1
|
Joshi S, Jan KM, Rumschitzki D. Pre-atherosclerotic flow and oncotically active solute transport across the arterial endothelium. J Theor Biol 2020; 499:110275. [PMID: 32275985 PMCID: PMC9776117 DOI: 10.1016/j.jtbi.2020.110275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 12/24/2022]
Abstract
Atherosclerosis starts with transmural (transwall) pressure-driven advective transport of blood-borne low-density lipoprotein (LDL) cholesterol across rare endothelial cell (EC) monolayer leaks into the arterial subendothelial intima (SI) wall layer where they can spread, bind to extracellular matrix and seed lesions. The local SI LDL concentration, which governs LDL's binding kinetics, depends on the overall diluting transmural liquid flow. Transmural pressures typically compress the SI at physiological pressures, which keeps this flow low. Nguyen et al. (2015) showed that aortic ECs express the water channel protein aquaporin-1 (AQP1) and the transEC (δP) portion of the transmural (ΔP) pressure difference drives, in parallel, water across AQP1s and plasma across interEC junctions. Since the lumen is isotonic, selective AQP1-mediated water flow should quickly render the ECs' lumen side hypertonic and the SI hypotonic; resulting transEC oncotic pressure differences, δπ, should oppose δP and quickly halt transEC flow. Yet Nguyen et al.'s (2015) transAQP1 flows persist for hours. To resolve this paradox, we extend our fluid filtration theory Joshi et al. (2015) to include mass transfer for oncotically active solutes like albumin. This addition nonlinearly couples mass transfer, fluid flow and wall mechanics. We simultaneously solve these problems at steady state. Surprisingly it finds that media layer filtration causes steady SI to exceed EC glycocalyx albumin concentration. Thus δπ reinforces rather than opposes δP, i.e., it sucks water from, rather than pushing water into the lumen from the SI. Endothelial AQP1s raise the overall driving force for flow across the EC above δP, most significantly at pressures too low to compress the SI, and they increase the ΔP needed for SI compression. This suggests the intriguing possibility that increasing EC AQP1 expression can raise this requisite compression pressure to physiological values. That is, increasing EC AQP1 may decompress the SI at physiological pressures, which would significantly increase SI thickness, flow and subsequently SI LDL dilution. This could retard LDL binding and delay preatherosclerotic lesion onset. The model also predicts that glycocalyx-degrading enzymes decrease overall transEC driving forces and thus lower, not raise, transmural water flux.
Collapse
Affiliation(s)
- Shripad Joshi
- Department of Chemical Engineering, City College of the City University of New York, NY, United States
| | - Kung-Ming Jan
- Department of Medicine, College of Physicians and Surgeons, Columbia University, NY, United States
| | - David Rumschitzki
- Department of Chemical Engineering, City College of the City University of New York, NY, United States,Department of Medicine, College of Physicians and Surgeons, Columbia University, NY, United States,Corresponding author at: Department of Chemical Engineering, City College of the City University of New York, NY, United States. , (D. Rumschitzki)
| |
Collapse
|
2
|
Human species-specific loss of CMP- N-acetylneuraminic acid hydroxylase enhances atherosclerosis via intrinsic and extrinsic mechanisms. Proc Natl Acad Sci U S A 2019; 116:16036-16045. [PMID: 31332008 DOI: 10.1073/pnas.1902902116] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) events due to atherosclerosis cause one-third of worldwide deaths and risk factors include physical inactivity, age, dyslipidemia, hypertension, diabetes, obesity, smoking, and red meat consumption. However, ∼15% of first-time events occur without such factors. In contrast, coronary events are extremely rare even in closely related chimpanzees in captivity, despite human-like CVD-risk-prone blood lipid profiles, hypertension, and mild atherosclerosis. Similarly, red meat-associated enhancement of CVD event risk does not seem to occur in other carnivorous mammals. Thus, heightened CVD risk may be intrinsic to humans, and genetic changes during our evolution need consideration. Humans exhibit a species-specific deficiency of the sialic acid N-glycolylneuraminic acid (Neu5Gc), due to pseudogenization of cytidine monophosphate-N-acetylneuraminic acid (Neu5Ac) hydroxylase (CMAH), which occurred in hominin ancestors ∼2 to 3 Mya. Ldlr -/- mice with human-like Cmah deficiency fed a sialic acids (Sias)-free high-fat diet (HFD) showed ∼1.9-fold increased atherogenesis over Cmah wild-type Ldlr -/- mice, associated with elevated macrophage cytokine expression and enhanced hyperglycemia. Human consumption of Neu5Gc (from red meat) acts as a "xeno-autoantigen" via metabolic incorporation into endogenous glycoconjugates, as interactions with circulating anti-Neu5Gc "xeno-autoantibodies" potentiate chronic inflammation ("xenosialitis"). Cmah -/- Ldlr -/- mice immunized with Neu5Gc-bearing antigens to generate human-like anti-Neu5Gc antibodies suffered a ∼2.4-fold increased atherosclerosis on a Neu5Gc-rich HFD, compared with Neu5Ac-rich or Sias-free HFD. Lesions in Neu5Gc-immunized and Neu5Gc-rich HFD-fed Cmah -/- Ldlr -/- mice were more advanced but unexplained by lipoprotein or glucose changes. Human evolutionary loss of CMAH likely contributes to atherosclerosis predisposition via multiple intrinsic and extrinsic mechanisms, and future studies could consider this more human-like model.
