1
|
Borén J, Packard CJ, Binder CJ. Apolipoprotein B-containing lipoproteins in atherogenesis. Nat Rev Cardiol 2025:10.1038/s41569-024-01111-0. [PMID: 39743565 DOI: 10.1038/s41569-024-01111-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Apolipoprotein B (apoB) is the main structural protein of LDLs, triglyceride-rich lipoproteins and lipoprotein(a), and is crucial for their formation, metabolism and atherogenic properties. In this Review, we present insights into the role of apoB-containing lipoproteins in atherogenesis, with an emphasis on the mechanisms leading to plaque initiation and growth. LDL, the most abundant cholesterol-rich lipoprotein in plasma, is causally linked to atherosclerosis. LDL enters the artery wall by transcytosis and, in vulnerable regions, is retained in the subendothelial space by binding to proteoglycans via specific sites on apoB. A maladaptive response ensues. This response involves modification of LDL particles, which promotes LDL retention and the release of bioactive lipid products that trigger inflammatory responses in vascular cells, as well as adaptive immune responses. Resident and recruited macrophages take up modified LDL, leading to foam cell formation and ultimately cell death due to inadequate cellular lipid handling. Accumulation of dead cells and cholesterol crystallization are hallmarks of the necrotic core of atherosclerotic plaques. Other apoB-containing lipoproteins, although less abundant, have substantially greater atherogenicity per particle than LDL. These lipoproteins probably contribute to atherogenesis in a similar way to LDL but might also induce additional pathogenic mechanisms. Several targets for intervention to reduce the rate of atherosclerotic lesion initiation and progression have now been identified, including lowering plasma lipoprotein levels and modulating the maladaptive responses in the artery wall.
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Yamamoto K, Scilabra SD, Bonelli S, Jensen A, Scavenius C, Enghild JJ, Strickland DK. Novel insights into the multifaceted and tissue-specific roles of the endocytic receptor LRP1. J Biol Chem 2024; 300:107521. [PMID: 38950861 PMCID: PMC11325810 DOI: 10.1016/j.jbc.2024.107521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Receptor-mediated endocytosis provides a mechanism for the selective uptake of specific molecules thereby controlling the composition of the extracellular environment and biological processes. The low-density lipoprotein receptor-related protein 1 (LRP1) is a widely expressed endocytic receptor that regulates cellular events by modulating the levels of numerous extracellular molecules via rapid endocytic removal. LRP1 also participates in signalling pathways through this modulation as well as in the interaction with membrane receptors and cytoplasmic adaptor proteins. LRP1 SNPs are associated with several diseases and conditions such as migraines, aortic aneurysms, cardiopulmonary dysfunction, corneal clouding, and bone dysmorphology and mineral density. Studies using Lrp1 KO mice revealed a critical, nonredundant and tissue-specific role of LRP1 in regulating various physiological events. However, exactly how LRP1 functions to regulate so many distinct and specific processes is still not fully clear. Our recent proteomics studies have identified more than 300 secreted proteins that either directly interact with LRP1 or are modulated by LRP1 in various tissues. This review will highlight the remarkable ability of this receptor to regulate secreted molecules in a tissue-specific manner and discuss potential mechanisms underpinning such specificity. Uncovering the depth of these "hidden" specific interactions modulated by LRP1 will provide novel insights into a dynamic and complex extracellular environment that is involved in diverse biological and pathological processes.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.
| | - Simone D Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy
| | - Simone Bonelli
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Anders Jensen
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Soto Y, Hernández A, Sarduy R, Brito V, Marleau S, Vine DF, Vázquez AM, Proctor SD. Monoclonal Antibody chP3R99 Reduces Subendothelial Retention of Atherogenic Lipoproteins in Insulin-Resistant Rats: Acute Treatment Versus Long-Term Protection as an Idiotypic Vaccine for Atherosclerosis. J Am Heart Assoc 2024; 13:e032419. [PMID: 38934863 PMCID: PMC11255714 DOI: 10.1161/jaha.123.032419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 05/15/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Atherosclerosis is triggered by the retention of apolipoprotein B-containing lipoproteins by proteoglycans. In addition to low-density lipoprotein, remnant lipoproteins have emerged as pivotal contributors to this pathology, particularly in the context of insulin resistance and diabetes. We have previously reported antiatherogenic properties of a monoclonal antibody (chP3R99) that recognizes sulfated glycosaminoglycans on arterial proteoglycans. METHODS AND RESULTS Solid-phase assays demonstrated that chP3R99 effectively blocked >50% lipoprotein binding to chondroitin sulfate and vascular extracellular matrix in vitro. The preperfusion of chP3R99 (competitive effect) resulted in specific antibody-arterial accumulation and reduced fluorescent lipoprotein retention by ~60% in insulin resistant JCR:LA-cp rats. This competitive reduction was dose dependent (25-250 μg/mL), effectively decreasing deposition of cholesterol associated with lipoproteins. In a 5-week vaccination study in insulin resistant rats with (200 μg subcutaneously, once a week), chP3R99 reduced arterial lipoprotein retention, and was associated with the production of antichondroitin sulfate antibodies (Ab3) able to accumulate in the arteries (dot-blot). Neither the intravenous inoculation of chP3R99 (4.5 mg/kg), nor the immunization with this antibody displayed adverse effects on lipid or glucose metabolism, insulin resistance, liver function, blood cell indices, or inflammation pathways in JCR:LA-cp rats. CONCLUSIONS Both acute (passive) and long-term administration (idiotypic cascade) of chP3R99 antibody reduced low-density lipoprotein and remnant lipoprotein interaction with proteoglycans in an insulin-resistant setting. These findings support the innovative approach of targeting proatherogenic lipoprotein retention by chP3R99 as a passive therapy or as an idiotypic vaccine for atherosclerosis.
Collapse
Affiliation(s)
- Yosdel Soto
- Department of ImmunobiologyDirection of Immunology and ImmunotherapyCentre for Molecular ImmunologyHavanaCuba
- Metabolic and Cardiovascular Disease LaboratoryGroup on Molecular and Cell Biology of LipidsAlberta Diabetes and Mazankowski Heart InstitutesUniversity of AlbertaEdmontonABCanada
| | - Arletty Hernández
- Department of ImmunobiologyDirection of Immunology and ImmunotherapyCentre for Molecular ImmunologyHavanaCuba
| | - Roger Sarduy
- Department of ImmunobiologyDirection of Immunology and ImmunotherapyCentre for Molecular ImmunologyHavanaCuba
| | - Victor Brito
- Department of ImmunobiologyDirection of Immunology and ImmunotherapyCentre for Molecular ImmunologyHavanaCuba
| | - Sylvie Marleau
- Faculté de PharmacieUniversité de MontréalMontréalQCCanada
| | - Donna F. Vine
- Metabolic and Cardiovascular Disease LaboratoryGroup on Molecular and Cell Biology of LipidsAlberta Diabetes and Mazankowski Heart InstitutesUniversity of AlbertaEdmontonABCanada
| | - Ana M. Vázquez
- Innovation and Managing DirectionCenter for Molecular ImmunologyHavanaCuba
| | - Spencer D. Proctor
- Metabolic and Cardiovascular Disease LaboratoryGroup on Molecular and Cell Biology of LipidsAlberta Diabetes and Mazankowski Heart InstitutesUniversity of AlbertaEdmontonABCanada
| |
Collapse
|
4
|
Geh EN, Swertfeger DK, Sexmith H, Heink A, Tarapore P, Melchior JT, Davidson WS, Shah AS. A novel assay to measure low-density lipoproteins binding to proteoglycans. PLoS One 2024; 19:e0291632. [PMID: 38295021 PMCID: PMC10830033 DOI: 10.1371/journal.pone.0291632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 09/04/2023] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND The binding of low-density lipoprotein (LDL) to proteoglycans (PGs) in the extracellular matrix (ECM) of the arterial intima is a key initial step in the development of atherosclerosis. Although many techniques have been developed to assess this binding, most of the methods are labor-intensive and technically challenging to standardize across research laboratories. Thus, sensitive, and reproducible assay to detect LDL binding to PGs is needed to screen clinical populations for atherosclerosis risk. OBJECTIVES The aim of this study was to develop a quantitative, and reproducible assay to evaluate the affinity of LDL towards PGs and to replicate previously published results on LDL-PG binding. METHODS Immunofluorescence microscopy was performed to visualize the binding of LDL to PGs using mouse vascular smooth muscle (MOVAS) cells. An in-cell ELISA (ICE) was also developed and optimized to quantitatively measure LDL-PG binding using fixed MOVAS cells cultured in a 96-well format. RESULTS We used the ICE assay to show that, despite equal APOB concentrations, LDL isolated from adults with cardiovascular disease bound to PG to a greater extent than LDL isolated from adults without cardiovascular disease (p<0.05). CONCLUSION We have developed an LDL-PG binding assay that is capable of detecting differences in PG binding affinities despite equal APOB concentrations. Future work will focus on candidate apolipoproteins that enhance or diminish this interaction.
Collapse
Affiliation(s)
- Esmond N. Geh
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center & the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Debi K. Swertfeger
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center & the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Hannah Sexmith
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center & the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Anna Heink
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center & the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Pheruza Tarapore
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - John T. Melchior
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, United States of America
- Department of Neurology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - W. Sean Davidson
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Amy Sanghavi Shah
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center & the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| |
Collapse
|
5
|
den Hartigh LJ, May KS, Zhang XS, Chait A, Blaser MJ. Serum amyloid A and metabolic disease: evidence for a critical role in chronic inflammatory conditions. Front Cardiovasc Med 2023; 10:1197432. [PMID: 37396595 PMCID: PMC10311072 DOI: 10.3389/fcvm.2023.1197432] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/15/2023] [Indexed: 07/04/2023] Open
Abstract
Serum amyloid A (SAA) subtypes 1-3 are well-described acute phase reactants that are elevated in acute inflammatory conditions such as infection, tissue injury, and trauma, while SAA4 is constitutively expressed. SAA subtypes also have been implicated as playing roles in chronic metabolic diseases including obesity, diabetes, and cardiovascular disease, and possibly in autoimmune diseases such as systemic lupus erythematosis, rheumatoid arthritis, and inflammatory bowel disease. Distinctions between the expression kinetics of SAA in acute inflammatory responses and chronic disease states suggest the potential for differentiating SAA functions. Although circulating SAA levels can rise up to 1,000-fold during an acute inflammatory event, elevations are more modest (∼5-fold) in chronic metabolic conditions. The majority of acute-phase SAA derives from the liver, while in chronic inflammatory conditions SAA also derives from adipose tissue, the intestine, and elsewhere. In this review, roles for SAA subtypes in chronic metabolic disease states are contrasted to current knowledge about acute phase SAA. Investigations show distinct differences between SAA expression and function in human and animal models of metabolic disease, as well as sexual dimorphism of SAA subtype responses.
