1
|
Waugh CM, Mousavizadeh R, Lee J, Screen HRC, Scott A. Mild hypercholesterolemia impacts achilles sub-tendon mechanical properties in young rats. BMC Musculoskelet Disord 2023; 24:282. [PMID: 37046262 PMCID: PMC10091839 DOI: 10.1186/s12891-023-06375-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Hypercholesterolemia is associated with tendon pathology, but the reasons underpinning this relationship are not well understood. Cholesterol can accumulate in the tendon non-collagenous matrix which may affect both global and local tissue mechanics. Changes to the local strain environment within tendon may have significant implications for mechanosensitive tenocytes. Here, we investigated the association between elevated blood cholesterol and presence of tendon lipids in the Achilles tendon. We expected lipids to be localised in the proteoglycan-rich inter-sub-tendon matrix (ISTM), therefore we also sought to examine the impact of this on the biomechanical and viscoelastic properties of the ISTM. METHODS The Achilles tendons of 32 young wild-type (SD) and 32 apolipoprotein E knock-out rats (ApoE-/-) were harvested at 15.6 ± 2.3 weeks of age. 32 specimens underwent histological examination to assess the distribution of lipids throughout sub-tendons and ISTM. The remaining specimens were prepared for biomechanical testing, where the ISTM between the gastrocnemius and soleus sub-tendons was subjected to shear load mechanical testing. A sub-set of tests were video recorded to enable a strain analysis. RESULTS ApoE-/- serum cholesterol was double that of SD rats (mean 2.25 vs. 1.10 mg/ml, p < 0.001) indicating a relatively mild hypercholesterolemia phenotype. Nonetheless, we found histological evidence of esterified lipids in the ISTM and unesterified lipids in the sub-tendons, although the location or intensity of staining was not appreciably different between rat strains. Despite a lack of observable histological differences in lipid content between groups, there were significant differences in the mechanical and viscoelastic behaviour of the Achilles sub-tendon matrix. CONCLUSION Even slightly elevated cholesterol may result in subtle changes to tendon biomechanical properties and hence injury risk. The young age of our cohort and the mild phenotype of our ApoE-/- rats are likely to have limited our findings and so we also conclude that the ApoE-/- rat model is not well suited for investigating the biomechanical impact of tendon xanthomas on Achilles sub-tendon function.
Collapse
Affiliation(s)
- Charlie M Waugh
- Dept. Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver BC, Canada.
- School of Engineering and Materials Science, Queen Mary, University of London, London, U.K..
| | - Rouhollah Mousavizadeh
- Dept. Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver BC, Canada
| | - Jenny Lee
- Dept. Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver BC, Canada
| | - Hazel R C Screen
- School of Engineering and Materials Science, Queen Mary, University of London, London, U.K
| | - Alexander Scott
- Dept. Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver BC, Canada
| |
Collapse
|
2
|
Xiang P, Blanchard V, Francis GA. Smooth Muscle Cell—Macrophage Interactions Leading to Foam Cell Formation in Atherosclerosis: Location, Location, Location. Front Physiol 2022; 13:921597. [PMID: 35795646 PMCID: PMC9251363 DOI: 10.3389/fphys.2022.921597] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Cholesterol-overloaded cells or “foam cells” in the artery wall are the biochemical hallmark of atherosclerosis, and are responsible for much of the growth, inflammation and susceptibility to rupture of atherosclerotic lesions. While it has previously been thought that macrophages are the main contributor to the foam cell population, recent evidence indicates arterial smooth muscle cells (SMCs) are the source of the majority of foam cells in both human and murine atherosclerosis. This review outlines the timeline, site of appearance and proximity of SMCs and macrophages with lipids in human and mouse atherosclerosis, and likely interactions between SMCs and macrophages that promote foam cell formation and removal by both cell types. An understanding of these SMC-macrophage interactions in foam cell formation and regression is expected to provide new therapeutic targets to reduce the burden of atherosclerosis for the prevention of coronary heart disease, stroke and peripheral vascular disease.
Collapse
|
3
|
Mao L, Yin R, Yang L, Zhao D. Role of advanced glycation end products on vascular smooth muscle cells under diabetic atherosclerosis. Front Endocrinol (Lausanne) 2022; 13:983723. [PMID: 36120471 PMCID: PMC9470882 DOI: 10.3389/fendo.2022.983723] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease and leading cause of cardiovascular diseases. The progression of AS is a multi-step process leading to high morbidity and mortality. Hyperglycemia, dyslipidemia, advanced glycation end products (AGEs), inflammation and insulin resistance which strictly involved in diabetes are closely related to the pathogenesis of AS. A growing number of studies have linked AGEs to AS. As one of the risk factors of cardiac metabolic diseases, dysfunction of VSMCs plays an important role in AS pathogenesis. AGEs are increased in diabetes, participate in the occurrence and progression of AS through multiple molecular mechanisms of vascular cell injury. As the main functional cells of vascular, vascular smooth muscle cells (VSMCs) play different roles in each stage of atherosclerotic lesions. The interaction between AGEs and receptor for AGEs (RAGE) accelerates AS by affecting the proliferation and migration of VSMCs. In addition, increasing researches have reported that AGEs promote osteogenic transformation and macrophage-like transformation of VSMCs, and affect the progression of AS through other aspects such as autophagy and cell cycle. In this review, we summarize the effect of AGEs on VSMCs in atherosclerotic plaque development and progression. We also discuss the AGEs that link AS and diabetes mellitus, including oxidative stress, inflammation, RAGE ligands, small noncoding RNAs.
Collapse
Affiliation(s)
| | | | | | - Dong Zhao
- *Correspondence: Longyan Yang, ; Dong Zhao,
| |
Collapse
|
4
|
Lipid accumulation and novel insight into vascular smooth muscle cells in atherosclerosis. J Mol Med (Berl) 2021; 99:1511-1526. [PMID: 34345929 DOI: 10.1007/s00109-021-02109-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 06/03/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022]
Abstract
Atherosclerosis is a chronic and progressive process. It is the most important pathological basis of cardiovascular disease and stroke. Vascular smooth muscle cells (VSMCs) are an essential cell type in atherosclerosis. Previous studies have revealed that VSMCs undergo phenotypic transformation in atherosclerosis to participate in the retention of atherogenic lipoproteins as well as the formation of the fibrous cap and the underlying necrotic core in plaques. The emergence of lineage-tracing studies indicates that the function and number of VSMCs in plaques have been greatly underestimated. In addition, recent studies have revealed that VSMCs make up at least 50% of the foam cell population in human and mouse atherosclerotic lesions. Therefore, understanding the formation of lipid-loaded VSMCs and their regulatory mechanisms is critical to elucidate the pathogenesis of atherosclerosis and to explore potential therapeutic targets. Moreover, combination of many complementary technologies such as lineage tracing, single-cell RNA sequencing (scRNA-seq), flow cytometry, and mass cytometry (CyTOF) with immunostaining has been performed to further understand the complex VSMC function. Correct identification of detrimental and beneficial processes may reveal successful therapeutic treatments targeting VSMCs and their derivatives during atherosclerosis. The purpose of this review is to summarize the process of lipid-loaded VSMC formation in atherosclerosis and to describe novel insight into VSMCs gained by using multiple advanced methods.
Collapse
|
5
|
Shrestha P, Adepu S, Vivès RR, Masri RE, Klooster A, Kaptein F, Dam W, Bakker SJL, van Goor H, van de Sluis B, van den Born J. Hypercholesterolemia in Progressive Renal Failure Is Associated with Changes in Hepatic Heparan Sulfate - PCSK9 Interaction. J Am Soc Nephrol 2021; 32:1371-1388. [PMID: 33758009 PMCID: PMC8259657 DOI: 10.1681/asn.2020091376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 02/04/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Dyslipidemia is an important risk factor in CKD. The liver clears triglyceride-rich lipoproteins (TRL) via LDL receptor (LDLR), LDLR-related protein-1 (LRP-1), and heparan sulfate proteoglycans (HSPGs), mostly syndecan-1. HSPGs also facilitate LDLR degradation by proprotein convertase subtilisin/kexin type 9 (PCSK9). Progressive renal failure affects the structure and activity of hepatic lipoprotein receptors, PCSK9, and plasma cholesterol. METHODS Uninephrectomy- and aging-induced CKD in normotensive Wistar rats and hypertensive Munich-Wistar-Frömter (MWF) rats. RESULTS Compared with 22-week-old sex- and strain-matched rats, 48-week-old uninephrectomized Wistar-CKD and MWF-CKD rats showed proteinuria, increased plasma creatinine, and hypercholesterolemia (all P<0.05), which were most apparent in hypertensive MWF-CKD rats. Hepatic PCSK9 expression increased in both CKD groups (P<0.05), with unusual sinusoidal localization, which was not seen in 22-week-old rats. Heparan sulfate (HS) disaccharide analysis, staining with anti-HS mAbs, and mRNA expression of HS polymerase exostosin-1 (Ext-1), revealed elongated HS chains in both CKD groups. Solid-phase competition assays showed that the PCSK9 interaction with heparin-albumin (HS-proteoglycan analogue) was critically dependent on polysaccharide chain length. VLDL binding to HS from CKD livers was reduced (P<0.05). Proteinuria and plasma creatinine strongly associated with plasma cholesterol, PCSK9, and HS changes. CONCLUSIONS Progressive CKD induces hepatic HS elongation, leading to increased interaction with PCSK9. This might reduce hepatic lipoprotein uptake and thereby induce dyslipidemia in CKD. Therefore, PCSK9/HS may be a novel target to control dyslipidemia.
