron
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Tanya Bennet
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Elyn M. Rowe
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Mehwish Anwer
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Cheryl L. Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Karen C. Cheung
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Electrical & Computer Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
39
|
Bioengineered in vitro models of leukocyte-vascular interactions. Biochem Soc Trans 2021; 49:693-704. [PMID: 33843967 DOI: 10.1042/bst20200620] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 03/12/2021] [Accepted: 03/18/2021] [Indexed: 01/13/2023]
Abstract
Leukocytes continuously circulate our body through the blood and lymphatic vessels. To survey invaders or abnormalities and defend our body against them, blood-circulating leukocytes migrate from the blood vessels into the interstitial tissue space (leukocyte extravasation) and exit the interstitial tissue space through draining lymphatic vessels (leukocyte intravasation). In the process of leukocyte trafficking, leukocytes recognize and respond to multiple biophysical and biochemical cues in these vascular microenvironments to determine adequate migration and adhesion pathways. As leukocyte trafficking is an essential part of the immune system and is involved in numerous immune diseases and related immunotherapies, researchers have attempted to identify the key biophysical and biochemical factors that might be responsible for leukocyte migration, adhesion, and trafficking. Although intravital live imaging of in vivo animal models has been remarkably advanced and utilized, bioengineered in vitro models that recapitulate complicated in vivo vascular structure and microenvironments are needed to better understand leukocyte trafficking since these in vitro models better allow for spatiotemporal analyses of leukocyte behaviors, decoupling of interdependent biological factors, better controlling of experimental parameters, reproducible experiments, and quantitative cellular analyses. This review discusses bioengineered in vitro model systems that are developed to study leukocyte interactions with complex microenvironments of blood and lymphatic vessels. This review focuses on the emerging concepts and methods in generating relevant biophysical and biochemical cues. Finally, the review concludes with expert perspectives on the future research directions for investigating leukocyte and vascular biology using the in vitro models.
Collapse
|
40
|
Martins BR, Pinto TS, da Costa Fernandes CJ, Bezerra F, Zambuzzi WF. PI3K/AKT signaling drives titanium-induced angiogenic stimulus. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:18. [PMID: 33506378 PMCID: PMC7840643 DOI: 10.1007/s10856-020-06473-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 12/10/2020] [Indexed: 05/15/2023]
Abstract
Although osseointegration and clinical success of titanium (Ti)-implanted materials depend on neovascularization in the reactional peri-implant tissue, very little has been achieved considering the Ti-molecules release on the behavior of endothelial cells. To address this issue, we challenged endothelial cells (HUVECs) with Ti-enriched medium obtained from two types of commercial titanium surfaces [presenting or not dual-acid etching (DAE)] up to 72 h to allow molecular machinery analysis. Our data show that the Ti-enriched medium provokes significant stimulus of angiogenesis-related machinery in endothelial cells by upexpressing VEGFR1, VEGFR2, VEGF, eNOS, and iNOS genes, while the PI3K/Akt signaling pathway was also significantly enhanced. As PI3K/AKT signaling was related to angiogenesis in response to vascular endothelial growth factor (VEGF), we addressed the importance of PI3K/Akt upon Ti-enriched medium responses by concomitantly treating the cells with wortmannin, a well-known PI3K inhibitor. Wortmannin suppressed the angiogenic factors, because VEGF, VEGFR1, and eNOS genes were downregulated in those cells, highlighting the importance of PI3K/AKT signaling on driving angiogenic phenotype and angiogenesis performance within the peri-implant tissue reaction. In conjunction, these data reinforce that titanium-implantable devices modify the metabolism of surrounding cells, such as endothelial cells, probably coupling osteogenesis and angiogenesis processes in peri-implant tissue and then contributing to successfully osseointegration of biomedical titanium-based devices.