Collapse
Affiliation(s)
- Laura J. den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Karolline S. May
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| | - Alan Chait
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Martin J. Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
6
|
Reed E, Fellows A, Lu R, Rienks M, Schmidt L, Yin X, Duregotti E, Brandt M, Krasemann S, Hartmann K, Barallobre-Barreiro J, Addison O, Cuello F, Hansen A, Mayr M. Extracellular Matrix Profiling and Disease Modelling in Engineered Vascular Smooth Muscle Cell Tissues. Matrix Biol Plus 2022; 16:100122. [PMID: 36193159 PMCID: PMC9526190 DOI: 10.1016/j.mbplus.2022.100122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/22/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
Aortic smooth muscle cells (SMCs) have an intrinsic role in regulating vessel homeostasis and pathological remodelling. In two-dimensional (2D) cell culture formats, however, SMCs are not embedded in their physiological extracellular matrix (ECM) environment. To overcome the limitations of conventional 2D SMC cultures, we established a 3D in vitro model of engineered vascular smooth muscle cell tissues (EVTs). EVTs were casted from primary murine aortic SMCs by suspending a SMC-fibrin master mix between two flexible silicon-posts at day 0 before prolonged culture up to 14 days. Immunohistochemical analysis of EVT longitudinal sections demonstrated that SMCs were aligned, viable and secretory. Mass spectrometry-based proteomics analysis of murine EVT lysates was performed and identified 135 matrisome proteins. Proteoglycans, including the large aggregating proteoglycan versican, accumulated within EVTs by day 7 of culture. This was followed by the deposition of collagens, elastin-binding proteins and matrix regulators up to day 14 of culture. In contrast to 2D SMC controls, accumulation of versican occurred in parallel to an increase in versikine, a cleavage product mediated by proteases of the A Disintegrin and Metalloproteinase with Thrombospondin motifs (ADAMTS) family. Next, we tested the response of EVTs to stimulation with transforming growth factor beta-1 (TGFβ-1). EVTs contracted in response to TGFβ-1 stimulation with altered ECM composition. In contrast, treatment with the pharmacological activin-like kinase inhibitor (ALKi) SB 431542 suppressed ECM secretion. As a disease stimulus, we performed calcification assays. The ECM acts as a nidus for calcium phosphate deposition in the arterial wall. We compared the onset and extent of calcification in EVTs and 2D SMCs cultured under high calcium and phosphate conditions for 7 days. Calcified EVTs displayed increased tissue stiffness by up to 30 % compared to non-calcified controls. Unlike the rapid calcification of SMCs in 2D cultures, EVTs sustained expression of the calcification inhibitor matrix Gla protein and allowed for better discrimination of the calcification propensity between independent biological replicates. In summary, EVTs are an intuitive and versatile model to investigate ECM synthesis and turnover by SMCs in a 3D environment. Unlike conventional 2D cultures, EVTs provide a more relevant pathophysiological model for retention of the nascent ECM produced by SMCs.
Collapse
Key Words
- 2D, Two-dimensional
- 3D cell culture
- 3D, Three-dimensional
- ADAMTS, A disintegrin and metalloproteinase with thrombospondin motifs
- ALKi, Activin-like kinase inhibitor
- Calcification
- ECM
- ECM, Extracellular matrix
- EHT, Engineered heart tissue
- EVT, Engineered vascular smooth muscle cell tissue
- LC-MS/MS, Liquid chromatography with tandem mass spectrometry
- Proteomics
- SMC, Smooth muscle cell
- Smooth muscle cells
- TCP, Tissue culture polystyrene
- TGFβ-1, Transforming growth factor beta-1
- Tissue engineering
Collapse
Affiliation(s)
- Ella Reed
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Adam Fellows
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Ruifang Lu
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Marieke Rienks
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Lukas Schmidt
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Xiaoke Yin
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Elisa Duregotti
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Mona Brandt
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, University Medical Center Hamburg-Eppendorf, Germany
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Kristin Hartmann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Javier Barallobre-Barreiro
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Owen Addison
- Centre of Oral, Clinical & Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, University Medical Center Hamburg-Eppendorf, Germany
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, University Medical Center Hamburg-Eppendorf, Germany
| | - Manuel Mayr
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| |
Collapse
|
7
|
Khamis RY, Hartley A, Caga-Anan M, Pandey SS, Marceddu C, Kojima C, Chang SH, Boyle JJ, Johnson JL, Björkbacka H, Guo L, Finn AV, Virmani R, Nilsson J, Haskard DO. Monoclonal Autoantibody Against a Cryptic Epitope on Tissue-Adherent Low-Density Lipoprotein for Molecular Imaging in Atherosclerosis. JACC Cardiovasc Imaging 2022; 15:1458-1470. [PMID: 35926905 DOI: 10.1016/j.jcmg.2022.02.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/04/2022] [Accepted: 02/09/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Antibody-based constructs for molecular imaging and therapeutic delivery provide promising opportunities for the diagnosis and treatment of atherosclerosis. OBJECTIVES The authors aimed to generate and characterize immunoglobulin (Ig)G monoclonal autoantibodies in atherosclerosis for targeting of novel molecular determinants. METHODS The authors created hybridomas from an unimmunized low-density lipoprotein (LDL) receptor-deficient (Ldlr-/-) mouse and selected an IgG2b isotype autoantibody, LO9, for further characterization. RESULTS LO9 reacted well with native LDL bound to immobilized matrix components and less well to oxidized LDL. LO9 binding to immobilized native LDL was not neutralized by fluid-phase native LDL, indicating an adhesion-dependent epitope. The authors localized the epitope to a 20 amino-acid peptide sequence (P5) in the globular amino-terminus of apolipoprotein B. LO9 reacted with antigen in mouse atherosclerosis and in both human stable and ruptured coronary atherosclerosis. Furthermore, in vivo near-infrared fluorescence molecular tomographic imaging, and ex vivo confocal microscopy showed that intravenously injected LO9 localized beneath endothelium of the aortic arch in Ldlr-/- mice, in the vicinity of macrophages. CONCLUSIONS The authors believe LO9 is the first example of an IgG autoantibody that reacts with a native LDL epitope revealed by adherence to tissue matrix. Antibodies against adherent native LDL have potential as molecular targeting agents for imaging of and therapeutic delivery to atherosclerosis.
Collapse
Affiliation(s)
- Ramzi Y Khamis
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Adam Hartley
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Mikhail Caga-Anan
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Samata S Pandey
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Cinzia Marceddu
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Chiari Kojima
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Shang-Hung Chang
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Division of Cardiology, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Joseph J Boyle
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jason L Johnson
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | | | - Liang Guo
- CVPath Institute, Gaithersburg, Maryland, USA
| | | | | | - Jan Nilsson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Dorian O Haskard
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
8
|
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: 21] [Impact Index Per Article: 7.0] [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
|
9
|
Glise L, Rutberg M, Håversen L, Levin MC, Levin M, Jeppsson A, Borén J, Fogelstrand P. pH-Dependent Protonation of Histidine Residues Is Critical for Electrostatic LDL (Low-Density Lipoprotein) Binding to Human Coronary Arteries. Arterioscler Thromb Vasc Biol 2022; 42:1037-1047. [PMID: 35652335 DOI: 10.1161/atvbaha.122.317868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The initiating step in atherogenesis is the electrostatic binding of LDL (low-density lipoprotein) to proteoglycan glycosaminoglycans in the arterial intima. However, although proteoglycans are widespread throughout the intima of most coronary artery segments, LDL is not evenly distributed, indicating that LDL retention is not merely dependent on the presence of proteoglycans. We aim to identify factors that promote the interaction between LDL and the vessel wall of human coronary arteries. METHODS We developed an ex vivo model to investigate binding of human-labeled LDL to human coronary artery sections without the interference of cellular processes. RESULTS By staining consecutive sections of human coronary arteries, we found strong staining of sulfated glycosaminoglycans throughout the arterial intima, whereas endogenous LDL deposits were focally distributed. Ex vivo binding of LDL was uniform in all intimal areas with sulfated glycosaminoglycans. However, lowering the pH from 7.4 to 6.5 triggered a 35-fold increase in LDL binding. The pH-dependent binding was abolished by pretreating LDL with diethyl-pyrocarbonate, which blocks the protonation of histidine residues, or cyclohexanedione, which inhibits the positive charge of site B on LDL. Thus, both histidine protonation and site B are required for strong electrostatic LDL binding to the intima. CONCLUSIONS This study identifies histidine protonation as an important component for electrostatic LDL binding to human coronary arteries. Our findings show that the local pH will have a profound impact on LDL's affinity for sulfated glycosaminoglycans, which may influence the retention and accumulation pattern of LDL in the arterial vasculature.
Collapse
Affiliation(s)
- Lars Glise
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| | - Mikael Rutberg
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| | - Liliana Håversen
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| | - Malin C Levin
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| | - Max Levin
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| | - Anders Jeppsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.).,Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden (A.J.)
| | - Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.).,Sahlgrenska University Hospital/Wallenberg Laboratory, Gothenburg, Sweden (J.B.)
| | - Per Fogelstrand
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| |
Collapse
|
10
|
HDL as Bidirectional Lipid Vectors: Time for New Paradigms. Biomedicines 2022; 10:biomedicines10051180. [PMID: 35625916 PMCID: PMC9138557 DOI: 10.3390/biomedicines10051180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 02/06/2023] Open
Abstract
The anti-atherogenic properties of high-density lipoproteins (HDL) have been explained mainly by reverse cholesterol transport (RCT) from peripheral tissues to the liver. The RCT seems to agree with most of the negative epidemiological correlations between HDL cholesterol levels and coronary artery disease. However, therapies designed to increase HDL cholesterol failed to reduce cardiovascular risk, despite their capacity to improve cholesterol efflux, the first stage of RCT. Therefore, the cardioprotective role of HDL may not be explained by RCT, and it is time for new paradigms about the physiological function of these lipoproteins. It should be considered that the main HDL apolipoprotein, apo AI, has been highly conserved throughout evolution. Consequently, these lipoproteins play an essential physiological role beyond their capacity to protect against atherosclerosis. We propose HDL as bidirectional lipid vectors carrying lipids from and to tissues according to their local context. Lipid influx mediated by HDL appears to be particularly important for tissue repair right on site where the damage occurs, including arteries during the first stages of atherosclerosis. In contrast, the HDL-lipid efflux is relevant for secretory cells where the fusion of intracellular vesicles drastically enlarges the cytoplasmic membrane with the potential consequence of impairment of cell function. In such circumstances, HDL could deliver some functional lipids and pick up not only cholesterol but an integral part of the membrane in excess, restoring the viability of the secretory cells. This hypothesis is congruent with the beneficial effects of HDL against atherosclerosis as well as with their capacity to induce insulin secretion and merits experimental exploration.