Collapse
Affiliation(s)
- Pragyi Shrestha
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Saritha Adepu
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Romain R. Vivès
- University Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS), Commissariat à l’Energie Atomique (CEA), Institute of Structural Biology, Grenoble, France
| | - Rana El Masri
- University Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS), Commissariat à l’Energie Atomique (CEA), Institute of Structural Biology, Grenoble, France
| | - Astrid Klooster
- Department of Pathology, Pathology Friesland, Leeuwarden, The Netherlands
| | - Fleur Kaptein
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wendy Dam
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J. L. Bakker
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Harry van Goor
- Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bart van de Sluis
- Department Pediatrics, Section of Molecular Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jacob van den Born
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Steffen HLM, Anderson JLC, Poot ML, Lei Y, Connelly MA, Bakker SJL, Öörni K, Tietge UJF. Proteoglycan binding as proatherogenic function metric of apoB-containing lipoproteins and chronic kidney graft failure. J Lipid Res 2021; 62:100083. [PMID: 33939983 PMCID: PMC8173310 DOI: 10.1016/j.jlr.2021.100083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 04/14/2021] [Indexed: 12/31/2022] Open
Abstract
Lipoprotein-proteoglycan binding is an early key event in atherosclerotic lesion formation and thus conceivably could play a major role in vasculopathy-driven chronic graft failure and cardiovascular mortality in renal transplant recipients. The present study investigated whether lipoprotein-proteoglycan binding susceptibility (LPBS) of apoB-containing lipoproteins and levels of the classical atherosclerosis biomarker LDL-C were associated with cardiovascular mortality (n = 130) and graft failure (n = 73) in 589 renal transplant recipients who were followed up from at least 1 year after transplantation for 9.5 years. At baseline, LPBS was significantly higher in patients who subsequently developed graft failure than in those with a surviving graft (1.68 ± 0.93 vs. 1.46 ± 0.49 nmol/mmol, P = 0.001). Cox regression analysis showed an association between LPBS and chronic graft failure in an age- and sex-adjusted model (hazard ratio: 1.45; 95% CI, 1.14-1.85; P = 0.002), but no association was observed with cardiovascular mortality. LDL-C levels were not associated with graft failure or cardiovascular mortality. This study shows that measurement of cholesterol retention outperformed the traditionally used quantitative parameter of LDL-C levels in predicting graft failure, suggesting a higher relevance of proatherogenic function than the quantity of apoB-containing lipoproteins in chronic kidney graft failure.
Collapse
Affiliation(s)
- Hannah L M Steffen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Josephine L C Anderson
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Margot L Poot
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yu Lei
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, NC, USA
| | - Stephan J L Bakker
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland; Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Uwe J F Tietge
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
7
|
Allahverdian S, Ortega C, Francis GA. Smooth Muscle Cell-Proteoglycan-Lipoprotein Interactions as Drivers of Atherosclerosis. Handb Exp Pharmacol 2020; 270:335-358. [PMID: 33340050 DOI: 10.1007/164_2020_364] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In humans, smooth muscle cells (SMCs) are the main cell type in the artery medial layer, in pre-atherosclerotic diffuse thickening of the intima, and in all stages of atherosclerotic lesion development. SMCs secrete the proteoglycans responsible for the initial binding and retention of atherogenic lipoproteins in the artery intima, with this retention driving foam cell formation and subsequent stages of atherosclerosis. In this chapter we review current knowledge of the extracellular matrix generated by SMCs in medial and intimal arterial layers, their relationship to atherosclerotic lesion development and stabilization, how these findings correlate with mouse models of atherosclerosis, and potential therapies aimed at targeting the SMC matrix-lipoprotein interaction for atherosclerosis prevention.
Collapse
Affiliation(s)
- Sima Allahverdian
- Department of Medicine, Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Carleena Ortega
- Department of Medicine, Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Gordon A Francis
- Department of Medicine, Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
8
|
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: 606] [Impact Index Per Article: 121.2] [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
|
9
|
Wight TN. A role for proteoglycans in vascular disease. Matrix Biol 2018; 71-72:396-420. [PMID: 29499356 PMCID: PMC6110991 DOI: 10.1016/j.matbio.2018.02.019] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 12/15/2022]
Abstract
The content of proteoglycans (PGs) is low in the extracellular matrix (ECM) of vascular tissue, but increases dramatically in all phases of vascular disease. Early studies demonstrated that glycosaminoglycans (GAGs) including chondroitin sulfate (CS), dermatan sulfate (DS), keratan sulfate (KS) and heparan sulfate (HS) accumulate in vascular lesions in both humans and in animal models in areas of the vasculature that are susceptible to disease initiation (such as at branch points) and are frequently coincident with lipid deposits. Later studies showed the GAGs were covalently attached to specific types of core proteins that accumulate in vascular lesions. These molecules include versican (CSPG), biglycan and decorin (DS/CSPGs), lumican and fibromodulin (KSPGs) and perlecan (HSPG), although other types of PGs are present, but in lesser quantities. While the overall molecular design of these macromolecules is similar, there is tremendous structural diversity among the different PG families creating multiple forms that have selective roles in critical events that form the basis of vascular disease. PGs interact with a variety of different molecules involved in disease pathogenesis. For example, PGs bind and trap serum components that accumulate in vascular lesions such as lipoproteins, amyloid, calcium, and clotting factors. PGs interact with other ECM components and regulate, in part, ECM assembly and turnover. PGs interact with cells within the lesion and alter the phenotypes of both resident cells and cells that invade the lesion from the circulation. A number of therapeutic strategies have been developed to target specific PGs involved in key pathways that promote vascular disease. This review will provide a historical perspective of this field of research and then highlight some of the evidence that defines the involvement of PGs and their roles in the pathogenesis of vascular disease.
Collapse
Affiliation(s)
- Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, United States.
| |
Collapse
|
10
|
Afroz R, Cao Y, Rostam MA, Ta H, Xu S, Zheng W, Osman N, Kamato D, Little PJ. Signalling pathways regulating galactosaminoglycan synthesis and structure in vascular smooth muscle: Implications for lipoprotein binding and atherosclerosis. Pharmacol Ther 2018; 187:88-97. [DOI: 10.1016/j.pharmthera.2018.02.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
11
|
Brito V, Mellal K, Zoccal KF, Soto Y, Ménard L, Sarduy R, Faccioli LH, Ong H, Vázquez AM, Marleau S. Atheroregressive Potential of the Treatment with a Chimeric Monoclonal Antibody against Sulfated Glycosaminoglycans on Pre-existing Lesions in Apolipoprotein E-Deficient Mice. Front Pharmacol 2017; 8:782. [PMID: 29163168 PMCID: PMC5672559 DOI: 10.3389/fphar.2017.00782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/16/2017] [Indexed: 02/02/2023] Open
Abstract
The retention of lipoprotein particles in the intima, in particular to glycosaminoglycan side chains of proteoglycans, is a critical step in atherosclerosis initiation. Administration of chP3R99, a chimeric mouse/human monoclonal antibody inducing an anti-idiotypic network response against glycosaminoglycans was previously shown to prevent atherosclerotic lesion progression, yet its effect in the late-stage progression of lesions remains unknown. This study investigated the effect of chP3R99 at a late stage of disease development in apolipoprotein E-deficient mice and the vascular mechanisms involved. Male apolipoprotein E-deficient mice were fed a high-fat high-cholesterol diet from 4 to 19 weeks old, at which time mice were fed normal chow and 5 doses of chP3R99 (50 μg) or isotype-matched IgG (hR3) were administered subcutaneously weekly for the first 3 administrations, then at weeks 24 and 26 before sacrifice (week 28). Lesions progression was reduced by 88% in treated mice with no change in total plasma cholesterol levels, yet with increased sera reactivity to chP3R99 idiotype and heparin, suggesting the induction of an anti-idiotype antibody cascade against glycosaminoglycans, which was likely related with the atheroprotective effect. chP3R99 treatment initiated regression in a significant number of mice. Circulating levels of interleukin-6 were reduced along with a striking diminution of inflammatory cell accumulation in the vessel wall, and of VCAM-1 labeling in vivo. The ratio of IL-10/iNOS gene expression in aortas increased in chP3R99-treated mice. In conclusion, our results show that treatment with chP3R99 reduces vascular inflammatory burden and halts lesion progression with potential for regression in the late phase of the disease in atherosclerotic mice, and support the therapeutic intervention against glycosaminoglycans as a novel strategy to reverse atherosclerosis.
