1
|
Zhang L, Feng Q, Kong W. ECM Microenvironment in Vascular Homeostasis: New Targets for Atherosclerosis. Physiology (Bethesda) 2024; 39:0. [PMID: 38984789 DOI: 10.1152/physiol.00028.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/05/2024] [Accepted: 03/23/2024] [Indexed: 07/11/2024] Open
Abstract
Alterations in vascular extracellular matrix (ECM) components, interactions, and mechanical properties influence both the formation and stability of atherosclerotic plaques. This review discusses the contribution of the ECM microenvironment in vascular homeostasis and remodeling in atherosclerosis, highlighting Cartilage oligomeric matrix protein (COMP) and its degrading enzyme ADAMTS7 as examples, and proposes potential avenues for future research aimed at identifying novel therapeutic targets for atherosclerosis based on the ECM microenvironment.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qianqian Feng
- 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
|
Shao X, Hou X, Zhang X, Zhang R, Zhu R, Qi H, Zheng J, Guo X, Feng R. Integrated single-cell RNA-seq analysis reveals the vital cell types and dynamic development signature of atherosclerosis. Front Physiol 2023; 14:1118239. [PMID: 37089432 PMCID: PMC10117136 DOI: 10.3389/fphys.2023.1118239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/06/2023] [Indexed: 04/03/2023] Open
Abstract
Introduction: In the development of atherosclerosis, the remodeling of blood vessels is a key process involving plaque formation and rupture. So far, most reports mainly believe that macrophages, smooth muscle cells, and endothelial cells located at the intima and media of artery play the key role in this process. Few studies had focused on whether fibroblasts located at adventitia are involved in regulating disease process.Methods and results: In this study, we conducted in-depth analysis of single-cell RNA-seq data of the total of 18 samples from healthy and atherosclerotic arteries. This study combines several analysis methods including transcription regulator network, cell-cell communication network, pseudotime trajectory, gene set enrichment analysis, and differential expression analysis. We found that SERPINF1 is highly expressed in fibroblasts and is involved in the regulation of various signaling pathways.Conclusion: Our research reveals a potential mechanism of atherosclerosis, SERPINF1 regulates the formation and rupture of plaques through the Jak-STAT signaling pathway, which may provide new insights into the pathological study of disease. Moreover, we suggest that SRGN and IGKC as potential biomarkers for unstable arterial plaques.
Collapse
Affiliation(s)
- Xiuli Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Xiuyang Hou
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Xiaolin Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ruijia Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Rongli Zhu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - He Qi
- Department of Medical Biotechnology, Liaoning Vocational College of Medicine, Shenyang, China
| | - Jianling Zheng
- Department of Medical Biotechnology, Liaoning Vocational College of Medicine, Shenyang, China
| | - Xiaoling Guo
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rui Feng
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
- *Correspondence: Rui Feng,
| |
Collapse
|
3
|
Dregoesc MI, Ţigu AB, Bekkering S, van der Heijden CDCC, Bolboacǎ SD, Joosten LAB, Visseren FLJ, Netea MG, Riksen NP, Iancu AC. Relation Between Plasma Proteomics Analysis and Major Adverse Cardiovascular Events in Patients With Stable Coronary Artery Disease. Front Cardiovasc Med 2022; 9:731325. [PMID: 35211520 PMCID: PMC8861429 DOI: 10.3389/fcvm.2022.731325] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 01/17/2022] [Indexed: 11/22/2022] Open
Abstract
Objective Despite the advances in the control of traditional risk factors, coronary artery disease (CAD) remains the greatest cause of morbidity and mortality. Our aim was to establish the relation between plasma proteomics analysis and the risk of cardiovascular events in patients with stable CAD. Materials and Methods Patients with stable CAD and documented coronary atherosclerosis were screened for inclusion. Using proximity extension assays, 177 plasma proteins were simultaneously measured. The endpoint consisted of the first major adverse cardiovascular event (MACE) and was the composite of cardiovascular death, acute coronary syndrome, stroke, transient ischemic attack, or acute limb ischemia at 18 months follow-up. Cox proportional-hazards regression with adjustment for multiple comparisons was used to identify biomarkers for the outcomes of interest. Results The cohort consisted of 229 patients. Six mediators were associated with MACE (p < 0.001). For these markers, the risk of MACE was calculated: tumor necrosis factor receptor superfamily member 13B (HR = 1.65; 95% CI: 1.30–2.10), C-C motif chemokine-3 (HR = 1.57; 95% CI: 1.23–1.98), decorin (HR = 1.65; 95% CI: 1.26–2.16), fibroblast growth factor-23 (HR = 1.56; 95% CI: 1.23–1.99), tumor necrosis factor-related apoptosis-inducing ligand-receptor 2 (TRAIL-R2) (HR = 1.61; 95% CI: 1.23–2.11), and tumor necrosis factor receptor superfamily member 10A (HR = 1.69; 95% CI: 1.25–2.29). Except for TRAIL-R2, the other proteins were associated with MACE independent of age, sex, diabetes mellitus, or estimated glomerular filtration rate. Conclusions In patients with stable CAD, five novel biomarkers were identified as independent risk factors for adverse outcomes. Novel biomarkers could represent pharmacological targets for the prevention of adverse cardiovascular events.
Collapse
Affiliation(s)
- Mihaela Ioana Dregoesc
- Department of Cardiology, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Adrian Bogdan Ţigu
- Medfuture—The Research Center for Advanced Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Siroon Bekkering
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia
- Department of Internal Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Charlotte D. C. C. van der Heijden
- Department of Internal Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Sorana Daniela Bolboacǎ
- Department of Medical Informatics and Biostatistics, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Leo A. B. Joosten
- Department of Internal Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Frank L. J. Visseren
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Niels P. Riksen
- Department of Internal Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Niels P. Riksen
| | - Adrian Corneliu Iancu
- Department of Cardiology, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- *Correspondence: Adrian Corneliu Iancu
| |
Collapse
|
4
|
Decorin counteracts disease progression in mice with recessive dystrophic epidermolysis bullosa. Matrix Biol 2018; 81:3-16. [PMID: 30528862 DOI: 10.1016/j.matbio.2018.12.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 11/20/2022]
Abstract
Loss-of-function mutations in the gene encoding type VII collagen underlie recessive dystrophic epidermolysis bullosa (RDEB), a disease characterized by skin and mucosal blistering, impaired wound healing, and diffuse dermal inflammation and fibrosis. Transforming growth factor-β signaling plays a crucial role in determining RDEB fibrotic microenvironment that leads to the development of disabling secondary disease manifestations, including hand and foot deformities. Experimental findings indicate that expression levels of decorin, a small leucine-rich proteoglycan and an endogenous TGF-β inhibitor, can modulate RDEB disease phenotype by contrasting dermal fibroblast fibrotic behavior. In this study, the ability of decorin to modify RDEB course was investigated by systemically treating RDEB mice with a lentivirus expressing human decorin. Overexpressed decorin was able to enhance survival, and to limit digit contraction and the development of paw deformities. These effects were associated with decreased TGF-β1 levels and TGF-β signaling activation. Fibrotic traits were strongly reduced in paw skin and also attenuated in the non-chronically injured back skin. However, the expression of pro-inflammatory proteins was not decreased in both paw and back skin. Our findings confirm TGF-β role in promoting fibrosis and disease progression in RDEB, and show that decorin counteracts disease manifestations by inhibiting TGF-β activation. More generally, our data indicate that modifying extracellular matrix composition is an option to improve RDEB disease course.