Collapse
|
11
|
Ali L, Safan A, Kamran S, Akhtar N, Elalamy O. Acute Thromboembolic Ischemic Stroke From Complex Aortic Arch Plaque. Cureus 2021; 13:e16977. [PMID: 34540387 PMCID: PMC8423320 DOI: 10.7759/cureus.16977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2021] [Indexed: 12/04/2022] Open
Abstract
Atherosclerosis is a systemic pathologic process, may involve aorta and is important cause of systemic embolization. The risk of embolism is increased for mobile and complex aortic plaques that are >4 mm thick. The most common manifestations are stroke, transient ischemic attack (TIA) and peripheral embolization. Imaging modalities used include transesophageal echocardiogram (TEE), CT angiography and magnetic resonance angiography (MRA). The mainstays of medical treatment are antiplatelets and statin. The role of anticoagulation is reserved for plaques with thrombotic component. There were two patients who presented with large acute ischemic stroke with high grade, floating aortic arch thrombus and complex aortic arch plaques. In one of cases, after 10-day follow-up CT aortic angiography showed completely resolved thrombus after being treated with IV tissue plasminogen activator (TPA) followed by low molecular weight heparin (LMWH). The risk of embolism depends on size of aortic plaques and mobility. TEE is modality of choice for thoracic aortic plaques. Aortic plaques >4 mm are independent predictors of recurrent ischemic stroke. There are limited data available for off-label use of intravenous thrombolysis and mechanical thrombectomy (MT) in presence of aortic arch thrombus in acute ischemic strokes. These two case reports help in recognition of aortic arch complex plaques as independent risk factor for recurrent stroke. The right patients may consider about the use of intravenous alteplase and MT performed via trans-brachial access after excluding aortic dissection and aneurysm. In future, multicenter, randomized controlled trials will be required for safety of IV TPA and MT.
Collapse
Affiliation(s)
- Liaquat Ali
- Neurology, Hamad General Hospital, Doha, QAT
| | - Abeer Safan
- Neurology, Hamad Medical Corporation, Doha, QAT
| | | | | | | |
Collapse
|
12
|
Small Leucine-Rich Proteoglycans (SLRPs) in the Retina. Int J Mol Sci 2021; 22:ijms22147293. [PMID: 34298915 PMCID: PMC8305803 DOI: 10.3390/ijms22147293] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 01/17/2023] Open
Abstract
Retinal diseases such as age-related macular degeneration (AMD), retinopathy of prematurity (ROP), and diabetic retinopathy (DR) are the leading causes of visual impairment worldwide. There is a critical need to understand the structural and cellular components that play a vital role in the pathophysiology of retinal diseases. One potential component is the family of structural proteins called small leucine-rich proteoglycans (SLRPs). SLRPs are crucial in many fundamental biological processes involved in the maintenance of retinal homeostasis. They are present within the extracellular matrix (ECM) of connective and vascular tissues and contribute to tissue organization and modulation of cell growth. They play a vital role in cell–matrix interactions in many upstream signaling pathways involved in fibrillogenesis and angiogenesis. In this comprehensive review, we describe the expression patterns and function of SLRPs in the retina, including Biglycan and Decorin from class I; Fibromodulin, Lumican, and a Proline/arginine-rich end leucine-rich repeat protein (PRELP) from class II; Opticin and Osteoglycin/Mimecan from class III; and Chondroadherin (CHAD), Tsukushi and Nyctalopin from class IV.
Collapse
|
13
|
The Importance of Lipoprotein Lipase Regulation in Atherosclerosis. Biomedicines 2021; 9:biomedicines9070782. [PMID: 34356847 PMCID: PMC8301479 DOI: 10.3390/biomedicines9070782] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 02/07/2023] Open
Abstract
Lipoprotein lipase (LPL) plays a major role in the lipid homeostasis mainly by mediating the intravascular lipolysis of triglyceride rich lipoproteins. Impaired LPL activity leads to the accumulation of chylomicrons and very low-density lipoproteins (VLDL) in plasma, resulting in hypertriglyceridemia. While low-density lipoprotein cholesterol (LDL-C) is recognized as a primary risk factor for atherosclerosis, hypertriglyceridemia has been shown to be an independent risk factor for cardiovascular disease (CVD) and a residual risk factor in atherosclerosis development. In this review, we focus on the lipolysis machinery and discuss the potential role of triglycerides, remnant particles, and lipolysis mediators in the onset and progression of atherosclerotic cardiovascular disease (ASCVD). This review details a number of important factors involved in the maturation and transportation of LPL to the capillaries, where the triglycerides are hydrolyzed, generating remnant lipoproteins. Moreover, LPL and other factors involved in intravascular lipolysis are also reported to impact the clearance of remnant lipoproteins from plasma and promote lipoprotein retention in capillaries. Apolipoproteins (Apo) and angiopoietin-like proteins (ANGPTLs) play a crucial role in regulating LPL activity and recent insights into LPL regulation may elucidate new pharmacological means to address the challenge of hypertriglyceridemia in atherosclerosis development.
Collapse
|
14
|
Abstract
The a disintegrin-like and metalloproteinase with thrombospondin motif (ADAMTS) family comprises 19 proteases that regulate the structure and function of extracellular proteins in the extracellular matrix and blood. The best characterized cardiovascular role is that of ADAMTS-13 in blood. Moderately low ADAMTS-13 levels increase the risk of ischeamic stroke and very low levels (less than 10%) can cause thrombotic thrombocytopenic purpura (TTP). Recombinant ADAMTS-13 is currently in clinical trials for treatment of TTP. Recently, new cardiovascular roles for ADAMTS proteases have been discovered. Several ADAMTS family members are important in the development of blood vessels and the heart, especially the valves. A number of studies have also investigated the potential role of ADAMTS-1, -4 and -5 in cardiovascular disease. They cleave proteoglycans such as versican, which represent major structural components of the arteries. ADAMTS-7 and -8 are attracting considerable interest owing to their implication in atherosclerosis and pulmonary arterial hypertension, respectively. Mutations in the ADAMTS19 gene cause progressive heart valve disease and missense variants in ADAMTS6 are associated with cardiac conduction. In this review, we discuss in detail the evidence for these and other cardiovascular roles of ADAMTS family members, their proteolytic substrates and the potential molecular mechanisms involved.
Collapse
Affiliation(s)
- Salvatore Santamaria
- Centre for Haematology, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Rens de Groot
- Centre for Haematology, Imperial College London, Du Cane Road, London W12 0NN, UK.,Institute of Cardiovascular Science, University College London, 51 Chenies Mews, London WC1E 6HX, UK
| |
Collapse
|
15
|
Hu Y, Meuret C, Martinez A, Yassine HN, Nedelkov D. Distinct patterns of apolipoprotein C-I, C-II, and C-III isoforms are associated with markers of Alzheimer's disease. J Lipid Res 2020; 62:100014. [PMID: 33518512 PMCID: PMC7859854 DOI: 10.1194/jlr.ra120000919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023] Open
Abstract
Apolipoproteins C-I, C-II, and C-III interact with ApoE to regulate lipoprotein metabolism and contribute to Alzheimer's disease pathophysiology. In plasma, apoC-I and C-II exist as truncated isoforms, while apoC-III exhibits multiple glycoforms. This study aimed to 1) delineate apoC-I, C-II, and C-III isoform profiles in cerebrospinal fluid (CSF) and plasma in a cohort of nondemented older individuals (n = 61), and 2) examine the effect of APOE4 on these isoforms and their correlation with CSF Aβ42, a surrogate of brain amyloid accumulation. The isoforms of the apoCs were immunoaffinity enriched and measured with MALDI-TOF mass spectrometry, revealing a significantly higher percentage of truncated apoC-I and apoC-II in CSF compared with matched plasma, with positive correlation between CSF and plasma. A greater percentage of monosialylated and disialylated apoC-III isoforms was detected in CSF, accompanied by a lower percentage of the two nonsialylated apoC-III isoforms, with significant linear correlations between CSF and plasma. Furthermore, a greater percentage of truncated apoC-I in CSF and apoC-II in plasma and CSF was observed in individuals carrying at least one APOE Ɛ4 allele. Increased apoC-I and apoC-II truncations were associated with lower CSF Aβ42. Finally, monosialylated apoC-III was lower, and disialylated apoC-III greater in the CSF of Ɛ4 carriers. Together, these results reveal distinct patterns of the apoCs isoforms in CSF, implying CSF-specific apoCs processing. These patterns were accentuated in APOE Ɛ4 allele carriers, suggesting an association between APOE4 genotype and Alzheimer's disease pathology with apoCs processing and function in the brain.
Collapse
Affiliation(s)
| | | | - Ashley Martinez
- University of Southern California, Los Angeles, California, USA
| | | | | |
Collapse
|
16
|
Wang Y, Zhang Y, Wei J, Du W, Ding J, Zhang Y, Zhang N, Mao M, Liu P. Vitamin D for inflammation biomarkers in coronary artery disease: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e21407. [PMID: 32756139 PMCID: PMC7402871 DOI: 10.1097/md.0000000000021407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Coronary artery disease (CAD) is a clinically common coronary heart disease. Vitamin D might be beneficial in CAD patients through its favorable effects on inflammation biomarkers. This study will be performed to examine the effects of Vitamin D supplementation on inflammatory markers in CAD patients. METHODS We will search the electronical databases and hand-searching journals or reference lists. The study screening and data extraction will be carried out by 2 investigators independently. The primary outcome is inflammatory biomarkers of peripheral blood. Secondary outcomes are triglyceride, total cholesterol, high-density lipoprotein cholesterol levels, low-density lipoprotein cholesterol, and blood pressure. Review Manager (RevMan, version 5.3; The Nordic Cochrane Centre, Copenhagen, Denmark: http://community.cochrane.org) V.5.3 software will be used to compute the data. RESULTS The results of the study will provide a reliable evidence to assess the efficacy of Vitamin D supplement on inflammation biomarkers of CAD patients. CONCLUSION The conclusion of our systematic review will answer whether Vitamin D is an effective intervention to relieve inflammation of CAD patients. ETHICS Because all of the data used in this review has been published, this review does not require ethical approval. REGISTRATION NUMBER INPLASY202060072.
Collapse
Affiliation(s)
- Yiru Wang
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine
| | - Yifan Zhang
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine
| | - Jing Wei
- Department of Traditional Chinese Medicine, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Wenting Du
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine
| | - Jie Ding
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine
| | - Yiyi Zhang
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine
| | - Na Zhang
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine
| | - Meijiao Mao
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine
| | - Ping Liu
- Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine
| |
Collapse
|
17
|
Ma Z, Mao C, Jia Y, Fu Y, Kong W. Extracellular matrix dynamics in vascular remodeling. Am J Physiol Cell Physiol 2020; 319:C481-C499. [PMID: 32579472 DOI: 10.1152/ajpcell.00147.2020] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Vascular remodeling is the adaptive response to various physiological and pathophysiological alterations that are closely related to aging and vascular diseases. Understanding the mechanistic regulation of vascular remodeling may be favorable for discovering potential therapeutic targets and strategies. The extracellular matrix (ECM), including matrix proteins and their degradative metalloproteases, serves as the main component of the microenvironment and exhibits dynamic changes during vascular remodeling. This process involves mainly the altered composition of matrix proteins, metalloprotease-mediated degradation, posttranslational modification of ECM proteins, and altered topographical features of the ECM. To date, adequate studies have demonstrated that ECM dynamics also play a critical role in vascular remodeling in various diseases. Here, we review these related studies, summarize how ECM dynamics control vascular remodeling, and further indicate potential diagnostic biomarkers and therapeutic targets in the ECM for corresponding vascular diseases.