Collapse
Affiliation(s)
- Victor Brito
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada.,Division of Immunobiology, Center of Molecular Immunology, Havana, Cuba
| | - Katia Mellal
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Karina F Zoccal
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada.,Department of Clinical Analysis, Toxicology and Bromatology, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Yosdel Soto
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada.,Division of Immunobiology, Center of Molecular Immunology, Havana, Cuba
| | - Liliane Ménard
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Roger Sarduy
- Division of Immunobiology, Center of Molecular Immunology, Havana, Cuba
| | - Lucia H Faccioli
- Department of Clinical Analysis, Toxicology and Bromatology, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Huy Ong
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Ana M Vázquez
- Division of Immunobiology, Center of Molecular Immunology, Havana, Cuba
| | - Sylvie Marleau
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
12
|
Dubland JA, Francis GA. So Much Cholesterol: the unrecognized importance of smooth muscle cells in atherosclerotic foam cell formation. Curr Opin Lipidol 2016; 27:155-61. [PMID: 26836481 DOI: 10.1097/mol.0000000000000279] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Smooth muscle cells (SMCs) form the thickened intimal layer in atherosclerosis-prone arteries in early life, and provide the initial site for retention and uptake of atherogenic lipoproteins. Here we review current knowledge regarding the importance of SMCs in the deposition of cholesterol in atherosclerotic plaque. RECENT FINDINGS SMCs were found to comprise at least 50% of total foam cells in human coronary artery atherosclerosis, and exhibit a selective loss of expression of the cholesterol efflux promoter ATP-binding cassette transporter A1. Cholesterol loading induced a loss of SMC gene expression and an increase in macrophage and proinflammatory marker expression by cultured mouse and human arterial SMCs, with reversal of these effects upon removal of the excess cholesterol. Mice engineered to track all cells of SMC lineage indicated that, at most, SMCs make up about one-third of total cells in atherosclerotic plaque in these animals. SUMMARY SMCs appear to be the origin of the majority of foam cells in human atherosclerotic plaque. Recent studies suggest a renaissance of research on the role of SMCs in atherosclerosis is needed to make the next leap forward in the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Joshua A Dubland
- Division of Endocrinology and Metabolism, Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
13
|
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
|
14
|
Bowler MA, Merryman WD. In vitro models of aortic valve calcification: solidifying a system. Cardiovasc Pathol 2015; 24:1-10. [PMID: 25249188 PMCID: PMC4268061 DOI: 10.1016/j.carpath.2014.08.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 07/21/2014] [Accepted: 08/07/2014] [Indexed: 12/21/2022] Open
Abstract
Calcific aortic valve disease (CAVD) affects 25% of people over 65, and the late-stage stenotic state can only be treated with total valve replacement, requiring 85,000 surgeries annually in the US alone (University of Maryland Medical Center, 2013, http://umm.edu/programs/services/heart-center-programs/cardiothoracic-surgery/valve-surgery/facts). As CAVD is an age-related disease, many of the affected patients are unable to undergo the open-chest surgery that is its only current cure. This challenge motivates the elucidation of the mechanisms involved in calcification, with the eventual goal of alternative preventative and therapeutic strategies. There is no sufficient animal model of CAVD, so we turn to potential in vitro models. In general, in vitro models have the advantages of shortened experiment time and better control over multiple variables compared to in vivo models. As with all models, the hypothesis being tested dictates the most important characteristics of the in vivo physiology to recapitulate. Here, we collate the relevant pieces of designing and evaluating aortic valve calcification so that investigators can more effectively draw significant conclusions from their results.
Collapse
Affiliation(s)
- Meghan A Bowler
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212.
| |
Collapse
|
15
|
Usman A, Ribatti D, Sadat U, Gillard JH. From Lipid Retention to Immune-Mediate Inflammation and Associated Angiogenesis in the Pathogenesis of Atherosclerosis. J Atheroscler Thromb 2015; 22:739-49. [DOI: 10.5551/jat.30460] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Ammara Usman
- University Department of Radiology, Cambridge University Hospitals NHS Foundation Trust
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, National Cancer Institute “Giovanni Paolo II”
| | - Umar Sadat
- Cambridge Vascular Unit, Cambridge University Hospitals NHS Foundation Trust
| | - Jonathan H Gillard
- University Department of Radiology, Cambridge University Hospitals NHS Foundation Trust
| |
Collapse
|
16
|
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: 1.5] [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]
|
17
|
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: 2.9] [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
|
18
|
Fratta Pasini A, Stranieri C, Pasini A, Vallerio P, Mozzini C, Solani E, Cominacini M, Cominacini L, Garbin U. Lysophosphatidylcholine and carotid intima-media thickness in young smokers: a role for oxidized LDL-induced expression of PBMC lipoprotein-associated phospholipase A2? PLoS One 2013; 8:e83092. [PMID: 24358251 PMCID: PMC3866188 DOI: 10.1371/journal.pone.0083092] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 11/08/2013] [Indexed: 12/13/2022] Open
Abstract
Background Although cigarette smoking has been associated with carotid intima-media thickness (CIMT) the mechanisms are yet not completely known. Lysophosphatidylcholine (lysoPC), a main product of lipoprotein-associated phospholipase A2 (Lp-PLA2) activity, appears to be a major determinant of the pro-atherogenic properties of oxidized LDL (oxLDL) and to induce proteoglycan synthesis, a main player in intimal thickening. In this study we assessed whether cigarette smoking-induced oxidative stress may influence plasma Lp-PLA2 and lysoPC and Lp-PLA2 expression in peripheral blood mononuclear cells (PBMC), as well as the relationship between lysoPC and CIMT. Methods/Results 45 healthy smokers and 45 age and sex-matched subjects participated in this study. Smokers, compared to non-smokers, showed increased plasma concentrations of oxLDL, Lp-PLA2 and lysoPC together with up-regulation of Lp-PLA2 (mRNA and protein) expression in PBMC (P<0.001). Plasma Lp-PLA2 positively correlated with both lysoPC (r=0.639, P<0.001) and PBMC mRNA Lp-PLA2 (r=0.484, P<0.001) in all subjects. Moreover CIMT that was higher in smokers (P<0.001), positively correlated with lysoPC (r=0.55, P<0.001). Then in invitro study we demonstrated that both oxLDL (at concentrations similar to those found in smoker’s serum) and oxidized phospholipids contained in oxLDL, were able to up-regulate mRNA Lp-PLA2 in PBMC. This effect was likely due, at least in part, to the enrichment in oxidized phospholipids found in PBMC after exposure to oxLDL. Our results also showed that in human aortic smooth muscle cells lysoPC, at concentrations similar to those found in smokers, increased the expression of biglycan and versican, two main proteoglycans. Conclusions In smokers a further effect of raised oxidative stress is the up-regulation of Lp-PLA2 expression in PBMC with subsequent increase of plasma Lp-PLA2 and lysoPC. Moreover the correlation between lysoPC and CIMT together with the finding that lysoPC up-regulates proteoglycan synthesis suggests that lysoPC may be a link between smoking and intimal thickening.
Collapse
Affiliation(s)
- Anna Fratta Pasini
- Section of Internal Medicine D, Department of Medicine, University of Verona, Verona, Italy
- * E-mail:
| | - Chiara Stranieri
- Section of Internal Medicine D, Department of Medicine, University of Verona, Verona, Italy
| | - Andrea Pasini
- Section of Internal Medicine D, Department of Medicine, University of Verona, Verona, Italy
| | - Paola Vallerio
- Section of Internal Medicine D, Department of Medicine, University of Verona, Verona, Italy
| | - Chiara Mozzini
- Section of Internal Medicine D, Department of Medicine, University of Verona, Verona, Italy
| | - Erika Solani
- Section of Internal Medicine D, Department of Medicine, University of Verona, Verona, Italy
| | - Mattia Cominacini
- Section of Internal Medicine D, Department of Medicine, University of Verona, Verona, Italy
| | - Luciano Cominacini
- Section of Internal Medicine D, Department of Medicine, University of Verona, Verona, Italy
| | - Ulisse Garbin
- Section of Internal Medicine D, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
19
|
Kaga E, Karademir B, Baykal AT, Ozer NK. Identification of differentially expressed proteins in atherosclerotic aorta and effect of vitamin E. J Proteomics 2013; 92:260-73. [DOI: 10.1016/j.jprot.2013.06.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 06/06/2013] [Accepted: 06/09/2013] [Indexed: 10/26/2022]
|
20
|
de Gonzalo-Calvo D, Revuelta-López E, Llorente-Cortés V. [Basic mechanisms. Regulation and clearance of lipoproteins that contain apolipoprotein B]. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2013; 25:194-200. [PMID: 23768652 DOI: 10.1016/j.arteri.2013.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 05/17/2013] [Indexed: 06/02/2023]
Affiliation(s)
- David de Gonzalo-Calvo
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, Barcelona, España
| | | | | |
Collapse
|
21
|
Song R, Zeng Q, Ao L, Yu JA, Cleveland JC, Zhao KS, Fullerton DA, Meng X. Biglycan Induces the Expression of Osteogenic Factors in Human Aortic Valve Interstitial Cells via Toll-Like Receptor-2. Arterioscler Thromb Vasc Biol 2012; 32:2711-20. [DOI: 10.1161/atvbaha.112.300116] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Rui Song
- From the Department of Surgery, University of Colorado Denver, Aurora, CO (R.S., Q.Z., L.A., J.A.Y., J.C.C., D.A.F., X.M.); and Department of Pathophysiology (R.S., K.Z.), Guangdong Key Laboratory of Shock and Microcirculation Research, and Department of Cardiology (Q.Z.), Laboratory for Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingchun Zeng