Collapse
|
3
|
Alkhalil M, Choudhury RP. Current concepts in atherosclerosis. Indian J Thorac Cardiovasc Surg 2018; 34:198-205. [PMID: 33060939 PMCID: PMC7525593 DOI: 10.1007/s12055-018-0699-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/06/2018] [Accepted: 07/12/2018] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis is a complex disease process. It is increasingly recognised that both lipoprotein retention and inflammatory cellular components are intricately related in the initiation and development of atherosclerotic plaque. LDL-c (cholesterol) has been long established as a cause for atherosclerosis; additionally, inflammatory cells such as monocytes and subsequently foam cells have also been directly linked to the progression of atherosclerotic disease. Emerging data suggest that structures outside vascular intima and media are also closely related to atherosclerosis. Perivascular adipose tissue (PVAT) may be a determinant of the inflammatory status of the atherosclerotic plaque. All these features are becoming extremely relevant as therapies against atherosclerosis are targeting both lipid retention and inflammation. Recently, there has been some success in these novel therapies, such as the proprotein convertase subtilisin-kexin type 9 (PCSK-9) inhibitor evolocumab and the interleukin-1ß neutralising antibody, canakinumab, in reducing cardiovascular events when added to standard therapy such as statin. This review will discuss the pathogenesis of atherosclerosis, including some novel features, and its management using new anti-atherosclerotic drugs.
Collapse
Affiliation(s)
- Mohammad Alkhalil
- Acute Vascular Imaging Centre, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU UK.,Cardiology Department, Royal Victoria Hospital, Belfast, UK
| | - Robin P Choudhury
- Acute Vascular Imaging Centre, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU UK
| |
Collapse
|
4
|
Curcio CA. Antecedents of Soft Drusen, the Specific Deposits of Age-Related Macular Degeneration, in the Biology of Human Macula. Invest Ophthalmol Vis Sci 2018; 59:AMD182-AMD194. [PMID: 30357337 PMCID: PMC6733529 DOI: 10.1167/iovs.18-24883] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AMD pathobiology was irreversibly changed by the recent discovery of extracellular cholesterol-containing deposits in the subretinal space, between the photoreceptors and retinal pigment epithelium (RPE), called subretinal drusenoid deposits (SDDs). SDDs strikingly mirror the topography of rod photoreceptors in human macula, raising the question of whether an equivalent process results in a deposition related to foveal cones. Herein we propose that AMD's pathognomonic lesion-soft drusen and basal linear deposit (BLinD, same material, diffusely distributed)-is the leading candidate. Epidemiologic, clinical, and histologic data suggest that these deposits are most abundant in the central macula, under the fovea. Strong evidence presented in a companion article supports the idea that the dominant ultrastructural component is large apolipoprotein B,E-containing lipoproteins, constitutively secreted by RPE. Lipoprotein fatty acids are dominated by linoleate (implicating diet) rather than docosahexaenoate (implicating photoreceptors); we seek within the retina cellular relationships and dietary drivers to explain soft druse topography. The delivery of xanthophyll pigments to highly evolved and numerous Müller cells in the human fovea, through RPE, is one strong candidate, because Müller cells are the main reservoir of these pigments, which replenish from diet. We propose that the evolution of neuroglial relations and xanthophyll delivery that underlie exquisite human foveal vision came with a price, that is, soft drusen and sequela, long after our reproductive years.