Collapse
Affiliation(s)
- Bruna Rodrigues Martins
- Institute of Biosciences of Botucatu, Department of Chemical and Biological Sciences, UNESP - São Paulo State University, Botucatu, São Paulo, Brazil
| | - Thais Silva Pinto
- Institute of Biosciences of Botucatu, Department of Chemical and Biological Sciences, UNESP - São Paulo State University, Botucatu, São Paulo, Brazil
| | - Célio Junior da Costa Fernandes
- Institute of Biosciences of Botucatu, Department of Chemical and Biological Sciences, UNESP - São Paulo State University, Botucatu, São Paulo, Brazil
| | - Fábio Bezerra
- Institute of Biosciences of Botucatu, Department of Chemical and Biological Sciences, UNESP - São Paulo State University, Botucatu, São Paulo, Brazil
| | - Willian Fernando Zambuzzi
- Institute of Biosciences of Botucatu, Department of Chemical and Biological Sciences, UNESP - São Paulo State University, Botucatu, São Paulo, Brazil.
| |
Collapse
|
41
|
Pewowaruk R, Lamers L, Roldán-Alzate A. Longitudinal Evolution of Pulmonary Artery Wall Shear Stress in a Swine Model of Pulmonary Artery Stenosis and Stent Interventions. Ann Biomed Eng 2021; 49:1477-1492. [PMID: 33398618 DOI: 10.1007/s10439-020-02696-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/05/2020] [Indexed: 11/30/2022]
Abstract
Branch pulmonary artery stenosis (PAS) commonly occurs in congenital heart disease and it has previously been hypothesized that in branch PAS the pulmonary arteries (PAs) remodel their lumen diameter to maintain constant wall shear stress (WSS). We quantified the longitudinal progression of PA WSS in a swine model of unilateral PAS and two different intervention time courses to test this hypothesis. To quantify WSS in the entire pulmonary tree we used 4D Flow MRI for the large-proximal PAs and a structured tree model for the small-distal PAs. Our results only partially supported the hypothesis that in branch PAS the PAs remodel their lumen diameter to maintain WSS homeostasis. Proximal PA WSS was similar between groups at the final study time-point but WSS of mid-sized (5 mm to 500 μm) PA segments was found to be different between the sham and LPAS groups. This suggests that WSS homeostasis may only be achieved for the large-proximal PAs. Additionally, our results do not show WSS homeostasis being achieved over shorter periods of time suggesting that any potential WSS dependent changes in PA lumen diameter were a long-term remodeling response rather than a short-term vasodilation response. Future studies should confirm if these findings hold true in humans and investigate the impacts of WSS at different levels of the pulmonary tree on growth.
Collapse
Affiliation(s)
- Ryan Pewowaruk
- Biomedical Engineering, University of Wisconsin - Madison, Madison, WI, USA
| | - Luke Lamers
- Pediatrics, Division of Cardiology, University of Wisconsin - Madison, Madison, WI, USA
| | - Alejandro Roldán-Alzate
- Biomedical Engineering, University of Wisconsin - Madison, Madison, WI, USA. .,Mechanical Engineering, University of Wisconsin - Madison, Madison, WI, USA. .,Radiology, University of Wisconsin - Madison, Madison, WI, USA. .,Wisconsin Institutes for Medical Research, 1111 Highland Ave, Madison, WI, 53792-4108, USA.
| |
Collapse
|
42
|
Pillalamarri N, Patnaik S, Piskin S, Gueldner P, Finol E. Ex Vivo Regional Mechanical Characterization of Porcine Pulmonary Arteries. EXPERIMENTAL MECHANICS 2021; 61:285-303. [PMID: 33814554 PMCID: PMC8011683 DOI: 10.1007/s11340-020-00678-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 10/22/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Regional mechanical characterization of pulmonary arteries can be useful in the development of computational models of pulmonary arterial mechanics. OBJECTIVE We performed a biomechanical and microstructural characterization study of porcine pulmonary arteries, inclusive of the main, left, and right pulmonary arteries (MPA, LPA, and RPA, respectively). METHODS The specimens were initially stored at -20°C and allowed to thaw for 12-24 hours prior to testing. Each artery was further subdivided into proximal, middle, and distal regions, leading to ten location-based experimental groups. Planar equibiaxial tensile testing was performed to evaluate the mechanical behavior of the specimens, from which we calculated the stress at the maximum strain (S 55), tensile modulus (TM), anisotropy index (AI), and strain energy in terms of area under the stress-strain curve (AUC). Histological quantification was performed to evaluate the area fraction of elastin and collagen content, intima-media thickness (IMT), and adventitial thickness (AT). The constitutive material behavior of each group was represented by a five-constant Holzapfel-Gasser-Ogden model. RESULTS The specimens exhibited non-linear stress-strain characteristics across all groups. The MPA exhibited the highest mean wall stress and TM in the longitudinal and circumferential directions, while the bifurcation region yielded the highest values of AI and AUC. All regions revealed a higher stiffness in the longitudinal direction compared to the circumferential direction, suggesting a degree of anisotropy that is believed to be within the margin of experimental uncertainty. Collagen content was found to be the highest in the MPA and decreased significantly at the bifurcation, LPA and RPA. Elastin content did not yield such significant differences amongst the ten groups. The MPA had the highest IMT, which decreased concomitantly to the distal LPA and RPA. No significant differences were found in the AT amongst the ten groups. CONCLUSION The mechanical properties of porcine pulmonary arteries exhibit strong regional dissimilarities, which can be used to inform future studies of high fidelity finite element models.