- From the Department of Surgery, University of Colorado Denver, Aurora, CO (R.S., Q.Z., L.A., J.A.Y., J.C.C., D.A.F., X.M.); and Department of Pathophysiology (R.S., K.Z.), Guangdong Key Laboratory of Shock and Microcirculation Research, and Department of Cardiology (Q.Z.), Laboratory for Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lihua Ao
- From the Department of Surgery, University of Colorado Denver, Aurora, CO (R.S., Q.Z., L.A., J.A.Y., J.C.C., D.A.F., X.M.); and Department of Pathophysiology (R.S., K.Z.), Guangdong Key Laboratory of Shock and Microcirculation Research, and Department of Cardiology (Q.Z.), Laboratory for Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jessica A. Yu
- From the Department of Surgery, University of Colorado Denver, Aurora, CO (R.S., Q.Z., L.A., J.A.Y., J.C.C., D.A.F., X.M.); and Department of Pathophysiology (R.S., K.Z.), Guangdong Key Laboratory of Shock and Microcirculation Research, and Department of Cardiology (Q.Z.), Laboratory for Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Joseph C. Cleveland
- From the Department of Surgery, University of Colorado Denver, Aurora, CO (R.S., Q.Z., L.A., J.A.Y., J.C.C., D.A.F., X.M.); and Department of Pathophysiology (R.S., K.Z.), Guangdong Key Laboratory of Shock and Microcirculation Research, and Department of Cardiology (Q.Z.), Laboratory for Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke-seng Zhao
- From the Department of Surgery, University of Colorado Denver, Aurora, CO (R.S., Q.Z., L.A., J.A.Y., J.C.C., D.A.F., X.M.); and Department of Pathophysiology (R.S., K.Z.), Guangdong Key Laboratory of Shock and Microcirculation Research, and Department of Cardiology (Q.Z.), Laboratory for Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - David A. Fullerton
- From the Department of Surgery, University of Colorado Denver, Aurora, CO (R.S., Q.Z., L.A., J.A.Y., J.C.C., D.A.F., X.M.); and Department of Pathophysiology (R.S., K.Z.), Guangdong Key Laboratory of Shock and Microcirculation Research, and Department of Cardiology (Q.Z.), Laboratory for Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xianzhong Meng
- From the Department of Surgery, University of Colorado Denver, Aurora, CO (R.S., Q.Z., L.A., J.A.Y., J.C.C., D.A.F., X.M.); and Department of Pathophysiology (R.S., K.Z.), Guangdong Key Laboratory of Shock and Microcirculation Research, and Department of Cardiology (Q.Z.), Laboratory for Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Röck K, Meusch M, Fuchs N, Tigges J, Zipper P, Fritsche E, Krutmann J, Homey B, Reifenberger J, Fischer JW. Estradiol protects dermal hyaluronan/versican matrix during photoaging by release of epidermal growth factor from keratinocytes. J Biol Chem 2012; 287:20056-69. [PMID: 22493503 DOI: 10.1074/jbc.m112.353151] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Hyaluronan (HA) and versican are key components of the dermis and are responsive to ultraviolet (UV)B-induced remodeling. The aim of this study was to explore the molecular mechanisms mediating the effects of estrogen (E(2)) on HA-rich extracellular matrix during photoaging. Hairless skh-1 mice were irradiated with UVB (three times, 1 minimal erythema dose (80 mJ/cm(2)), weekly) for 10 weeks, and endogenous sex hormone production was abrogated by ovariectomy. Subcutaneous substitution of E(2) by means of controlled-release pellets caused a strong increase in the dermal HA content in both irradiated and nonirradiated skin. The increase in dermal HA correlated with induction of HA synthase HAS3 by E(2). Expression of splice variant 2 of the HA-binding proteoglycan versican was also increased by E(2). In search of candidate mediators of these effects, it was found that E(2) strongly induced the expression of epidermal growth factor (EGF) in UVB-irradiated epidermis in vivo and in keratinocytes in vitro. EGF in turn up-regulated the expression of HAS3 and versican V2 in dermal fibroblasts. HAS3 knockdown by shRNA caused inhibition of fibroblast proliferation. Furthermore, HAS3 and versican V2 induction by E(2) correlated positively with proliferation in vivo. In addition, the accumulation of inflammatory macrophages, expression of inducible cyclooxygenase 2, as well as proinflammatory monocyte chemotactic protein 1 were decreased in response to E(2) in the dermis. Collectively, these data suggest that E(2) treatment increases the amount of dermal HA and versican V2 via paracrine release of EGF, which may be implicated in the pro-proliferative and anti-inflammatory effects of E(2) during photoaging.
Collapse
Affiliation(s)
- Katharina Röck
- Institut für Pharmakologie and Klinische Pharmakologie, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Chang MY, Chan CK, Braun KR, Green PS, O'Brien KD, Chait A, Day AJ, Wight TN. Monocyte-to-macrophage differentiation: synthesis and secretion of a complex extracellular matrix. J Biol Chem 2012; 287:14122-35. [PMID: 22351750 PMCID: PMC3340194 DOI: 10.1074/jbc.m111.324988] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Although monocyte- and macrophage-derived molecules are known to promote extracellular matrix (ECM) disruption and destabilization, it is less appreciated that they also synthesize molecules contributing to ECM formation, stabilization, and function. We have identified and characterized the synthesis of proteoglycans and related proteins, some not previously known to be associated with macrophages. Proteoglycan extracts of [35S]sulfate- and 35S-trans amino acid-radiolabeled culture media from THP-1 monocytes induced to differentiate by treatment with phorbol myristate acetate revealed three major proteins of ∼25, 90, and 100 kDa following chondroitin ABC lyase digestion. The 25-kDa protein was predominant for monocytes, whereas the 90- and 100-kDa proteins were predominant for macrophages. Tandem mass spectrometry identified (i) the 25-kDa core protein as serglycin, (ii) the 90-kDa core protein as inter-α-inhibitor heavy chain 2 (IαIHC2), and (iii) the 100-kDa core as amyloid precursor-like protein 2 (APLP2). Differentiation was also associated with (i) a >500-fold increase in mRNA for TNF-stimulated gene-6, an essential cofactor for heavy chain-mediated matrix stabilization; (ii) a >800-fold increase in mRNA for HAS2, which is responsible for hyaluronan synthesis; and (iii) a 3-fold increase in mRNA for versican, which interacts with hyaluronan. Biochemical evidence is also presented for an IαIHC2-APLP2 complex, and immunohistochemical staining of human atherosclerotic lesions demonstrates similar staining patterns for APLP2 and IαIHC2 with macrophages, whereas serglycin localizes to the underlying glycosaminoglycan-rich region. These findings indicate that macrophages synthesize many of the molecules participating in ECM formation and function, suggesting a novel role for these molecules in the differentiation of macrophages in the development of atherosclerosis.
Collapse
Affiliation(s)
- Mary Y Chang
- Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Fogelstrand P, Borén J. Retention of atherogenic lipoproteins in the artery wall and its role in atherogenesis. Nutr Metab Cardiovasc Dis 2012; 22:1-7. [PMID: 22176921 DOI: 10.1016/j.numecd.2011.09.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 09/27/2011] [Indexed: 02/07/2023]
Abstract
AIMS In this review, we discuss the mechanisms behind the binding of low-density lipoproteins (LDL) to the arterial wall and how this interaction might be targeted to prevent atherosclerosis. DATA SYNTHESIS An increasing body of evidence shows that accumulation of LDL in the vessel wall is a critical step in the development of atherosclerosis. The retained lipoproteins subsequently provoke an inflammatory response that ultimately leads to atherosclerosis. In the arterial wall, LDL binds ionically to proteoglycans in the extracellular matrix. In particular, proteoglycans with elongated glycosaminoglycan (GAG) chains seem to play a crucial role in this process. CONCLUSIONS The LDL-proteoglycan interaction is a highly regulated process that might provide new therapeutic targets against cardiovascular disease.
Collapse
Affiliation(s)
- P Fogelstrand
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, University of Gothenburg, 41345 Gothenburg, Sweden.
| | | |
Collapse
|
25
|
Merrilees MJ, Beaumont BW, Braun KR, Thomas AC, Kang I, Hinek A, Passi A, Wight TN. Neointima formed by arterial smooth muscle cells expressing versican variant V3 is resistant to lipid and macrophage accumulation. Arterioscler Thromb Vasc Biol 2011; 31:1309-16. [PMID: 21441139 DOI: 10.1161/atvbaha.111.225573] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Extracellular matrix (ECM) of neointima formed following angioplasty contains elevated levels of versican, loosely arranged collagen, and fragmented deposits of elastin, features associated with lipid and macrophage accumulation. ECM with a low versican content, compact structure, and increased elastic fiber content can be achieved by expression of versican variant V3, which lacks chondroitin sulfate glycosaminoglycans. We hypothesized that V3-expressing arterial smooth muscle cells (ASMC) can be used to form a neointima resistant to lipid and macrophage accumulation associated with hypercholesterolemia. METHODS AND RESULTS ASMC transduced with V3 cDNA were seeded into ballooned rabbit carotid arteries, and animals were fed a chow diet for 4 weeks, followed by a cholesterol-enriched diet for 4 weeks, achieving plasma cholesterol levels of 20 to 25 mmol/L. V3 neointimae at 8 weeks were compact, multilayered, and elastin enriched. They were significantly thinner (57%) than control neointimae; contained significantly more elastin (118%), less collagen (22%), and less lipid (76%); and showed significantly reduced macrophage infiltration (85%). Mechanistic studies demonstrated that oxidized low-density lipoprotein stimulated the formation of a monocyte-binding ECM, which was inhibited in the presence of V3 expressing ASMC. CONCLUSION These results demonstrate that expression of V3 in vessel wall creates an elastin-rich neointimal matrix that in the presence of hyperlipidemia is resistant to lipid deposition and macrophage accumulation.