Collapse
Affiliation(s)
- Christine A Curcio
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
5
|
Ference BA, Ginsberg HN, Graham I, Ray KK, Packard CJ, Bruckert E, Hegele RA, Krauss RM, Raal FJ, Schunkert H, Watts GF, Borén J, Fazio S, Horton JD, Masana L, Nicholls SJ, Nordestgaard BG, van de Sluis B, Taskinen MR, Tokgözoglu L, Landmesser U, Laufs U, Wiklund O, Stock JK, Chapman MJ, Catapano AL. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur Heart J 2018; 38:2459-2472. [PMID: 28444290 PMCID: PMC5837225 DOI: 10.1093/eurheartj/ehx144] [Citation(s) in RCA: 2139] [Impact Index Per Article: 356.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/08/2017] [Indexed: 12/15/2022] Open
Abstract
Aims To appraise the clinical and genetic evidence that low-density lipoproteins (LDLs) cause atherosclerotic cardiovascular disease (ASCVD). Methods and results We assessed whether the association between LDL and ASCVD fulfils the criteria for causality by evaluating the totality of evidence from genetic studies, prospective epidemiologic cohort studies, Mendelian randomization studies, and randomized trials of LDL-lowering therapies. In clinical studies, plasma LDL burden is usually estimated by determination of plasma LDL cholesterol level (LDL-C). Rare genetic mutations that cause reduced LDL receptor function lead to markedly higher LDL-C and a dose-dependent increase in the risk of ASCVD, whereas rare variants leading to lower LDL-C are associated with a correspondingly lower risk of ASCVD. Separate meta-analyses of over 200 prospective cohort studies, Mendelian randomization studies, and randomized trials including more than 2 million participants with over 20 million person-years of follow-up and over 150 000 cardiovascular events demonstrate a remarkably consistent dose-dependent log-linear association between the absolute magnitude of exposure of the vasculature to LDL-C and the risk of ASCVD; and this effect appears to increase with increasing duration of exposure to LDL-C. Both the naturally randomized genetic studies and the randomized intervention trials consistently demonstrate that any mechanism of lowering plasma LDL particle concentration should reduce the risk of ASCVD events proportional to the absolute reduction in LDL-C and the cumulative duration of exposure to lower LDL-C, provided that the achieved reduction in LDL-C is concordant with the reduction in LDL particle number and that there are no competing deleterious off-target effects. Conclusion Consistent evidence from numerous and multiple different types of clinical and genetic studies unequivocally establishes that LDL causes ASCVD.
Collapse
Affiliation(s)
- Brian A Ference
- Division of Translational Research and Clinical Epidemiology, Division of Cardiovascular Medicine, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Henry N Ginsberg
- Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA
| | | | - Kausik K Ray
- Department of Primary Care and Public Health, Imperial Centre for Cardiovascular Disease Prevention, Imperial College, London, UK
| | - Chris J Packard
- College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| | - Eric Bruckert
- INSERM UMRS1166, Department of Endocrinology-Metabolism, ICAN - Institute of CardioMetabolism and Nutrition, AP-HP, Hôpital de la Pitié, Paris, France
| | - Robert A Hegele
- Department of Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Ronald M Krauss
- Department of Atherosclerosis Research, Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Frederick J Raal
- Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Technische Universität München, Munich 80636, Germany.,Deutsches Zentrum für Herz und Kreislauferkrankungen (DZHK), Partner Site Munich Heart Alliance, Munich 81377, Germany
| | - Gerald F Watts
- Lipid Disorders Clinic, Centre for Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Jan Borén
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Sergio Fazio
- Department of Medicine, Center for Preventive Cardiology of the Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Jay D Horton
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Luis Masana
- Research Unit of Lipids and Atherosclerosis, University Rovira i Virgili, C. Sant Llorenç 21, Reus 43201, Spain
| | - Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry and The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.,The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Denmark
| | - Bart van de Sluis
- Department of Pediatrics, Molecular Genetics Section, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen AV 9713, The Netherlands
| | - Marja-Riitta Taskinen
- Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity, Heart and Lung Centre, University of Helsinki, Helsinki, Finland
| | | | - Ulf Landmesser
- Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA.,INSERM UMRS1166, Department of Endocrinology-Metabolism, ICAN - Institute of CardioMetabolism and Nutrition, AP-HP, Hôpital de la Pitié, Paris, France.,Department of Cardiology, Charité-Universitätsmedizin Berlin (CBF), Hindenburgdamm 30, Berlin 12203, Germany
| | - Ulrich Laufs
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Homburg, Saar, Germany
| | - Olov Wiklund
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jane K Stock
- European Atherosclerosis Society, Gothenburg, Sweden
| | - M John Chapman
- INSERM, Dyslipidemia and Atherosclerosis Research, and University of Pierre and Marie Curie, Pitié-Sâlpetrière University Hospital, Paris, France