Collapse
Affiliation(s)
- N.R. Pillalamarri
- University of Texas at San Antonio, Department of Mechanical Engineering, San Antonio, TX
| | - S.S. Patnaik
- University of Texas at San Antonio, Department of Mechanical Engineering, San Antonio, TX
| | - S. Piskin
- University of Texas at San Antonio, Department of Mechanical Engineering, San Antonio, TX
- Istinye University, Department of Mechanical Engineering, Zeytinburnu, Istanbul, Turkey
| | - P. Gueldner
- University of Texas at San Antonio, Department of Biomedical Engineering, San Antonio, TX
| | - E.A. Finol
- University of Texas at San Antonio, Department of Mechanical Engineering, San Antonio, TX
- University of Texas at San Antonio, UTSA/UTHSA Joint Graduate Program in Biomedical Engineering, San Antonio, TX
| |
Collapse
|
43
|
Honda H, Igaki M, Komatsu M, Tanaka S. Seasonal variations on endothelium-dependent flow-mediated vasodilation in adults with type 2 diabetes and nondiabetic adults with hypertension and/or dyslipidaemia who perform regular exercise. Endocrinol Diabetes Metab 2021; 4:e00168. [PMID: 33532610 PMCID: PMC7831209 DOI: 10.1002/edm2.168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/14/2020] [Accepted: 06/20/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction Endothelium-dependent flow-mediated dilation (FMD) of the brachial artery often changes seasonally. We aimed to examine the association between the seasonal variation on FMD and regular exercise in adults with type 2 diabetes (T2D) and nondiabetic adults with hypertension and/or dyslipidaemia (non-T2D). Methods This retrospective study included 14 T2D and 17 non-T2D adults, who started to perform moderate-intensity aerobic exercise for 30-40 min/d at a hospital gym in 2006-2010 and maintained exercise performance at least 2 d/wk until the end of the observation period. We observed and analysed the data for 5 years (from March 2011 to February 2016). FMD, cardio-ankle vascular index (CAVI) and metabolic outcomes were compared among seasons in the T2D and non-T2D groups. Results The FMD values were lower in winter than in other seasons in both groups (all P < .01). The annual range of FMD was larger by 31% in the T2D group than in the non-T2D group (P < .05). The systolic blood pressure (BP) values were higher in winter than in other seasons in both groups (all P < .01), and the diastolic BP values were higher in winter than in summer in both groups (T2D: P < .05; non-T2D: P < .01). CAVI and other outcomes did not change seasonally. Conclusions Flow-mediated vasodilation showed seasonal variation in T2D adults, even if they performed exercise regularly for a long period of time. Additionally, we found that the annual range of FMD might increase with the presence of T2D.