Collapse
Affiliation(s)
- Zihan Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Chenfeng Mao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Yiting Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
18
|
Khosravi M, Poursaleh A, Ghasempour G, Farhad S, Najafi M. The effects of oxidative stress on the development of atherosclerosis. Biol Chem 2020; 400:711-732. [PMID: 30864421 DOI: 10.1515/hsz-2018-0397] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is a cardiovascular disease (CVD) known widely world wide. Several hypothesizes are suggested to be involved in the narrowing of arteries during process of atherogenesis. The oxidative modification hypothesis is related to oxidative and anti-oxidative imbalance and is the most investigated. The aim of this study was to review the role of oxidative stress in atherosclerosis. Furthermore, it describes the roles of oxidative/anti-oxidative enzymes and compounds in the macromolecular and lipoprotein modifications and in triggering inflammatory events. The reactive oxygen (ROS) and reactive nitrogen species (RNS) are the most important endogenous sources produced by non-enzymatic and enzymatic [myeloperoxidase (MPO), nicotinamide adenine dinucleotide phosphate (NADH) oxidase and lipoxygenase (LO)] reactions that may be balanced with anti-oxidative compounds [glutathione (GSH), polyphenols and vitamins] and enzymes [glutathione peroxidase (Gpx), peroxiredoxins (Prdx), superoxide dismutase (SOD) and paraoxonase (PON)]. However, the oxidative and anti-oxidative imbalance causes the involvement of cellular proliferation and migration signaling pathways and macrophage polarization leads to the formation of atherogenic plaques. On the other hand, the immune occurrences and the changes in extra cellular matrix remodeling can develop atherosclerosis process.
Collapse
Affiliation(s)
- Mohsen Khosravi
- Biochemistry Department, Firoozabadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Adeleh Poursaleh
- Biochemistry Department, Firoozabadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Ghasem Ghasempour
- Biochemistry Department, Firoozabadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Shaikhnia Farhad
- Biochemistry Department, Firoozabadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Microbial Biotechnology Research Center, Biochemistry Department, Firoozabadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Scuruchi M, Potì F, Rodríguez-Carrio J, Campo GM, Mandraffino G. Biglycan and atherosclerosis: Lessons from high cardiovascular risk conditions. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158545. [PMID: 31672572 DOI: 10.1016/j.bbalip.2019.158545] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/26/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
Abstract
Atherosclerosis (ATH) is a chronic, dynamic, evolutive process involving morphological and structural subversion of artery walls, leading to the formation of atherosclerotic plaques. ATH generally initiates during the childhood, occurring as a result of a number of changes in the intima tunica and in the media of arteries. A key event occurring during the pathobiology of ATH is the accumulation of lipoproteins in the sub-intimal spaces mediated by extracellular matrix (ECM) molecules, especially by the chondroitin sulfate/dermatan sulfate (CS/DS) -containing proteoglycans (CS/DSPGs). Among them, the proteoglycan biglycan (BGN) is critically involved in the onset and progression of ATH and evidences show that BGN represents the missing link between the pro-atherogenic status induced by both traditional and non-traditional cardiovascular risk factors and the development and progression of vascular damage. In the light of these findings, the role of BGN in dyslipidemia, hypertension, cigarette smoking, diabetes, chronic kidney disease and inflammatory status is briefly analyzed and discussed in order to shed new light on the underlying mechanisms governing the association between BGN and ATH.
Collapse
Affiliation(s)
- Michele Scuruchi
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Francesco Potì
- Department of Medicine and Surgery-Unit of Neurosciences, University of Parma, Parma, Italy
| | - Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain; Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Oviedo, Spain; Bone and Mineral Research Unit, Instituto Reina Sofía de Investigación Nefrológica, REDinREN Del ISCIII, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Giuseppe Mandraffino
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.
| |
Collapse
|
20
|
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: 0.8] [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
|
21
|
Interaction of arterial proteoglycans with low density lipoproteins (LDLs): From theory to promising therapeutic approaches. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2019. [DOI: 10.1016/j.medntd.2019.100016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
22
|
Mittal N, Yoon SH, Enomoto H, Hiroshi M, Shimizu A, Kawakami A, Fujita M, Watanabe H, Fukuda K, Makino S. Versican is crucial for the initiation of cardiovascular lumen development in medaka (Oryzias latipes). Sci Rep 2019; 9:9475. [PMID: 31263118 PMCID: PMC6603046 DOI: 10.1038/s41598-019-45851-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 06/13/2019] [Indexed: 12/17/2022] Open
Abstract
Versican is an evolutionary conserved extracellular matrix proteoglycan, and versican expression loss in mice results in embryonic lethality owing to cardiovascular defects. However, the in utero development of mammals limits our understanding of the precise role of versican during cardiovascular development. Therefore, the use of evolutionarily distant species that develop ex utero is more suitable for studying the mechanistic basis of versican activity. We performed ENU mutagenesis screening to identify medaka mutants with defects in embryonic cardiovascular development. In this study, we described a recessive point mutation in the versican 3'UTR resulting in reduced versican protein expression. The fully penetrant homozygous mutant showed termination of cardiac development at the linear heart tube stage and exhibited absence of cardiac looping, a constricted outflow tract, and no cardiac jelly. Additionally, progenitor cells did not migrate from the secondary source towards the arterial pole of the linear heart tube, resulting in a constricted outflow tract. Furthermore, mutants lacked blood flow and vascular lumen despite continuous peristaltic heartbeats. These results enhance our understanding of the mechanistic basis of versican in cardiac development, and this mutant represents a novel genetic model to investigate the mechanisms of vascular tubulogenesis.
Collapse
Affiliation(s)
- Nishant Mittal
- Department of Cardiology, Keio University School of Medicine, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Sung Han Yoon
- Department of Interventional Cardiology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, AHSP A9229, Los Angeles, CA, 90048, USA
| | - Hirokazu Enomoto
- Department of Cardiology, Keio University School of Medicine, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Miyama Hiroshi
- Department of Cardiology, Keio University School of Medicine, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Atsushi Shimizu
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Atsushi Kawakami
- Department of Biological Information, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama, 226-8501, Japan
| | - Misato Fujita
- Department of Biological Science, Graduate School of Science, Kanagawa University, 2946 Tsuchiya, Hiratsuka-Shi, Kanagawa-Ken, 259-1293, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-, Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shinji Makino
- Department of Cardiology, Keio University School of Medicine, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan.
- Keio University Health Centre, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
23
|
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: 107] [Impact Index Per Article: 15.3] [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
|
24
|
Townsend D, Hughes E, Akien G, Stewart KL, Radford SE, Rochester D, Middleton DA. Epigallocatechin-3-gallate remodels apolipoprotein A-I amyloid fibrils into soluble oligomers in the presence of heparin. J Biol Chem 2018; 293:12877-12893. [PMID: 29853648 PMCID: PMC6102129 DOI: 10.1074/jbc.ra118.002038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/25/2018] [Indexed: 11/06/2022] Open
Abstract
Amyloid deposits of WT apolipoprotein A-I (apoA-I), the main protein component of high-density lipoprotein, accumulate in atherosclerotic plaques where they may contribute to coronary artery disease by increasing plaque burden and instability. Using CD analysis, solid-state NMR spectroscopy, and transmission EM, we report here a surprising cooperative effect of heparin and the green tea polyphenol (-)-epigallocatechin-3-gallate (EGCG), a known inhibitor and modulator of amyloid formation, on apoA-I fibrils. We found that heparin, a proxy for glycosaminoglycan (GAG) polysaccharides that co-localize ubiquitously with amyloid in vivo, accelerates the rate of apoA-I formation from monomeric protein and associates with insoluble fibrils. Mature, insoluble apoA-I fibrils bound EGCG (KD = 30 ± 3 μm; Bmax = 40 ± 3 μm), but EGCG did not alter the kinetics of apoA-I amyloid assembly from monomer in the presence or absence of heparin. EGCG selectively increased the mobility of specific backbone and side-chain sites of apoA-I fibrils formed in the absence of heparin, but the fibrils largely retained their original morphology and remained insoluble. By contrast, fibrils formed in the presence of heparin were mobilized extensively by the addition of equimolar EGCG, and the fibrils were remodeled into soluble 20-nm-diameter oligomers with a largely α-helical structure that were nontoxic to human umbilical artery endothelial cells. These results argue for a protective effect of EGCG on apoA-I amyloid associated with atherosclerosis and suggest that EGCG-induced remodeling of amyloid may be tightly regulated by GAGs and other amyloid co-factors in vivo, depending on EGCG bioavailability.
Collapse
Affiliation(s)
- David Townsend
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB
| | - Eleri Hughes
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB
| | - Geoffrey Akien
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB
| | - Katie L Stewart
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - David Rochester
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB
| | | |
Collapse
|
25
|
Nahon JE, Hoekstra M, Havik SR, Van Santbrink PJ, Dallinga-Thie GM, Kuivenhoven JA, Geerling JJ, Van Eck M. Proteoglycan 4 regulates macrophage function without altering atherosclerotic lesion formation in a murine bone marrow-specific deletion model. Atherosclerosis 2018; 274:120-127. [DOI: 10.1016/j.atherosclerosis.2018.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 11/15/2022]
|
26
|
Ganjali S, Momtazi-Borojeni AA, Banach M, Kovanen PT, Gotto AM, Sahebkar A. HDL functionality in familial hypercholesterolemia: effects of treatment modalities and pharmacological interventions. Drug Discov Today 2018; 23:171-180. [DOI: 10.1016/j.drudis.2017.09.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/02/2017] [Accepted: 09/25/2017] [Indexed: 01/14/2023]
|
27
|
Lehti S, Nguyen SD, Belevich I, Vihinen H, Heikkilä HM, Soliymani R, Käkelä R, Saksi J, Jauhiainen M, Grabowski GA, Kummu O, Hörkkö S, Baumann M, Lindsberg PJ, Jokitalo E, Kovanen PT, Öörni K. Extracellular Lipids Accumulate in Human Carotid Arteries as Distinct Three-Dimensional Structures and Have Proinflammatory Properties. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:525-538. [PMID: 29154769 DOI: 10.1016/j.ajpath.2017.09.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 09/11/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022]
Abstract
Lipid accumulation is a key characteristic of advancing atherosclerotic lesions. Herein, we analyzed the ultrastructure of the accumulated lipids in endarterectomized human carotid atherosclerotic plaques using three-dimensional (3D) electron microscopy, a method never used in this context before. 3D electron microscopy revealed intracellular lipid droplets and extracellular lipoprotein particles. Most of the particles were aggregated, and some connected to needle-shaped or sheet-like cholesterol crystals. Proteomic analysis of isolated extracellular lipoprotein particles revealed that apolipoprotein B is their main protein component, indicating their origin from low-density lipoprotein, intermediate-density lipoprotein, very-low-density lipoprotein, lipoprotein (a), or chylomicron remnants. The particles also contained small exchangeable apolipoproteins, complement components, and immunoglobulins. Lipidomic analysis revealed differences between plasma lipoproteins and the particles, thereby indicating involvement of lipolytic enzymes in their generation. Incubation of human monocyte-derived macrophages with the isolated extracellular lipoprotein particles or with plasma lipoproteins that had been lipolytically modified in vitro induced intracellular lipid accumulation and triggered inflammasome activation in them. Taken together, extracellular lipids accumulate in human carotid plaques as distinct 3D structures that include aggregated and fused lipoprotein particles and cholesterol crystals. The particles originate from plasma lipoproteins, show signs of lipolytic modifications, and associate with cholesterol crystals. By inducing intracellular cholesterol accumulation (ie, foam cell formation) and inflammasome activation, the extracellular lipoprotein particles may actively enhance atherogenesis.