Collapse
|
5
|
Yang Y, Wu QH, Li Y, Gao PJ. Association of SLRPs with carotid artery atherosclerosis in essential hypertensive patients. J Hum Hypertens 2018; 32:564-571. [DOI: 10.1038/s41371-018-0077-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/03/2018] [Accepted: 05/04/2018] [Indexed: 01/26/2023]
|
6
|
Zhang W, Ge Y, Cheng Q, Zhang Q, Fang L, Zheng J. Decorin is a pivotal effector in the extracellular matrix and tumour microenvironment. Oncotarget 2018; 9:5480-5491. [PMID: 29435195 PMCID: PMC5797066 DOI: 10.18632/oncotarget.23869] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/27/2017] [Indexed: 12/02/2022] Open
Abstract
Decorin (DCN), an extracellular matrix (ECM) protein, belongs to the small leucine-rich proteoglycan family. As a pluripotent molecule, DCN regulates the bioactivities of cell growth factors and participates in ECM assembly. Accumulating evidence has shown that DCN acts as a ligand of various cytokines and growth factors by directly or indirectly interacting with the corresponding signalling molecules involved in cell growth, differentiation, proliferation, adhesion and metastasis and that DCN especially plays vital roles in cancer cell proliferation, spread, pro-inflammatory processes and anti-fibrillogenesis. The multifunctional nature of DCN thus enables it to be a potential therapeutic agent for a variety of diseases and shows good prospects for clinical and research applications. DCN, an extracellular matrix (ECM) protein that belongs to the small leucine-rich proteoglycan family, is widely distributed and plays multifunctional roles in the stroma and epithelial cells. Originally, DCN was known as an effective collagen-binding partner for fibrillogenesis [1] and to modulate key biomechanical parameters of tissue integrity in the tendon, skin and cornea [2]; thus, it was named decorin (DCN). Since being initially cloned in 1986, DCN was discovered to be a structural constituent of the ECM [3]. However, the paradigm has been shifted; it has become increasingly evident that in addition to being a matrix structural protein, DCN affects a wide range of biological processes, including cell growth, differentiation, proliferation, adhesion, spread and migration, and regulates inflammation and fibrillogenesis [4–7]. Two main themes for DCN functions have emerged: maintenance of cellular structure and regulation of signal transduction pathways, culminating in anti-tumourigenic effects. Here, we review the interaction network of DCN and emphasize the biological correlations between these interactions, some of which are expected to be therapeutic intervention targets.
Collapse
Affiliation(s)
- Wen Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Yan Ge
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qian Cheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Qi Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for The Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
7
|
Kokkinopoulos I, Wong MM, Potter CMF, Xie Y, Yu B, Warren DT, Nowak WN, Le Bras A, Ni Z, Zhou C, Ruan X, Karamariti E, Hu Y, Zhang L, Xu Q. Adventitial SCA-1 + Progenitor Cell Gene Sequencing Reveals the Mechanisms of Cell Migration in Response to Hyperlipidemia. Stem Cell Reports 2017; 9:681-696. [PMID: 28757161 PMCID: PMC5549964 DOI: 10.1016/j.stemcr.2017.06.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 01/08/2023] Open
Abstract
Adventitial progenitor cells, including SCA-1+ and mesenchymal stem cells, are believed to be important in vascular remodeling. It has been shown that SCA-1+ progenitor cells are involved in neointimal hyperplasia of vein grafts, but little is known concerning their involvement in hyperlipidemia-induced atherosclerosis. We employed single-cell sequencing technology on primary adventitial mouse SCA-1+ cells from wild-type and atherosclerotic-prone (ApoE-deficient) mice and found that a group of genes controlling cell migration and matrix protein degradation was highly altered. Adventitial progenitors from ApoE-deficient mice displayed an augmented migratory potential both in vitro and in vivo. This increased migratory ability was mimicked by lipid loading to SCA-1+ cells. Furthermore, we show that lipid loading increased miRNA-29b expression and induced sirtuin-1 and matrix metalloproteinase-9 levels to promote cell migration. These results provide direct evidence that blood cholesterol levels influence vascular progenitor cell function, which could be a potential target cell for treatment of vascular disease.
Collapse
Affiliation(s)
- Ioannis Kokkinopoulos
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Mei Mei Wong
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Claire M F Potter
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Yao Xie
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Baoqi Yu
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Derek T Warren
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Witold N Nowak
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Alexandra Le Bras
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Zhichao Ni
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Chao Zhou
- John Moorhead Research Laboratory, Centre for Nephrology, University College London, Rowland Hill Street, London NW3 2PF, UK
| | - Xiongzhong Ruan
- John Moorhead Research Laboratory, Centre for Nephrology, University College London, Rowland Hill Street, London NW3 2PF, UK
| | - Eirini Karamariti
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Yanhua Hu
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China.