Collapse
Affiliation(s)
- Hiroto Honda
- Department of Physical TherapyFaculty of Health SciencesAino UniversityHigashiodaJapan
| | - Makoto Igaki
- Department of RehabilitationToyooka Hospital Hidaka Medical CenterToyookaJapan
| | - Motoaki Komatsu
- Department of Internal MedicineToyooka Hospital Hidaka Medical CenterToyookaJapan
| | - Shin‐ichiro Tanaka
- Department of Internal MedicineToyooka Hospital Hidaka Medical CenterToyookaJapan
| |
Collapse
|
44
|
Effect of moderate-intensity seated exercise on the management of metabolic outcomes in hypertensive individuals with or without exercise habits. J Exerc Sci Fit 2020; 19:51-56. [PMID: 33224205 PMCID: PMC7658705 DOI: 10.1016/j.jesf.2020.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 09/24/2020] [Accepted: 09/26/2020] [Indexed: 11/21/2022] Open
Abstract
Background We aimed to evaluate the effect of moderate-intensity seated exercise on metabolic outcomes in hypertensive individuals with or without exercise habits. Methods Forty-two hypertensive individuals volunteered for this study and were classified into 3 groups by their habits and place for moderate-intensity exercise prior to this study: NONE (<2 days/week or no exercise; n = 13), HOME (≥30 min/day and ≥2 days/week at home; n = 15), and GYM (≥30 min/day and ≥2 days/week at a hospital gym; n = 14). They performed their daily activities as usual and seated exercise (stepping and stepping with trunk rotation; a range of 11–13 on the Borg rating of perceived exertion scale) for at least 15 min/day and at least 3 days/week for 12 weeks. Results Thirty-five participants (age: 67.7 ± 5.9 years) completed the study, and there was no difference among the 3 groups regarding weekly exercise. The homeostasis model assessment of insulin resistance (HOMA-IR) value in the NONE group was significantly higher than that in the GYM group at baseline (p < 0.05), but it decreased significantly after 12 weeks (from 2.2 ± 0.8 to 1.7 ± 0.7, p < 0.05). Changes in HOMA-IR in the NONE group after 12 weeks was greater than that in the HOME and GYM groups (both p < 0.01). The HOME and GYM groups showed no significant changes in any of the variables. Conclusion Moderate-intensity seated exercise may be an effective strategy to improve insulin resistance in hypertensive individuals without exercise habits.
Collapse
|
45
|
Li S, Yang Z, Li M, Zhu JZ, Zhang XQ. A bare-metal stents treatment of arterial injuries at the joint levels. Chin Med J (Engl) 2020; 133:2625-2627. [PMID: 32991366 PMCID: PMC7722550 DOI: 10.1097/cm9.0000000000001103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Indexed: 12/02/2022] Open
Affiliation(s)
- Shuai Li
- Postgraduate Department, Shandong First Medical University, Taian, Shandong 271016, China
| | - Zhen Yang
- Department of Nuclear Medicine, PLA 960th Hospital, Jinan, Shandong 250031, China
| | - Min Li
- Department of Nuclear Medicine, PLA 960th Hospital, Jinan, Shandong 250031, China
| | - Jian-Zhong Zhu
- Department of Medical Imaging, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, China
| | - Xi-Quan Zhang
- Department of Interventional Vascular, PLA 960th Hospital, Zibo, Shandong 255300, China
| |
Collapse
|
46
|
Walther BK, Mojiri A, Ohayon J, Cooke JP, Pettigrew RI. Caveats on modeling of nuclear biomechanics. Mol Biol Cell 2020; 31:2421-2422. [PMID: 33054638 PMCID: PMC7851855 DOI: 10.1091/mbc.e20-05-0281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Brandon K Walther
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030.,Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843
| | - Anahita Mojiri
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030
| | - Jacques Ohayon
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030.,Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843.,Savoie Mont-Blanc University, Polytech Annecy-Chambéry, 73376 Le Bourget du Lac, France, & Laboratory TIMC-IMAG-UGA, CNRS UMR 5525, 38706 Grenoble, France
| | - John P Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030.,Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843.,Houston Methodist Hospital, 6565 Fannin St. Houston, TX 77030
| | - Roderic I Pettigrew
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843.,Houston Methodist Hospital, 6565 Fannin St. Houston, TX 77030
| |
Collapse
|
47
|
Bosch Rué E, Delgado LM, Gil FJ, Perez RA. Direct extrusion of individually encapsulated endothelial and smooth muscle cells mimicking blood vessel structures and vascular native cell alignment. Biofabrication 2020; 13. [PMID: 32998120 DOI: 10.1088/1758-5090/abbd27] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/30/2020] [Indexed: 12/17/2022]
Abstract
Cardiovascular diseases (CVDs) are considered the principal cause of worldwide death, being atherosclerosis the main etiology. Up to now, the predominant treatment for CVDs has been bypass surgery from autologous source. However, due to previous harvest or the type of disease, this is not always an option. For this reason, tissue engineering blood vessels (TEBV) emerged as an alternative graft source for blood vessel replacement. In order to develop a TEBV, it should mimic the architecture of a native blood vessel encapsulating the specific vascular cells in their respective layers with native alignment, and with appropriate mechanical stability. Here, we propose the extrusion of two different cell encapsulating hydrogels, mainly alginate and collagen, and a sacrificial polymer, through a triple coaxial nozzle, which in contact with a crosslinking solution allows the formation of bilayered hollow fibers, mimicking the architecture of native blood vessels. Prior to extrusion, the innermost cell encapsulating hydrogel was loaded with human umbilical vein endothelial cells (HUVECs), whereas the outer hydrogel was loaded with human aortic smooth muscle cells (HASMCs). The size of the TEVB could be controlled by changing the injection speed, presenting homogeneity between the constructs. The obtained structures were robust, allowing its manipulation as well as the perfusion of liquids. Both cell types presented high rates of survival after the extrusion process as well as after 20 days in culture (over 90%). Additionally, a high percentage of HASMC and HUVEC were aligned perpendicular and parallel to the TEBV, respectively, in their own layers, resembling the physiological arrangement found in vivo. Our approach enables the rapid formation of TEBV-like structures presenting high cell viability and allowing proliferation and natural alignment of vascular cells.
