1
|
Zhang L, Zhou J, Kong W. Extracellular matrix in vascular homeostasis and disease. Nat Rev Cardiol 2025; 22:333-353. [PMID: 39743560 DOI: 10.1038/s41569-024-01103-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 01/04/2025]
Abstract
The extracellular matrix is an essential component and constitutes a dynamic microenvironment of the vessel wall with an indispensable role in vascular homeostasis and disease. From early development through to ageing, the vascular extracellular matrix undergoes various biochemical and biomechanical alterations in response to diverse environmental cues and exerts precise regulatory control over vessel remodelling. Advances in novel technologies that enable the comprehensive evaluation of extracellular matrix components and cell-matrix interactions have led to the emergence of therapeutic strategies that specifically target this fine-tuned network. In this Review, we explore various aspects of extracellular matrix biology in vascular development, disorders and ageing, emphasizing the effect of the extracellular matrix on disease initiation and progression. Additionally, we provide an overview of the potential therapeutic implications of targeting the extracellular matrix microenvironment in vascular diseases.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| |
Collapse
|
2
|
Ben Hassine A, Petit C, Thomas M, Mundweiler S, Guignandon A, Avril S. Gene expression modulation in human aortic smooth muscle cells under induced physiological mechanical stretch. Sci Rep 2024; 14:31147. [PMID: 39732782 DOI: 10.1038/s41598-024-82495-4] [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: 06/04/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
In this study, we investigated gene expression in vitro of human primary Aortic smooth muscle cells (AoSMCs) in response to 9% physiological dynamic stretch over a 4 to 72-h timeframe using RT-qPCR. AoSMC were derived from primary culture and were exposed to continuous cycles of stretch and relaxation at 1 Hz by a computer-controlled Flex Jr.™ Tension System. Unstretched control AoSMCs were simultaneously cultured in the same dishes. Our results revealed a rapid and significant upregulation of specific genes (COL1A1, FBN1, LAMA5, TGFBR1 and TGFBR2) within the initial 4 h for AoSMCs subjected to dynamic stretching, whilst control cells did not respond within the same 4 h. The upregulated genes were the ones associated with extracellular matrix (ECM) fibrillogenesis and regulation of traction forces. Interestingly, stretched cells maintained stable gene expression between 4 and 72 h, whilst control cells exhibited variations over time in the absence of mechanical cues. These findings shed light on the essential role played by pulsatile stretches in the regulation of gene expressions by AoSMCs and the intricate processes governing their mechanobiological function, paving the way for further investigations in cardiovascular health.
Collapse
Affiliation(s)
- Amira Ben Hassine
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Claudie Petit
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Mireille Thomas
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Stéphanie Mundweiler
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Alain Guignandon
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Stéphane Avril
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France.
| |
Collapse
|
3
|
Farach-Carson MC, Wu D, França CM. Proteoglycans in Mechanobiology of Tissues and Organs: Normal Functions and Mechanopathology. PROTEOGLYCAN RESEARCH 2024; 2:e21. [PMID: 39584146 PMCID: PMC11584024 DOI: 10.1002/pgr2.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/03/2024] [Indexed: 11/26/2024]
Abstract
Proteoglycans (PGs) are a diverse class of glycoconjugates that serve critical functions in normal mechanobiology and mechanopathology. Both the protein cores and attached glycosaminoglycan (GAG) chains function in mechanically-sensitive processes, and loss of either can contribute to development of pathological conditions. PGs function as key components of the extracellular matrix (ECM) where they can serve as mechanosensors in mechanosensitive tissues including bone, cartilage, tendon, blood vessels and soft organs. The mechanical properties of these tissues depend on the presence and function of PGs, which play important roles in tissue elasticity, osmolarity and pressure sensing, and response to physical activity. Tissue responses depend on cell surface mechanoreceptors that include integrins, CD44, voltage sensitive ion channels, transient receptor potential (TRP) and piezo channels. PGs contribute to cell and molecular interplay in wound healing, fibrosis, and cancer, where they transduce the mechanical properties of the ECM and influence the progression of various context-specific conditions and diseases. The PGs that are most important in mechanobiology vary depending on the tissue and its functions and functional needs. Perlecan, for example, is important in the mechanobiology of basement membranes, cardiac and skeletal muscle, while aggrecan plays a primary role in the mechanical properties of cartilage and joints. A variety of techniques have been used to study the mechanobiology of PGs, including atomic force microscopy, mouse knockout models, and in vitro cell culture experiments with 3D organoid models. These studies have helped to elucidate the tissue-specific roles that PGs play in cell-level mechanosensing and tissue mechanics. Overall, the study of PGs in mechanobiology is yielding fundamental new concepts in the molecular basis of mechanosensing that can open the door to the development of new treatments for a host of conditions related to mechanopathology.
Collapse
Affiliation(s)
- Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX 77005
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX 77005
| | - Cristiane Miranda França
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, Portland, OR, 97201
- Knight Cancer Precision Biofabrication Hub, Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97201
| |
Collapse
|
4
|
Gao Y, Jiang Z, Xu B, Mo R, Li S, Jiang Y, Zhao D, Cao W, Chen B, Tian M, Tan Q. Evaluation of topical methylene blue nanoemulsion for wound healing in diabetic mice. PHARMACEUTICAL BIOLOGY 2023; 61:1462-1473. [PMID: 37691404 PMCID: PMC10496548 DOI: 10.1080/13880209.2023.2254341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023]
Abstract
CONTEXT Diabetic wounds (DW) are a complication of diabetes and slow wound healing is the main manifestation. Methylene blue (MB) has been shown to exhibit therapeutic effects on diabetes-related diseases. OBJECTIVE To investigate the mechanisms of action of MB-nanoemulsion (NE) in the treatment of DW. MATERIALS AND METHODS The concentration of MB-NE used in the in vivo and in vitro experiments was 0.1 mg/mL. Streptozocin-induced diabetic mice were used as models. The mice were separated into nondiabetic, diabetic, MB-NE treated, and NE-treated groups. Intervention of high glucose-induced human umbilical vein endothelial cells using MB-NE. The mechanism by which MB-NE promotes DW healing is investigated by combining histological analysis, immunofluorescence analysis, TUNEL and ROS assays and western blotting. RESULTS In diabetic mice, the MB-NE accelerated DW healing (p < 0.05), promoted the expression of endothelial cell markers (α-SMA, CD31 and VEGF) (p < 0.05), and reduced TUNEL levels. In vitro, MB accelerated the migration rate of cells (p < 0.05); promoted the expression of CD31, VEGF, anti-apoptotic protein Bcl2 (p < 0.05) and decreased the expression of the pro-apoptotic proteins cleaved caspase-3 and Bax (p < 0.05). MB upregulated the expression of Nrf2, catalase, HO-1 and SOD2 (p < 0.05). In addition, MB reduced the immunofluorescence intensity of TUNEL and ROS in cells and reduced apoptosis. The therapeutic effect of MB was attenuated after treatment with an Nrf2 inhibitor (ML385). DISCUSSION AND CONCLUSION This study provides a foundation for the application of MB-NE in the treatment of DW.
Collapse
Affiliation(s)
- Yu Gao
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Zhounan Jiang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Xu
- Hubei Xiangyang Central Hospital, Xiangyang, China
| | - Ran Mo
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Shiyan Li
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yanan Jiang
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Demei Zhao
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wangbin Cao
- Nanjing Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Bin Chen
- Institute of Plant Resources and Chemistry, Nanjing Research Institute for Comprehensive Utilization of Wild Plants, Nanjing, China
| | - Meng Tian
- Department of Plastic Surgery, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qian Tan
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Tasci O, Dogan K. Evaluation of tumour necrosis factor alpha-stimulated gene-6 and fibroblast growth factor-2 levels in patients diagnosed with multi-system inflammatory syndrome in children. Cardiol Young 2023; 33:1086-1091. [PMID: 36918343 DOI: 10.1017/s1047951123000355] [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: 03/16/2023]
Abstract
Investigations are still ongoing about the pathophysiology of multi-system inflammatory syndrome in children, which can progress with serious morbidity and mortality after COVID-19 infection. In this study, we aimed to investigate whether fibroblast growth factor-2 and tumour necrosis factor alpha-stimulated gene-6 levels play a role in the diagnosis of the disease and on cardiac involvement. Twenty-three patients (11 girls, 12 boys) and 26 healthy controls (10 girls, 16 boys) were included in the study. The mean age of the patient and control group was 8.45 ± 2.43 and 10.73 ± 4.27 years, respectively. There was no difference between the fibroblast growth factor-2 and tumour necrosis factor alpha-stimulated gene-6 levels of the patient and control groups. When the patients with myocardial involvement in the patient group were compared with the patients without myocardial involvement in terms of fibroblast growth factor-2 and tumour necrosis factor alpha-stimulated gene-6 levels, no difference was found between these groups. The correlation of fibroblast growth factor-2 and tumour necrosis factor alpha-stimulated gene-6 levels with other laboratory parameters was investigated in the patient group. Fibroblast growth factor-2 was moderately inversely correlated with white blood cell count (r = -0.541, p = 0.008), absolute neutrophil count (r = -0.502, p = 0.015) and C-reactive protein (r = -0.528, p = 0.010). Fibroblast growth factor-2 was strongly inversely correlated with erythrocyte sedimentation rate (r = -0.694, p =<0.001). Our data show that fibroblast growth factor-2 and tumour necrosis factor alpha stimulated gene-6 do not provide sufficient information about diagnosis and cardiac involvement in multi-system inflammatory syndrome in children.
Collapse
Affiliation(s)
- Onur Tasci
- Sivas Numune Hospital, Department of Pediatric Cardiology, Sivas, Turkey
| | - Kubra Dogan
- Sivas Numune Hospital, Department of Biochemistry, Sivas, Turkey
| |
Collapse
|
6
|
van der Have O, Mead TJ, Westöö C, Peruzzi N, Mutgan AC, Norvik C, Bech M, Struglics A, Hoetzenecker K, Brunnström H, Westergren‐Thorsson G, Kwapiszewska G, Apte SS, Tran‐Lundmark K. Aggrecan accumulates at sites of increased pulmonary arterial pressure in idiopathic pulmonary arterial hypertension. Pulm Circ 2023; 13:e12200. [PMID: 36824691 PMCID: PMC9941846 DOI: 10.1002/pul2.12200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Expansion of extracellular matrix occurs in all stages of pulmonary angiopathy associated with pulmonary arterial hypertension (PAH). In systemic arteries, dysregulation and accumulation of the large chondroitin-sulfate proteoglycan aggrecan is associated with swelling and disruption of vessel wall homeostasis. Whether aggrecan is present in pulmonary arteries, and its potential roles in PAH, has not been thoroughly investigated. Here, lung tissue from 11 patients with idiopathic PAH was imaged using synchrotron radiation phase-contrast microcomputed tomography (TOMCAT beamline, Swiss Light Source). Immunohistochemistry for aggrecan core protein in subsequently sectioned lung tissue demonstrated accumulation in PAH compared with failed donor lung controls. RNAscope in situ hybridization indicated ACAN expression in vascular endothelium and smooth muscle cells. Based on qualitative histological analysis, aggrecan localizes to cellular, rather than fibrotic or collagenous, lesions. Interestingly, ADAMTS15, a potential aggrecanase, was upregulated in pulmonary arteries in PAH. Aligning traditional histological analysis with three-dimensional renderings of pulmonary arteries from synchrotron imaging identified aggrecan in lumen-reducing lesions containing loose, cell-rich connective tissue, at sites of intrapulmonary bronchopulmonary shunting, and at sites of presumed elevated pulmonary blood pressure. Our findings suggest that ACAN expression may be an early response to injury in pulmonary angiopathy and supports recent work showing that dysregulation of aggrecan turnover is a hallmark of arterial adaptations to altered hemodynamics. Whether cause or effect, aggrecan and aggrecanase regulation in PAH are potential therapeutic targets.