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK.
| |
Collapse
|
8
|
Wilson P, Drennon K, Tannock LR. Regulation of Vascular Proteoglycan Synthesis by Metabolic Factors Associated with Diabetes. J Investig Med 2016; 55:18-25. [PMID: 17441408 DOI: 10.2310/6650.2007.05067] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Diabetes is associated with accelerated atherosclerosis, but the mechanisms responsible for this are not known. Proteoglycans have been shown to play a critical role in the initiation of atherosclerosis owing to their ability to bind and retain atherogenic lipoproteins in the artery wall. Proteoglycan structure and composition are altered in atherosclerotic lesions compared with adjacent normal regions of the artery wall, and this is exaggerated in diabetes. The purpose of this study was to determine if metabolic factors associated with diabetes lead to altered proteoglycan structure and composition. METHODS Vascular smooth muscle cells, endothelial cells, and macrophages were exposed to normal (5.6 mmol/L) or high (25 mmol/L) glucose levels, various insulin and free fatty acid levels, and the cytokines transforming growth factor beta (TGF-beta1) and platelet-derived growth factor, alone or in combination, and proteoglycan synthesis was determined. RESULTS Glucose concentrations, insulin, and free fatty acids did not alter proteoglycan synthesis, size, or relative distribution. The effect of TGF-beta to increase biglycan and versican synthesis, increase sulfate incorporation, and increase the size of the secreted proteoglycans was not altered by the ambient glucose level in the culture medium, nor did high glucose increase levels of active TGF-beta. CONCLUSION Vascular proteoglycan synthesis is not affected by metabolic factors associated with diabetes. We suggest that elevated TGF-beta levels in diabetes are responsible for the altered proteoglycan synthesis observed in diabetes.
Collapse
Affiliation(s)
- Patricia Wilson
- Division of Endocrinology and Molecular Medicine, Department of Medicine, University of Kentucky, Lexington, KY 40536-0200, USA
| | | | | |
Collapse
|
9
|
Ueda K, Yoshimura K, Yamashita O, Harada T, Morikage N, Hamano K. Possible dual role of decorin in abdominal aortic aneurysm. PLoS One 2015; 10:e0120689. [PMID: 25781946 PMCID: PMC4362951 DOI: 10.1371/journal.pone.0120689] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/25/2015] [Indexed: 02/08/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is characterized by chronic inflammation, which leads to pathological remodeling of the extracellular matrix. Decorin, a small leucine-rich repeat proteoglycan, has been suggested to regulate inflammation and stabilize the extracellular matrix. Therefore, the present study investigated the role of decorin in the pathogenesis of AAA. Decorin was localized in the aortic adventitia under normal conditions in both mice and humans. AAA was induced in mice using CaCl2 treatment. Initially, decorin protein levels decreased, but as AAA progressed decorin levels increased in all layers. Local administration of exogenous decorin prevented the development of CaCl2-induced AAA. However, decorin was highly expressed in the degenerative lesions of human AAA walls, and this expression positively correlated with matrix metalloproteinase (MMP)-9 expression. In cell culture experiments, the addition of decorin inhibited secretion of MMP-9 in vascular smooth muscle cells, but had the opposite effect in macrophages. The results suggest that decorin plays a dual role in AAA. Adventitial decorin in normal aorta may protect against the development of AAA, but macrophages expressing decorin in AAA walls may facilitate the progression of AAA by up-regulating MMP-9 secretion.
Collapse
Affiliation(s)
- Koshiro Ueda
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755–8505, Japan
| | - Koichi Yoshimura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755–8505, Japan
- Graduate School of Health and Welfare, Yamaguchi Prefectural University, Yamaguchi, 753–8502, Japan
- * E-mail:
| | - Osamu Yamashita
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755–8505, Japan
| | - Takasuke Harada
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755–8505, Japan
| | - Noriyasu Morikage
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755–8505, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755–8505, Japan
| |
Collapse
|
10
|
Moretto P, Karousou E, Viola M, Caon I, D'Angelo ML, De Luca G, Passi A, Vigetti D. Regulation of hyaluronan synthesis in vascular diseases and diabetes. J Diabetes Res 2015; 2015:167283. [PMID: 25834831 PMCID: PMC4365328 DOI: 10.1155/2015/167283] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/20/2015] [Accepted: 03/01/2015] [Indexed: 02/06/2023] Open
Abstract
Cell microenvironment has a critical role determining cell fate and modulating cell responses to injuries. Hyaluronan (HA) is a ubiquitous extracellular matrix glycosaminoglycan that can be considered a signaling molecule. In fact, interacting with several cell surface receptors can deeply shape cell behavior. In vascular biology, HA triggers smooth muscle cells (SMCs) dedifferentiation which contributes to vessel wall thickening. Furthermore, HA is able to modulate inflammation by altering the adhesive properties of endothelial cells. In hyperglycemic conditions, HA accumulates in vessels and can contribute to the diabetic complications at micro- and macrovasculature. Due to the pivotal role in favoring atherogenesis and neointima formation after injuries, HA could be a new target for cardiovascular pathologies. This review will focus on the recent findings regarding the regulation of HA synthesis in human vascular SMCs. In particular, the effects of the intracellular HA substrates availability, adenosine monophosphate-activated protein kinase (AMPK), and protein O-GlcNAcylation on the main HA synthetic enzyme (i.e., HAS2) will be discussed.
Collapse
Affiliation(s)
- Paola Moretto
- Department of Surgical and Morphological Sciences, University of Insubria, 21100 Varese, Italy
| | - Evgenia Karousou
- Department of Surgical and Morphological Sciences, University of Insubria, 21100 Varese, Italy
| | - Manuela Viola
- Department of Surgical and Morphological Sciences, University of Insubria, 21100 Varese, Italy
| | - Ilaria Caon
- Department of Surgical and Morphological Sciences, University of Insubria, 21100 Varese, Italy
| | - Maria Luisa D'Angelo
- Department of Surgical and Morphological Sciences, University of Insubria, 21100 Varese, Italy
| | - Giancarlo De Luca
- Department of Surgical and Morphological Sciences, University of Insubria, 21100 Varese, Italy
| | - Alberto Passi
- Department of Surgical and Morphological Sciences, University of Insubria, 21100 Varese, Italy
| | - Davide Vigetti
- Department of Surgical and Morphological Sciences, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
11
|
Osteoglycin deficiency does not affect atherosclerosis in mice. Atherosclerosis 2014; 237:418-25. [DOI: 10.1016/j.atherosclerosis.2014.09.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 09/16/2014] [Accepted: 09/17/2014] [Indexed: 01/04/2023]
|
12
|
Tannock LR. Vascular proteoglycans and atherosclerosis: not over yet. Atherosclerosis 2014; 237:435-6. [PMID: 25463070 DOI: 10.1016/j.atherosclerosis.2014.08.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 08/19/2014] [Indexed: 02/05/2023]
Affiliation(s)
- Lisa R Tannock
- Division of Endocrinology and Molecular Medicine, University of Kentucky, Lexington, KY 40536, USA; Department of Veterans Affairs, Lexington, KY, USA.