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan and IRCCS Multimedica, Milan, Italy
| |
Collapse
|
6
|
Wijesinghe P, Johansen NJ, Curatolo A, Sampson DD, Ganss R, Kennedy BF. Ultrahigh-Resolution Optical Coherence Elastography Images Cellular-Scale Stiffness of Mouse Aorta. Biophys J 2018; 113:2540-2551. [PMID: 29212007 DOI: 10.1016/j.bpj.2017.09.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/22/2017] [Accepted: 09/19/2017] [Indexed: 01/08/2023] Open
Abstract
Cellular-scale imaging of the mechanical properties of tissue has helped to reveal the origins of disease; however, cellular-scale resolution is not readily achievable in intact tissue volumes. Here, we demonstrate volumetric imaging of Young's modulus using ultrahigh-resolution optical coherence elastography, and apply it to characterizing the stiffness of mouse aortas. We achieve isotropic resolution of better than 15 μm over a 1-mm lateral field of view through the entire depth of an intact aortic wall. We employ a method of quasi-static compression elastography that measures volumetric axial strain and uses a compliant, transparent layer to measure surface axial stress. This combination is used to estimate Young's modulus throughout the volume. We demonstrate differentiation by stiffness of individual elastic lamellae and vascular smooth muscle. We observe stiffening of the aorta in regulator of G protein signaling 5-deficient mice, a model that is linked to vascular remodeling and fibrosis. We observe increased stiffness with proximity to the heart, as well as regions with micro-structural and micro-mechanical signatures characteristic of fibrous and lipid-rich tissue. High-resolution imaging of Young's modulus with optical coherence elastography may become an important tool in vascular biology and in other fields concerned with understanding the role of mechanics within the complex three-dimensional architecture of tissue.
Collapse
Affiliation(s)
- Philip Wijesinghe
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia; Optical+Biomedical Engineering Laboratory, School of Electrical, Electronic and Computer Engineering, The University of Western Australia, Perth, Western Australia, Australia.
| | - Niloufer J Johansen
- Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia; Research Department, St John of God Subiaco Hospital, Subiaco, Western Australia, Australia
| | - Andrea Curatolo
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia; School of Electrical, Electronic and Computer Engineering, The University of Western Australia, Perth, Western Australia, Australia
| | - David D Sampson
- Optical+Biomedical Engineering Laboratory, School of Electrical, Electronic and Computer Engineering, The University of Western Australia, Perth, Western Australia, Australia; Centre for Microscopy, Characterisation and Analysis, The University of Western Australia, Perth, Western Australia, Australia
| | - Ruth Ganss
- Vascular Biology and Stromal Targeting, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Brendan F Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia; School of Electrical, Electronic and Computer Engineering, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
7
|
Toussaint J, Raval CB, Nguyen T, Fadaifard H, Joshi S, Wolberg G, Quarfordt S, Jan KM, Rumschitzki DS. Chronic hypertension increases aortic endothelial hydraulic conductivity by upregulating endothelial aquaporin-1 expression. Am J Physiol Heart Circ Physiol 2017; 313:H1063-H1073. [PMID: 28733452 PMCID: PMC5792199 DOI: 10.1152/ajpheart.00651.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 07/14/2017] [Accepted: 07/14/2017] [Indexed: 01/22/2023]
Abstract
Numerous studies have examined the role of aquaporins in osmotic water transport in various systems, but virtually none have focused on the role of aquaporin in hydrostatically driven water transport involving mammalian cells save for our laboratory's recent study of aortic endothelial cells. Here, we investigated aquaporin-1 expression and function in the aortic endothelium in two high-renin rat models of hypertension, the spontaneously hypertensive genetically altered Wistar-Kyoto rat variant and Sprague-Dawley rats made hypertensive by two-kidney, one-clip Goldblatt surgery. We measured aquaporin-1 expression in aortic endothelial cells from whole rat aortas by quantitative immunohistochemistry and function by measuring the pressure-driven hydraulic conductivities of excised rat aortas with both intact and denuded endothelia on the same vessel. We used them to calculate the effective intimal hydraulic conductivity, which is a combination of endothelial and subendothelial components. We observed well-correlated enhancements in aquaporin-1 expression and function in both hypertensive rat models as well as in aortas from normotensive rats whose expression was upregulated by 2 h of forskolin treatment. Upregulated aquaporin-1 expression and function may be a response to hypertension that critically determines conduit artery vessel wall viability and long-term susceptibility to atherosclerosis.NEW & NOTEWORTHY The aortic endothelia of two high-renin hypertensive rat models express greater than two times the aquaporin-1 and, at low pressures, have greater than two times the endothelial hydraulic conductivity of normotensive rats. Data are consistent with theory predicting that higher endothelial aquaporin-1 expression raises the critical pressure for subendothelial intima compression and for artery wall hydraulic conductivity to drop.