Collapse
Affiliation(s)
- Satu Lehti
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Su D Nguyen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Ilya Belevich
- Electron Microscopy Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Helena Vihinen
- Electron Microscopy Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Hanna M Heikkilä
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Rabah Soliymani
- Clinical Proteomics Core Facility, Medicum-Biochemistry and Developmental Biology, School of Medicine, University of Helsinki, Helsinki, Finland
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit, Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Jani Saksi
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Matti Jauhiainen
- National Institute for Health and Welfare, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Gregory A Grabowski
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Kiniksa Pharmaceuticals, Ltd., Wellesley, Massachusetts
| | - Outi Kummu
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Sohvi Hörkkö
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland; Medical Research Center and Nordlab Oulu, University Hospital and University of Oulu, Oulu, Finland
| | - Marc Baumann
- Clinical Proteomics Core Facility, Medicum-Biochemistry and Developmental Biology, School of Medicine, University of Helsinki, Helsinki, Finland
| | - Perttu J Lindsberg
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland; Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eija Jokitalo
- Electron Microscopy Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Petri T Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland; Helsinki University Lipidomics Unit, Department of Biosciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
28
|
Elevated circulating TGF-β is not the cause of increased atherosclerosis development in biglycan deficient mice. Atherosclerosis 2017; 268:68-75. [PMID: 29182988 DOI: 10.1016/j.atherosclerosis.2017.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 10/14/2017] [Accepted: 11/09/2017] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS Vascular biglycan contributes to atherosclerosis development and increased biglycan expression correlates with increased atherosclerosis. However, mice deficient in biglycan have either no reduction in atherosclerosis or an unexpected increase in atherosclerosis. Biglycan deficient mice have systemically elevated TGF-β, likely due to lack of sequestration of TGF-β in the extracellular matrix. The purpose of this study was to determine if prevention of TGF-β elevations in biglycan deficient mice affected atherosclerosis development. METHODS Biglycan deficient mice were crossed to Ldlr deficient mice. Diabetes was induced via streptozotocin and all mice were fed a high cholesterol diet. Diabetic biglycan wild type and biglycan deficient Ldlr deficient mice were injected with the TGF-β neutralizing antibody 1D11 or the irrelevant control antibody 13C4. RESULTS Biglycan deficient mice had significantly elevated plasma TGF-β levels, which was further increased by diabetes, and significantly increased atherosclerosis. There was a significant correlation between TGF-β concentrations and atherosclerosis. However, despite nearly complete suppression of plasma TGF-β levels in mice treated with the TGF-β neutralizing antibody 1D11, there was no significant difference in atherosclerosis between mice with elevated TGF-β levels and mice with suppressed TGF-β levels. CONCLUSIONS The increased atherosclerosis in biglycan deficient mice does not appear to be due to elevations in TGF-β.
Collapse
|
29
|
Ujueta F, Weiss EN, Shah B, Sedlis SP. Effect of Percutaneous Coronary Intervention on Survival in Patients with Stable Ischemic Heart Disease. Curr Cardiol Rep 2017; 19:17. [PMID: 28213668 DOI: 10.1007/s11886-017-0821-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW This study aims to determine if percutaneous coronary intervention (PCI) does improve survival in stable ischemic heart disease (SIHD). RECENT FINDINGS The International Study of Comparative Health Effectiveness with Medical and Invasive Approaches (ISCHEMIA) trial will evaluate patients with moderate to severe ischemia and will be the largest randomized trial of an initial management strategy of coronary revascularization (percutaneous or surgical) versus optimal medical therapy alone for SIHD. Although the ISCHEMIA trial may show a benefit with upfront coronary revascularization in this high-risk population, cardiac events after PCI are largely caused by plaque rupture in segments outside of the original stented segment. Furthermore, given the robust data from prior randomized trials, which showed no survival benefit with PCI, and the likelihood that the highest risk patients in ISCHEMIA will be treated with surgery, it is unlikely that the ISCHEMIA trial will show a survival benefit particular to PCI. RECENT FINDINGS Although PCI relieves symptoms, the evidence base indicates that it does not prolong survival in SIHD.
Collapse
Affiliation(s)
- Francisco Ujueta
- Department of Medicine Division of Cardiology New York VA Healthcare Network and New York University School of Medicine, 423 East 23rd Street, New York, NY, 10010, USA
| | - Ephraim N Weiss
- Department of Medicine Division of Cardiology New York VA Healthcare Network and New York University School of Medicine, 423 East 23rd Street, New York, NY, 10010, USA
| | - Binita Shah
- Department of Medicine Division of Cardiology New York VA Healthcare Network and New York University School of Medicine, 423 East 23rd Street, New York, NY, 10010, USA
| | - Steven P Sedlis
- Department of Medicine Division of Cardiology New York VA Healthcare Network and New York University School of Medicine, 423 East 23rd Street, New York, NY, 10010, USA.
| |
Collapse
|
30
|
Rosú SA, Toledo L, Urbano BF, Sanchez SA, Calabrese GC, Tricerri MA. Learning from Synthetic Models of Extracellular Matrix; Differential Binding of Wild Type and Amyloidogenic Human Apolipoprotein A-I to Hydrogels Formed from Molecules Having Charges Similar to Those Found in Natural GAGs. Protein J 2017. [PMID: 28634774 DOI: 10.1007/s10930-017-9728-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Among other components of the extracellular matrix (ECM), glycoproteins and glycosaminoglycans (GAGs) have been strongly associated to the retention or misfolding of different proteins inducing the formation of deposits in amyloid diseases. The composition of these molecules is highly diverse and a key issue seems to be the equilibrium between physiological and pathological events. In order to have a model in which the composition of the matrix could be finely controlled, we designed and synthesized crosslinked hydrophilic polymers, the so-called hydrogels varying the amounts of negative charges and hydroxyl groups that are prevalent in GAGs. We checked and compared by fluorescence techniques the binding of human apolipoprotein A-I and a natural mutant involved in amyloidosis to the hydrogel scaffolds. Our results indicate that both proteins are highly retained as long as the negative charge increases, and in addition it was shown that the mutant is more retained than the Wt, indicating that the retention of specific proteins in the ECM could be part of the pathogenicity. These results show the importance of the use of these polymers as a model to get deep insight into the studies of proteins within macromolecules.
Collapse
Affiliation(s)
- Silvana A Rosú
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET, La Plata, Buenos Aires, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120 S/N La Plata, 1900, La Plata, Buenos Aires, Argentina
| | - Leandro Toledo
- Departamento de Polímeros, Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile
| | - Bruno F Urbano
- Departamento de Polímeros, Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile
| | - Susana A Sanchez
- Departamento de Polímeros, Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile
| | - Graciela C Calabrese
- Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 954, Primer Piso, C1113AAD, Buenos Aires, Argentina
| | - M Alejandra Tricerri
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET, La Plata, Buenos Aires, Argentina. .,Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120 S/N La Plata, 1900, La Plata, Buenos Aires, Argentina.
| |
Collapse
|
31
|
Ganjali S, Momtazi AA, Banach M, Kovanen PT, Stein EA, Sahebkar A. HDL abnormalities in familial hypercholesterolemia: Focus on biological functions. Prog Lipid Res 2017; 67:16-26. [DOI: 10.1016/j.plipres.2017.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/25/2017] [Accepted: 05/10/2017] [Indexed: 02/07/2023]
|
32
|
The arterial microenvironment: the where and why of atherosclerosis. Biochem J 2017; 473:1281-95. [PMID: 27208212 DOI: 10.1042/bj20150844] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 02/15/2016] [Indexed: 12/11/2022]
Abstract
The formation of atherosclerotic plaques in the large and medium sized arteries is classically driven by systemic factors, such as elevated cholesterol and blood pressure. However, work over the past several decades has established that atherosclerotic plaque development involves a complex coordination of both systemic and local cues that ultimately determine where plaques form and how plaques progress. Although current therapeutics for atherosclerotic cardiovascular disease primarily target the systemic risk factors, a large array of studies suggest that the local microenvironment, including arterial mechanics, matrix remodelling and lipid deposition, plays a vital role in regulating the local susceptibility to plaque development through the regulation of vascular cell function. Additionally, these microenvironmental stimuli are capable of tuning other aspects of the microenvironment through collective adaptation. In this review, we will discuss the components of the arterial microenvironment, how these components cross-talk to shape the local microenvironment, and the effect of microenvironmental stimuli on vascular cell function during atherosclerotic plaque formation.
Collapse
|
33
|
Reimann C, Brangsch J, Colletini F, Walter T, Hamm B, Botnar RM, Makowski MR. Molecular imaging of the extracellular matrix in the context of atherosclerosis. Adv Drug Deliv Rev 2017; 113:49-60. [PMID: 27639968 DOI: 10.1016/j.addr.2016.09.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/01/2016] [Accepted: 09/07/2016] [Indexed: 12/25/2022]
Abstract
This review summarizes the current status of molecular imaging of the extracellular matrix (ECM) in the context of atherosclerosis. Apart from cellular components, the ECM of the atherosclerotic plaque plays a relevant role during the initiation of atherosclerosis and its' subsequent progression. Important structural and signaling components of the ECM include elastin, collagen and fibrin. However, the ECM not only plays a structural role in the arterial wall but also interacts with different cell types and has important biological signaling functions. Molecular imaging of the ECM has emerged as a new diagnostic tool to characterize biological aspects of atherosclerotic plaques, which cannot be characterized by current clinically established imaging techniques, such as X-ray angiography. Different types of molecular probes can be detected in vivo by imaging modalities such as magnetic resonance imaging (MRI), positron emission tomography (PET) and single photon emission computed tomography (SPECT). The modality specific signaling component of the molecular probe provides information about its spatial location and local concentration. The successful introduction of molecular imaging into clinical practice and guidelines could open new pathways for an earlier detection of disease processes and a better understanding of the disease state on a biological level. Quantitative in vivo molecular parameters could also contribute to the development and evaluation of novel cardiovascular therapeutic interventions and the assessment of response to treatment.