| |
Collapse
|
13
|
Hsieh LTH, Nastase MV, Zeng-Brouwers J, Iozzo RV, Schaefer L. Soluble biglycan as a biomarker of inflammatory renal diseases. Int J Biochem Cell Biol 2014; 54:223-35. [PMID: 25091702 DOI: 10.1016/j.biocel.2014.07.020] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 12/20/2022]
Abstract
Chronic renal inflammation is often associated with a progressive accumulation of various extracellular matrix constituents, including several members of the small leucine-rich proteoglycan (SLRP) gene family. It is becoming increasingly evident that the matrix-unbound SLRPs strongly regulate the progression of inflammation and fibrosis. Soluble SLRPs are generated either via partial proteolytic processing of collagenous matrices or by de novo synthesis evoked by stress or injury. Liberated SLRPs can then bind to and activate Toll-like receptors, thus modulating downstream inflammatory signaling. Preclinical animal models and human studies have recently identified soluble biglycan as a key initiator and regulator of various inflammatory renal diseases. Biglycan, generated by activated macrophages, can enter the circulation and its elevated levels in plasma and renal parenchyma correlate with unfavorable renal function and outcome. In this review, we will focus on the critical role of soluble biglycan in inflammatory signaling in various renal disorders. Moreover, we will provide new data implicating proinflammatory effects of soluble decorin in unilateral ureteral obstruction. Finally, we will critically evaluate the potential application of soluble biglycan vis-à-vis other SLRPs (decorin, lumican and fibromodulin) as a promising target and novel biomarker of inflammatory renal diseases.
Collapse
Affiliation(s)
- Louise Tzung-Harn Hsieh
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Madalina-Viviana Nastase
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Jinyang Zeng-Brouwers
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany.
| |
Collapse
|
14
|
Chuang CY, Degendorfer G, Davies MJ. Oxidation and modification of extracellular matrix and its role in disease. Free Radic Res 2014; 48:970-89. [DOI: 10.3109/10715762.2014.920087] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
15
|
Odorisio T, Di Salvio M, Orecchia A, Di Zenzo G, Piccinni E, Cianfarani F, Travaglione A, Uva P, Bellei B, Conti A, Zambruno G, Castiglia D. Monozygotic twins discordant for recessive dystrophic epidermolysis bullosa phenotype highlight the role of TGF-β signalling in modifying disease severity. Hum Mol Genet 2014; 23:3907-22. [DOI: 10.1093/hmg/ddu102] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
16
|
Nastase MV, Iozzo RV, Schaefer L. Key roles for the small leucine-rich proteoglycans in renal and pulmonary pathophysiology. Biochim Biophys Acta Gen Subj 2014; 1840:2460-70. [PMID: 24508120 DOI: 10.1016/j.bbagen.2014.01.035] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 01/28/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Small leucine-rich proteoglycans (SLRPs) are molecules that have signaling roles in a multitude of biological processes. In this respect, SLRPs play key roles in the evolution of a variety of diseases throughout the human body. SCOPE OF REVIEW We will critically review current developments in the roles of SLRPs in several types of disease of the kidney and lungs. Particular emphasis will be given to the roles of decorin and biglycan, the best characterized members of the SLRP gene family. MAJOR CONCLUSIONS In both renal and pulmonary disorders, SLRPs are essential elements that regulate several pathophysiological processes including fibrosis, inflammation and tumor progression. Decorin has remarkable antifibrotic and antitumorigenic properties and is considered a valuable potential treatment of these diseases. Biglycan can modulate inflammatory processes in lung and renal inflammation and is a potential target in the treatment of inflammatory conditions. GENERAL SIGNIFICANCE SLRPs can serve as either treatment targets or as potential treatment in renal or lung disease. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Madalina V Nastase
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
17
|
Hiebert PR, Boivin WA, Zhao H, McManus BM, Granville DJ. Perforin and granzyme B have separate and distinct roles during atherosclerotic plaque development in apolipoprotein E knockout mice. PLoS One 2013; 8:e78939. [PMID: 24205352 PMCID: PMC3811993 DOI: 10.1371/journal.pone.0078939] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/17/2013] [Indexed: 01/04/2023] Open
Abstract
The granzyme B/perforincytotoxic pathway is a well established mechanism of initiating target cell apoptosis. Previous studies have suggested a role for the granzyme B/perforin cytotoxic pathway in vulnerable atherosclerotic plaque formation. In the present study, granzyme B deficiency resulted in reduced atherosclerotic plaque development in the descending aortas of apolipoprotein E knockout mice fed a high fat diet for 30 weeks while perforindeficiency resulted in greater reduction in plaque development with significantly less plaque area than granzyme Bdeficient mice. In contrast to the descending aorta, no significant change in plaque size was observed in aortic roots from either granzyme Bdeficient or perforindeficient apolipoprotein E knockout mice. However, atherosclerotic plaques in the aortic roots did exhibit significantly more collagen in granzyme B, but not perforin deficient mice. Together these results suggest significant, yet separate roles for granzyme B and perforin in the pathogenesis of atherosclerosis that go beyond the traditional apoptotic pathway with additional implications in plaque development, stability and remodelling of extracellular matrix.
Collapse
Affiliation(s)
- Paul R. Hiebert
- UBC James Hogg Research Centre at the Institute for Heart + Lung Health, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wendy A. Boivin
- UBC James Hogg Research Centre at the Institute for Heart + Lung Health, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hongyan Zhao
- UBC James Hogg Research Centre at the Institute for Heart + Lung Health, St. Paul’s Hospital, Vancouver, British Columbia, Canada
| | - Bruce M. McManus
- UBC James Hogg Research Centre at the Institute for Heart + Lung Health, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - David J. Granville
- UBC James Hogg Research Centre at the Institute for Heart + Lung Health, St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
18
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
19
|
Mangat R, Warnakula S, Borthwick F, Hassanali Z, Uwiera RRE, Russell JC, Cheeseman CI, Vine DF, Proctor SD. Arterial retention of remnant lipoproteins ex vivo is increased in insulin resistance because of increased arterial biglycan and production of cholesterol-rich atherogenic particles that can be improved by ezetimibe in the JCR:LA-cp rat. J Am Heart Assoc 2012; 1:e003434. [PMID: 23316299 PMCID: PMC3541624 DOI: 10.1161/jaha.112.003434] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 08/14/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND Literature supports the "response-to-retention" hypothesis-that during insulin resistance, impaired metabolism of remnant lipoproteins can contribute to accelerated cardiovascular disease progression. We used the JCR:LA-cp rat model of metabolic syndrome (MetS) to determine the extent of arterial accumulation of intestinal-derived remnants ex vivo and potential mechanisms that contribute to exacerbated cholesterol deposition in insulin resistance. METHODS AND RESULTS Arteries from control and MetS (insulin-resistant) JCR:LA-cp rats were perfused ex vivo with Cy5-labeled remnant lipoproteins, and their arterial retention was quantified by confocal microscopy. Arterial proteoglycans were isolated from control and MetS rats at 6, 12, and 32 weeks of age. There was a significant increase in the arterial retention of remnants and in associated cholesterol accumulation in MetS rats as compared to control rats. Mechanistic studies reveal that increased cholesterol deposition is a result of greater arterial biglycan content; longer glycosaminoglycans and increased production of cholesterol-rich intestinal-derived remnants, as compared to controls. Additionally, perfusion of vessels treated with ezetimibe, alone or in combination with simvastatin, with remnants isolated from the respective treatment group reduced ex vivo arterial retention of remnant-derived cholesterol ex vivo as compared to untreated controls. CONCLUSIONS Increased progression of atherosclerotic cardiovascular disease in MetS and type 2 diabetes mellitus might be explained in part by an increase in the arterial retention of cholesterol-rich remnants. Furthermore, ezetimibe alone or in combination treatment with simvastatin could be beneficial in ameliorating atherosclerotic cardiovascular disease in insulin resistance and MetS.