Collapse
Affiliation(s)
- Elia Bosch Rué
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, C/ Josep Trueta, sn, Barcelona, Barcelona, 08018, SPAIN
| | - Luis M Delgado
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Catalunya, SPAIN
| | - F Javier Gil
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Catalunya, SPAIN
| | - Roman A Perez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Catalunya, SPAIN
| |
Collapse
|
48
|
MiRNAs, lncRNAs, and circular RNAs as mediators in hypertension-related vascular smooth muscle cell dysfunction. Hypertens Res 2020; 44:129-146. [DOI: 10.1038/s41440-020-00553-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/20/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022]
|
49
|
Leopold JA. Endocrine Delivery of MicroRNA-210: A Trusted Traveler That Mediates Pulmonary Hypertension. Circ Res 2020; 127:693-695. [PMID: 32790524 DOI: 10.1161/circresaha.120.317625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jane A Leopold
- From the Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
50
|
Liang J, Gu S, Mao X, Tan Y, Wang H, Li S, Zhou Y. Endothelial Cell Morphology Regulates Inflammatory Cells Through MicroRNA Transferred by Extracellular Vesicles. Front Bioeng Biotechnol 2020; 8:369. [PMID: 32509739 PMCID: PMC7248333 DOI: 10.3389/fbioe.2020.00369] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/02/2020] [Indexed: 01/13/2023] Open
Abstract
Vascular inflammation plays an important role in the pathogenesis and the development of cardiovascular diseases such as arteriosclerosis and restenosis, and the dysfunction of endothelial cells (ECs) may result in the activation of monocytes and other inflammatory cells. ECs exhibit an elongated morphology in the straight part of arteries but a cobblestone shape near the pro-atherogenic region such as branch bifurcation. Although the effects of hemodynamic forces on ECs have been widely studied, it is not clear whether the EC morphology affects its own function and thus the inflammatory response of monocytes. Here we showed that elongated ECs cultured on poly-(dimethyl siloxane) membrane surface with microgrooves significantly suppressed the activation of the monocytes in co-culture, in comparison to ECs with a cobblestone shape. The transfer of EC-conditioned medium to monocytes had the same effect, suggesting that soluble factors were involved in EC-monocyte communication. Further investigation demonstrated that elongated ECs upregulated the expression of anti-inflammatory microRNAs, especially miR-10a. Moreover, miR-10a was found in the extracellular vesicles (EVs) released by ECs and transferred to monocytes, and the inhibition of EV secretion from ECs repressed the upregulation of miR-10a. Consistently, the inhibition of miR-10a expression in ECs reduced their anti-inflammatory effect on monocytes. These results reveal that the EC morphology can regulate inflammatory response through EVs, which provides a basis for the design and the optimization of biomaterials for vascular tissue engineering.
Collapse
Affiliation(s)
- Jiaqi Liang
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Med-X Research Institution, Shanghai Jiao Tong University, Shanghai, China
| | - Shuangying Gu
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Med-X Research Institution, Shanghai Jiao Tong University, Shanghai, China
| | - Xiuli Mao
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Med-X Research Institution, Shanghai Jiao Tong University, Shanghai, China
| | - Yiling Tan
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Med-X Research Institution, Shanghai Jiao Tong University, Shanghai, China
| | - Huanli Wang
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Med-X Research Institution, Shanghai Jiao Tong University, Shanghai, China
| | - Song Li
- Department of Bioengineering, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yue Zhou
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Med-X Research Institution, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|