Collapse
Affiliation(s)
- Mervyn J Merrilees
- Department of Anatomy with Radiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Yan J, Stringer SE, Hamilton A, Charlton-Menys V, Götting C, Müller B, Aeschlimann D, Alexander MY. Decorin GAG synthesis and TGF-β signaling mediate Ox-LDL-induced mineralization of human vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2011; 31:608-15. [PMID: 21205989 DOI: 10.1161/atvbaha.110.220749] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Decorin and oxidized low-density lipoprotein (Ox-LDL) independently induce osteogenic differentiation of vascular smooth muscle cells (VSMCs). We aimed to determine whether decorin glycosaminoglycan (GAG) chain synthesis contributes to Ox-LDL-induced differentiation and calcification of human VSMCs in vitro. METHODS AND RESULTS Human VSMCs treated with Ox-LDL to induce oxidative stress showed increased alkaline phosphatase (ALP) activity, accelerated mineralization, and a difference in both decorin GAG chain biosynthesis and CS/DS structure compared with untreated controls. Ox-LDL increased mRNA abundance of both xylosyltransferase (XT)-I, the key enzyme responsible for GAG chain biosynthesis and Msx2, a marker of osteogenic differentiation. Furthermore, downregulation of XT-I expression using small interfering RNA blocked Ox-LDL-induced VSMC mineralization. Adenoviral-mediated overexpression of decorin, but not a mutated unglycanated form, accelerated mineralization of VSMCs, suggesting GAG chain addition on decorin is crucial for the process of differentiation. The decorin-induced VSMC osteogenic differentiation involved activation of the transforming growth factor (TGF)-β pathway, because it was attenuated by blocking of TGF-β receptor signaling and because decorin overexpression potentiated phosphorylation of the downstream signaling molecule smad2. CONCLUSIONS These studies provide direct evidence that oxidative stress-mediated decorin GAG chain synthesis triggers TGF-β signaling and mineralization of VSMCs in vitro.
Collapse
Affiliation(s)
- Jianyun Yan
- Cardiovascular Research Group, University of Manchester, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Anggraeni VY, Emoto N, Yagi K, Mayasari DS, Nakayama K, Izumikawa T, Kitagawa H, Hirata KI. Correlation of C4ST-1 and ChGn-2 expression with chondroitin sulfate chain elongation in atherosclerosis. Biochem Biophys Res Commun 2011; 406:36-41. [DOI: 10.1016/j.bbrc.2011.01.096] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 01/26/2011] [Indexed: 10/18/2022]
|
28
|
Stephens EH, Saltarrelli JG, Baggett LS, Nandi I, Kuo JJ, Davis AR, Olmsted-Davis EA, Reardon MJ, Morrisett JD, Grande-Allen KJ. Differential proteoglycan and hyaluronan distribution in calcified aortic valves. Cardiovasc Pathol 2010; 20:334-42. [PMID: 21185747 DOI: 10.1016/j.carpath.2010.10.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2010] [Revised: 09/09/2010] [Accepted: 10/21/2010] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND While the prevalence of calcified aortic valve disease continues to rise and no pharmacological treatments exist, little is known regarding the pathogenesis of the disease. Proteoglycans and the glycosaminoglycan hyaluronan are involved in calcification in arteriosclerosis and their characterization in calcified aortic valves may lend insight into the pathogenesis of the disease. METHODS Fourteen calcified aortic valves removed during valve replacement surgery were immunohistochemically stained for the proteoglycans decorin, biglycan, and versican, as well as the glycosaminoglycan hyaluronan. Staining intensity was evaluated in the following regions of interest: center of calcified nodule, edge of nodule, tissue directly surrounding the nodule; center and tissue surrounding small "prenodules"; and fibrosa layer of normal regions of the leaflet distanced from the nodule. RESULTS Decorin, biglycan, and versican, as well as hyaluronan, were abundantly present immediately surrounding the calcified nodules, but minimally within the nodule itself. Expression of decorin and biglycan in and surrounding prenodules was greater than in the edge and center regions of mature nodules. The levels of expression of the proteoglycans and hyaluronan were highly correlated with one another in the different regions of the valve. CONCLUSIONS The three proteoglycans and hyaluronan demonstrated distinctive localization relative to nodules within calcified aortic valves, where they likely mediate lipid retention, cell proliferation, and extracellular matrix remodeling, and motivate further study. Comparisons between expression of these components in mature nodules and prenodules suggest distinct roles for these components in nodule progression, especially in the tissues surrounding the nodules.
Collapse
|
29
|
Getachew R, Ballinger ML, Burch ML, Reid JJ, Khachigian LM, Wight TN, Little PJ, Osman N. PDGF beta-receptor kinase activity and ERK1/2 mediate glycosaminoglycan elongation on biglycan and increases binding to LDL. Endocrinology 2010; 151:4356-67. [PMID: 20610572 DOI: 10.1210/en.2010-0027] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The initiation of atherosclerosis involves the subendothelial retention of lipoproteins by proteoglycans (PGs). Structural characteristics of glycosaminoglycan (GAG) chains on PGs influence lipoprotein binding and are altered adversely by platelet-derived growth factor (PDGF). The signaling pathway for PDGF-mediated GAG elongation via the PDGF receptor (PDGFR) was investigated. In human vascular smooth muscle cells, PDGF significantly increased (35)S-sulfate incorporation into PGs and GAG chain size. PGs from PDGF-stimulated cells showed increased binding low-density lipoprotein (P < 0.001) in gel mobility shift assays. Knockdown of PDGFRbeta using small interfering RNA demonstrated that PDGF mediated changes in PGs via PDGFRbeta. GAG synthesis and hyperelongation was blocked by inhibition of receptor tyrosine kinase autophosphorylation site Tyr857 activity using Ki11502 or imatinib. Downstream signaling to GAG hyperelongation was mediated through ERK MAPK and not phosphatidylinositol-3 kinase or phospholipase Cgamma. In high-fat-fed apolipoprotein E(-/-) mice, inhibition of PDGFRbeta activity by imatinib reduced aortic total lipid staining area by 35% (P < 0.05). Inhibition of PDGFRbeta tyrosine kinase activity leads to inhibition of GAG synthesis on vascular PGs and aortic lipid area in vivo. PDGFRbeta and its signaling pathways are potential targets for novel therapeutic agents to prevent the earliest stages atherosclerosis.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Apolipoproteins E/genetics
- Apolipoproteins E/metabolism
- Benzamides
- Biglycan
- Cells, Cultured
- Dietary Fats/administration & dosage
- Extracellular Matrix Proteins/metabolism
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Glycosaminoglycans/metabolism
- Humans
- Imatinib Mesylate
- Lipids/analysis
- Lipoproteins, LDL/metabolism
- Male
- Mice
- Mice, Knockout
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Phosphorylation/drug effects
- Piperazines/pharmacology
- Platelet-Derived Growth Factor/pharmacology
- Protein Binding/drug effects
- Protein Kinase Inhibitors/pharmacology
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/metabolism
- Proteoglycans/metabolism
- Pyrimidines/pharmacology
- RNA Interference
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Robel Getachew
- Diabetes and Cell Biology Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, St. Kilda Road Central, Melbourne, Victoria 8008, Australia
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Oörni K, Kovanen PT. Lipoprotein modification by secretory phospholipase A(2) enzymes contributes to the initiation and progression of atherosclerosis. Curr Opin Lipidol 2009; 20:421-7. [PMID: 19593123 DOI: 10.1097/mol.0b013e32832fa14d] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW Secretory phospholipase A2s (sPLA2s) are considered to be important enzymes in the initiation and progression of atherosclerosis. In this review, we discuss the various mechanisms by which the direct action of the sPLA2s on LDL particles in the arterial intima may contribute to atherogenesis. RECENT FINDINGS A wealth of evidence, both in vitro and in vivo, supports a role for the sPLA2s in atherogenesis. Very recently, systemic inhibition of sPLA2s was found to reduce measures of arterial inflammation. The mechanisms behind this inhibition, however, are largely unknown. Here, we discuss the consequences of sPLA2 action on LDL in the arterial intima and address the recent findings regarding the effects of the lipolytic products of sPLA2, lysophosphatidylcholine, and fatty acids on intimal cells. LDL modified by sPLA2 can accumulate in the arterial intima both extracellularly and intracellularly. Importantly, the lipolytic products promote atherosclerosis by monocyte/macrophage recruitment, by enhancing the production of proretentive molecules by vascular smooth muscle cells, and by inducing cell death. SUMMARY Recent findings on sPLA2s support the idea that the enzymes contribute to human atherogenesis not only as initiating agents but also in maintaining plaque inflammation.
Collapse
|
31
|
Ballinger ML, Osman N, Hashimura K, de Haan JB, Jandeleit-Dahm K, Allen T, Tannock LR, Rutledge JC, Little PJ. Imatinib inhibits vascular smooth muscle proteoglycan synthesis and reduces LDL binding in vitro and aortic lipid deposition in vivo. J Cell Mol Med 2009; 14:1408-18. [PMID: 19754668 PMCID: PMC3033015 DOI: 10.1111/j.1582-4934.2009.00902.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The ‘response to retention’ hypothesis of atherogenesis proposes that proteoglycans bind and retain low-density lipoproteins (LDL) in the vessel wall. Platelet-derived growth factor (PDGF) is strongly implicated in atherosclerosis and stimulates proteoglycan synthesis. Here we investigated the action of the PDGF receptor inhibitor imatinib on PDGF-mediated proteoglycan biosynthesis in vitro, lipid deposition in the aortic wall in vivo and the carotid artery ex vivo. In human vSMCs, imatinib inhibited PDGF mediated 35S-SO4 incorporation into proteoglycans by 31% (P < 0.01) and inhibited PDGF-mediated size increases in both chemically cleaved and xyloside associated glycosaminoglycan (GAG) chains by 19%, P < 0.05 and 27%, P < 0.05, respectively. Imatinib decreased PDGF stimulation of the 6:4 position sulphation ratio of disaccharides. The half maximal saturation value for LDL binding for proteoglycans from PDGF stimulated cells in the presence of imatinib was approximately 2.5-fold higher than for PDGF treatment alone. In high fat fed ApoE−/– mice, imatinib reduced total lipid staining area by ∼31% (P < 0.05). Carotid artery lipid accumulation in imatinib treated mice was also reduced. Furthermore, we demonstrate that imatinib inhibits phosphorylation of tyrosine 857, the autophosphorylation site of the PDGF receptor, in vSMCs. Thus imatinib inhibits GAG synthesis on vascular proteoglycans and reduces LDL binding in vitro and in vivo and this effect is mediated via the PDGF receptor. These findings validate a novel mechanism to prevent cardiac disease.