Collapse
Affiliation(s)
| | | | | | - Thula Walter
- Department of Radiology, Charité, Berlin, Germany
| | - Bernd Hamm
- Department of Radiology, Charité, Berlin, Germany
| | - Rene M Botnar
- King's College London, Division of Imaging Sciences, United Kingdom; Wellcome Trust and EPSRC Medical Engineering Center, United Kingdom; BHF Centre of Excellence, King's College London, London, United Kingdom; NIHR Biomedical Research Centre, King's College London, London, United Kingdom
| | - Marcus R Makowski
- Department of Radiology, Charité, Berlin, Germany; King's College London, Division of Imaging Sciences, United Kingdom.
| |
Collapse
|
34
|
Choi HY, Hafiane A, Schwertani A, Genest J. High-Density Lipoproteins: Biology, Epidemiology, and Clinical Management. Can J Cardiol 2017; 33:325-333. [DOI: 10.1016/j.cjca.2016.09.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 09/18/2016] [Accepted: 09/19/2016] [Indexed: 01/29/2023] Open
|
35
|
Hara T, Yoshida E, Shinkai Y, Yamamoto C, Fujiwara Y, Kumagai Y, Kaji T. Biglycan Intensifies ALK5-Smad2/3 Signaling by TGF-β 1 and Downregulates Syndecan-4 in Cultured Vascular Endothelial Cells. J Cell Biochem 2017; 118:1087-1096. [PMID: 27585241 PMCID: PMC6221004 DOI: 10.1002/jcb.25721] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022]
Abstract
Proteoglycans are macromolecules that consist of a core protein and one or more glycosaminoglycan side chains. A small leucine‐rich dermatan sulfate proteoglycan, biglycan, is one of the predominant types of proteoglycans synthesized by vascular endothelial cells; however, the physiological functions of biglycan are not completely understood. In the present study, bovine aortic endothelial cells in culture were transfected with small interfering RNAs for biglycan, and the expression of other proteoglycans was examined. Transforming growth factor‐β1 signaling was also investigated, because the interaction of biglycan with cytokines has been reported. Biglycan was found to form a complex with either transforming growth factor‐β1 or the transforming growth factor‐β1 type I receptor, ALK5, and to intensify the phosphorylation of Smad2/3, resulting in a lower expression of the transmembrane heparan sulfate proteoglycan, syndecan‐4. This is the first report to clarify the function of biglycan as a regulatory molecule of the ALK5–Smad2/3 TGF‐β1 signaling pathway that mediates the suppression of syndecan‐4 expression in vascular endothelial cells. J. Cell. Biochem. 118: 1087–1096, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Takato Hara
- Faculty of Pharmaceutical Sciences, Department of Environmental Health, Tokyo University of Science, 2641 Yamazaki, Noda, 278-8510, Japan
| | - Eiko Yoshida
- Faculty of Pharmaceutical Sciences, Department of Environmental Health, Tokyo University of Science, 2641 Yamazaki, Noda, 278-8510, Japan
| | - Yasuhiro Shinkai
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan
| | - Chika Yamamoto
- Faculty of Pharmaceutical Sciences, Department of Environmental Health, Toho University, 2-2-1 Miyama, Funabashi, 274-8510, Japan
| | - Yasuyuki Fujiwara
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, 192-0392, Japan
| | - Yoshito Kumagai
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan
| | - Toshiyuki Kaji
- Faculty of Pharmaceutical Sciences, Department of Environmental Health, Tokyo University of Science, 2641 Yamazaki, Noda, 278-8510, Japan
| |
Collapse
|
36
|
Abstract
The vasculature is essential for proper organ function. Many pathologies are directly and indirectly related to vascular dysfunction, which causes significant morbidity and mortality. A common pathophysiological feature of diseased vessels is extracellular matrix (ECM) remodelling. Analysing the protein composition of the ECM by conventional antibody-based techniques is challenging; alternative splicing or post-translational modifications, such as glycosylation, can mask epitopes required for antibody recognition. By contrast, proteomic analysis by mass spectrometry enables the study of proteins without the constraints of antibodies. Recent advances in proteomic techniques make it feasible to characterize the composition of the vascular ECM and its remodelling in disease. These developments may lead to the discovery of novel prognostic and diagnostic markers. Thus, proteomics holds potential for identifying ECM signatures to monitor vascular disease processes. Furthermore, a better understanding of the ECM remodelling processes in the vasculature might make ECM-associated proteins more attractive targets for drug discovery efforts. In this review, we will summarize the role of the ECM in the vasculature. Then, we will describe the challenges associated with studying the intricate network of ECM proteins and the current proteomic strategies to analyse the vascular ECM in metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- M Lynch
- King's British Heart Foundation Centre, King's College London, London, UK
| | | | | | - M Mayr
- King's British Heart Foundation Centre, King's College London, London, UK.
| |
Collapse
|
37
|
Hsu JJ, Lim J, Tintut Y, Demer LL. Cell-matrix mechanics and pattern formation in inflammatory cardiovascular calcification. Heart 2016; 102:1710-1715. [PMID: 27406839 DOI: 10.1136/heartjnl-2016-309667] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/20/2016] [Indexed: 12/24/2022] Open
Abstract
Calcific diseases of the cardiovascular system, such as atherosclerotic calcification and calcific aortic valve disease, are widespread and clinically significant, causing substantial morbidity and mortality. Vascular cells, like bone cells, interact with their matrix substrate through molecular signals, and through biomechanical signals, such as traction forces transmitted from cytoskeleton to matrix. The interaction of contractile vascular cells with their matrix may be one of the most important factors controlling pathological mineralisation of the artery wall and cardiac valves. In many respects, the matricrine and matrix mechanical changes in calcific vasculopathy and valvulopathy resemble those occurring in embryonic bone development and normal bone mineralisation. The matrix proteins provide a microenvironment for propagation of crystal growth and provide mechanical cues to the cells that direct differentiation. Small contractions of the cytoskeleton may tug on integrin links to sites on matrix proteins, and thereby sense the stiffness, possibly through deformation of binding proteins causing release of differentiation factors such as products of the members of the transforming growth factor-β superfamily. Inflammation and matrix characteristics are intertwined: inflammation alters the matrix such as through matrix metalloproteinases, while matrix mechanical properties affect cellular sensitivity to inflammatory cytokines. The adhesive properties of the matrix also regulate self-organisation of vascular cells into patterns through reaction-diffusion phenomena and left-right chirality. In this review, we summarise the roles of extracellular matrix proteins and biomechanics in the development of inflammatory cardiovascular calcification.
Collapse
Affiliation(s)
- Jeffrey J Hsu
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Jina Lim
- Department of Pediatrics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Yin Tintut
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, California, USA Department of Physiology, University of California, Los Angeles (UCLA), Los Angeles, California, USA Department of Orthopaedic Surgery, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Linda L Demer
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, California, USA Department of Physiology, University of California, Los Angeles (UCLA), Los Angeles, California, USA Department of Bioengineering, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| |
Collapse
|
38
|
Ollikainen E, Tulamo R, Lehti S, Lee-Rueckert M, Hernesniemi J, Niemelä M, Ylä-Herttuala S, Kovanen PT, Frösen J. Smooth Muscle Cell Foam Cell Formation, Apolipoproteins, and ABCA1 in Intracranial Aneurysms: Implications for Lipid Accumulation as a Promoter of Aneurysm Wall Rupture. J Neuropathol Exp Neurol 2016; 75:689-99. [PMID: 27283327 DOI: 10.1093/jnen/nlw041] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Saccular intracranial aneurysm (sIA) aneurysm causes intracranial hemorrhages that are associated with high mortality. Lipid accumulation and chronic inflammation occur in the sIA wall. A major mechanism for lipid clearance from arteries is adenosine triphosphate-binding cassette A1 (ABCA1)-mediated lipid efflux from foam cells to apolipoprotein A-I (apoA-I). We investigated the association of wall degeneration, inflammation, and lipid-related parameters in tissue samples of 16 unruptured and 20 ruptured sIAs using histology and immunohistochemistry. Intracellular lipid accumulation was associated with wall remodeling (p = 0.005) and rupture (p = 0.020). Foam cell formation was observed in smooth muscle cells, in addition to CD68- and CD163-positive macrophages. Macrophage infiltration correlated with intracellular lipid accumulation and apolipoproteins, including apoA-I. ApoA-I correlated with markers of lipid accumulation and wall degeneration (p = 0.01). ApoA-I-positive staining colocalized with ABCA1-positive cells particularly in sIAs with high number of smooth muscle cells (p = 0.003); absence of such colocalization was associated with wall degeneration (p = 0.017). Known clinical risk factors for sIA rupture correlated inversely with apoA-I. We conclude that lipid accumulation associates with sIA wall degeneration and risk of rupture, possibly via formation of foam cells and subsequent loss of mural cells. Reduced removal of lipids from the sIA wall via ABCA1-apoA-I pathway may contribute to this process.
Collapse
Affiliation(s)
- Eliisa Ollikainen
- From the Biomedicum, Neurosurgery Research Group, Helsinki, Finland (EO, RT, JH, MN, JF); Biomedicum, Wihuri Research Institute, Helsinki, Finland (EO, SL, ML-R, PTK); Department of Vascular Surgery, Helsinki University Central Hospital, Helsinki, Finland (RT); Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland (JH, MN); Department of Molecular Medicine, AIV-Institute, Kuopio, Finland, University of Eastern Finland (SY-H); Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland (JF); and Hemorrhagic Brain Pathology Research Group, Kuopio University Hospital, Kuopio, Finland (JF)
| | - Riikka Tulamo
- From the Biomedicum, Neurosurgery Research Group, Helsinki, Finland (EO, RT, JH, MN, JF); Biomedicum, Wihuri Research Institute, Helsinki, Finland (EO, SL, ML-R, PTK); Department of Vascular Surgery, Helsinki University Central Hospital, Helsinki, Finland (RT); Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland (JH, MN); Department of Molecular Medicine, AIV-Institute, Kuopio, Finland, University of Eastern Finland (SY-H); Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland (JF); and Hemorrhagic Brain Pathology Research Group, Kuopio University Hospital, Kuopio, Finland (JF)
| | - Satu Lehti
- From the Biomedicum, Neurosurgery Research Group, Helsinki, Finland (EO, RT, JH, MN, JF); Biomedicum, Wihuri Research Institute, Helsinki, Finland (EO, SL, ML-R, PTK); Department of Vascular Surgery, Helsinki University Central Hospital, Helsinki, Finland (RT); Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland (JH, MN); Department of Molecular Medicine, AIV-Institute, Kuopio, Finland, University of Eastern Finland (SY-H); Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland (JF); and Hemorrhagic Brain Pathology Research Group, Kuopio University Hospital, Kuopio, Finland (JF)
| | - Miriam Lee-Rueckert
- From the Biomedicum, Neurosurgery Research Group, Helsinki, Finland (EO, RT, JH, MN, JF); Biomedicum, Wihuri Research Institute, Helsinki, Finland (EO, SL, ML-R, PTK); Department of Vascular Surgery, Helsinki University Central Hospital, Helsinki, Finland (RT); Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland (JH, MN); Department of Molecular Medicine, AIV-Institute, Kuopio, Finland, University of Eastern Finland (SY-H); Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland (JF); and Hemorrhagic Brain Pathology Research Group, Kuopio University Hospital, Kuopio, Finland (JF)
| | - Juha Hernesniemi
- From the Biomedicum, Neurosurgery Research Group, Helsinki, Finland (EO, RT, JH, MN, JF); Biomedicum, Wihuri Research Institute, Helsinki, Finland (EO, SL, ML-R, PTK); Department of Vascular Surgery, Helsinki University Central Hospital, Helsinki, Finland (RT); Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland (JH, MN); Department of Molecular Medicine, AIV-Institute, Kuopio, Finland, University of Eastern Finland (SY-H); Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland (JF); and Hemorrhagic Brain Pathology Research Group, Kuopio University Hospital, Kuopio, Finland (JF)