Collapse
Affiliation(s)
- Oscar van der Have
- Department of Experimental Medical Science, Faculty of MedicineLund UniversityLundSweden
| | - Timothy J. Mead
- Department of Biomedical EngineeringCleveland Clinic Lerner Research InstituteClevelandOhioUSA
| | - Christian Westöö
- Department of Experimental Medical Science, Faculty of MedicineLund UniversityLundSweden
| | - Niccolò Peruzzi
- Department of Experimental Medical Science, Faculty of MedicineLund UniversityLundSweden
- Department of Medical Radiation Physics, Clinical Sciences LundLund UniversityLundSweden
| | - Ayse C. Mutgan
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Division of Physiology, Otto Loewi Research CenterMedical University GrazGrazAustria
| | - Christian Norvik
- Department of Experimental Medical Science, Faculty of MedicineLund UniversityLundSweden
| | - Martin Bech
- Department of Medical Radiation Physics, Clinical Sciences LundLund UniversityLundSweden
| | - André Struglics
- Department of Clinical Sciences Lund, Orthopaedics, Faculty of MedicineLund UniversityLundSweden
| | | | - Hans Brunnström
- Department of Clinical Sciences Lund, Division of Pathology, Faculty of MedicineLund UniversityLundSweden
- Department of Genetics and PathologyDivision of Laboratory MedicineLundSweden
| | - Gunilla Westergren‐Thorsson
- Department of Experimental Medical Science, Faculty of MedicineLund UniversityLundSweden
- Wallenberg Center for Molecular MedicineLund UniversityLundSweden
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Division of Physiology, Otto Loewi Research CenterMedical University GrazGrazAustria
- Institute for Lung HealthJustus Liebig UniversityGiessenGermany
| | - Suneel S. Apte
- Department of Biomedical EngineeringCleveland Clinic Lerner Research InstituteClevelandOhioUSA
| | - Karin Tran‐Lundmark
- Department of Experimental Medical Science, Faculty of MedicineLund UniversityLundSweden
- Wallenberg Center for Molecular MedicineLund UniversityLundSweden
- The Pediatric Heart CenterSkåne University HospitalLundSweden
| |
Collapse
|
7
|
Tokgoz A, Wang S, Sastry P, Sun C, Figg NL, Huang Y, Bennett MR, Sinha S, Gillard JH, Sutcliffe MPF, Teng Z. Association of Collagen, Elastin, Glycosaminoglycans, and Macrophages With Tissue Ultimate Material Strength and Stretch in Human Thoracic Aortic Aneurysms: A Uniaxial Tension Study. J Biomech Eng 2022; 144:101001. [PMID: 35274123 DOI: 10.1115/1.4054060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Indexed: 11/08/2022]
Abstract
Fiber structures and pathological features, e.g., inflammation and glycosaminoglycan (GAG) deposition, are the primary determinants of aortic mechanical properties which are associated with the development of an aneurysm. This study is designed to quantify the association of tissue ultimate strength and extensibility with the structural percentage of different components, in particular, GAG, and local fiber orientation. Thoracic aortic aneurysm (TAA) tissues from eight patients were collected. Ninety-six tissue strips of thickened intima, media, and adventitia were prepared for uni-extension tests and histopathological examination. Area ratios of collagen, elastin, macrophage and GAG, and collagen fiber dispersion were quantified. Collagen, elastin, and GAG were layer-dependent and the inflammatory burden in all layers was low. The local GAG ratio was negatively associated with the collagen ratio (r2 = 0.173, p < 0.05), but positively with elastin (r2 = 0.037, p < 0.05). Higher GAG deposition resulted in larger local collagen fiber dispersion in the media and adventitia, but not in the intima. The ultimate stretch in both axial and circumferential directions was exclusively associated with elastin ratio (axial: r2 = 0.186, p = 0.04; circumferential: r2 = 0.175, p = 0.04). Multivariate analysis showed that collagen and GAG contents were both associated with ultimate strength in the circumferential direction, but not with the axial direction (collagen: slope = 27.3, GAG: slope = -18.4, r2 = 0.438, p = 0.002). GAG may play important roles in TAA material strength. Their deposition was found to be associated positively with the local collagen fiber dispersion and negatively with ultimate strength in the circumferential direction.
Collapse
Affiliation(s)
- Aziz Tokgoz
- Department of Engineering, University of Cambridge, Cambridge CB2 1TN, UK
| | - Shuo Wang
- Department of Radiology, University of Cambridge, Cambridge CB2 1TN, UK; Digital Medical Research Center, School of Basic Medical Sciences, Fudan University, Shanghai 200437, China; Shanghai Key Laboratory of MICCAI, Shanghai, China
| | - Priya Sastry
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK
| | - Chang Sun
- Department of Radiology, University of Cambridge, Cambridge CB2 1TN, UK
| | - Nichola L Figg
- Digital Medical Research Center, School of Basic Medical Sciences, Fudan University, Shanghai 200437, China
| | - Yuan Huang
- Department of Radiology, University of Cambridge, Cambridge CB2 1TN, UK; Centre for Mathematical and Statistical Analysis of Multimodal Clinical Imaging, University of Cambridge, Cambridge CB2 1TN, UK
| | - Martin R Bennett
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK
| | - Sanjay Sinha
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK
| | | | - Michael P F Sutcliffe
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK
| | - Zhongzhao Teng
- Department of Engineering, University of Cambridge, Cambridge CB2 1TN, UK; Department of Radiology, University of Cambridge, Level 5, Box 218, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK; Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China; Nanjing Jingsan Medical Science and Technology, Ltd., Jiangsu, China
| |
Collapse
|
8
|
Maselli D, Garoffolo G, Cassanmagnago GA, Vono R, Ruiter MS, Thomas AC, Madeddu P, Pesce M, Spinetti G. Mechanical Strain Induces Transcriptomic Reprogramming of Saphenous Vein Progenitors. Front Cardiovasc Med 2022; 9:884031. [PMID: 35711359 PMCID: PMC9197233 DOI: 10.3389/fcvm.2022.884031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/28/2022] [Indexed: 11/23/2022] Open
Abstract
Intimal hyperplasia is the leading cause of graft failure in aortocoronary bypass grafts performed using human saphenous vein (SV). The long-term consequences of the altered pulsatile stress on the cells that populate the vein wall remains elusive, particularly the effects on saphenous vein progenitors (SVPs), cells resident in the vein adventitia with a relatively wide differentiation capacity. In the present study, we performed global transcriptomic profiling of SVPs undergoing uniaxial cyclic strain in vitro. This type of mechanical stimulation is indeed involved in the pathology of the SV. Results showed a consistent stretch-dependent gene regulation in cyclically strained SVPs vs. controls, especially at 72 h. We also observed a robust mechanically related overexpression of Adhesion Molecule with Ig Like Domain 2 (AMIGO2), a cell surface type I transmembrane protein involved in cell adhesion. The overexpression of AMIGO2 in stretched SVPs was associated with the activation of the transforming growth factor β pathway and modulation of intercellular signaling, cell-cell, and cell-matrix interactions. Moreover, the increased number of cells expressing AMIGO2 detected in porcine SV adventitia using an in vivo arterialization model confirms the upregulation of AMIGO2 protein by the arterial-like environment. These results show that mechanical stress promotes SVPs' molecular phenotypic switching and increases their responsiveness to extracellular environment alterations, thus prompting the targeting of new molecular effectors to improve the outcome of bypass graft procedure.
Collapse
Affiliation(s)
- Davide Maselli
- IRCCS MultiMedica, Milan, Italy
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Gloria Garoffolo
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Giada Andrea Cassanmagnago
- IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | | | - Matthijs S. Ruiter
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Anita C. Thomas
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Paolo Madeddu
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Gaia Spinetti
- IRCCS MultiMedica, Milan, Italy
- *Correspondence: Gaia Spinetti
| |
Collapse
|
9
|
Swiatlowska P, Sit B, Feng Z, Marhuenda E, Xanthis I, Zingaro S, Ward M, Zhou X, Xiao Q, Shanahan C, Jones GE, Yu CH, Iskratsch T. Pressure and stiffness sensing together regulate vascular smooth muscle cell phenotype switching. SCIENCE ADVANCES 2022; 8:eabm3471. [PMID: 35427166 PMCID: PMC9012473 DOI: 10.1126/sciadv.abm3471] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 03/01/2022] [Indexed: 06/14/2023]
Abstract
Vascular smooth muscle cells (VSMCs) play a central role in the progression of atherosclerosis, where they switch from a contractile to a synthetic phenotype. Because of their role as risk factors for atherosclerosis, we sought here to systematically study the impact of matrix stiffness and (hemodynamic) pressure on VSMCs. Thereby, we find that pressure and stiffness individually affect the VSMC phenotype. However, only the combination of hypertensive pressure and matrix compliance, and as such mechanical stimuli that are prevalent during atherosclerosis, leads to a full phenotypic switch including the formation of matrix-degrading podosomes. We further analyze the molecular mechanism in stiffness and pressure sensing and identify a regulation through different but overlapping pathways culminating in the regulation of the actin cytoskeleton through cofilin. Together, our data show how different pathological mechanical signals combined but through distinct pathways accelerate a phenotypic switch that will ultimately contribute to atherosclerotic disease progression.
Collapse
Affiliation(s)
- Pamela Swiatlowska
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Brian Sit
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, UK
- School of Biomedical Sciences, Hong Kong University, Hong Kong, Hong Kong
| | - Zhen Feng
- School of Biomedical Sciences, Hong Kong University, Hong Kong, Hong Kong
| | - Emilie Marhuenda
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Ioannis Xanthis
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Simona Zingaro
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, UK
| | - Matthew Ward
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Xinmiao Zhou
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Qingzhong Xiao
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Cathy Shanahan
- School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| | - Gareth E. Jones
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, UK
| | - Cheng-han Yu
- School of Biomedical Sciences, Hong Kong University, Hong Kong, Hong Kong
| | - Thomas Iskratsch
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, UK
| |
Collapse
|
10
|
Vang S, Cochran P, Sebastian Domingo J, Krick S, Barnes JW. The Glycobiology of Pulmonary Arterial Hypertension. Metabolites 2022; 12:metabo12040316. [PMID: 35448503 PMCID: PMC9026683 DOI: 10.3390/metabo12040316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive pulmonary vascular disease of complex etiology. Cases of PAH that do not receive therapy after diagnosis have a low survival rate. Multiple reports have shown that idiopathic PAH, or IPAH, is associated with metabolic dysregulation including altered bioavailability of nitric oxide (NO) and dysregulated glucose metabolism. Multiple processes such as increased proliferation of pulmonary vascular cells, angiogenesis, apoptotic resistance, and vasoconstriction may be regulated by the metabolic changes demonstrated in PAH. Recent reports have underscored similarities between metabolic abnormalities in cancer and IPAH. In particular, increased glucose uptake and altered glucose utilization have been documented and have been linked to the aforementioned processes. We were the first to report a link between altered glucose metabolism and changes in glycosylation. Subsequent reports have highlighted similar findings, including a potential role for altered metabolism and aberrant glycosylation in IPAH pathogenesis. This review will detail research findings that demonstrate metabolic dysregulation in PAH with an emphasis on glycobiology. Furthermore, this report will illustrate the similarities in the pathobiology of PAH and cancer and highlight the novel findings that researchers have explored in the field.
Collapse
|
11
|
Grégory Franck. Role of mechanical stress and neutrophils in the pathogenesis of plaque erosion. Atherosclerosis 2020; 318:60-69. [PMID: 33190807 DOI: 10.1016/j.atherosclerosis.2020.11.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/05/2020] [Accepted: 11/03/2020] [Indexed: 02/05/2023]
Abstract
Mechanical stress is a well-recognized driver of plaque rupture. Likewise, investigating the role of mechanical forces in plaque erosion has recently begun to provide some important insights, yet the knowledge is by far less advanced. The most significant example is that of shear stress, which has early been proposed as a possible driver for focal endothelial death and denudation. Recent findings using optical coherence tomography, computational sciences and mechanical models show that plaque erosion occurs most likely around atheromatous plaque throats with specific stress pattern. In parallel, we have recently shown that neutrophil-dependent inflammation promotes plaque erosion, possibly through a noxious action on ECs. Most importantly, spontaneous thrombosis - associated or not with EC denudation - can be impacted by hemodynamics, and it is now established that neutrophils promote thrombosis and platelet activation, highlighting a potential relationship between, mechanical stress, inflammation, and EC loss in the setting of coronary plaque erosion. Here, we review our current knowledge regarding the implication of both mechanical stress and neutrophils, and we discuss their implication in the promotion of plaque erosion via EC loss and thrombosis.
Collapse
Affiliation(s)
- Grégory Franck
- Inserm LVTS U1148. CHU Bichat, 46 Rue Henri Huchard, 75018, Paris, France.
| |
Collapse
|
12
|
Large-deformation strain energy density function for vascular smooth muscle cells. J Biomech 2020; 111:110005. [DOI: 10.1016/j.jbiomech.2020.110005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/29/2020] [Accepted: 08/21/2020] [Indexed: 01/03/2023]
|
13
|
Detection of TSG-6-like protein in human corneal epithelium. Simultaneous presence with CD44 and hyaluronic acid. J Fr Ophtalmol 2020; 43:879-883. [PMID: 32829938 DOI: 10.1016/j.jfo.2020.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/23/2020] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Tumor necrosis factor-inducible gene 6 protein (TSG-6) is member of the hyaluronan-binding protein family (hyaladherins) to which CD44 also belongs. Inflammatory mediators such as tumor necrosis factor α (TNF-α) and interleukin-1 (IL-1) stimulate TSG-6 production. Recently, however, externally applied TSG-6 has been shown to be effective in the treatment of inflammatory dry eye. On the other hand, it is still unknown whether TSG-6 is naturally present in human corneal epithelium. MATERIAL AND METHODS Corneal sections of 15 eyes enucleated for posterior segment uveal melanoma were immunohistochemically stained for hyaluronic acid (HA), CD44, and TSG-6. RESULTS Throughout the corneal epithelium of all sections, CD44 and hyaluronic acid were detected most intensely in the basal epithelial layer. Whereas the presence of HA was intense even in the cytoplasm of the cells, CD44 was located predominantly at the cell membranes. The intensity of the specific staining decreased towards the surface, where CD44 was barely detectable. Hyaluronic acid was, on the other hand, detectable in the extracellular matrix and cells, even at the surface. TSG-6 like immunoreactivity was detected in all sections in a pattern similar to CD44 but much more distinct and intense, with a marked localization in the cell membranes and intercellular spaces, i.e., extracellular matrix. TSG-6 like immunoreactivity was clearly detectable through all cell layers of the corneal epithelium. All control sections were negative. DISCUSSION Tumor necrosis factor-inducible gene 6 (TSG-6)- like protein is present in human corneal epithelium. It might be a natural component of this tissue which is constantly exposed and mechanically traumatized, and displays localization with similarities to that of CD44. The immunohistological detection of HA as major component of the ECM and epithelial tissue only confirms the results of earlier studies. However, the simultaneous presence and colocalization of CD44 and TSG-6, both HA-binding proteins, requires further investigation of the individual role, regulation and interaction of this system. CONCLUSION The detection of TSG-6 in human corneal epithelium in the absence of inflammation underlines the importance of normal mechanical forces on the gene expression and regulation of this protein in ocular surface tissues. Given the relationship between inflammation and the protein, TSG-6 may be a major unknown and underestimated player in the regulation of the inflammation encountered in the presence of ocular surface desiccation and dry eye disease.