Collapse
Affiliation(s)
- Mandy L Ballinger
- Diabetes & Cell Biology, BakerIDI Heart & Diabetes Institute, Prahran, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Transcriptional and posttranscriptional regulators of biglycan in cardiac fibroblasts. Basic Res Cardiol 2009; 105:99-108. [PMID: 19701788 DOI: 10.1007/s00395-009-0049-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 07/15/2009] [Accepted: 07/22/2009] [Indexed: 12/13/2022]
Abstract
Biglycan, a small leucine-rich proteoglycan, is essential for scar formation and preservation of hemodynamic function after myocardial infarction, as shown in biglycan-knockout mice. Because of this important role in cardiac pathophysiology, we aimed to identify regulators of biglycan expression and posttranslational modifications in cardiac fibroblasts. Cardiac fibroblasts were isolated from neonatal Wistar-Kyoto rats and used in the first passage. Expression of biglycan was analyzed after metabolic labeling with [(35)S]-sulfate by SDS-polyacrylamide gel electrophoresis and molecular sieve chromatography; mRNA expression was examined by Northern analysis and real-time RT-PCR. Serum, thrombin, transforming growth factor beta1 (TGFbeta 1) and platelet-derived growth factor BB (PDGF-BB) strongly increased [(35)S]-labeled proteoglycan levels. Tumor necrosis factor alpha further increased the stimulatory effect of PDGF-BB. PDGF-BB increased glycosaminoglycan (GAG) chain length as shown by molecular sieve chromatography after beta-elimination to release GAG chains. Nitric oxide was the only negative regulator of biglycan as evidenced by marked downregulation in response to DETA-NO ((Z)-1-[2-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate), a long acting nitric oxide donor and SNAP (S-nitroso-N-acetyl-l,l-penicillamine), which completely inhibited PDGF-BB-induced secretion of total [(35)S]-labeled proteoglycans and biglycan mRNA expression. Of note, the molecular weight of biglycan GAG chains was even further increased by NO donors compared to control and PDGF-BB stimulation. The current results suggest that in cardiac fibroblasts, biglycan is induced by a variety of stimuli including serum, thrombin and growth factors such as PDGF-BB and TGFbeta1. This response is counteracted by NO and enhanced by TNFalpha. Interestingly, both up- and downregulation were associated with posttranslational increase of GAG chain length.
Collapse
|
33
|
Yang SNY, Osman N, Burch ML, Little PJ. Factors affecting proteoglycan synthesis and structure that modify the interaction with lipoproteins. ACTA ACUST UNITED AC 2009. [DOI: 10.2217/clp.09.37] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
34
|
Ballinger ML, Ivey ME, Osman N, Thomas WG, Little PJ. Endothelin-1 activates ETA receptors on human vascular smooth muscle cells to yield proteoglycans with increased binding to LDL. Atherosclerosis 2009; 205:451-7. [DOI: 10.1016/j.atherosclerosis.2009.01.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 01/15/2009] [Accepted: 01/15/2009] [Indexed: 11/28/2022]
|
35
|
Yang SNY, Burch ML, Getachew R, Ballinger ML, Osman N, Little PJ. Growth factor-mediated hyper-elongation of glycosaminoglycan chains on biglycan requires transcription and translation. Arch Physiol Biochem 2009; 115:147-54. [PMID: 19580379 DOI: 10.1080/13813450903110754] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The mechanism through which growth factors cause glycosaminoglycan (GAG) hyper-elongation is unclear. We have investigated the role of transcription and translation on the GAG hyper-elongation effect of platelet-derived growth factor (PDGF) in human vascular smooth muscle cells (VSMCs). To determine if the response involves specific signalling pathways or the process of GAG hyper-elongation we have also investigated the effects of epidermal growth factor (EGF), transforming growth factor-beta (TGF-beta) and thrombin. We report that both actinomycin D and cycloheximide completely abolished the ability of PDGF to stimulate radiosulphate incorporation and GAG elongation into secreted proteoglycans, and to increase the size of xyloside GAGs. Blocking de novo protein synthesis completely prevented the action of all growth factors tested to elongate GAG chains. These results lay a foundation for further investigation into the genes and proteins implicated in this response.
Collapse
Affiliation(s)
- Sundy N Y Yang
- BakerIDI Heart and Diabetes Institute, Diabetes and Cell Biology Laboratory, Melbourne, VIC, 3004, Australia
| | | | | | | | | | | |
Collapse
|
36
|
Shetty R, Pibarot P, Audet A, Janvier R, Dagenais F, Perron J, Couture C, Voisine P, Després JP, Mathieu P. Lipid-mediated inflammation and degeneration of bioprosthetic heart valves. Eur J Clin Invest 2009; 39:471-80. [PMID: 19490057 DOI: 10.1111/j.1365-2362.2009.02132.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND The durability of bioprosthetic valves is limited by structural valve degeneration (SVD) leading to bioprostheses (BPs) stenosis or regurgitation. We hypothesized that a lipid-mediated inflammatory mechanism is involved in the SVD of BPs. MATERIAL AND METHODS Eighteen Freestyle stentless BP valves were explanted for SVD at a mean time of 5.9 +/- 3 years after implantation and were analysed by immunohistochemistry and transmission electron microscopy (TEM). RESULTS The mean age of the patients was 65 +/- 8 years and there were 11 male and seven female patients. Two of the 18 BPs had macroscopic calcification, whereas the other valves had minimal or no macroscopic calcification. Tears at the commissures leading to regurgitation was present in 16 BPs. Immunohistochemistry showed the presence of oxidized low-density lipoprotein (ox-LDL) and glycosaminoglycans in the fibrosa layer of 13 BPs. Areas with ox-LDL were infiltrated by macrophages (CD68(+)) co-expressing the scavenger receptor CD36 and metalloproteinase-9 (MMP-9). Zymogram showed the active form of MMP-9 within explanted BPs. EM studies revealed the presence of lipid-laden cells featuring foam cells and fragmented collagen. Nonimplanted control BPs obtained from the manufacturer (n = 4) had no evidence of lipid accumulation, inflammatory cell infiltration or expression of MMP9 within the leaflets. CONCLUSIONS These results support the concept that lipid-mediated inflammatory mechanisms may contribute to the SVD of BPs. These findings suggest that modification of atherosclerotic risk factors with the use of behavioural or pharmacological interventions could help to reduce the incidence of SVD.
Collapse
Affiliation(s)
- R Shetty
- Laval University, Quebec, QC, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
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
|
38
|
Little PJ, Ballinger ML, Burch ML, Osman N. Biosynthesis of natural and hyperelongated chondroitin sulfate glycosaminoglycans: new insights into an elusive process. Open Biochem J 2008; 2:135-42. [PMID: 19238187 PMCID: PMC2627520 DOI: 10.2174/1874091x00802010135] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 09/10/2008] [Accepted: 09/14/2008] [Indexed: 11/24/2022] Open
Abstract
Proteoglycans are important components of the extracellular matrix of all tissues. Proteoglycans are comprised of a core protein and one or more covalently attached glycosaminoglycan (GAG) chains. The major chondroitin sulfate (CS) and dermatan sulfate (DS) proteoglycans are aggrecan, versican, biglycan and decorin. Cells synthesize GAGs of natural or basal lengths and the GAG chains are subject to considerable growth factor, hormonal and metabolic regulation to yield longer GAG chains with altered structure and function. The mechanism by which the CS/DS GAG chains are polymerized is unknown. Recent work has identified several monosaccharide transferases which when co-expressed yield GAG polymers and the length of the polymers depends upon the pair of enzymes coexpressed. The further extension of these chains is regulated by signaling pathways. Inhibition of these latter pathways may be a therapeutic target to prevent the elongation which is associated with increased binding of atherogenic lipids and the disease process of atherosclerosis.
Collapse
Affiliation(s)
- Peter J Little
- Diabetes and Cell Biology Laboratory, Vascular and Hypertension Division, BakerIDI Heart and Diabetes Institute, Melbourne, VIC, Australia 3004 and
| | | | | | | |
Collapse
|
39
|
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
|
40
|
Abstract
PURPOSE OF REVIEW To outline a role for the dermatan sulfate proteoglycan biglycan and specifically its growth factor modified form having elongated glycosaminoglycan chains as being a primary initiator of atherosclerosis. RECENT FINDINGS Antiatherosclerotic therapies have mostly targeted epidemiologically identified, experimentally confirmed risk factors. The efficacy of such therapies is less than optimal, and rates of cardiovascular disease remain stubbornly high. A variety of targets have been actively pursued, but as yet no new therapy has emerged that specifically targets the vessel wall. One area concerns the role of proteoglycans in the trapping of atherogenic lipoproteins as an early and initiating step in atherogenesis. On the basis of studies in human coronary arteries, the prime proteoglycan for lipoprotein retention is biglycan. The glycosaminoglycan chains on biglycan are subject to regulation that yields several structural changes, but most prominently elongation of the chains to form 'hyperelongated biglycan'. Multiple animal studies and a recent human disorder study have demonstrated the colocalization of atherogenic lipoproteins with biglycan in atherosclerotic lesions. Moreover, in the human atherosclerosis, the deposition of lipid appears to precede the chronic inflammatory response typical of atherosclerotic lesions. SUMMARY The process of biglycan-associated glycosaminoglycan elongation represents a novel potential therapeutic target worthy of full investigation for the prevention of atherosclerosis.