Collapse
Affiliation(s)
- Rabban Mangat
- Metabolic and Cardiovascular Diseases Laboratory, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
XU YUANZHI, ZHAO KAIJUN, YANG ZHIGANG, ZHANG YUHUI, ZHANG YONGWEI, HONG BO, LIU JIANMIN. Decreased plasma decorin levels following acute ischemic stroke: Correlation with MMP-2 and differential expression in TOAST subtypes. Mol Med Rep 2012; 6:1319-24. [DOI: 10.3892/mmr.2012.1108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 08/01/2012] [Indexed: 11/06/2022] Open
|
21
|
Dynamic Reduction of Plasma Decorin Following Ischemic Stroke: A Pilot Study. Neurochem Res 2012; 37:1843-8. [DOI: 10.1007/s11064-012-0787-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/29/2012] [Accepted: 04/24/2012] [Indexed: 10/28/2022]
|
22
|
Abstract
Atherosclerotic cardiovascular disease is a major cause of morbidity and mortality in the Western world. Despite tremendous strides in understandings its pathogenesis, it still remains a challenge because of gaps in our understanding of its initiation, progression and complications leading to the clinical syndromes of angina, acute coronary syndrome, cerebrovascular disease and peripheral vascular disease. Recent studies have provided impetus on the shift from models of atherosclerosis based on cellular interactions to models where the important role of extracellular matrix is recognized. Proteoglycans, especially those belonging to the small leucine-rich proteoglycan family of which decorin is a representative example, have come under close scrutiny for their role in atherogenesis. There is evidence from in vitro and in vivo animal models as well as humans to suggest an important role of decorin in attenuating progression of atherosclerosis. Decorin distribution in different blood vessels has been shown to inversely correlate with the tendency to develop atherosclerosis. Decorin seems to interact closely with different cellular components of the plaque milieu, thereby suggesting its role in influencing atherogenesis at different steps. Here we review the current understanding of the role of decorin in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Sandeep Singla
- Division of Cardiovascular Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA.
| | | | | | | |
Collapse
|
23
|
Abstract
Research over the past 2 decades provides ample evidence that small leucine-rich proteoglycans (SLRPs; such as decorin, biglycan, fibromodulin, and lumican) of the extracellular matrix are deeply involved in the regulation of inflammatory and fibrotic renal disorders. Initial efforts in SLRP research focused on the interaction between decorin and TGF-β because it had been unequivocally demonstrated that decorin treatment exerts beneficial effects in fibrotic disorders involving TGF-β overproduction in the kidney. This was followed by a paradigm shift in our understanding of SLRP biology, with new evidence showing that in addition to their role as structural matrix components, soluble SLRPs also act as signaling molecules regulating various complex biologic processes in a molecule- and cell-specific manner. With the identification of SLRP-derived endogenous ligands of Toll-like receptors, the general question regarding the mechanisms of SLRP-derived signaling in pathogen-dependent and independent renal inflammation arose. This led to the fascinating concept of SLRPs as autonomous triggers of sterile renal inflammation in response to renal stress or injury. This review focuses on the key biologic roles of SLRPs in the normal and diseased kidney with special emphasis on newly described signaling events triggered by these proteoglycans.
Collapse
Affiliation(s)
- Liliana Schaefer
- Pharmazentrum Frankfurt, Institut fur Allgemeine Pharmakologie und Toxikologie, Klinikum der JW Goethe-Universität Frankfurt am Main, Haus 74, Z. 3.108a, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
24
|
Hageman J, Herrema H, Groen AK, Kuipers F. A role of the bile salt receptor FXR in atherosclerosis. Arterioscler Thromb Vasc Biol 2010; 30:1519-28. [PMID: 20631352 DOI: 10.1161/atvbaha.109.197897] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This study reviews current insights into the role of bile salts and bile salt receptors on the progression and regression of atherosclerosis. Bile salts have emerged as important modifiers of lipid and energy metabolism. At the molecular level, bile salts regulate lipid and energy homeostasis mainly via the bile salt receptors FXR and TGR5. Activation of FXR has been shown to improve plasma lipid profiles, whereas Fxr(-/-) mice have increased plasma triglyceride and very-low-density lipoprotein levels. Nevertheless, high-density lipoprotein cholesterol levels are increased in these mice, suggesting that FXR has both anti- and proatherosclerotic properties. Interestingly, there is increasing evidence for a role of FXR in "nonclassical" bile salt target tissues, eg, vasculature and macrophages. In these tissues, FXR has been shown to influence vascular tension and regulate the unloading of cholesterol from foam cells, respectively. Recent publications have provided insight into the antiinflammatory properties of FXR in atherosclerosis. Bile salt signaling via TGR5 might regulate energy homeostasis, which could serve as an attractive target to increase energy expenditure and weight loss. Interventions aiming to increase cholesterol turnover (eg, by bile salt sequestration) significantly improve plasma lipid profiles and diminish atherosclerosis in animal models. Bile salt metabolism and bile salt signaling pathways represent attractive therapeutic targets for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jurre Hageman
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University Medical Center Groningen, Hanzeplein 1, 9713 EZ Groningen, The Netherlands.