Collapse
|
14
|
van Setten GB. Impact of Attrition, Intercellular Shear in Dry Eye Disease: When Cells are Challenged and Neurons are Triggered. Int J Mol Sci 2020; 21:E4333. [PMID: 32570730 PMCID: PMC7352662 DOI: 10.3390/ijms21124333] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/02/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022] Open
Abstract
The mechanical component in the pathophysiology of dry eye disease (DED) deserves attention as an important factor. The lubrication deficit induced impaired mechano-transduction of lid pressure to the ocular surfaces may lead to the dysregulation of homeostasis in the epithelium, with sensations of pain and secondary inflammation. Ocular pain is possibly the first sign of attrition and may occur in the absence of visible epithelial damage. Attrition is a process which involves the constant or repeated challenge of ocular surface tissues by mechanical shear forces; it is enhanced by the thinning of corneal epithelium in severe DED. As a highly dynamic process leading to pain and neurogenic inflammation, the identification of the impact of attrition and its potential pathogenic role could add a new perspective to the current more tear film-oriented models of ocular surface disease. Treatment of DED addressing lubrication deficiencies and inflammation should also consider the decrease of attrition in order to stimulate epithelial recovery and neural regeneration. The importance of hyaluronic acid, its molecular characteristics, the extracellular matrix and autoregulative mechanisms in this process is outlined. The identification of the attrition and recognition of its impact in dry eye pathophysiology could contribute to a better understanding of the disease and optimized treatment regimens.
Collapse
Affiliation(s)
- Gysbert-Botho van Setten
- Department of Clinical Neuroscience (CNS), Karolinska Institutet, 11282 Stockholm, Sweden; ; Tel.: +46-8-672-3298
- St Eriks Eye Hospital, 11282 Stockholm, Sweden
| |
Collapse
|
15
|
Amirfakhryan H. Vaccination against atherosclerosis: An overview. Hellenic J Cardiol 2020; 61:78-91. [DOI: 10.1016/j.hjc.2019.07.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023] Open
|
16
|
Atherosclerosis and the Capillary Network; Pathophysiology and Potential Therapeutic Strategies. Cells 2019; 9:cells9010050. [PMID: 31878229 PMCID: PMC7016600 DOI: 10.3390/cells9010050] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis and associated ischemic organ dysfunction represent the number one cause of mortality worldwide. While the key drivers of atherosclerosis, arterial hypertension, hypercholesterolemia and diabetes mellitus, are well known disease entities and their contribution to the formation of atherosclerotic plaques are intensively studied and well understood, less effort is put on the effect of these disease states on microvascular structure an integrity. In this review we summarize the pathological changes occurring in the vascular system in response to prolonged exposure to these major risk factors, with a particular focus on the differences between these pathological alterations of the vessel wall in larger arteries as compared to the microcirculation. Furthermore, we intend to highlight potential therapeutic strategies to improve microvascular function during atherosclerotic vessel disease.
Collapse
|
17
|
Zun PS, Narracott AJ, Chiastra C, Gunn J, Hoekstra AG. Location-Specific Comparison Between a 3D In-Stent Restenosis Model and Micro-CT and Histology Data from Porcine In Vivo Experiments. Cardiovasc Eng Technol 2019; 10:568-582. [PMID: 31531821 PMCID: PMC6863796 DOI: 10.1007/s13239-019-00431-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 09/07/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Coronary artery restenosis is an important side effect of percutaneous coronary intervention. Computational models can be used to better understand this process. We report on an approach for validation of an in silico 3D model of in-stent restenosis in porcine coronary arteries and illustrate this approach by comparing the modelling results to in vivo data for 14 and 28 days post-stenting. METHODS This multiscale model includes single-scale models for stent deployment, blood flow and tissue growth in the stented vessel, including smooth muscle cell (SMC) proliferation and extracellular matrix (ECM) production. The validation procedure uses data from porcine in vivo experiments, by simulating stent deployment using stent geometry obtained from micro computed tomography (micro-CT) of the stented vessel and directly comparing the simulation results of neointimal growth to histological sections taken at the same locations. RESULTS Metrics for comparison are per-strut neointimal thickness and per-section neointimal area. The neointimal area predicted by the model demonstrates a good agreement with the detailed experimental data. For 14 days post-stenting the relative neointimal area, averaged over all vessel sections considered, was 20 ± 3% in vivo and 22 ± 4% in silico. For 28 days, the area was 42 ± 3% in vivo and 41 ± 3% in silico. CONCLUSIONS The approach presented here provides a very detailed, location-specific, validation methodology for in silico restenosis models. The model was able to closely match both histology datasets with a single set of parameters. Good agreement was obtained for both the overall amount of neointima produced and the local distribution. It should be noted that including vessel curvature and ECM production in the model was paramount to obtain a good agreement with the experimental data.
Collapse
Affiliation(s)
- P S Zun
- Institute for Informatics, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands.
- Biomechanics Laboratory, Department of Biomedical Engineering, Erasmus Medical Center, Rotterdam, The Netherlands.
- National Center for Cognitive Technologies, ITMO University, Saint Petersburg, Russia.
| | - A J Narracott
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
| | - C Chiastra
- Laboratory of Biological Structure Mechanics (LaBS), Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
- PoliToBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - J Gunn
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
| | - A G Hoekstra
- Institute for Informatics, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Basatemur GL, Jørgensen HF, Clarke MCH, Bennett MR, Mallat Z. Vascular smooth muscle cells in atherosclerosis. Nat Rev Cardiol 2019; 16:727-744. [PMID: 31243391 DOI: 10.1038/s41569-019-0227-9] [Citation(s) in RCA: 699] [Impact Index Per Article: 116.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2019] [Indexed: 02/08/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are a major cell type present at all stages of an atherosclerotic plaque. According to the 'response to injury' and 'vulnerable plaque' hypotheses, contractile VSMCs recruited from the media undergo phenotypic conversion to proliferative synthetic cells that generate extracellular matrix to form the fibrous cap and hence stabilize plaques. However, lineage-tracing studies have highlighted flaws in the interpretation of former studies, revealing that these studies had underestimated both the content and functions of VSMCs in plaques and have thus challenged our view on the role of VSMCs in atherosclerosis. VSMCs are more plastic than previously recognized and can adopt alternative phenotypes, including phenotypes resembling foam cells, macrophages, mesenchymal stem cells and osteochondrogenic cells, which could contribute both positively and negatively to disease progression. In this Review, we present the evidence for VSMC plasticity and summarize the roles of VSMCs and VSMC-derived cells in atherosclerotic plaque development and progression. Correct attribution and spatiotemporal resolution of clinically beneficial and detrimental processes will underpin the success of any therapeutic intervention aimed at VSMCs and their derivatives.
Collapse
Affiliation(s)
- Gemma L Basatemur
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Helle F Jørgensen
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Murray C H Clarke
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Martin R Bennett
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK.
- INSERM U970, Paris Cardiovascular Research Center, Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
19
|
Hayashi N, Sato T, Yumoto M, Kokabu S, Fukushima Y, Kawata Y, Kajihara T, Mizuno Y, Mizuno Y, Kawakami T, Kirita T, Hayata T, Noda M, Yoda T. Cyclic stretch induces decorin expression via yes-associated protein in tenocytes: A possible mechanism for hyperplasia in masticatory muscle tendon-aponeurosis hyperplasia. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY, MEDICINE, AND PATHOLOGY 2019. [DOI: 10.1016/j.ajoms.2018.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
20
|
Dyck GJB, Raj P, Zieroth S, Dyck JRB, Ezekowitz JA. The Effects of Resveratrol in Patients with Cardiovascular Disease and Heart Failure: A Narrative Review. Int J Mol Sci 2019; 20:ijms20040904. [PMID: 30791450 PMCID: PMC6413130 DOI: 10.3390/ijms20040904] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 02/03/2019] [Accepted: 02/06/2019] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease (CVD) is the main cause of death globally and responsible for the second highest number of deaths in Canada. Medical advancements in the treatment of CVD have led to patients living longer with CVD but often progressing to another condition called heart failure (HF). As a result, HF has emerged in the last decade as a major medical concern. Fortunately, various “traditional” pharmacotherapies for HF exist and have shown success in reducing HF-associated mortality. However, to augment the treatment of patients with CVD and/or HF, alternative pharmacotherapies using nutraceuticals have also shown promise in the prevention and treatment of these two conditions. One of these natural compounds considered to potentially help treat HF and CVD and prevent their development is resveratrol. Herein, we review the clinical findings of resveratrol’s ability to be used as an effective treatment to potentially help treat HF and CVD. This will allow us to gain a more fulsome appreciation for the effects of resveratrol in the health outcomes of specific patient populations who have various disorders that constitute CVD.
Collapse
Affiliation(s)
- Garrison J B Dyck
- Canadian VIGOUR Centre, Mazankowski Alberta Heart Institute, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| | - Pema Raj
- St Boniface Hospital, Department of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada.
| | - Shelley Zieroth
- St Boniface Hospital, Department of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada.
| | - Jason R B Dyck
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada.
| | - Justin A Ezekowitz
- Canadian VIGOUR Centre, Mazankowski Alberta Heart Institute, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
21
|
TNFα-stimulated protein 6 (TSG-6) reduces lung inflammation in an experimental model of bronchopulmonary dysplasia. Pediatr Res 2019; 85:390-397. [PMID: 30538263 DOI: 10.1038/s41390-018-0250-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 11/15/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND Inflammation is a key factor in the pathogenesis of bronchopulmonary dysplasia (BPD). Tumor necrosis factor-stimulated protein 6 (TSG-6) is a glycoprotein that modulates inflammation. Here we tested the hypothesis that intra-tracheal (IT) administration of an adenovirus overexpressing TSG-6 (AdTSG-6) would decrease inflammation and restore lung structure in experimental BPD. METHODS Newborn Sprague-Dawley rats exposed to normoxia (RA) or hyperoxia (85% O2) from postnatal day (P) 1-P14 were randomly assigned to receive IT AdTSG-6 or placebo (PL) on P3. The effect of IT AdTSG-6 on lung inflammation, alveolarization, angiogenesis, apoptosis, pulmonary vascular remodeling, and pulmonary hypertension were evaluated on P14. Data were analyzed by two-way ANOVA. RESULTS TSG-6 mRNA was significantly increased in pups who received IT AdTSG-6. Compared to RA, hyperoxia PL-treated pups had increased NF-kβ activation and lung inflammation. In contrast, IT AdTSG-6 hyperoxia-treated pups had decreased lung phosphorylated NF-kβ expression and markers of inflammation. This was accompanied by an improvement in alveolarization, angiogenesis, pulmonary vascular remodeling, and pulmonary hypertension. CONCLUSIONS IT AdTSG-6 decreases lung inflammation and improves lung structure in neonatal rats with experimental BPD. These findings suggest that therapies that increase lung TSG-6 expression may have beneficial effects in preterm infants with BPD.
Collapse
|
22
|
Reed MJ, Damodarasamy M, Pathan JL, Chan CK, Spiekerman C, Wight TN, Banks WA, Day AJ, Vernon RB, Keene CD. Increased Hyaluronan and TSG-6 in Association with Neuropathologic Changes of Alzheimer's Disease. J Alzheimers Dis 2019; 67:91-102. [PMID: 30507579 PMCID: PMC6398602 DOI: 10.3233/jad-180797] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Little is known about the extracellular matrix (ECM) during progression of AD pathology. Brain ECM is abundant in hyaluronan (HA), a non-sulfated glycosaminoglycan synthesized by HA synthases (HAS) 1-3 in a high molecular weight (MW) form that is degraded into lower MW fragments. We hypothesized that pathologic severity of AD is associated with increases in HA and HA-associated ECM molecules. To test this hypothesis, we assessed HA accumulation and size; HA synthases (HAS) 1-3; and the HA-stabilizing hyaladherin, TSG-6 in parietal cortex samples from autopsied research subjects with not AD (CERAD = 0, Braak = 0- II, n = 12-21), intermediate AD (CERAD = 2, Braak = III-IV, n = 13-18), and high AD (CERAD = 3, Braak = V-VI, n = 32-40) neuropathologic change. By histochemistry, HA was associated with deposits of amyloid and tau, and was also found diffusely in brain parenchyma, with overall HA quantity (measured by ELSA) significantly greater in brains with high AD neuropathology. Mean HA MW was similar among the samples. HAS2 and TSG-6 mRNA expression, and TSG-6 protein levels were significantly increased in high AD and both molecules were present in vasculature, NeuN-positive neurons, and Iba1-positive microglia. These results did not change when accounting for gender, advanced age (≥ 90 years versus <90 years), or the clinical diagnosis of dementia. Collectively, our results indicate a positive correlation between HA accumulation and AD neuropathology, and suggest a possible role for HA synthesis and metabolism in AD progression.