Collapse
Affiliation(s)
- Peter J Little
- Cell Biology of Diabetes Laboratory, Baker Heart Research Institute, Melbourne, Australia.
| | | | | |
Collapse
|
41
|
Nakashima Y, Wight TN, Sueishi K. Early atherosclerosis in humans: role of diffuse intimal thickening and extracellular matrix proteoglycans. Cardiovasc Res 2008; 79:14-23. [DOI: 10.1093/cvr/cvn099] [Citation(s) in RCA: 198] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
42
|
Doran AC, Meller N, McNamara CA. Role of smooth muscle cells in the initiation and early progression of atherosclerosis. Arterioscler Thromb Vasc Biol 2008; 28:812-9. [PMID: 18276911 DOI: 10.1161/atvbaha.107.159327] [Citation(s) in RCA: 602] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The initiation of atherosclerosis results from complex interactions of circulating factors and various cell types in the vessel wall, including endothelial cells, lymphocytes, monocytes, and smooth muscle cells (SMCs). Recent reviews highlight the role of activated endothelium and inflammatory cell recruitment in the initiation of and progression of early atherosclerosis. Yet, human autopsy studies, in vitro mechanistic studies, and in vivo correlative data suggest an important role for SMCs in the initiation of atherosclerosis. SMCs are the major producers of extracellular matrix within the vessel wall and in response to atherogenic stimuli can modify the type of matrix proteins produced. In turn, the type of matrix present can affect the lipid content of the developing plaque and the proliferative index of the cells that are adherent to it. SMCs are also capable of functions typically attributed to other cell types. Like macrophages, SMCs can express a variety of receptors for lipid uptake and can form foam-like cells, thereby participating in the early accumulation of plaque lipid. Like endothelial cells, SMCs can also express a variety of adhesion molecules such as vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 to which monocytes and lymphocytes can adhere and migrate into the vessel wall. In addition, through these adhesion molecules, SMCs can also stabilize these cells against apoptosis, thus contributing to the early cellularity of the lesion. Like many cells within the developing plaque, SMCs also produce many cytokines such as PDGF, transforming growth factor-beta, IFNgamma, and MCP-1, all of which contribute to the initiation and propagation of the inflammatory response to lipid. Recent advances in SMC-specific gene modulation have enhanced our ability to determine the role of SMCs in early atherogenesis.
Collapse
Affiliation(s)
- Amanda C Doran
- Cardiovascular Division/Department of Medicine, the Cardiovascular Research Center, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
43
|
McDonald TO, Gerrity RG, Jen C, Chen HJ, Wark K, Wight TN, Chait A, O'Brien KD. Diabetes and arterial extracellular matrix changes in a porcine model of atherosclerosis. J Histochem Cytochem 2007; 55:1149-57. [PMID: 17652266 PMCID: PMC3957530 DOI: 10.1369/jhc.7a7221.2007] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Patients with diabetes are at substantially increased risk for atherosclerosis and clinical cardiovascular events. Because arterial extracellular matrix contains several molecules, including biglycan, versican, hyaluronan, and elastin, that may affect plaque lipid retention and stability, we determined whether diabetes affects plaque content of these molecules in a porcine model of hyperlipidemia and diabetes. Coronary artery sections were studied from non-diabetic normolipidemic (n=11, N-NL), diabetic normolipidemic (n=10, DM-NL), non-diabetic hyperlipidemic (n=16, N-HL), and diabetic hyperlipidemic (n=15, DM-HL) animals. Hyaluronan, biglycan, versican, and apolipoprotein B (apoB) were detected with monospecific peptides or antisera, and elastin with Movat's pentachrome stain, and contents of each were quantified by computer-assisted morphometry. In the hyperlipidemic groups, diabetes was associated with a 4-fold increase in intimal area, with strong correlations between intimal area and immunostained areas for hyaluronan (R(2) = 0.83, p<0.0001), biglycan (R(2) = 0.72, p<0.0001), and apoB (R(2) = 0.23, p=0.0069). In contrast, median (interquartile range) intimal elastin content was significantly lower with diabetes [N-HL: 5.2% (2.4-8.2%) vs DM-HL: 1.5% (0.5-4.2%), p=0.01], and there was a strong negative correlation between intimal total and elastin areas (Spearman r = -0.62, p=0.001). In this porcine model, diabetes was associated with multiple extracellular matrix changes that have been associated with increased lesion instability, greater atherogenic lipoprotein retention, and accelerated atherogenesis.
Collapse
Affiliation(s)
- Thomas O. McDonald
- Divisions of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Ross G. Gerrity
- Department of Pathology, Medical College of Georgia, Augusta, Georgia
| | - Christy Jen
- Divisions of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Hao-Ji Chen
- Divisions of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Kathleen Wark
- Divisions of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Thomas N. Wight
- Hope Heart Program at the Benaroya Research Institute, Seattle, Washington
| | - Alan Chait
- Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| | - Kevin D. O'Brien
- Divisions of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
- Correspondence to: Kevin D. O'Brien, MD, Division of Cardiology, Box 356422, University of Washington, Seattle, WA 98195-6422. E-mail:
| |
Collapse
|
44
|
Nakashima Y, Fujii H, Sumiyoshi S, Wight TN, Sueishi K. Early human atherosclerosis: accumulation of lipid and proteoglycans in intimal thickenings followed by macrophage infiltration. Arterioscler Thromb Vasc Biol 2007; 27:1159-65. [PMID: 17303781 DOI: 10.1161/atvbaha.106.134080] [Citation(s) in RCA: 305] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The present study was designed to clarify the morphological features of early human atherosclerosis and to determine whether specific extracellular matrix proteoglycans play a role in early atherogenesis. METHODS AND RESULTS Step and serial sections were obtained from right coronary arteries with no or early atherosclerosis. Atherosclerosis was classified into 4 grades according to the amount of lipid deposition. Coronary arteries with Grade 0 showed diffuse intimal thickening (DIT) with no lipid deposits. The extracellular matrix proteoglycans, biglycan and decorin, were localized in the outer layer of DIT. Most cases of Grade 1 and Grade 2 exhibited fatty streaks with extracellular lipids colocalizing with biglycan and decorin in the outer layer of the intima. As lipid grades increased, macrophages increased in number and were present in the deeper layers. Most cases of Grade 3 exhibited pathologic intimal thickening (PIT) with extracellular lipids underneath a layer of foam cell macrophages. CONCLUSIONS In early human coronary atherosclerosis, fatty streaks develop via extracellular deposition of lipids associated with specific types of proteoglycans in the outer layer of preexisting DIT. As the amount of the lipid increases in fatty streaks, macrophages infiltrate toward the deposited lipid to form PIT with foam cells.
Collapse
|
45
|
Ballinger ML, Thomas MC, Nigro J, Ivey ME, Dilley RJ, Little PJ. Glycated and carboxy-methylated proteins do not directly activate human vascular smooth muscle cells. Kidney Int 2006; 68:2756-65. [PMID: 16316350 DOI: 10.1111/j.1523-1755.2005.00746.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Advanced glycation end products (AGEs) accumulate in patients with diabetes, particularly at sites of vascular damage and within atherosclerotic lesions, but whether they have direct actions on vascular smooth muscle cells (VSMCs) is controversial. METHODS AGEs were constructed and characterized by protein content, level of modification, fluorescence, and molecular size. Human VSMCs were derived from different vascular beds. Glucose consumption, de novo protein synthesis, and proteoglycan biosynthesis were measured using a colorimetric assay and metabolic radiolabeling. Receptor for AGEs (RAGE) expression was assessed by real-time reverse transcription-polymerase chain reaction (RT-PCR) and Western blot. RESULTS Treatment with AGEs under low or high glucose conditions showed no change in cellular glucose consumption or in cellular protein synthesis under low glucose conditions. Treatment of VSMCs with Nepsilon-(carboxymethyl)lysine in the presence of low glucose increased [35S]-sulfate incorporation into secreted proteoglycans by 72% (P < 0.001) and 67% (P < 0.001); however, the control proteins also increased [35S]-sulfate incorporation into proteoglycans by 56% (P < 0.01), with similar effects observed under high glucose conditions. Human VSMCs showed no difference in response to glycated and non-glycated protein. Protein and gene expression of RAGE in VSMC was approximately 50-fold lower compared to HMEC-1 and U937 cells, consistent with the immunohistochemical staining of RAGE in vivo. CONCLUSION VSMCs show very low levels of RAGE expression; thus, activation of VSMCs by AGEs does not occur. In diabetes, RAGE expression in VSM may increase to the extent that it becomes activated by AGEs in a manner that would contribute to the process of atherosclerosis.