| | | | | | | |
Collapse
|
25
|
Merline R, Schaefer RM, Schaefer L. The matricellular functions of small leucine-rich proteoglycans (SLRPs). J Cell Commun Signal 2009; 3:323-35. [PMID: 19809894 PMCID: PMC2778586 DOI: 10.1007/s12079-009-0066-2] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2009] [Accepted: 09/02/2009] [Indexed: 12/11/2022] Open
Abstract
The small leucine-rich proteoglycans (SLRPs) are biologically active components of the extracellular matrix (ECM), consisting of a protein core with leucine rich-repeat (LRR) motifs covalently linked to glycosaminoglycan (GAG) side chains. The diversity in composition resulting from the various combinations of protein cores substituted with one or more GAG chains along with their pericellular localization enables SLRPs to interact with a host of different cell surface receptors, cytokines, growth factors, and other ECM components, leading to modulation of cellular functions. SLRPs are capable of binding to: (i) different types of collagens, thereby regulating fibril assembly, organization, and degradation; (ii) Toll-like receptors (TLRs), complement C1q, and tumor necrosis factor-alpha (TNFalpha), regulating innate immunity and inflammation; (iii) epidermal growth factor receptor (EGF-R), insulin-like growth factor receptor (IGF-IR), and c-Met, influencing cellular proliferation, survival, adhesion, migration, tumor growth and metastasis as well as synthesis of other ECM components; (iv) low-density lipoprotein receptor-related protein (LRP-1) and TGF-beta, modulating cytokine activity and fibrogenesis; and (v) growth factors such as bone morphogenic protein (BMP-4) and Wnt-I-induced secreted protein-1 (WISP-1), controlling cell proliferation and differentiation. Thus, the ability of SLRPs, as ECM components, to directly or indirectly regulate cell-matrix crosstalk, resulting in the modulation of various biological processes, aptly qualifies these compounds as matricellular proteins.
Collapse
|
26
|
Recchia AG, Filice E, Pellegrino D, Dobrina A, Cerra MC, Maggiolini M. Endothelin-1 induces connective tissue growth factor expression in cardiomyocytes. J Mol Cell Cardiol 2008; 46:352-9. [PMID: 19111553 DOI: 10.1016/j.yjmcc.2008.11.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Revised: 11/24/2008] [Accepted: 11/29/2008] [Indexed: 10/21/2022]
Abstract
Endothelin (ET)-1 is a vasoconstrictor involved in cardiovascular diseases. Connective tissue growth factor/CCN2 (CTGF) is a fibrotic mediator overexpressed in human atherosclerotic lesions, myocardial infarction, and hypertension. In different cell types CTGF regulates cell proliferation/apoptosis, migration, and extracellular matrix (ECM) accumulation and plays important roles in angiogenesis, chondrogenesis, osteogenesis, tissue repair, cancer and fibrosis. In the present study, we investigated the ET-1 signaling which triggers CTGF expression in cultured adult mouse atrial-muscle HL-1 cells used as a model system. ET-1 activated the CTGF promoter and induced CTGF expression at both mRNA and protein levels. Real-time PCR analysis revealed CTGF induction also in isolated rat heart preparations perfused with ET-1. Several intracellular signals elicited by ET-1 via ET receptors and even Epidermal Growth Factor Receptor (EGFR) contributed to the up-regulation of CTGF, including ERK activation and induction of the AP-1 components c-fos and c-jun, as also evaluated by ChIP analysis. Moreover, in cells treated with ET-1 the expression of ECM component decorin was abolished by CTGF silencing, indicating that CTGF is involved in ET-1 induced ECM accumulation not only in a direct manner but also through downstream effectors. Collectively, our data indicate that CTGF could be a mediator of the profibrotic effects of ET-1 in cardiomyocytes. CTGF inhibitors should be considered in setting a comprehensive pharmacological approach towards ET-1 induced cardiovascular diseases.
Collapse
Affiliation(s)
- Anna Grazia Recchia
- Department of Pharmaco-Biology, Cell Biology, University of Calabria, 87036 Rende (CS), Italy
| | | | | | | | | | | |
Collapse
|
27
|
Tannock LR, King VL. Proteoglycan mediated lipoprotein retention: a mechanism of diabetic atherosclerosis. Rev Endocr Metab Disord 2008; 9:289-300. [PMID: 18584330 DOI: 10.1007/s11154-008-9078-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Accepted: 05/29/2008] [Indexed: 12/25/2022]
Abstract
The response to retention hypothesis outlines the initial stages of atherosclerotic lesion formation. The central theme of the hypothesis is that proteoglycan mediated lipoprotein retention plays a critical step in the initiation of atherosclerosis development. Recent research using human arterial specimens, transgenic mouse models and molecular biology techniques have added to our understanding of atherosclerosis development, and provided experimental data in support of the response to retention hypothesis. In this review we summarize the recent data, in particular that which addresses mechanisms by which diabetes can accelerate atherosclerosis formation, with a focus on proteoglycan-mediated LDL retention.
Collapse
Affiliation(s)
- Lisa R Tannock
- Department of Veterans Affairs, Lexington, KY, 40511, USA.
| | | |
Collapse
|
28
|
Wilson PG, Thompson JC, Webb NR, de Beer FC, King VL, Tannock LR. Serum amyloid A, but not C-reactive protein, stimulates vascular proteoglycan synthesis in a pro-atherogenic manner. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:1902-10. [PMID: 18974302 DOI: 10.2353/ajpath.2008.080201] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Inflammatory markers serum amyloid A (SAA) and C-reactive protein (CRP) are predictive of cardiac disease and are proposed to play causal roles in the development of atherosclerosis, in which the retention of lipoproteins by vascular wall proteoglycans is critical. The purpose of this study was to determine whether SAA and/or CRP alters vascular proteoglycan synthesis and lipoprotein retention in a pro-atherogenic manner. Vascular smooth muscle cells were stimulated with either SAA or CRP (1 to 100 mg/L) and proteoglycans were then isolated and characterized. SAA, but not CRP, increased proteoglycan sulfate incorporation by 50 to 100% in a dose-dependent manner (P < 0.0001), increased glycosaminoglycan chain length, and increased low-density lipoprotein (LDL) binding affinity (K(d), 29 microg/ml LDL versus 90 microg/ml LDL for SAA versus control proteoglycans; P < 0.005). Furthermore, SAA up-regulated biglycan via the induction of endogenous transforming growth factor (TGF)-beta. To determine whether SAA stimulated proteoglycan synthesis in vivo, ApoE(-/-) mice were injected with an adenovirus expressing human SAA-1, a null virus, or saline. Mice that received adenovirus expressing SAA had increased TGF-beta concentrations in plasma and increased aortic biglycan content compared with mice that received either null virus or saline. Thus, SAA alters vascular proteoglycans in a pro-atherogenic manner via the stimulation of TGF-beta and may play a causal role in the development of atherosclerosis.