Collapse
Affiliation(s)
- MJ Reed
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA
| | - M Damodarasamy
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA
| | - JL Pathan
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA
| | - CK Chan
- Matrix Biology Program, Benaroya Research Institute, Virginia Mason, Seattle, WA, USA
| | - C Spiekerman
- Center for Biomedical Statistics, Institute for Translational Health Sciences, University of Washington, Seattle, WA, USA
| | - TN Wight
- Matrix Biology Program, Benaroya Research Institute, Virginia Mason, Seattle, WA, USA
| | - WA Banks
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA
- VA Puget Sound Health Care System, Geriatric Research Education and Clinical Center, Seattle, WA, USA
| | - AJ Day
- Wellcome Trust Centre for Cell-Matrix Research, Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - RB Vernon
- Matrix Biology Program, Benaroya Research Institute, Virginia Mason, Seattle, WA, USA
| | - CD Keene
- Department of Pathology, Division of Neuropathology, University of Washington, Seattle, WA, USA
| |
Collapse
|
23
|
Contributions of Glycosaminoglycans to Collagen Fiber Recruitment in Constitutive Modeling of Arterial Mechanics. J Biomech 2018; 82:211-219. [PMID: 30415914 DOI: 10.1016/j.jbiomech.2018.10.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 10/20/2018] [Accepted: 10/23/2018] [Indexed: 01/08/2023]
Abstract
The contribution of glycosaminoglycans (GAGs) to the biological and mechanical functions of biological tissue has emerged as an important area of research. GAGs provide structural basis for the organization and assembly of extracellular matrix (ECM). The mechanics of tissue with low GAG content can be indirectly affected by the interaction of GAGs with collagen fibers, which have long been known to be one of the primary contributors to soft tissue mechanics. Our earlier study showed that enzymatic GAG depletion results in straighter collagen fibers that are recruited at lower levels of stretch, and a corresponding shift in earlier arterial stiffening (Mattson et al., 2016). In this study, the effect of GAGs on collagen fiber recruitment was studied through a structure-based constitutive model. The model incorporates structural information, such as fiber orientation distribution, content, and recruitment of medial elastin, medial collagen, and adventitial collagen fibers. The model was first used to study planar biaxial tensile stress-stretch behavior of porcine descending thoracic aorta. Changes in elastin and collagen fiber orientation distribution, and collagen fiber recruitment were then incorporated into the model in order to predict the stress-stretch behavior of GAG depleted tissue. Our study shows that incorporating early collagen fiber recruitment into the model predicts the stress-stretch response of GAG depleted tissue reasonably well (rms = 0.141); considering further changes of fiber orientation distribution does not improve the predicting capability (rms = 0.149). Our study suggests an important role of GAGs in arterial mechanics that should be considered in developing constitutive models.
Collapse
|
24
|
Bano F, Tammi MI, Kang DW, Harris EN, Richter RP. Single-Molecule Unbinding Forces between the Polysaccharide Hyaluronan and Its Binding Proteins. Biophys J 2018; 114:2910-2922. [PMID: 29925027 PMCID: PMC6026378 DOI: 10.1016/j.bpj.2018.05.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/30/2018] [Accepted: 05/07/2018] [Indexed: 12/11/2022] Open
Abstract
The extracellular polysaccharide hyaluronan (HA) is ubiquitous in all vertebrate tissues, where its various functions are encoded in the supramolecular complexes and matrices that it forms with HA-binding proteins (hyaladherins). In tissues, these supramolecular architectures are frequently subjected to mechanical stress, yet how this affects the intermolecular bonding is largely unknown. Here, we used a recently developed single-molecule force spectroscopy platform to analyze and compare the mechanical strength of bonds between HA and a panel of hyaladherins from the Link module superfamily, namely the complex of the proteoglycan aggrecan and cartilage link protein, the proteoglycan versican, the inflammation-associated protein TSG-6, the HA receptor for endocytosis (stabilin-2/HARE), and the HA receptor CD44. We find that the resistance to tensile stress for these hyaladherins correlates with the size of the HA-binding domain. The lowest mean rupture forces are observed for members of the type A subgroup (i.e., with the shortest HA-binding domains; TSG-6 and HARE). In contrast, the mechanical stability of the bond formed by aggrecan in complex with cartilage link protein (two members of the type C subgroup, i.e., with the longest HA-binding domains) and HA is equal or even superior to the high affinity streptavidin⋅biotin bond. Implications for the molecular mechanism of unbinding of HA⋅hyaladherin bonds under force are discussed, which underpin the mechanical properties of HA⋅hyaladherin complexes and HA-rich extracellular matrices.
Collapse
Affiliation(s)
- Fouzia Bano
- School of Biomedical Sciences, Faculty of Biological Sciences, School of Physics and Astronomy, Faculty of Mathematics and Physical Sciences, and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom; CIC biomaGUNE, Biosurfaces Laboratory, Donostia-San Sebastian, Spain
| | - Markku I Tammi
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - David W Kang
- Halozyme Therapeutics Inc., San Diego, California
| | - Edward N Harris
- Department of Biochemistry, University of Nebraska, Lincoln, Nebraska
| | - Ralf P Richter
- School of Biomedical Sciences, Faculty of Biological Sciences, School of Physics and Astronomy, Faculty of Mathematics and Physical Sciences, and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom; CIC biomaGUNE, Biosurfaces Laboratory, Donostia-San Sebastian, Spain.
| |
Collapse
|
25
|
Lord MS, Tang F, Rnjak-Kovacina J, Smith JGW, Melrose J, Whitelock JM. The multifaceted roles of perlecan in fibrosis. Matrix Biol 2018; 68-69:150-166. [PMID: 29475023 DOI: 10.1016/j.matbio.2018.02.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/11/2022]
Abstract
Perlecan, or heparan sulfate proteoglycan 2 (HSPG2), is a ubiquitous heparan sulfate proteoglycan that has major roles in tissue and organ development and wound healing by orchestrating the binding and signaling of mitogens and morphogens to cells in a temporal and dynamic fashion. In this review, its roles in fibrosis are reviewed by drawing upon evidence from tissue and organ systems that undergo fibrosis as a result of an uncontrolled response to either inflammation or traumatic cellular injury leading to an over production of a collagen-rich extracellular matrix. This review focuses on examples of fibrosis that occurs in lung, liver, kidney, skin, kidney, neural tissues and blood vessels and its link to the expression of perlecan in that particular organ system.
Collapse
Affiliation(s)
- Megan S Lord
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia.
| | - Fengying Tang
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia
| | | | - James G W Smith
- University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - James Melrose
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia; Raymond Purves Bone and Joint Research Laboratory, Kolling Institute Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia; Sydney Medical School, Northern, The University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| | - John M Whitelock
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia
| |
Collapse
|
26
|
Watanabe R, Sato Y, Ozawa N, Takahashi Y, Koba S, Watanabe T. Emerging Roles of Tumor Necrosis Factor-Stimulated Gene-6 in the Pathophysiology and Treatment of Atherosclerosis. Int J Mol Sci 2018; 19:E465. [PMID: 29401724 PMCID: PMC5855687 DOI: 10.3390/ijms19020465] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/22/2018] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
Tumor necrosis factor-stimulated gene-6 (TSG-6) is a 35-kDa glycoprotein that has been shown to exert anti-inflammatory effects in experimental models of arthritis, acute myocardial infarction, and acute cerebral infarction. Several lines of evidence have shed light on the pathophysiological roles of TSG-6 in atherosclerosis. TSG-6 suppresses inflammatory responses of endothelial cells, neutrophils, and macrophages as well as macrophage foam cell formation and vascular smooth muscle cell (VSMC) migration and proliferation. Exogenous TSG-6 infusion and endogenous TSG-6 attenuation with a neutralizing antibody for four weeks retards and accelerates, respectively, the development of aortic atherosclerotic lesions in ApoE-deficient mice. TSG-6 also decreases the macrophage/VSMC ratio (a marker of plaque instability) and promotes collagen fibers in atheromatous plaques. In patients with coronary artery disease (CAD), plasma TSG-6 levels are increased and TSG-6 is abundantly expressed in the fibrous cap within coronary atheromatous plaques, indicating that TSG-6 increases to counteract the progression of atherosclerosis and stabilize the plaque. These findings indicate that endogenous TSG-6 enhancement and exogenous TSG-6 replacement treatments are expected to emerge as new lines of therapy against atherosclerosis and related CAD. Therefore, this review provides support for the clinical utility of TSG-6 in the diagnosis and treatment of atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Rena Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Yuki Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Nana Ozawa
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Yui Takahashi
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan.
| | - Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| |
Collapse
|
27
|
Urner S, Kelly-Goss M, Peirce SM, Lammert E. Mechanotransduction in Blood and Lymphatic Vascular Development and Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 81:155-208. [PMID: 29310798 DOI: 10.1016/bs.apha.2017.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The blood and lymphatic vasculatures are hierarchical networks of vessels, which constantly transport fluids and, therefore, are exposed to a variety of mechanical forces. Considering the role of mechanotransduction is key for fully understanding how these vascular systems develop, function, and how vascular pathologies evolve. During embryonic development, for example, initiation of blood flow is essential for early vascular remodeling, and increased interstitial fluid pressure as well as initiation of lymph flow is needed for proper development and maturation of the lymphatic vasculature. In this review, we introduce specific mechanical forces that affect both the blood and lymphatic vasculatures, including longitudinal and circumferential stretch, as well as shear stress. In addition, we provide an overview of the role of mechanotransduction during atherosclerosis and secondary lymphedema, which both trigger tissue fibrosis.
Collapse
Affiliation(s)
- Sofia Urner
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Molly Kelly-Goss
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
28
|
Ruddy JM, Akerman AW, Kimbrough D, Nadeau EK, Stroud RE, Mukherjee R, Ikonomidis JS, Jones JA. Differential hypertensive protease expression in the thoracic versus abdominal aorta. J Vasc Surg 2016; 66:1543-1552. [PMID: 28034583 DOI: 10.1016/j.jvs.2016.07.120] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 07/24/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Hypertension (HTN), which is a major risk factor for cardiovascular morbidity and mortality, can drive pathologic remodeling of the macro- and microcirculation. Patterns of aortic pathology differ, however, suggesting regional heterogeneity of the pressure-sensitive protease systems triggering extracellular matrix remodeling in the thoracic (TA) and abdominal aortas (AA). This study tested the hypothesis that the expression of two major protease systems (matrix metalloproteinases [MMPs] and cathepsins) in the TA and AA would be differentially affected with HTN. METHODS Normotensive (BPN3) mice at 14-16 weeks of age underwent implantation of osmotic infusion pumps for 28-day angiotensin II (AngII) delivery (1.46 mg/kg/day; BPN3+AngII; n = 8) to induce HTN. The TA and AA were harvested to determine levels of MMP-2, MMP-9, and membrane type 1-MMP, and cathepsins S, K, and L were evaluated in age-matched BPN3 (n = 8) control and BPH2 spontaneously hypertensive mice (non-AngII pathway; n = 7). Blood pressure was monitored via CODA tail cuff plethysmography (Kent Scientific Corporation, Torrington, Conn). Quantitative real-time polymerase chain reaction and immunoblotting/zymography were used to measure MMP and cathepsin messenger RNA expression and protein abundance, respectively. Target protease values were compared within each aortic region via analysis of variance. RESULTS Following 28 days infusion, the BPN3+AngII mice had a 17% increase in systolic blood pressure, matching that of the BPH2 spontaneously hypertensive mice (both P < .05 vs BPN3). MMP-2 gene expression demonstrated an AngII-dependent increase in the TA (P < .05), but MMP-9 was not altered with HTN. Expression of tissue inhibitor of metalloproteinases-1 was markedly increased in TA of BPN3+AngII mice, but tissue inhibitor of metalloproteinases-2 demonstrated decreased expression in the AA of both hypertensive groups (P < .05). Only cathepsin K responded to AngII-induced HTN with significant elevation in the TA of those mice, but expression of cathepsin L and cystatin C was inhibited in AA of both hypertensive groups (P < .05). Apoptotic markers were not significantly elevated in any experimental group. CONCLUSIONS By using two different models of HTN, this study has identified pressure-dependent as well as AngII-dependent regional alterations in aortic gene expression of MMPs and cathepsins that may lead to differential remodeling responses in each of the aortic regions. Further studies will delineate mechanisms and may provide targeted therapies to attenuate down-stream aortic pathology based on demonstrated regional heterogeneity.