Collapse
MESH Headings
- Atherosclerosis/metabolism
- Cells, Cultured
- Gene Expression
- Glucose/metabolism
- Glycation End Products, Advanced/metabolism
- Glycation End Products, Advanced/pharmacology
- Glycolysis
- Glycosylation
- Humans
- Mammary Arteries/cytology
- Methylation
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Proteoglycans/biosynthesis
- Proteoglycans/metabolism
- Radial Artery/cytology
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Saphenous Vein/cytology
Collapse
Affiliation(s)
- Mandy L Ballinger
- Cell Biology of Diabetes Laboratory, Baker Heart Research Institute, Melbourne, The Alfred Hospital, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
46
|
Huang R, Merrilees MJ, Braun K, Beaumont B, Lemire J, Clowes AW, Hinek A, Wight TN. Inhibition of versican synthesis by antisense alters smooth muscle cell phenotype and induces elastic fiber formation in vitro and in neointima after vessel injury. Circ Res 2005; 98:370-7. [PMID: 16385080 DOI: 10.1161/01.res.0000202051.28319.c8] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The proteoglycan versican is implicated in several atherogenic events, including stimulation of vascular smooth muscle cell (VSMC) growth and migration, retention of lipoproteins, and promotion of thrombogenesis. A high content of intimal versican also correlates with a low content of elastin, suggesting an inhibitory role for versican in elastogenesis. To determine whether reduced production of versican can be used to enhance elastogenesis, we transduced Fischer rat VSMC (FRSMC) with a versican antisense sequence using the retroviral vector LXSN. Stable expression of versican antisense (LVaSN) significantly reduced versican production, induced a flattened morphology, reduced cell proliferation and migration, increased tropoelastin synthesis, increased elastin binding protein (S-Gal/EBP), and increased deposition of elastic fibers in long-term cultures. Add-back of chondroitin sulfate chains, or versican, decreased S-Gal/EBP and elastic fiber formation. LVaSN cells seeded into balloon catheter-injured rat carotid arteries formed neointimae containing low levels versican, increased amounts of S-Gal/EBP, and increased elastin deposits 7 days postinjury. At 4 weeks, neointimae formed from LVaSN cells were highly structured and contained multiple layers of elastic fibers and lamellae. These results indicate a central role for versican and its constituent chondroitin sulfate chains in controlling cell phenotype, elastogenesis, and intimal structure.
Collapse
Affiliation(s)
- Robert Huang
- Department of Anatomy with Radiology, The University of Aukland, New Zealand
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Chang MY, Han CY, Wight TN, Chait A. Antioxidants inhibit the ability of lysophosphatidylcholine to regulate proteoglycan synthesis. Arterioscler Thromb Vasc Biol 2005; 26:494-500. [PMID: 16357313 DOI: 10.1161/01.atv.0000200135.61362.27] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We previously have shown that lysophosphatidylcholine (lysoPC) regulates proteoglycan synthesis by vascular smooth muscle cells (SMCs). Given the accumulating evidence for reactive oxygen species (ROS) as mediators of a variety of effects of lysoPC, the present study evaluates the potential role of ROS as intermediate molecules in the regulation of proteoglycan synthesis by lysoPC. METHODS AND RESULTS LysoPC (10 micromol/L) was found to stimulate rapid and sustained generation of ROS by SMC, as indicated using a fluorescent probe for measuring intracellular oxidants and fluorescence-activated cell sorting. This was not associated with cytotoxicity, as evaluated by fluorescence microscopy using MitoTracker Red or propidium iodide, cell number, cell protein, or lactate dehydrogenase release. Pretreatment with catalase or superoxide dismutase, specific scavengers of hydrogen peroxide and superoxide, respectively, blocked the ability of lysoPC to stimulate both accumulation of ROS and proteoglycan synthesis. Most importantly, these enzymatic antioxidants prevented lysoPC from stimulating the synthesis of proteoglycans with enhanced lipoprotein-binding properties, as quantified by a gel shift binding assay. CONCLUSIONS These findings strongly suggest that ROS are key mediators in the ability of lysoPC to regulate proteoglycan synthesis and that these effects can be inhibited by antioxidants.
Collapse
Affiliation(s)
- Mary Y Chang
- Department of Medicine, University of Washington, Seattle, WA 98195-6426, USA.
| | | | | | | |
Collapse
|
48
|
Lindholm MW, Nilsson J, Moses J. Low density lipoprotein stimulation of human macrophage proteoglycan secretion. Biochem Biophys Res Commun 2005; 328:455-60. [PMID: 15694369 DOI: 10.1016/j.bbrc.2005.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Indexed: 11/19/2022]
Abstract
Lipoprotein trapping in arterial intima increases the risk for lipoprotein oxidation, foam cell formation, and inflammatory response in surrounding cells. Modified lipoproteins increase smooth muscle cell production of proteoglycans likely to retain lipoproteins in intimal extracellular matrix. We hypothesized that macrophage proteoglycan production is regulated in a similar manner, and characterized glycosaminoglycan side chains of secreted proteoglycans. Incubation with native low density lipoproteins (LDL) strongly stimulates total proteoglycan secretion in a time and concentration dependent manner. The main secretion product is small-sized (120 kDa) with unusually long galactosaminoglycan chains, predominantly chondroitin-6-O-sulfated. The effect appears specific for native LDL; oxidized LDL, very low density lipoproteins or phospholipid liposomes have only minor effects compared to control. These observations suggest that native LDL stimulate macrophages to secrete a chondroitin sulfate-rich proteoglycan moiety likely to have high capacity for vascular extracellular trapping of apolipoprotein B-containing lipoproteins.
Collapse
Affiliation(s)
- Marie Wickström Lindholm
- Department of Medicine, Lund University, Wallenberg Laboratory Plan 1, UMAS Ing. 46, 20502 Malmö, Sweden
| | | | | |
Collapse
|
49
|
O'Brien KD, Lewis K, Fischer JW, Johnson P, Hwang JY, Knopp EA, Kinsella MG, Barrett PHR, Chait A, Wight TN. Smooth muscle cell biglycan overexpression results in increased lipoprotein retention on extracellular matrix: implications for the retention of lipoproteins in atherosclerosis. Atherosclerosis 2005; 177:29-35. [PMID: 15488862 DOI: 10.1016/j.atherosclerosis.2004.07.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2003] [Revised: 05/13/2004] [Accepted: 07/02/2004] [Indexed: 10/26/2022]
Abstract
Lipoprotein retention on extracellular matrix (ECM) may play a central role in atherogenesis, and a specific extracellular matrix proteoglycan, biglycan, has been implicated in lipoprotein retention in human atherosclerosis. To test whether increased cellular biglycan expression results in increased retention of lipoproteins on ECM, rat aortic smooth muscle cells (SMCs) were transduced with a human biglycan cDNA-containing retroviral vector (LBSN) or with an empty retroviral vector (LXSN). To assess the importance of biglycan's glycosaminoglycan side chains in lipoprotein retention, ECM binding studies were also performed using RASMCs transduced with a retroviral vector encoding for a mutant, glycosaminoglycan-deficient biglycan (LBmutSN). Human biglycan mRNA and protein were confirmed in LBSN and LBmutSN RASMCs by Northern and Western blot analyses. HDL3+E binding to SMC ECM was increased significantly (as determined by 95% confidence intervals for binding curves) for LBSN as compared to either LXSN or LBmutSN cells; the increases for LBSN cell ECM were due primarily to an approximately 50% increase in binding sites (increased Bmax) versus LXSN cell ECM and of approximately 25% versus LBmutSN cell ECM. These results are consistent with the hypothesis that biglycan, through its glycosaminoglycan side chains, may mediate lipoprotein retention on atherosclerotic plaque ECM.
Collapse
Affiliation(s)
- Kevin D O'Brien
- Division of Cardiology, Box 356422, Department of Medicine, University of Washington, 1959N.E. Pacific St., Seattle, WA 98195-6422, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Nigro J, Ballinger ML, Dilley RJ, Jennings GLR, Wight TN, Little PJ. Fenofibrate modifies human vascular smooth muscle proteoglycans and reduces lipoprotein binding. Diabetologia 2004; 47:2105-13. [PMID: 15592811 DOI: 10.1007/s00125-004-1588-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2004] [Accepted: 07/19/2004] [Indexed: 01/22/2023]
Abstract
AIMS/HYPOTHESIS Vascular disease in type 2 diabetes is associated with an up-regulation of atherogenic growth factors, which stimulate matrix synthesis including proteoglycans. We have examined the direct actions of fenofibrate on human vascular smooth muscle cells (VSMCs) and have specifically investigated proteoglycan synthesis and binding to LDL. METHODS Proteoglycans synthesised by human VSMCs treated with fenofibrate (30 micromol/l) were assessed for binding to human LDL using a gel mobility shift assay, metabolically labelled with [(35)S]-sulphate and quantitated by cetylpyridinium chloride. They were then assessed for electrophoretic mobility by SDS-PAGE, for size by gel filtration, for sulphation pattern by fluorophore-assisted carbohydrate electrophoresis, and for glycosaminoglycan (GAG) composition by enzyme digestion. RESULTS Proteoglycans synthesised in the presence of fenofibrate showed an increase in the half-maximum saturation concentration of LDL from 36.8+/-12.4 microg/ml to 77.7+/-17 microg/ml under basal conditions, from 24.9+/-4.6 microg/ml to 39.1+/-6.1 microg/ml in the presence of TGF-beta1, and from 9.5+/-4.4 microg/ml to 31.1+/-3.4 microg/ml in the presence of platelet-derived growth factor/insulin. Fenofibrate treatment in the presence of TGF-beta1 inhibited the incorporation of [(35)S]-sulphate into secreted and cell-associated proteoglycans synthesised by human VSMCs by 59.2% (p<0.01) and 39.8% (p<0.01) respectively. The changes in sulphate incorporation following treatment with fenofibrate were associated with a concentration-related increase in the electrophoretic mobility due to a reduction in GAG length. There was no change in the sulphation pattern; however, there was an alteration in the disaccharide composition of the GAGs. CONCLUSIONS/INTERPRETATION Fenofibrate modifies the structure of vascular proteoglycans by reducing the length of the GAG chains and GAG composition, resulting in reduced binding to human LDL, a mechanism which may lead to a reduction of atherosclerosis and cardiovascular disease in people with diabetes treated with fenofibrate.
Collapse
Affiliation(s)
- J Nigro
- Cell Biology of Diabetes Laboratory, Baker Heart Research Institute, St. Kilda Rd Central, PO Box 6492, Melbourne, Victoria, 8008, Australia
| | | | | | | | | | | |
Collapse
|