Collapse
Affiliation(s)
- Patricia G Wilson
- Division of Endocrinology and Molecular Medicine, University of Kentucky, Lexington, KY 40536-0200, USA
| | | | | | | | | | | |
Collapse
|
29
|
Adiponectin as an inducer of decorin synthesis in cultured vascular smooth muscle cells. Life Sci 2008; 83:447-52. [DOI: 10.1016/j.lfs.2008.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Revised: 07/14/2008] [Accepted: 07/17/2008] [Indexed: 11/21/2022]
|
30
|
He F, Zhang Q, Kuruba R, Gao X, Li J, Li Y, Gong W, Jiang Y, Xie W, Li S. Upregulation of decorin by FXR in vascular smooth muscle cells. Biochem Biophys Res Commun 2008; 372:746-51. [PMID: 18514055 DOI: 10.1016/j.bbrc.2008.05.098] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Accepted: 05/20/2008] [Indexed: 12/31/2022]
Abstract
Decorin is a member of the family of small leucine-rich proteoglycans that are present in blood vessels and synthesized by vascular smooth muscle cells (VSMCs). Decorin plays complex roles in both normal vascular physiology and the pathogenesis of various types of vascular disorders. However, the mechanisms of regulation of decorin expression in vasculature are not clearly understood. Particularly little information is available about a role of nuclear receptors in the regulation of decorin expression. In the present study, we report that activation of vascular FXR by a specific ligand resulted in upregulation of decorin at the levels of both mRNA and protein. FXR appears to induce decorin expression at a transcriptional level because (1) upregulation of decorin mRNA expression was abolished by the treatment of a transcription inhibitor, actinomycin D; and (2) decorin promoter activity was significantly increased by activation of FXR. Functional analysis of human decorin promoter identified an imperfect inverted repeat DNA motif, IR8 (-2313TGGTCAtagtgtcaTGACCT-2294), as a likely FXR-responsive element that is involved in decorin regulation.
Collapse
MESH Headings
- Cells, Cultured
- DNA-Binding Proteins/agonists
- DNA-Binding Proteins/metabolism
- Decorin
- Extracellular Matrix Proteins/genetics
- Gene Expression Regulation
- Humans
- Isoxazoles/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Proteoglycans/genetics
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/agonists
- Receptors, Cytoplasmic and Nuclear/metabolism
- Repetitive Sequences, Nucleic Acid
- Response Elements/drug effects
- Sequence Analysis, DNA
- Transcription Factors/agonists
- Transcription Factors/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Fengtian He
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, 639 Salk Hall, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Urano A, Yamamoto C, Fujiwara Y, Kaji T. [Proteoglycan as a key molecule in atherosclerosis progression: characteristics of the structure and regulation of the synthesis]. YAKUGAKU ZASSHI 2008; 128:365-75. [PMID: 18311055 DOI: 10.1248/yakushi.128.365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Proteoglycans are macromolecules comprising a core protein and one or more glycosaminoglycan side chains. The macromolecules particularly derived from vascular smooth muscle cells accumulate in atherosclerotic vascular wall and are involved in the progression of vascular lesions. However, the functions of proteoglycans depend on the type of core proteins and microstructure of glycosaminoglycan chains, suggesting importance of the regulation of proteoglycan synthesis in vascular smooth muscle cells. Although the regulation of glycosaminoglycan chain formation is not clear, core protein synthesis is regulated by growth factors/cytokines, mechanical strain, coagulation factors, and other factors. Recently, we found that adiponectin, an adipose-specific plasma protein that exhibits antiatherogenic activities, regulates proteoglycan synthesis in vascular smooth muscle cells.
Collapse
Affiliation(s)
- Akiko Urano
- Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa City, Japan
| | | | | | | |
Collapse
|
32
|
Huang F, Thompson JC, Wilson PG, Aung HH, Rutledge JC, Tannock LR. Angiotensin II increases vascular proteoglycan content preceding and contributing to atherosclerosis development. J Lipid Res 2008; 49:521-30. [DOI: 10.1194/jlr.m700329-jlr200] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
33
|
Tabas I, Williams KJ, Borén J. Subendothelial lipoprotein retention as the initiating process in atherosclerosis: update and therapeutic implications. Circulation 2007; 116:1832-44. [PMID: 17938300 DOI: 10.1161/circulationaha.106.676890] [Citation(s) in RCA: 967] [Impact Index Per Article: 56.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The key initiating process in atherogenesis is the subendothelial retention of apolipoprotein B-containing lipoproteins. Local biological responses to these retained lipoproteins, including a chronic and maladaptive macrophage- and T-cell-dominated inflammatory response, promote subsequent lesion development. The most effective therapy against atherothrombotic cardiovascular disease to date--low density lipoprotein-lowering drugs--is based on the principle that decreasing circulating apolipoprotein B lipoproteins decreases the probability that they will enter and be retained in the subendothelium. Ongoing improvements in this area include more aggressive lowering of low-density lipoprotein and other atherogenic lipoproteins in the plasma and initiation of low-density lipoprotein-lowering therapy at an earlier age in at-risk individuals. Potential future therapeutic approaches include attempts to block the interaction of apolipoprotein B lipoproteins with the specific subendothelial matrix molecules that mediate retention and to interfere with accessory molecules within the arterial wall that promote retention such as lipoprotein lipase, secretory sphingomyelinase, and secretory phospholipase A2. Although not the primary focus of this review, therapeutic strategies that target the proatherogenic responses to retained lipoproteins and that promote the removal of atherogenic components of retained lipoproteins also hold promise. The finding that certain human populations of individuals who maintain lifelong low plasma levels of apolipoprotein B lipoproteins have an approximately 90% decreased risk of coronary artery disease gives hope that our further understanding of the pathogenesis of this leading killer could lead to its eradication.