Collapse
Affiliation(s)
- Jean Marie Ruddy
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC; Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC.
| | - Adam W Akerman
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| | - Denise Kimbrough
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| | - Elizabeth K Nadeau
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| | - Robert E Stroud
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| | - Rupak Mukherjee
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| | - John S Ikonomidis
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC; Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC
| | - Jeffrey A Jones
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
29
|
Chang YT, Chan CK, Eriksson I, Johnson PY, Cao X, Westöö C, Norvik C, Andersson-Sjöland A, Westergren-Thorsson G, Johansson S, Hedin U, Kjellén L, Wight TN, Tran-Lundmark K. Versican accumulates in vascular lesions in pulmonary arterial hypertension. Pulm Circ 2016; 6:347-59. [PMID: 27683612 DOI: 10.1086/686994] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a lethal condition for which there is no effective curative pharmacotherapy. PAH is characterized by vasoconstriction, wall thickening of pulmonary arteries, and increased vascular resistance. Versican is a chondroitin sulfate proteoglycan in the vascular extracellular matrix that accumulates following vascular injury and promotes smooth-muscle cell proliferation in systemic arteries. Here, we investigated whether versican may play a similar role in PAH. Paraffin-embedded lung sections from patients who underwent lung transplantation to treat PAH were used for immunohistochemistry. The etiologies of PAH in the subjects involved in this study were idiopathic PAH, scleroderma, and congenital heart disease (atrial septal defect) with left-to-right shunt. Independent of the underlying etiology, increased versican immunostaining was observed in areas of medial thickening, in neointima, and in plexiform lesions. Western blot of lung tissue lysates confirmed accumulation of versican in patients with PAH. Double staining for versican and CD45 showed only occasional colocalization in neointima of high-grade lesions and plexiform lesions. In vitro, metabolic labeling with [(35)S]sulfate showed that human pulmonary artery smooth-muscle cells (hPASMCs) produce mainly chondroitin sulfate glycosaminoglycans. In addition, hypoxia, but not cyclic stretch, was demonstrated to increase both versican messenger RNA expression and protein synthesis by hPASMCs. Versican accumulates in vascular lesions of PAH, and the amount of versican correlates more with lesion severity than with underlying etiology or inflammation. Hypoxia is a possible regulator of versican accumulation, which may promote proliferation of pulmonary smooth-muscle cells and vascular remodeling in PAH.
Collapse
Affiliation(s)
- Ya-Ting Chang
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Christina K Chan
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Inger Eriksson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Pamela Y Johnson
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Xiaofang Cao
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Christian Westöö
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Christian Norvik
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | - Staffan Johansson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Lena Kjellén
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Karin Tran-Lundmark
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
30
|
Moortgat P, Anthonissen M, Meirte J, Van Daele U, Maertens K. The physical and physiological effects of vacuum massage on the different skin layers: a current status of the literature. BURNS & TRAUMA 2016; 4:34. [PMID: 27660766 PMCID: PMC5027633 DOI: 10.1186/s41038-016-0053-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/23/2016] [Indexed: 11/10/2022]
Abstract
Vacuum massage is a non-invasive mechanical massage technique performed with a mechanical device that lifts the skin by means of suction, creates a skin fold and mobilises that skin fold. In the late 1970s, this therapy was introduced to treat traumatic or burn scars. Although vacuum massage was invented to treat burns and scars, one can find very little literature on the effects of this intervention. Therefore, the aim of this review is to present an overview of the available literature on the physical and physiological effects of vacuum massage on epidermal and dermal skin structures in order to find the underlying working mechanisms that could benefit the healing of burns and scars. The discussion contains translational analysis of the results and provides recommendations for future research on the topic. An extended search for publications was performed using PubMed, Web of Science and Google Scholar. Two authors independently identified and checked each study against the inclusion criteria. Nineteen articles were included in the qualitative synthesis. The two most reported physical effects of vacuum massage were improvement of the tissue hardness and the elasticity of the skin. Besides physical effects, a variety of physiological effects are reported in literature, for example, an increased number of fibroblasts and collagen fibres accompanied by an alteration of fibroblast phenotype and collagen orientation. Little information was found on the decrease of pain and itch due to vacuum massage. Although vacuum massage initially had been developed for the treatment of burn scars, this literature review found little evidence for the efficacy of this treatment. Variations in duration, amplitude or frequency of the treatment have a substantial influence on collagen restructuring and reorientation, thus implying possible beneficial influences on the healing potential by mechanotransduction pathways. Vacuum massage may release the mechanical tension associated with scar retraction and thus induce apoptosis of myofibroblasts. Suggestions for future research include upscaling the study design, investigating the molecular pathways and dose dependency, comparing effects in different stages of repair, including evolutive parameters and the use of more objective assessment tools.
Collapse
Affiliation(s)
- Peter Moortgat
- OSCARE, Organisation for Burns, Scar After-care and Research, Van Roiestraat 18, B-2170 Antwerp, Belgium
| | - Mieke Anthonissen
- OSCARE, Organisation for Burns, Scar After-care and Research, Van Roiestraat 18, B-2170 Antwerp, Belgium ; Department of Rehabilitation Sciences, KU Leuven, Tervuursevest 101, box 1500, 3001 Heverlee, Belgium
| | - Jill Meirte
- OSCARE, Organisation for Burns, Scar After-care and Research, Van Roiestraat 18, B-2170 Antwerp, Belgium ; Department of Rehabilitation Sciences and Physiotherapy, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Ulrike Van Daele
- Department of Rehabilitation Sciences and Physiotherapy, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Koen Maertens
- OSCARE, Organisation for Burns, Scar After-care and Research, Van Roiestraat 18, B-2170 Antwerp, Belgium ; Department of Clinical and Lifespan Psychology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| |
Collapse
|
31
|
Cowman MK, Schmidt TA, Raghavan P, Stecco A. Viscoelastic Properties of Hyaluronan in Physiological Conditions. F1000Res 2015; 4:622. [PMID: 26594344 PMCID: PMC4648226 DOI: 10.12688/f1000research.6885.1] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/17/2015] [Indexed: 02/01/2023] Open
Abstract
Hyaluronan (HA) is a high molecular weight glycosaminoglycan of the extracellular matrix (ECM), which is particularly abundant in soft connective tissues. Solutions of HA can be highly viscous with non-Newtonian flow properties. These properties affect the movement of HA-containing fluid layers within and underlying the deep fascia. Changes in the concentration, molecular weight, or even covalent modification of HA in inflammatory conditions, as well as changes in binding interactions with other macromolecules, can have dramatic effects on the sliding movement of fascia. The high molecular weight and the semi-flexible chain of HA are key factors leading to the high viscosity of dilute solutions, and real HA solutions show additional nonideality and greatly increased viscosity due to mutual macromolecular crowding. The shear rate dependence of the viscosity, and the viscoelasticity of HA solutions, depend on the relaxation time of the molecule, which in turn depends on the HA concentration and molecular weight. Temperature can also have an effect on these properties. High viscosity can additionally affect the lubricating function of HA solutions. Immobility can increase the concentration of HA, increase the viscosity, and reduce lubrication and gliding of the layers of connective tissue and muscle. Over time, these changes can alter both muscle structure and function. Inflammation can further increase the viscosity of HA-containing fluids if the HA is modified via covalent attachment of heavy chains derived from Inter-α-Inhibitor. Hyaluronidase hydrolyzes HA, thus reducing its molecular weight, lowering the viscosity of the extracellular matrix fluid and making outflow easier. It can also disrupt any aggregates or gel-like structures that result from HA being modified. Hyaluronidase is used medically primarily as a dispersion agent, but may also be useful in conditions where altered viscosity of the fascia is desired, such as in the treatment of muscle stiffness.
Collapse
Affiliation(s)
- Mary K Cowman
- Biomatrix Research Center, Department of Chemical and Biomolecular Engineering, Polytechnic School of Engineering, New York University, New York, NY, 10010, USA
| | - Tannin A Schmidt
- Faculty of Kinesiology & Schulich School of Engineering - Centre for Bioengineering Research & Education, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Preeti Raghavan
- Department of Rehabilitation Medicine, Rusk Rehabilitation, New York University School of Medicine, New York, NY, 10016, USA
| | - Antonio Stecco
- Department of Internal Medicine, University of Padova, Padua, 35100, Italy
| |
Collapse
|
32
|
Roccabianca S, Bellini C, Humphrey JD. Computational modelling suggests good, bad and ugly roles of glycosaminoglycans in arterial wall mechanics and mechanobiology. J R Soc Interface 2015; 11:20140397. [PMID: 24920112 DOI: 10.1098/rsif.2014.0397] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The medial layer of large arteries contains aggregates of the glycosaminoglycan hyaluronan and the proteoglycan versican. It is increasingly thought that these aggregates play important mechanical and mechanobiological roles despite constituting only a small fraction of the normal arterial wall. In this paper, we offer a new hypothesis that normal aggregates of hyaluronan and versican pressurize the intralamellar spaces, and thereby put into tension the radial elastic fibres that connect the smooth muscle cells to the elastic laminae, which would facilitate mechanosensing. This hypothesis is supported by novel computational simulations using two complementary models, a mechanistically based finite-element mixture model and a phenomenologically motivated continuum hyperelastic model. That is, the simulations suggest that normal aggregates of glycosaminoglycans/proteoglycans within the arterial media may play equally important roles in supporting (i.e. a structural role) and sensing (i.e. an instructional role) mechanical loads. Additional simulations suggest further, however, that abnormal increases in these aggregates, either distributed or localized, may over-pressurize the intralamellar units. We submit that these situations could lead to compromised mechanosensing, anoikis and/or reduced structural integrity, each of which represent fundamental aspects of arterial pathologies seen, for example, in hypertension, ageing and thoracic aortic aneurysms and dissections.
Collapse
Affiliation(s)
- S Roccabianca
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - C Bellini
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - J D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
33
|
Anderegg U, Simon JC, Averbeck M. More than just a filler - the role of hyaluronan for skin homeostasis. Exp Dermatol 2014; 23:295-303. [PMID: 24628940 DOI: 10.1111/exd.12370] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2014] [Indexed: 12/20/2022]
Abstract
In recent years, hyaluronan (HA) has become an increasingly attractive substance as a non-immunogenic filler and scaffolding material in cosmetic dermatology. Despite its wide use for skin augmentation and rejuvenation, relatively little is known about the molecular structures and interacting proteins of HA in normal and diseased skin. However, a comprehensive understanding of cutaneous HA homeostasis is required for future the development of HA-based applications for skin regeneration. This review provides an update on HA-based structures, expression, metabolism and its regulation, function and pharmacological targeting of HA in skin.
Collapse
Affiliation(s)
- Ulf Anderegg
- Department of Dermatology, Venerology and Allergology, University of Leipzig, Leipzig, Germany
| | | | | |
Collapse
|
34
|
Mandraffino G, Imbalzano E, Mamone F, Aragona C, Lo Gullo A, D'Ascola A, Alibrandi A, Cinquegrani A, Mormina E, Versace A, Basile G, Sardo M, Cinquegrani M, Carerj S, Saitta A. Biglycan expression in current cigarette smokers: A possible link between active smoking and atherogenesis. Atherosclerosis 2014; 237:471-9. [DOI: 10.1016/j.atherosclerosis.2014.10.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 10/16/2014] [Accepted: 10/17/2014] [Indexed: 01/31/2023]
|
35
|
Nichols S, Milner M, Meijer R, Carroll S, Ingle L. Variability of automated carotid intima-media thickness measurements by novice operators. Clin Physiol Funct Imaging 2014; 36:25-32. [PMID: 25216303 DOI: 10.1111/cpf.12189] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 08/15/2014] [Indexed: 02/01/2023]
Abstract
Carotid intima-media thickness (C-IMT) measurements provide a non-invasive assessment of subclinical atherosclerosis. The aim of the study was to assess the inter- and intra-observer variability of automated C-IMT measurements undertaken by two novice operators using the Panasonic CardioHealth Station. Participants were free from cardio-metabolic disease, and each underwent serial bilateral C-IMT ultrasound measurements. Immediate interoperator measurement variability was calculated by comparing initial measurements taken by two operators. Immediate retest variability was calculated from two consecutive measurements and longer term variability was assessed by conducting a further scan 1 week later. Fifty apparently healthy participants (n = 20 females), aged 26·2 ± 5·0 years, were recruited. Operator 1 recorded a median (interquartile range) right and left-sided C-IMT of 0·471 mm (0·072 mm) and 0·462 mm (0·047 mm). Female's right and left C-IMT were 0·442 mm (0·049 mm) and 0·451 mm (0·063 mm), respectively. The limits of agreement (LoA) for immediate interoperator variability were -0·063 to 0·056 mm (mean bias -0·003 mm). Operator 1's immediate retest intra-operator LoA were -0·057 to 0·046 mm (mean bias was -0·005 mm). One-week LoA were -0·057 to 0·050 mm (mean bias -0·003 mm). Operator 2 recorded median right and left-sided C-IMT of 0·467 mm (0·089 mm) and 0·458 mm (0·046 mm) for males, respectively, whilst female measurements were 0·441 mm (0·052 mm) and 0·444 mm (0·054 mm), respectively. Operator 2's intra-operator immediate retest LoA were -0·056 to 0·056 (mean bias <-0·001 mm). Intra-operator LoA at 1 week were -0·052 to 0·068 mm (mean bias 0·008 mm). Novice operators produce acceptable short-term and 1-week inter- and intra-operator C-IMT measurement variability in healthy, young to middle-aged adults using the Panasonic CardioHealth Station.