| | - Mika Niemelä
- From the Biomedicum, Neurosurgery Research Group, Helsinki, Finland (EO, RT, JH, MN, JF); Biomedicum, Wihuri Research Institute, Helsinki, Finland (EO, SL, ML-R, PTK); Department of Vascular Surgery, Helsinki University Central Hospital, Helsinki, Finland (RT); Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland (JH, MN); Department of Molecular Medicine, AIV-Institute, Kuopio, Finland, University of Eastern Finland (SY-H); Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland (JF); and Hemorrhagic Brain Pathology Research Group, Kuopio University Hospital, Kuopio, Finland (JF)
| | - Seppo Ylä-Herttuala
- From the Biomedicum, Neurosurgery Research Group, Helsinki, Finland (EO, RT, JH, MN, JF); Biomedicum, Wihuri Research Institute, Helsinki, Finland (EO, SL, ML-R, PTK); Department of Vascular Surgery, Helsinki University Central Hospital, Helsinki, Finland (RT); Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland (JH, MN); Department of Molecular Medicine, AIV-Institute, Kuopio, Finland, University of Eastern Finland (SY-H); Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland (JF); and Hemorrhagic Brain Pathology Research Group, Kuopio University Hospital, Kuopio, Finland (JF)
| | - Petri T Kovanen
- From the Biomedicum, Neurosurgery Research Group, Helsinki, Finland (EO, RT, JH, MN, JF); Biomedicum, Wihuri Research Institute, Helsinki, Finland (EO, SL, ML-R, PTK); Department of Vascular Surgery, Helsinki University Central Hospital, Helsinki, Finland (RT); Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland (JH, MN); Department of Molecular Medicine, AIV-Institute, Kuopio, Finland, University of Eastern Finland (SY-H); Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland (JF); and Hemorrhagic Brain Pathology Research Group, Kuopio University Hospital, Kuopio, Finland (JF)
| | - Juhana Frösen
- From the Biomedicum, Neurosurgery Research Group, Helsinki, Finland (EO, RT, JH, MN, JF); Biomedicum, Wihuri Research Institute, Helsinki, Finland (EO, SL, ML-R, PTK); Department of Vascular Surgery, Helsinki University Central Hospital, Helsinki, Finland (RT); Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland (JH, MN); Department of Molecular Medicine, AIV-Institute, Kuopio, Finland, University of Eastern Finland (SY-H); Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland (JF); and Hemorrhagic Brain Pathology Research Group, Kuopio University Hospital, Kuopio, Finland (JF)
| |
Collapse
|
39
|
Liang W, Ward LJ, Karlsson H, Ljunggren SA, Li W, Lindahl M, Yuan XM. Distinctive proteomic profiles among different regions of human carotid plaques in men and women. Sci Rep 2016; 6:26231. [PMID: 27198765 PMCID: PMC4873748 DOI: 10.1038/srep26231] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/28/2016] [Indexed: 11/20/2022] Open
Abstract
The heterogeneity of atherosclerotic tissue has limited comprehension in proteomic and metabolomic analyses. To elucidate the functional implications, and differences between genders, of atherosclerotic lesion formation we investigated protein profiles from different regions of human carotid atherosclerotic arteries; internal control, fatty streak, plaque shoulder, plaque centre, and fibrous cap. Proteomic analysis was performed using 2-DE with MALDI-TOF, with validation using nLC-MS/MS. Protein mapping of 2-DE identified 52 unique proteins, including 15 previously unmapped proteins, of which 41 proteins were confirmed by nLC-MS/MS analysis. Expression levels of 18 proteins were significantly altered in plaque regions compared to the internal control region. Nine proteins showed site-specific alterations, irrespective of gender, with clear associations to extracellular matrix remodelling. Five proteins display gender-specific alterations with 2-DE, with two alterations validated by nLC-MS/MS. Gender differences in ferritin light chain and transthyretin were validated using both techniques. Validation of immunohistochemistry confirmed significantly higher levels of ferritin in plaques from male patients. Proteomic analysis of different plaque regions has reduced the effects of plaque heterogeneity, and significant differences in protein expression are determined in specific regions and between genders. These proteomes have functional implications in plaque progression and are of importance in understanding gender differences in atherosclerosis.
Collapse
Affiliation(s)
- Wenzhao Liang
- Occupational and Environmental Medicine Center, and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Liam J Ward
- Occupational and Environmental Medicine Center, and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Division of Obstetrics and Gynaecology, and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Helen Karlsson
- Occupational and Environmental Medicine Center, and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Stefan A Ljunggren
- Occupational and Environmental Medicine Center, and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Wei Li
- Division of Obstetrics and Gynaecology, and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Mats Lindahl
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Xi-Ming Yuan
- Occupational and Environmental Medicine Center, and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
40
|
Fernández-Navarro J, Aldea P, de Hoz R, Salazar JJ, Ramírez AI, Rojas B, Gallego BI, Triviño A, Tejerina T, Ramírez JM. Neuroprotective Effects of Low-Dose Statins in the Retinal Ultrastructure of Hypercholesterolemic Rabbits. PLoS One 2016; 11:e0154800. [PMID: 27144842 PMCID: PMC4856380 DOI: 10.1371/journal.pone.0154800] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 04/19/2016] [Indexed: 11/19/2022] Open
Abstract
To evaluate the pleiotropic effects to statins, we analyze the qualitative and quantitative retinal changes in hypercholesterolemic rabbits after a low-dosage statin treatment. For this purpose, New Zealand rabbits were split into three groups: control (G0; n = 10), fed a standard diet; hypercholesterolemic (G1; n = 8), fed a 0.5% cholesterol-enriched diet for 8 months; and statins (G2; n = 8), fed a 0.5% cholesterol-enriched diet for 8 months, together with the administration of statin (pravastatin or fluvastatin sodium) at a dose of 2 mg / kg / day each diet. The retinas were analyzed by transmission electron microscopy and immunohistochemistry (glial fibrillary acidic protein). The retinal thickness of nuclear and plexiform layers were quantified in semi-thin sections. The results revealed that the low-statin-treated rabbits in comparison with the hypercholesterolemic group showed: i) a more preserved structure in all retinal layers; ii) a significant reduction in retinal thickness; iii) a decrease in cell death in the nuclear-and ganglion-cell layers; iv) a reduction of hydropic degeneration in the plexiform and nerve-fiber layers; v) a preservation of astrocytes and of the retinal area occupied by them; and vi) a better-preserved retinal vascular structure. Our findings indicate that low doses of statins can prevent retinal degeneration, acting on retinal macroglia, neurons and retinal vessels, despite that hypercholesterolemia remained unchanged. Thus, the pleiotropic effects of the statins may help safeguard the retinal ultrastructure.
Collapse
Affiliation(s)
- Judith Fernández-Navarro
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo. Universidad Complutense Madrid (UCM), Spain
| | - Pilar Aldea
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo. Universidad Complutense Madrid (UCM), Spain
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo. Universidad Complutense Madrid (UCM), Spain
- Facultad de Óptica y Optometría, UCM, Spain
| | - Juan J Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo. Universidad Complutense Madrid (UCM), Spain
- Facultad de Óptica y Optometría, UCM, Spain
| | - Ana I Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo. Universidad Complutense Madrid (UCM), Spain
- Facultad de Óptica y Optometría, UCM, Spain
| | - Blanca Rojas
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo. Universidad Complutense Madrid (UCM), Spain
- Facultad de Medicina, UCM, Spain
| | - Beatriz I. Gallego
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo. Universidad Complutense Madrid (UCM), Spain
- Facultad de Óptica y Optometría, UCM, Spain
| | - Alberto Triviño
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo. Universidad Complutense Madrid (UCM), Spain
- Facultad de Medicina, UCM, Spain
| | - Teresa Tejerina
- Department of Pharmacology, School of Medicine, Complutense University, Madrid, Spain
| | - José M. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo. Universidad Complutense Madrid (UCM), Spain
- Facultad de Medicina, UCM, Spain
- * E-mail:
| |
Collapse
|
41
|
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: 1.4] [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
|
42
|
Han CY, Tang C, Guevara ME, Wei H, Wietecha T, Shao B, Subramanian S, Omer M, Wang S, O'Brien KD, Marcovina SM, Wight TN, Vaisar T, de Beer MC, de Beer FC, Osborne WR, Elkon KB, Chait A. Serum amyloid A impairs the antiinflammatory properties of HDL. J Clin Invest 2015; 126:266-81. [PMID: 26642365 DOI: 10.1172/jci83475] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/29/2015] [Indexed: 01/25/2023] Open
Abstract
HDL from healthy humans and lean mice inhibits palmitate-induced adipocyte inflammation; however, the effect of the inflammatory state on the functional properties of HDL on adipocytes is unknown. Here, we found that HDL from mice injected with AgNO3 fails to inhibit palmitate-induced inflammation and reduces cholesterol efflux from 3T3-L1 adipocytes. Moreover, HDL isolated from obese mice with moderate inflammation and humans with systemic lupus erythematosus had similar effects. Since serum amyloid A (SAA) concentrations in HDL increase with inflammation, we investigated whether elevated SAA is a causal factor in HDL dysfunction. HDL from AgNO3-injected mice lacking Saa1.1 and Saa2.1 exhibited a partial restoration of antiinflammatory and cholesterol efflux properties in adipocytes. Conversely, incorporation of SAA into HDL preparations reduced antiinflammatory properties but not to the same extent as HDL from AgNO3-injected mice. SAA-enriched HDL colocalized with cell surface-associated extracellular matrix (ECM) of adipocytes, suggesting impaired access to the plasma membrane. Enzymatic digestion of proteoglycans in the ECM restored the ability of SAA-containing HDL to inhibit palmitate-induced inflammation and cholesterol efflux. Collectively, these findings indicate that inflammation results in a loss of the antiinflammatory properties of HDL on adipocytes, which appears to partially result from the SAA component of HDL binding to cell-surface proteoglycans, thereby preventing access of HDL to the plasma membrane.
Collapse
|
43
|
Martin-Rojas T, Mourino-Alvarez L, Alonso-Orgaz S, Rosello-Lleti E, Calvo E, Lopez-Almodovar LF, Rivera M, Padial LR, Lopez JA, de la Cuesta F, Barderas MG. iTRAQ proteomic analysis of extracellular matrix remodeling in aortic valve disease. Sci Rep 2015; 5:17290. [PMID: 26620461 PMCID: PMC4664895 DOI: 10.1038/srep17290] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 10/28/2015] [Indexed: 02/08/2023] Open
Abstract
Degenerative aortic stenosis (AS) is the most common worldwide cause of valve replacement. The aortic valve is a thin, complex, layered connective tissue with compartmentalized extracellular matrix (ECM) produced by specialized cell types, which directs blood flow in one direction through the heart. There is evidence suggesting remodeling of such ECM during aortic stenosis development. Thus, a better characterization of the role of ECM proteins in this disease would increase our understanding of the underlying molecular mechanisms. Aortic valve samples were collected from 18 patients which underwent aortic valve replacement (50% males, mean age of 74 years) and 18 normal control valves were obtained from necropsies (40% males, mean age of 69 years). The proteome of the samples was analyzed by 2D-LC MS/MS iTRAQ methodology. The results showed an altered expression of 13 ECM proteins of which 3 (biglycan, periostin, prolargin) were validated by Western blotting and/or SRM analyses. These findings are substantiated by our previous results demonstrating differential ECM protein expression. The present study has demonstrated a differential ECM protein pattern in individuals with AS, therefore supporting previous evidence of a dynamic ECM remodeling in human aortic valves during AS development.