Collapse
Affiliation(s)
- Ira Tabas
- Department of Medicine, Columbia University Medical Center, 630 W 168th St, New York, NY 10032, USA.
| | | | | |
Collapse
|
34
|
Hultgårdh-Nilsson A, Durbeej M. Role of the extracellular matrix and its receptors in smooth muscle cell function: implications in vascular development and disease. Curr Opin Lipidol 2007; 18:540-5. [PMID: 17885425 DOI: 10.1097/mol.0b013e3282ef77e9] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE OF REVIEW Cardiovascular disease affects millions of people worldwide, while the sarcoglycan deficient cardiomyopathies are rare disorders. One important common feature, however, is the vascular smooth muscle. Here we focus on the roles of extracellular matrix components and their receptors in the functions of vascular smooth muscle cells. RECENT FINDINGS Recent observations highlight the importance of integrins and the dystrophin-glycoprotein complex in development and cardiomyopathy. For example, integrin alpha4 and alpha7 subunits are important for distributing vascular smooth muscle cells during blood vessel development. Studies on delta-sarcoglycan deficient animals have revealed abnormal vascular smooth muscle proliferation and apoptosis. Furthermore, data suggest that perlecan, by affecting smooth muscle cell proliferation, participates in the atherosclerotic process. Overexpression of decorin leads to reduced progression of atherosclerosis and thrombospondin-1 has been implicated in regulation of smooth muscle cell contractility via inhibition of nitric oxide. Novel findings on versican suggest that the binding of versican to fibulin is of great importance for regulating smooth muscle cell function. SUMMARY By regulating migration, proliferation and apoptosis as well as extracellular matrix synthesis and assembly, proteoglycans, integrins and the dystrophin-glycoprotein complex may be of great importance both during development and in vascular disease.
Collapse
Affiliation(s)
- Anna Hultgårdh-Nilsson
- Vessel Wall Biology Unit, Sweden bMuscle Biology Unit, University of Lund, Lund, Sweden.
| | | |
Collapse
|
35
|
Williams KJ, Qiu G, Usui HK, Dunn SR, McCue P, Bottinger E, Iozzo RV, Sharma K. Decorin deficiency enhances progressive nephropathy in diabetic mice. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:1441-50. [PMID: 17884968 PMCID: PMC2043506 DOI: 10.2353/ajpath.2007.070079] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Decorin, a proteoglycan that inhibits active transforming growth factor-beta, is increased in diabetic nephropathy; however, its functional significance is unclear. In this study, we used low-dose streptozotocin to induce type 1 diabetes in wild-type (C57BL/6J Dcn(+/+)), Dcn(-/-), and Dcn(+/-) mice and studied the mice for up to 1 year of diabetes. Decorin gene dose had no effect on severity of diabetes; however, the Dcn(-/-) diabetic mice died significantly earlier than nondiabetic controls (57 versus 7.3% mortality). In contrast to wild-type diabetic mice, which failed to develop significant nephropathy, the Dcn(-/-) diabetic mice developed a significant increase in albuminuria and plasma creatinine and a concurrent decrease in circulating adiponectin levels. Interestingly, adiponectin levels at 6 months of diabetes were predictive of mortality in diabetic mice. Dcn(-/-) diabetic mice exhibited advanced glomerular lesions, including diffuse mesangial matrix accumulation and fibrin cap formation. By immunohistochemistry, Dcn(-/-) diabetic mice exhibited significant increases in glomerular transforming growth factor-beta, type I collagen, macrophage infiltration, and Nox4. We conclude that decorin is a natural protective factor against diabetic nephropathy and that the Dcn(-/-) diabetic mouse is a useful new model of progressive diabetic nephropathy.
Collapse
Affiliation(s)
- Kevin Jon Williams
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Yu W, Braz JC, Dutton AM, Prusakov P, Rekhter M. In vivo imaging of atherosclerotic plaques in apolipoprotein E deficient mice using nonlinear microscopy. JOURNAL OF BIOMEDICAL OPTICS 2007; 12:054008. [PMID: 17994896 DOI: 10.1117/1.2800337] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Structural proteins such as elastin and collagen can be readily imaged by using two-photon excitation and second-harmonic generation microscopic techniques, respectively, without physical or biochemical processing of the tissues. This time- and effort-saving advantage makes these imaging techniques convenient for determining the structural characteristics of blood vessels in vivo. Fibrillar collagen is a well-known element involved in the formation of atherosclerotic lesions. It is also an important component of the fibrous cap responsible for structural stability of atherosclerotic plaques. High resolution in vivo microscopic imaging and characterization of atherosclerotic lesions in animal models can be particularly useful for drug discovery. However, it is hindered by the limitations of regular microscope objectives to gain access of the tissues of interest and motional artifacts. We report a technique that facilitates in vivo microscopic imaging of carotid arteries of rodents using conventional microscope objectives, and at the same time avoids motional artifacts. As a result, collagen, elastin, leukocytes, cell nuclei, and neutral lipids can be visualized in three dimensions in live animals. We present and discuss in vivo imaging results using a flow cessation mouse model of accelerated atherosclerosis.
Collapse
Affiliation(s)
- Weiming Yu
- Indiana University School of Medicine, Department of Medicine, Nephrology Division, Indianapolis, Indiana 46202, USA.
| | | | | | | | | |
Collapse
|
37
|
Abstract
In the past several years, remarkable progress has been made in the understanding of the mechanisms of premature aging. These rare, genetic conditions offer valuable insights into the normal aging process and the complex biology of cardiovascular disease. Many of these advances have been made in the most dramatic of these disorders, Hutchinson–Gilford progeria syndrome. Although characterized by features of normal aging such as alopecia, skin wrinkling, and osteoporosis, patients with Hutchinson–Gilford progeria syndrome are affected by accelerated, premature arteriosclerotic disease that leads to heart attacks and strokes at a mean age of 13 years. In this review, we highlight recent advances in the biology of premature aging uncovered in Hutchinson–Gilford progeria syndrome and other accelerated aging syndromes, advances that provide insight into the mechanisms of cardiovascular diseases ranging from atherosclerosis to arrhythmias.
Collapse
Affiliation(s)
- Brian C Capell
- Genome Technology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892-2486, USA
| | | | | |
Collapse
|
38
|
Zschenker O, Illies T, Ameis D. Overexpression of lysosomal acid lipase and other proteins in atherosclerosis. J Biochem 2006; 140:23-38. [PMID: 16877765 DOI: 10.1093/jb/mvj137] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis is one of the major causes of morbidity and mortality in the western world. The existing data of elevated expression levels of proteins like DNA damage and DNA repair enzymes in human atherosclerotic plaques are reviewed. From the literature, the effect of overexpression of different proteins using adenoviral vectors or the model of transgenic mice on the development of atherosclerosis will be discussed. Special focus is placed on the lysosomal acid lipase (LAL), because LAL connects extra-cellular with intra-cellular lipid metabolism and is the only hydrolase for cleavage of cholesteryl esters delivered to the lysosomes. Patients with a deficiency of LAL show an accumulation of lipids in the cells and develop pre-mature atherosclerosis. To answer the question of the influence of LAL in atherosclerosis if overexpressed, we show for the first time data of transgenic mice overexpressing LAL and the effect on the lipid level.
Collapse
Affiliation(s)
- Oliver Zschenker
- Medical Center, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | | | | |
Collapse
|