Collapse
Affiliation(s)
- S Nichols
- Department of Sport, Health & Exercise Science, University of Hull, Kingston-upon-Hull, UK
| | - M Milner
- Department of Sport, Health & Exercise Science, University of Hull, Kingston-upon-Hull, UK
| | - R Meijer
- Julius Center for Health Sciences and Primary Care, University Medical Center, Utrecht, The Netherlands
| | - S Carroll
- Department of Sport, Health & Exercise Science, University of Hull, Kingston-upon-Hull, UK
| | - L Ingle
- Department of Sport, Health & Exercise Science, University of Hull, Kingston-upon-Hull, UK
| |
Collapse
|
36
|
Kanazawa T, Nakagami G, Minematsu T, Yamane T, Huang L, Mugita Y, Noguchi H, Mori T, Sanada H. Biological responses of three-dimensional cultured fibroblasts by sustained compressive loading include apoptosis and survival activity. PLoS One 2014; 9:e104676. [PMID: 25102054 PMCID: PMC4125229 DOI: 10.1371/journal.pone.0104676] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 07/16/2014] [Indexed: 12/11/2022] Open
Abstract
Pressure ulcers are characterized by chronicity, which results in delayed wound healing due to pressure. Early intervention for preventing delayed healing due to pressure requires a prediction method. However, no study has reported the prediction of delayed healing due to pressure. Therefore, this study focused on biological response-based molecular markers for the establishment of an assessment technology to predict delayed healing due to pressure. We tested the hypothesis that sustained compressive loading applied to three dimensional cultured fibroblasts leads to upregulation of heat shock proteins (HSPs), CD44, hyaluronan synthase 2 (HAS2), and cyclooxygenase 2 (COX2) along with apoptosis via disruption of adhesion. First, sustained compressive loading was applied to fibroblast-seeded collagen sponges. Following this, collagen sponge samples and culture supernatants were collected for apoptosis and proliferation assays, gene expression analysis, immunocytochemistry, and quantification of secreted substances induced by upregulation of mRNA and protein level. Compared to the control, the compressed samples demonstrated that apoptosis was induced in a time- and load- dependent manner; vinculin and stress fiber were scarce; HSP90α, CD44, HAS2, and COX2 expression was upregulated; and the concentrations of HSP90α, hyaluronan (HA), and prostaglandin E2 (PGE2) were increased. In addition, the gene expression of antiapoptotic Bcl2 was significantly increased in the compressed samples compared to the control. These results suggest that compressive loading induces not only apoptosis but also survival activity. These observations support that HSP90α, HA, and, PGE2 could be potential molecular markers for prediction of delayed wound healing due to pressure.
Collapse
Affiliation(s)
- Toshiki Kanazawa
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Gojiro Nakagami
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeo Minematsu
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takumi Yamane
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Nutritional Sciences, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Lijuan Huang
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuko Mugita
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Noguchi
- Department of Life Support Technology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taketoshi Mori
- Department of Life Support Technology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiromi Sanada
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
37
|
The effects of vitamin E and omega-3 PUFAs on endothelial function among adolescents with metabolic syndrome. BIOMED RESEARCH INTERNATIONAL 2014; 2014:906019. [PMID: 25136638 PMCID: PMC4127288 DOI: 10.1155/2014/906019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 06/20/2014] [Indexed: 01/17/2023]
Abstract
Aim. The present study aims to explore the effects of vitamin E and omega-3 on endothelial function indicators among adolescents with metabolic syndrome. Method. In a randomized, double blind, and placebo-controlled trial, 90 young individuals, aged 10 to 18 years, with metabolic syndrome were randomly assigned to receive either vitamin E tablets (400 IU/day) or omega-3 tablets (2.4 gr/day) or placebo. For assessing endothelial functional state, the serum level of vascular endothelial growth factor (VEGF) was measured by ELISA test. Results. The use of omega-3 supplementation for eight weeks led to significant increase in serum HDL level compared with the group treated with vitamin E or placebo group. In this regard, no significant correlations were found between the change in VEGF and baseline levels of other markers including anthropometric indices and serum lipids. Omega-3 could significantly reduce VEGF with the presence of other baseline variables (Beta = −12.55; P = 0.012). Conclusion. The administration of omega-3 can effectively improve endothelial function in adolescents with metabolic syndrome by reducing the level of serum VEGF, as a major index for atherosclerosis progression and endothelial destabilization. Omega-3 can be proposed as a VEGF antagonist for improving endothelial function in metabolic syndrome. The clinical implications of our findings should be assessed in future studies.
Collapse
|
38
|
Heusch G, Libby P, Gersh B, Yellon D, Böhm M, Lopaschuk G, Opie L. Cardiovascular remodelling in coronary artery disease and heart failure. Lancet 2014; 383:1933-43. [PMID: 24831770 PMCID: PMC4330973 DOI: 10.1016/s0140-6736(14)60107-0] [Citation(s) in RCA: 562] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Remodelling is a response of the myocardium and vasculature to a range of potentially noxious haemodynamic, metabolic, and inflammatory stimuli. Remodelling is initially functional, compensatory, and adaptive but, when sustained, progresses to structural changes that become self-perpetuating and pathogenic. Remodelling involves responses not only of the cardiomyocytes, endothelium, and vascular smooth muscle cells, but also of interstitial cells and matrix. In this Review we characterise the remodelling processes in atherosclerosis, vascular and myocardial ischaemia-reperfusion injury, and heart failure, and we draw attention to potential avenues for innovative therapeutic approaches, including conditioning and metabolic strategies.
Collapse
Affiliation(s)
- Gerd Heusch
- Institut für Pathophysiologie, Universitätsklinikum Essen, Essen, Germany
| | - Peter Libby
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bernard Gersh
- Division of Cardiovascular Diseases, Mayo Clinic, and Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Derek Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Michael Böhm
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | - Gary Lopaschuk
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Lionel Opie
- Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
39
|
Thompson JC, Tang T, Wilson PG, Yoder MH, Tannock LR. Increased atherosclerosis in mice with increased vascular biglycan content. Atherosclerosis 2014; 235:71-5. [PMID: 24816040 DOI: 10.1016/j.atherosclerosis.2014.03.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 03/12/2014] [Accepted: 03/31/2014] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The response to retention hypothesis of atherogenesis proposes that atherosclerosis is initiated via the retention of atherogenic lipoproteins by vascular proteoglycans. Co-localization studies suggest that of all the vascular proteoglycans, biglycan is the one most closely co-localized with LDL. The goal of this study was to determine if over-expression of biglycan in hyperlipidemic mice would increase atherosclerosis development. METHODS Transgenic mice were developed by expressing biglycan under control of the smooth muscle actin promoter, and were crossed to the LDL receptor deficient (C57BL/6 background) atherosclerotic mouse model. Biglycan transgenic and non-transgenic control mice were fed an atherogenic Western diet for 4-12 weeks. RESULTS LDL receptor deficient mice overexpressing biglycan under control of the smooth muscle alpha actin promoter had increased atherosclerosis development that correlated with vascular biglycan content. CONCLUSION Increased vascular biglycan content predisposes to increased lipid retention and increased atherosclerosis development.
Collapse
Affiliation(s)
- Joel C Thompson
- Division of Endocrinology and Molecular Medicine, Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Tao Tang
- Division of Endocrinology and Molecular Medicine, Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Patricia G Wilson
- Division of Endocrinology and Molecular Medicine, Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Meghan H Yoder
- Division of Endocrinology and Molecular Medicine, Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Lisa R Tannock
- Division of Endocrinology and Molecular Medicine, Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA; Department of Veterans Affairs, Lexington, KY, USA.
| |
Collapse
|
40
|
Galmiche G, Pizard A, Gueret A, El Moghrabi S, Ouvrard-Pascaud A, Berger S, Challande P, Jaffe IZ, Labat C, Lacolley P, Jaisser F. Smooth muscle cell mineralocorticoid receptors are mandatory for aldosterone-salt to induce vascular stiffness. Hypertension 2014; 63:520-526. [PMID: 24296280 PMCID: PMC4446717 DOI: 10.1161/hypertensionaha.113.01967] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 10/28/2013] [Indexed: 01/12/2023]
Abstract
Arterial stiffness is recognized as a risk factor for many cardiovascular diseases. Aldosterone via its binding to and activation of the mineralocorticoid receptors (MRs) is a main regulator of blood pressure by controlling renal sodium reabsorption. Although both clinical and experimental data indicate that MR activation by aldosterone is involved in arterial stiffening, the molecular mechanism is not known. In addition to the kidney, MR is expressed in both endothelial and vascular smooth muscle cells (VSMCs), but the specific contribution of the VSMC MR to aldosterone-induced vascular stiffness remains to be explored. To address this question, we generated a mouse model with conditional inactivation of the MR in VSMC (MR(SMKO)). MR(SMKO) mice show no alteration in renal sodium handling or vascular structure, but they have decreased blood pressure when compared with control littermate mice. In vivo at baseline, large vessels of mutant mice presented with normal elastic properties, whereas carotids displayed a smaller diameter when compared with those of the control group. As expected after aldosterone/salt challenge, the arterial stiffness increased in control mice; however, it remained unchanged in MR(SMKO) mice, without significant modification in vascular collagen/elastin ratio. Instead, we found that the fibronectin/α5-subunit integrin ratio is profoundly altered in MR(SMKO) mice because the induction of α5 expression by aldosterone/salt challenge is prevented in mice lacking VSMC MR. Altogether, our data reveal in the aldosterone/salt hypertension model that MR activation specifically in VSMC leads to the arterial stiffening by modulation of cell-matrix attachment proteins independent of major vascular structural changes.
Collapse
MESH Headings
- Aldosterone/toxicity
- Animals
- Blood Pressure/drug effects
- Disease Models, Animal
- Hypertension/chemically induced
- Hypertension/metabolism
- Hypertension/physiopathology
- Male
- Mice
- Mice, Transgenic
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Receptors, Mineralocorticoid/metabolism
- Signal Transduction
- Sodium Chloride, Dietary/toxicity
- Vascular Stiffness/drug effects
Collapse
Affiliation(s)
- Guillaume Galmiche
- Centre de Recherche des Cordeliers, Université Pierre et Marie, Inserm U872 Équipe 1, Paris, France (G.G., S.E.M., F.J.); Inserm U1116, Université de Lorraine, Vandoeuvre-lès-Nancy, France (A.P., C.L., P.L.); Inserm U1096, Rouen, France (A.G., A.O.-P.); German Cancer Research Center, Heidelberg, Germany (S.B.); Université Pierre et Marie Curie, Paris 06, France (P.C.); CNRS, UMR 7190, Paris, France (P.C.); Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.); and Centre for Clinical Investigation, Inserm U9501, CHU Brabois, Vandoeuvre-lès-Nancy, France (A.P., F.J.)
| | - Anne Pizard
- Centre de Recherche des Cordeliers, Université Pierre et Marie, Inserm U872 Équipe 1, Paris, France (G.G., S.E.M., F.J.); Inserm U1116, Université de Lorraine, Vandoeuvre-lès-Nancy, France (A.P., C.L., P.L.); Inserm U1096, Rouen, France (A.G., A.O.-P.); German Cancer Research Center, Heidelberg, Germany (S.B.); Université Pierre et Marie Curie, Paris 06, France (P.C.); CNRS, UMR 7190, Paris, France (P.C.); Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.); and Centre for Clinical Investigation, Inserm U9501, CHU Brabois, Vandoeuvre-lès-Nancy, France (A.P., F.J.)
| | - Alexandre Gueret
- Centre de Recherche des Cordeliers, Université Pierre et Marie, Inserm U872 Équipe 1, Paris, France (G.G., S.E.M., F.J.); Inserm U1116, Université de Lorraine, Vandoeuvre-lès-Nancy, France (A.P., C.L., P.L.); Inserm U1096, Rouen, France (A.G., A.O.-P.); German Cancer Research Center, Heidelberg, Germany (S.B.); Université Pierre et Marie Curie, Paris 06, France (P.C.); CNRS, UMR 7190, Paris, France (P.C.); Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.); and Centre for Clinical Investigation, Inserm U9501, CHU Brabois, Vandoeuvre-lès-Nancy, France (A.P., F.J.)
| | - Soumaya El Moghrabi
- Centre de Recherche des Cordeliers, Université Pierre et Marie, Inserm U872 Équipe 1, Paris, France (G.G., S.E.M., F.J.); Inserm U1116, Université de Lorraine, Vandoeuvre-lès-Nancy, France (A.P., C.L., P.L.); Inserm U1096, Rouen, France (A.G., A.O.-P.); German Cancer Research Center, Heidelberg, Germany (S.B.); Université Pierre et Marie Curie, Paris 06, France (P.C.); CNRS, UMR 7190, Paris, France (P.C.); Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.); and Centre for Clinical Investigation, Inserm U9501, CHU Brabois, Vandoeuvre-lès-Nancy, France (A.P., F.J.)