Collapse
Affiliation(s)
- Tatiana Martin-Rojas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | - Laura Mourino-Alvarez
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | - Sergio Alonso-Orgaz
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | - Esther Rosello-Lleti
- Cardiocirculatory Unit, Health Research Institute, Hospital La Fe, Valencia, Spain
| | | | | | - Miguel Rivera
- Cardiocirculatory Unit, Health Research Institute, Hospital La Fe, Valencia, Spain
| | - Luis R Padial
- Department of Cardiology, Hospital Virgen de la Salud, SESCAM, Toledo, Spain
| | | | - Fernando de la Cuesta
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | - Maria G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| |
Collapse
|
44
|
Hultgårdh-Nilsson A, Borén J, Chakravarti S. The small leucine-rich repeat proteoglycans in tissue repair and atherosclerosis. J Intern Med 2015; 278:447-61. [PMID: 26477596 PMCID: PMC4616156 DOI: 10.1111/joim.12400] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Proteoglycans consist of a protein core with one or more covalently attached glycosaminoglycan (GAG) side chains and have multiple roles in the initiation and progression of atherosclerosis. Here we discuss the potential and known functions of a group of small leucine-rich repeat proteoglycans (SLRPs) in atherosclerosis. We focus on five SLRPs, decorin, biglycan, lumican, fibromodulin and PRELP, because these have been detected in atherosclerotic plaques or demonstrated to have a role in animal models of atherosclerosis. Decorin and biglycan are modified post-translationally by substitution with chondroitin/dermatan sulphate GAGs, whereas lumican, fibromodulin and PRELP have keratan sulphate side chains, and the core proteins have leucine-rich repeat (LRR) motifs that are characteristic of the LRR superfamily. The chondroitin/dermatan sulphate GAG side chains have been implicated in lipid retention in atherosclerosis. The core proteins are discussed here in the context of (i) interactions with collagens and their implications in tissue integrity, fibrosis and wound repair and (ii) interactions with growth factors, cytokines, pathogen-associated molecular patterns and cell surface receptors that impact normal physiology and disease processes such as inflammation, innate immune responses and wound healing (i.e. processes that are all important in plaque development and progression). Thus, studies of these SLRPs in the context of wound healing are providing clues about their functions in early stages of atherosclerosis to plaque vulnerability and cardiovascular disease at later stages. Understanding of signal transduction pathways regulated by the core protein interactions is leading to novel roles and therapeutic potential for these proteins in wound repair and atherosclerosis.
Collapse
Affiliation(s)
| | - J Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - S Chakravarti
- Departments of Medicine, Ophthalmology and Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
45
|
Mao Y, Zlatic CO, Griffin MDW, Howlett GJ, Todorova N, Yarovsky I, Gooley PR. Hydrogen/Deuterium Exchange and Molecular Dynamics Analysis of Amyloid Fibrils Formed by a D69K Charge-Pair Mutant of Human Apolipoprotein C-II. Biochemistry 2015. [DOI: 10.1021/acs.biochem.5b00535] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yu Mao
- Department of Biochemistry and
Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Courtney O. Zlatic
- Department of Biochemistry and
Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Michael D. W. Griffin
- Department of Biochemistry and
Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Geoffrey J. Howlett
- Department of Biochemistry and
Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Nevena Todorova
- Health Innovations
Research Institute, RMIT University, GPO
Box 2476, Melbourne, Victoria 3001, Australia
| | - Irene Yarovsky
- Health Innovations
Research Institute, RMIT University, GPO
Box 2476, Melbourne, Victoria 3001, Australia
| | - Paul R. Gooley
- Department of Biochemistry and
Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
46
|
Massaro M, Martinelli R, Gatta V, Scoditti E, Pellegrino M, Carluccio MA, Calabriso N, Buonomo T, Stuppia L, Storelli C, De Caterina R. Transcriptome-based identification of new anti-inflammatory and vasodilating properties of the n-3 fatty acid docosahexaenoic acid in vascular endothelial cell under proinflammatory conditions [corrected]. PLoS One 2015; 10:e0129652. [PMID: 26114549 PMCID: PMC4482638 DOI: 10.1371/journal.pone.0129652] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 05/12/2015] [Indexed: 01/01/2023] Open
Abstract
Scope High intakes of n-3 fatty acids exert anti-inflammatory effects and cardiovascular protection, but the underlying molecular basis is incompletely defined. By genome-wide analysis we searched for novel effects of docosahexaenoic acid (DHA) on gene expression and pathways in human vascular endothelium under pro-inflammatory conditions. Methods and Results Human umbilical vein endothelial cells were treated with DHA and then stimulated with interleukin(IL)-1β. Total RNA was extracted, and gene expression examined by DNA microarray. DHA alone altered the expression of 188 genes, decreasing 92 and increasing 96. IL-1β changed the expression of 2031 genes, decreasing 997 and increasing 1034. Treatment with DHA before stimulation significantly affected the expression of 116 IL-1β-deregulated genes, counter-regulating the expression of 55 genes among those decreased and of 61 among those increased. Functional and network analyses identified immunological, inflammatory and metabolic pathways as the most affected. Newly identified DHA-regulated genes are involved in stemness, cellular growth, cardiovascular system function and cancer, and included cytochrome p450 4F2(CYP4F2), transforming growth factor(TGF)-β2, Cluster of Differentiation (CD)47, caspase recruitment domain(CARD)11 and phosphodiesterase(PDE)5α. Conclusions Endothelial exposure to DHA regulates novel genes and related pathways. Such unbiased identification should increase our understanding of mechanisms by which n-3 fatty acids affect human diseases.
Collapse
Affiliation(s)
- Marika Massaro
- National Research Council (CNR), Institute of Clinical Physiology, Lecce, Italy
| | - Rosanna Martinelli
- CEINGE Biotecnologie Avanzate, Naples, Italy
- Department of Medicine and Surgery of Salerno University, Salerno, Italy
| | - Valentina Gatta
- “Gabriele d’Annunzio” University and Center of Excellence on Aging, Chieti, Italy
| | - Egeria Scoditti
- National Research Council (CNR), Institute of Clinical Physiology, Lecce, Italy
| | - Mariangela Pellegrino
- National Research Council (CNR), Institute of Clinical Physiology, Lecce, Italy
- Department of Biological and Environmental Science and Technology (Disteba), University of Salento, Lecce, Italy
| | | | - Nadia Calabriso
- National Research Council (CNR), Institute of Clinical Physiology, Lecce, Italy
| | | | - Liborio Stuppia
- “Gabriele d’Annunzio” University and Center of Excellence on Aging, Chieti, Italy
| | - Carlo Storelli
- Department of Biological and Environmental Science and Technology (Disteba), University of Salento, Lecce, Italy
| | - Raffaele De Caterina
- “Gabriele d’Annunzio” University and Center of Excellence on Aging, Chieti, Italy
- Fondazione Toscana “Gabriele Monasterio”, Pisa, Italy
- * E-mail:
| |
Collapse
|
47
|
Abstract
Coronary artery disease (CAD) is the leading cause of death in the United States. Although CAD was formerly considered a lipid accumulation-mediated disease, it has now been clearly shown to involve an ongoing inflammatory response. Advances in basic science research have established the crucial role of inflammation in mediating all stages of CAD. Today, there is convincing evidence that multiple interrelated immune mechanisms interact with metabolic risk factors to initiate, promote, and ultimately activate lesions in the coronary arteries. This review aims to provide current evidence pertaining to the role of inflammation in the pathogenesis of CAD and discusses the impact of inflammatory markers and their modification on clinical outcomes.
Collapse
|
48
|
|
49
|
Witos J, Samuelsson J, Cilpa-Karhu G, Metso J, Jauhiainen M, Riekkola ML. Partial filling affinity capillary electrophoresis including adsorption energy distribution calculations--towards reliable and feasible biomolecular interaction studies. Analyst 2015; 140:3175-82. [PMID: 25751597 DOI: 10.1039/c5an00210a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In this work, a method to study and analyze the interaction data in free solution by exploiting partial filling affinity capillary electrophoresis (PF-ACE) followed by adsorption energy distribution calculations (AED) prior model fit to adsorption isotherms will be demonstrated. PF-ACE-AED approach allowed the possibility to distinguish weak and strong interactions of the binding processes between the most common apolipoprotein E protein isoforms (apoE2, apoE3, apoE4) of high density lipoprotein (HDL) and apoE-containing HDL2 with major glycosaminoglycan (GAG) chain of proteoglycans (PGs), chondroitin-6-sulfate (C6S). The AED analysis clearly revealed the heterogeneity of the binding processes. The major difference was that they were heterogeneous with two different adsorption sites for apoE2 and apoE4 isoforms, whereas interestingly for apoE3 and apoE-containing HDL2, the binding was homogeneous (one site) adsorption process. Moreover, our results allowed the evaluation of differences in the binding process strengths giving the following order with C6S: apoE-containing HDL2 > apoE2 > apoE4 > apoE3. In addition, the affinity constant values determined could be compared with those obtained in our previous studies for the interactions between apoE isoforms and another important GAG chain of PGs - dermatan sulfate (DS). The success of the combination of AED calculations prior to non-linear adsorption isotherm model fit with PF-ACE when the concentration range was extended, confirmed the power of the system in the clarification of the heterogeneity of biological processes studied.
Collapse
Affiliation(s)
- Joanna Witos
- Laboratory of Analytical Chemistry, Department of Chemistry, P.O. Box 55, FIN-00014 University of Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
50
|
The small leucine-rich proteoglycan BGN accumulates in CADASIL and binds to NOTCH3. Transl Stroke Res 2015; 6:148-55. [PMID: 25578324 DOI: 10.1007/s12975-014-0379-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/09/2014] [Accepted: 11/18/2014] [Indexed: 10/24/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is an inherited form of cerebral small vessel disease caused by mutations in conserved residues of NOTCH3. Affected arteries of CADASIL feature fibrosis and accumulation of NOTCH3. A variety of collagen subtypes (types I, III, IV, and VI) have been identified in fibrotic CADASIL vessels. Biglycan (BGN) and decorin (DCN) are class I members of the small leucine-rich proteoglycan (SLRP) family that regulate collagen fibril size. Because DCN has been shown to deposit in arteries in cerebral small vessel disease, we tested whether BGN accumulates in arteries of CADASIL brains. BGN was strongly expressed in both small penetrating and leptomeningeal arteries of CADASIL brain. BGN protein was localized to all three layers of arteries (intima, media, and adventitia). Substantially, more immunoreactivity was observed in CADASIL brains compared to controls. Immunoblotting of brain lysates showed a fourfold increase in CADASIL brains (compared to controls). Messenger RNA encoding BGN was also increased in CADASIL and was localized by in situ hybridization to all three vascular layers in CADASIL. Human cerebrovascular smooth muscle cells exposed to purified NOTCH3 ectodomain upregulated BGN, DCN, and COL4A1 through mechanisms that are sensitive to rapamycin, a potent mTOR inhibitor. In addition, BGN protein interacted directly with NOTCH3 protein in cell culture and in direct protein interaction assays. In conclusion, BGN is a CADASIL-enriched protein that potentially accumulates in vessels by mTOR-mediated transcriptional activation and/or post-translational accumulation via protein interactions with NOTCH3 and collagen.
Collapse
|