| | - Antoine Ouvrard-Pascaud
- Centre de Recherche des Cordeliers, Université Pierre et Marie, Inserm U872 Équipe 1, Paris, France (G.G., S.E.M., F.J.); Inserm U1116, Université de Lorraine, Vandoeuvre-lès-Nancy, France (A.P., C.L., P.L.); Inserm U1096, Rouen, France (A.G., A.O.-P.); German Cancer Research Center, Heidelberg, Germany (S.B.); Université Pierre et Marie Curie, Paris 06, France (P.C.); CNRS, UMR 7190, Paris, France (P.C.); Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.); and Centre for Clinical Investigation, Inserm U9501, CHU Brabois, Vandoeuvre-lès-Nancy, France (A.P., F.J.)
| | - Stefan Berger
- Centre de Recherche des Cordeliers, Université Pierre et Marie, Inserm U872 Équipe 1, Paris, France (G.G., S.E.M., F.J.); Inserm U1116, Université de Lorraine, Vandoeuvre-lès-Nancy, France (A.P., C.L., P.L.); Inserm U1096, Rouen, France (A.G., A.O.-P.); German Cancer Research Center, Heidelberg, Germany (S.B.); Université Pierre et Marie Curie, Paris 06, France (P.C.); CNRS, UMR 7190, Paris, France (P.C.); Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.); and Centre for Clinical Investigation, Inserm U9501, CHU Brabois, Vandoeuvre-lès-Nancy, France (A.P., F.J.)
| | - Pascal Challande
- Centre de Recherche des Cordeliers, Université Pierre et Marie, Inserm U872 Équipe 1, Paris, France (G.G., S.E.M., F.J.); Inserm U1116, Université de Lorraine, Vandoeuvre-lès-Nancy, France (A.P., C.L., P.L.); Inserm U1096, Rouen, France (A.G., A.O.-P.); German Cancer Research Center, Heidelberg, Germany (S.B.); Université Pierre et Marie Curie, Paris 06, France (P.C.); CNRS, UMR 7190, Paris, France (P.C.); Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.); and Centre for Clinical Investigation, Inserm U9501, CHU Brabois, Vandoeuvre-lès-Nancy, France (A.P., F.J.)
| | - Iris Z Jaffe
- Centre de Recherche des Cordeliers, Université Pierre et Marie, Inserm U872 Équipe 1, Paris, France (G.G., S.E.M., F.J.); Inserm U1116, Université de Lorraine, Vandoeuvre-lès-Nancy, France (A.P., C.L., P.L.); Inserm U1096, Rouen, France (A.G., A.O.-P.); German Cancer Research Center, Heidelberg, Germany (S.B.); Université Pierre et Marie Curie, Paris 06, France (P.C.); CNRS, UMR 7190, Paris, France (P.C.); Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.); and Centre for Clinical Investigation, Inserm U9501, CHU Brabois, Vandoeuvre-lès-Nancy, France (A.P., F.J.)
| | - Carlos Labat
- Centre de Recherche des Cordeliers, Université Pierre et Marie, Inserm U872 Équipe 1, Paris, France (G.G., S.E.M., F.J.); Inserm U1116, Université de Lorraine, Vandoeuvre-lès-Nancy, France (A.P., C.L., P.L.); Inserm U1096, Rouen, France (A.G., A.O.-P.); German Cancer Research Center, Heidelberg, Germany (S.B.); Université Pierre et Marie Curie, Paris 06, France (P.C.); CNRS, UMR 7190, Paris, France (P.C.); Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.); and Centre for Clinical Investigation, Inserm U9501, CHU Brabois, Vandoeuvre-lès-Nancy, France (A.P., F.J.)
| | - Patrick Lacolley
- Centre de Recherche des Cordeliers, Université Pierre et Marie, Inserm U872 Équipe 1, Paris, France (G.G., S.E.M., F.J.); Inserm U1116, Université de Lorraine, Vandoeuvre-lès-Nancy, France (A.P., C.L., P.L.); Inserm U1096, Rouen, France (A.G., A.O.-P.); German Cancer Research Center, Heidelberg, Germany (S.B.); Université Pierre et Marie Curie, Paris 06, France (P.C.); CNRS, UMR 7190, Paris, France (P.C.); Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.); and Centre for Clinical Investigation, Inserm U9501, CHU Brabois, Vandoeuvre-lès-Nancy, France (A.P., F.J.)
| | - Frédéric Jaisser
- Centre de Recherche des Cordeliers, Université Pierre et Marie, Inserm U872 Équipe 1, Paris, France (G.G., S.E.M., F.J.); Inserm U1116, Université de Lorraine, Vandoeuvre-lès-Nancy, France (A.P., C.L., P.L.); Inserm U1096, Rouen, France (A.G., A.O.-P.); German Cancer Research Center, Heidelberg, Germany (S.B.); Université Pierre et Marie Curie, Paris 06, France (P.C.); CNRS, UMR 7190, Paris, France (P.C.); Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.); and Centre for Clinical Investigation, Inserm U9501, CHU Brabois, Vandoeuvre-lès-Nancy, France (A.P., F.J.)
| |
Collapse
|
41
|
Dinardo CL, Venturini G, Zhou EH, Watanabe IS, Campos LCG, Dariolli R, da Motta-Leal-Filho JM, Carvalho VM, Cardozo KHM, Krieger JE, Alencar AM, Pereira AC. Variation of mechanical properties and quantitative proteomics of VSMC along the arterial tree. Am J Physiol Heart Circ Physiol 2014; 306:H505-16. [DOI: 10.1152/ajpheart.00655.2013] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are thought to assume a quiescent and homogeneous mechanical behavior after arterial tree development phase. However, VSMCs are known to be molecularly heterogeneous in other aspects and their mechanics may play a role in pathological situations. Our aim was to evaluate VSMCs from different arterial beds in terms of mechanics and proteomics, as well as investigate factors that may influence this phenotype. VSMCs obtained from seven arteries were studied using optical magnetic twisting cytometry (both in static state and after stretching) and shotgun proteomics. VSMC mechanical data were correlated with anatomical parameters and ultrastructural images of their vessels of origin. Femoral, renal, abdominal aorta, carotid, mammary, and thoracic aorta exhibited descending order of stiffness (G, P < 0.001). VSMC mechanical data correlated with the vessel percentage of elastin and amount of surrounding extracellular matrix (ECM), which decreased with the distance from the heart. After 48 h of stretching simulating regional blood flow of elastic arteries, VSMCs exhibited a reduction in basal rigidity. VSMCs from the thoracic aorta expressed a significantly higher amount of proteins related to cytoskeleton structure and organization vs. VSMCs from the femoral artery. VSMCs are heterogeneous in terms of mechanical properties and expression/organization of cytoskeleton proteins along the arterial tree. The mechanical phenotype correlates with the composition of ECM and can be modulated by cyclic stretching imposed on VSMCs by blood flow circumferential stress.
Collapse
Affiliation(s)
- Carla Luana Dinardo
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Gabriela Venturini
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Enhua H. Zhou
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts
| | - Ii Sei Watanabe
- Institute of Biomedical Sciences, Department of Anatomy, University of São Paulo, São Paulo, Brazil
| | | | - Rafael Dariolli
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | | | | | | | - José Eduardo Krieger
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | | | | |
Collapse
|
42
|
Chow MJ, Choi M, Yun SH, Zhang Y. The effect of static stretch on elastin degradation in arteries. PLoS One 2013; 8:e81951. [PMID: 24358135 PMCID: PMC3864902 DOI: 10.1371/journal.pone.0081951] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 10/18/2013] [Indexed: 11/19/2022] Open
Abstract
Previously we have shown that gradual changes in the structure of elastin during an elastase treatment can lead to important transition stages in the mechanical behavior of arteries. However, in vivo arteries are constantly being loaded due to systolic and diastolic pressures and so understanding the effects of loading on the enzymatic degradation of elastin in arteries is important. With biaxial tensile testing, we measured the mechanical behavior of porcine thoracic aortas digested with a mild solution of purified elastase (5 U/mL) in the presence of a static stretch. Arterial mechanical properties and biochemical composition were analyzed to assess the effects of mechanical stretch on elastin degradation. As elastin is being removed, the dimensions of the artery increase by more than 20% in both the longitude and circumference directions. Elastin assays indicate a faster rate of degradation when stretch was present during the digestion. A simple exponential decay fitting confirms the time constant for digestion with stretch (0.11 ± 0.04 h(-1)) is almost twice that of digestion without stretch (0.069 ± 0.028 h(-1)). The transition from J-shaped to S-shaped stress vs. strain behavior in the longitudinal direction generally occurs when elastin content is reduced by about 60%. Multiphoton image analysis confirms the removal/fragmentation of elastin and also shows that the collagen fibers are closely intertwined with the elastin lamellae in the medial layer. After removal of elastin, the collagen fibers are no longer constrained and become disordered. Release of amorphous elastin during the fragmentation of the lamellae layers is observed and provides insights into the process of elastin degradation. Overall this study reveals several interesting microstructural changes in the extracellular matrix that could explain the resulting mechanical behavior of arteries with elastin degradation.
Collapse
Affiliation(s)
- Ming-Jay Chow
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Myunghwan Choi
- Wellman Center for Photomedicine, Harvard Medical School and Massachusetts General Hospital, Cambridge, Massachusetts, United States of America
- Graduate School of Nanoscience and Technology (WCU), Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Seok Hyun Yun
- Wellman Center for Photomedicine, Harvard Medical School and Massachusetts General Hospital, Cambridge, Massachusetts, United States of America
| | - Yanhang Zhang
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts, United States of America
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
43
|
Hutter R, Huang L, Speidl WS, Giannarelli C, Trubin P, Bauriedel G, Klotman ME, Fuster V, Badimon JJ, Klotman PE. Novel small leucine-rich repeat protein podocan is a negative regulator of migration and proliferation of smooth muscle cells, modulates neointima formation, and is expressed in human atheroma. Circulation 2013; 128:2351-63. [PMID: 24043300 DOI: 10.1161/circulationaha.113.004634] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Smooth muscle cell (SMC) migration and proliferation critically influence the clinical course of vascular disease. We tested the effect of the novel small leucine-rich repeat protein podocan on SMC migration and proliferation using a podocan-deficient mouse in combination with a model of arterial injury and aortic explant SMC culture. In addition, we examined the effect of overexpression of the human form of podocan on human SMCs and tested for podocan expression in human atherosclerosis. In all these conditions, we concomitantly evaluated the Wnt-TCF (T-cell factor) pathway. METHODS AND RESULTS Podocan was strongly and selectively expressed in arteries of wild-type mice after injury. Podocan-deficient mice showed increased arterial lesion formation compared with wild-type littermates in response to injury (P<0.05). Also, SMC proliferation was increased in arteries of podocan-deficient mice compared with wild-type (P<0.05). In vitro, migration and proliferation were increased in podocan-deficient SMCs and were normalized by transfection with the wild-type podocan gene (P<0.05). In addition, upregulation of the Wnt-TCF pathway was found in SMCs of podocan-deficient mice both in vitro and in vivo. On the other hand, podocan overexpression in human SMCs significantly reduced SMC migration and proliferation, inhibiting the Wnt-TCF pathway. Podocan and a Wnt-TCF pathway marker were differently expressed in human coronary restenotic versus primary lesions. CONCLUSIONS Podocan appears to be a potent negative regulator of the migration and proliferation of both murine and human SMCs. The lack of podocan results in excessive arterial repair and prolonged SMC proliferation, which likely is mediated by the Wnt-TCF pathway.
Collapse
Affiliation(s)
- Randolph Hutter
- Departments of Medicine and Cardiology, Mount Sinai School of Medicine, New York, NY (R.H., L.H., W.S.S., C.G., P.T., V.F., J.J.B.); Department of Cardiology, Elisabeth Klinikum, Schmalkalden, Germany (G.B.); Department of Medicine, Duke University, Durham, NC (M.E.K.); and Department of Medicine, Baylor College of Medicine, Houston, TX (P.E.K.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Understanding strain-induced collagen matrix development in engineered cardiovascular tissues from gene expression profiles. Cell Tissue Res 2013; 352:727-37. [DOI: 10.1007/s00441-013-1573-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 01/17/2013] [Indexed: 12/24/2022]
|
45
|
Selwaness M, van den Bouwhuijsen QJ, Verwoert GC, Dehghan A, Mattace-Raso FU, Vernooij M, Franco OH, Hofman A, van der Lugt A, Wentzel JJ, Witteman JC. Blood Pressure Parameters and Carotid Intraplaque Hemorrhage as Measured by Magnetic Resonance Imaging. Hypertension 2013; 61:76-81. [DOI: 10.1161/hypertensionaha.112.198267] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Mariana Selwaness
- From the Departments of Epidemiology (M.S., Q.J.A.v.d.B., G.C.V., A.D., F.U.S.M.-R., M.V., O.H.F., A.H., J.C.M.W.), Biomedical Engineering (M.S., J.J.W.), Radiology (M.S., Q.J.A.v.d.B., M.V., A.v.d.L.), and Internal Medicine (G.C.V., F.U.S.M.-R.), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Quirijn J.A. van den Bouwhuijsen
- From the Departments of Epidemiology (M.S., Q.J.A.v.d.B., G.C.V., A.D., F.U.S.M.-R., M.V., O.H.F., A.H., J.C.M.W.), Biomedical Engineering (M.S., J.J.W.), Radiology (M.S., Q.J.A.v.d.B., M.V., A.v.d.L.), and Internal Medicine (G.C.V., F.U.S.M.-R.), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Germaine C. Verwoert
- From the Departments of Epidemiology (M.S., Q.J.A.v.d.B., G.C.V., A.D., F.U.S.M.-R., M.V., O.H.F., A.H., J.C.M.W.), Biomedical Engineering (M.S., J.J.W.), Radiology (M.S., Q.J.A.v.d.B., M.V., A.v.d.L.), and Internal Medicine (G.C.V., F.U.S.M.-R.), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Abbas Dehghan
- From the Departments of Epidemiology (M.S., Q.J.A.v.d.B., G.C.V., A.D., F.U.S.M.-R., M.V., O.H.F., A.H., J.C.M.W.), Biomedical Engineering (M.S., J.J.W.), Radiology (M.S., Q.J.A.v.d.B., M.V., A.v.d.L.), and Internal Medicine (G.C.V., F.U.S.M.-R.), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Francesco U.S. Mattace-Raso
- From the Departments of Epidemiology (M.S., Q.J.A.v.d.B., G.C.V., A.D., F.U.S.M.-R., M.V., O.H.F., A.H., J.C.M.W.), Biomedical Engineering (M.S., J.J.W.), Radiology (M.S., Q.J.A.v.d.B., M.V., A.v.d.L.), and Internal Medicine (G.C.V., F.U.S.M.-R.), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Meike Vernooij
- From the Departments of Epidemiology (M.S., Q.J.A.v.d.B., G.C.V., A.D., F.U.S.M.-R., M.V., O.H.F., A.H., J.C.M.W.), Biomedical Engineering (M.S., J.J.W.), Radiology (M.S., Q.J.A.v.d.B., M.V., A.v.d.L.), and Internal Medicine (G.C.V., F.U.S.M.-R.), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Oscar H. Franco
- From the Departments of Epidemiology (M.S., Q.J.A.v.d.B., G.C.V., A.D., F.U.S.M.-R., M.V., O.H.F., A.H., J.C.M.W.), Biomedical Engineering (M.S., J.J.W.), Radiology (M.S., Q.J.A.v.d.B., M.V., A.v.d.L.), and Internal Medicine (G.C.V., F.U.S.M.-R.), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Albert Hofman
- From the Departments of Epidemiology (M.S., Q.J.A.v.d.B., G.C.V., A.D., F.U.S.M.-R., M.V., O.H.F., A.H., J.C.M.W.), Biomedical Engineering (M.S., J.J.W.), Radiology (M.S., Q.J.A.v.d.B., M.V., A.v.d.L.), and Internal Medicine (G.C.V., F.U.S.M.-R.), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Aad van der Lugt
- From the Departments of Epidemiology (M.S., Q.J.A.v.d.B., G.C.V., A.D., F.U.S.M.-R., M.V., O.H.F., A.H., J.C.M.W.), Biomedical Engineering (M.S., J.J.W.), Radiology (M.S., Q.J.A.v.d.B., M.V., A.v.d.L.), and Internal Medicine (G.C.V., F.U.S.M.-R.), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jolanda J. Wentzel
- From the Departments of Epidemiology (M.S., Q.J.A.v.d.B., G.C.V., A.D., F.U.S.M.-R., M.V., O.H.F., A.H., J.C.M.W.), Biomedical Engineering (M.S., J.J.W.), Radiology (M.S., Q.J.A.v.d.B., M.V., A.v.d.L.), and Internal Medicine (G.C.V., F.U.S.M.-R.), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jacqueline C.M. Witteman
- From the Departments of Epidemiology (M.S., Q.J.A.v.d.B., G.C.V., A.D., F.U.S.M.-R., M.V., O.H.F., A.H., J.C.M.W.), Biomedical Engineering (M.S., J.J.W.), Radiology (M.S., Q.J.A.v.d.B., M.V., A.v.d.L.), and Internal Medicine (G.C.V., F.U.S.M.-R.), Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
46
|
Mechanical properties of the extracellular matrix of the aorta studied by enzymatic treatments. Biophys J 2012; 102:1731-7. [PMID: 22768928 DOI: 10.1016/j.bpj.2012.03.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 03/08/2012] [Accepted: 03/20/2012] [Indexed: 01/24/2023] Open
Abstract
The microarchitecture of different components of the extracellular matrix (ECM) is crucial to our understanding of the properties of a tissue. In the study presented here, we used a top-down approach to understand how the interplay among different fibers determines the mechanical properties of real tissues. By selectively removing different elements of the arterial wall, we were able to measure the contribution of the different constituents of the ECM to the mechanical properties of the whole tissue. Changes in the network structure were imaged with the use of two-photon microscopy. We used an atomic force microscope to measure changes in the mechanical properties by performing nanoindentation experiments. We show that although the removal of a key element of the ECM reduced the local stiffness by up to 50 times, the remaining tissue still formed a coherent network. We also show how this method can be extended to study the effects of cells on real tissues. This new (to our knowledge) way of studying the ECM will not only help physicists gain a better understanding of biopolymers, it will be a valuable tool for biomedical researchers studying processes such as wound healing and cervix ripening.
Collapse
|
47
|
Theuma P, Fonseca VA. Inflammation, insulin resistance, and atherosclerosis. Metab Syndr Relat Disord 2012; 2:105-13. [PMID: 18370641 DOI: 10.1089/met.2004.2.105] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Until recently, atherosclerosis was thought to be a passive process of lipid deposition in the arterial wall, followed by progressive occlusion of the lumen, and finally plaque rupture and thrombosis. Recent data suggest the contrary-atherosclerosis is a dynamic process developing over many years, characterized by active uptake of lipids and smooth muscle proliferation, "molding" of plaque, and subject to the influence of many environmental and genetic factors. Central to these processes, both at initiation and propagation, are factors associated with inflammation. Insulin resistance (IR), the underlying cause of type 2 diabetes mellitus (DM), is also associated with elevated levels of inflammatory factors, such as C reactive protein (CRP), plasminogen activator inhibitor-1 (PAI-1), and fibrinogen. Recent studies indicate that these same factors precede and predict DM. These findings have led to the notion that the strong association of IR/DM with cardiovascular disease (CVD) may be through inflammation pathways. In this article, we review what is known about the association of inflammation with IR and atherosclerosis. We show that many of the same inflammatory factors associated with IR are present in atherosclerosis. We also discuss the underlying determinants of inflammation in these conditions.
Collapse
Affiliation(s)
- Pierre Theuma
- Department of Medicine, Section of Endocrinology, Tulane University Health Sciences Center, New Orleans, Louisiana
| | | |
Collapse
|
48
|
Subbotin VM. Neovascularization of coronary tunica intima (DIT) is the cause of coronary atherosclerosis. Lipoproteins invade coronary intima via neovascularization from adventitial vasa vasorum, but not from the arterial lumen: a hypothesis. Theor Biol Med Model 2012; 9:11. [PMID: 22490844 PMCID: PMC3492120 DOI: 10.1186/1742-4682-9-11] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 03/18/2012] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND An accepted hypothesis states that coronary atherosclerosis (CA) is initiated by endothelial dysfunction due to inflammation and high levels of LDL-C, followed by deposition of lipids and macrophages from the luminal blood into the arterial intima, resulting in plaque formation. The success of statins in preventing CA promised much for extended protection and effective therapeutics. However, stalled progress in pharmaceutical treatment gives a good reason to review logical properties of the hypothesis underlining our efforts, and to reconsider whether our perception of CA is consistent with facts about the normal and diseased coronary artery. ANALYSIS To begin with, it must be noted that the normal coronary intima is not a single-layer endothelium covering a thin acellular compartment, as claimed in most publications, but always appears as a multi-layer cellular compartment, or diffuse intimal thickening (DIT), in which cells are arranged in many layers. If low density lipoprotein cholesterol (LDL-C) invades the DIT from the coronary lumen, the initial depositions ought to be most proximal to blood, i.e. in the inner DIT. The facts show that the opposite is true, and lipids are initially deposited in the outer DIT. This contradiction is resolved by observing that the normal DIT is always avascular, receiving nutrients by diffusion from the lumen, whereas in CA the outer DIT is always neovascularized from adventitial vasa vasorum. The proteoglycan biglycan, confined to the outer DIT in both normal and diseased coronary arteries, has high binding capacity for LDL-C. However, the normal DIT is avascular and biglycan-LDL-C interactions are prevented by diffusion distance and LDL-C size (20 nm), whereas in CA, biglycan in the outer DIT can extract lipoproteins by direct contact with the blood. These facts lead to the single simplest explanation of all observations: (1) lipid deposition is initially localized in the outer DIT; (2) CA often develops at high blood LDL-C levels; (3) apparent CA can develop at lowered blood LDL-C levels. This mechanism is not unique to the coronary artery: for instance, the normally avascular cornea accumulates lipoproteins after neovascularization, resulting in lipid keratopathy. HYPOTHESIS Neovascularization of the normally avascular coronary DIT by permeable vasculature from the adventitial vasa vasorum is the cause of LDL deposition and CA. DIT enlargement, seen in early CA and aging, causes hypoxia of the outer DIT and induces neovascularization. According to this alternative proposal, coronary atherosclerosis is not related to inflammation and can occur in individuals with normal circulating levels of LDL, consistent with research findings.
Collapse
|
49
|
Leali D, Inforzato A, Ronca R, Bianchi R, Belleri M, Coltrini D, Di Salle E, Sironi M, Norata GD, Bottazzi B, Garlanda C, Day AJ, Presta M. Long pentraxin 3/tumor necrosis factor-stimulated gene-6 interaction: a biological rheostat for fibroblast growth factor 2-mediated angiogenesis. Arterioscler Thromb Vasc Biol 2012; 32:696-703. [PMID: 22267482 PMCID: PMC3551298 DOI: 10.1161/atvbaha.111.243998] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Angiogenesis is regulated by the balance between pro- and antiangiogenic factors and by extracellular matrix protein interactions. Fibroblast growth factor 2 (FGF2) is a major proangiogenic inducer inhibited by the interaction with the soluble pattern recognition receptor long pentraxin 3 (PTX3). PTX3 is locally coexpressed with its ligand tumor necrosis factor-stimulated gene-6 (TSG-6), a secreted glycoprotein that cooperates with PTX3 in extracellular matrix assembly. Here, we characterized the effect of TSG-6 on PTX3/FGF2 interaction and FGF2-mediated angiogenesis. METHODS AND RESULTS Solid phase binding and surface plasmon resonance assays show that TSG-6 and FGF2 bind the PTX3 N-terminal domain with similar affinity. Accordingly, TSG-6 prevents FGF2/PTX3 interaction and suppresses the inhibition exerted by PTX3 on heparan sulfate proteoglycan/FGF2/FGF receptor complex formation and on FGF2-dependent angiogenesis in vitro and in vivo. Also, endogenous PTX3 exerts an inhibitory effect on vascularization induced by FGF2 in a murine subcutaneous Matrigel plug assay, the inhibition being abolished in Ptx3-null mice or by TSG-6 treatment in wild-type animals. CONCLUSION TSG-6 reverts the inhibitory effects exerted by PTX3 on FGF2-mediated angiogenesis through competition of FGF2/PTX3 interaction. This may provide a novel mechanism to control angiogenesis in those pathological settings characterized by the coexpression of TSG-6 and PTX3, in which the relative levels of these proteins may fine-tune the angiogenic activity of FGF2.
Collapse
Affiliation(s)
- Daria Leali
- Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Wight TN, Potter-Perigo S. The extracellular matrix: an active or passive player in fibrosis? Am J Physiol Gastrointest Liver Physiol 2011; 301:G950-5. [PMID: 21512158 PMCID: PMC3233785 DOI: 10.1152/ajpgi.00132.2011] [Citation(s) in RCA: 217] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fibrosis is characterized by excessive accumulation of collagen and other extracellular matrix (ECM) components, and this process has been likened to aberrant wound healing. The early phases of wound healing involve the formation of a provisional ECM containing fibrin, fibrinogen, and fibronectin. Fibroblasts occupy this matrix and proliferate in response to activators elaborated by leukocytes that have migrated into the wound and are retained by the ECM. This coincides with the appearance of the myofibroblast, a specialized form of fibroblast whose differentiation is primarily driven by cytokines, such as transforming growth factor-β (TGF-β), and by mechanical tension. When these signals are reduced, as when TGF-β secretion is reduced, or as in scar shrinkage, myofibroblasts undergo apoptosis, resulting in a collagen-rich, cell-poor scar. Retention of myofibroblasts in fibrosis has been described as the result of imbalanced cytokine signaling, especially with respect to levels of activated TGF-β. ECM components can regulate myofibroblast persistence directly, since this phenotype is dependent on extracellular hyaluronan, tenascin-C, and the fibronectin splice variant containing the "extra domain A," and also, indirectly, through retention of TGF-β-secreting cells such as eosinophils. Thus the ECM is actively involved in both cellular and extracellular events that lead to fibrosis. Targeting components of the ECM as cells respond to injury and inflammatory stimuli holds promise as a means to avoid development of fibrosis and direct the wound-healing process toward reestablishment of a healthy equilibrium.
Collapse
Affiliation(s)
- Thomas N. Wight
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington
| | - Susan Potter-Perigo
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington
| |
Collapse
|