Collapse
Affiliation(s)
- Jimmy Toussaint
- 1Department of Chemical Engineering, City College of the City University of New York, New York, New York; ,4Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts;
| | - Chirag Bharavi Raval
- 1Department of Chemical Engineering, City College of the City University of New York, New York, New York; ,2Department of Biomedical Engineering, City College of the City University of New York, New York, New York;
| | - Tieuvi Nguyen
- 2Department of Biomedical Engineering, City College of the City University of New York, New York, New York;
| | - Hadi Fadaifard
- 3Department of Computer Science, City College of the City University of New York, New York, New York;
| | - Shripad Joshi
- 1Department of Chemical Engineering, City College of the City University of New York, New York, New York;
| | - George Wolberg
- 3Department of Computer Science, City College of the City University of New York, New York, New York;
| | - Steven Quarfordt
- 1Department of Chemical Engineering, City College of the City University of New York, New York, New York;
| | - Kung-ming Jan
- 5Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York; and
| | - David S. Rumschitzki
- 1Department of Chemical Engineering, City College of the City University of New York, New York, New York; ,5Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York; and ,6Biology (Molecular, Cellular, and Developmental Biology) and Chemistry (Biophysics) Departments, The Graduate School and University Center, City University of New York, New York, New York
| |
Collapse
|
8
|
Daugherty A, Tall AR, Daemen MJ, Falk E, Fisher EA, García-Cardeña G, Lusis AJ, Owens AP, Rosenfeld ME, Virmani R. Recommendation on Design, Execution, and Reporting of Animal Atherosclerosis Studies: A Scientific Statement From the American Heart Association. Circ Res 2017; 121:e53-e79. [DOI: 10.1161/res.0000000000000169] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.
Collapse
|
9
|
Daugherty A, Tall AR, Daemen MJAP, Falk E, Fisher EA, García-Cardeña G, Lusis AJ, Owens AP, Rosenfeld ME, Virmani R. Recommendation on Design, Execution, and Reporting of Animal Atherosclerosis Studies: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2017; 37:e131-e157. [PMID: 28729366 DOI: 10.1161/atv.0000000000000062] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.
Collapse
|
10
|
Ismail EN, Ruberti JW, Malek G. Quick-freeze/deep-etch electron microscopy visualization of the mouse posterior pole. Exp Eye Res 2017. [PMID: 28629927 DOI: 10.1016/j.exer.2017.06.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The mouse is one of the most commonly used mammalian systems to study human diseases. In particular it has been an invaluable tool to model a multitude of ocular pathologies affecting the posterior pole. The aim of this study was to create a comprehensive map of the ultrastructure of the mouse posterior pole using the quick-freeze/deep-etch method (QFDE). QFDE can produce detailed three-dimensional images of tissue structure and macromolecular moieties, without many of the artifacts introduced by structure-altering post-processing methods necessary to perform conventional transmission electron microscopy (cTEM). A total of 18 eyes from aged C57BL6/J mice were enucleated and the posterior poles were processed, either intact or with the retinal pigment epithelium (RPE) cell layer removed, for imaging by either QFDE or cTEM. QFDE images were correlated with cTEM cross-sections and en face images through the outer retina. Nicely preserved outer retinal architecture was observed with both methods, however, QFDE provided excellent high magnification imaging, with greater detail, of the apical, central, and basal planes of the RPE. Furthermore, key landmarks within Bruch's membrane, choriocapillaris, choroid and sclera were characterized and identified. In this study we developed methods for preparing the outer retina of the mouse for evaluation with QFDE and provide a map of the ultrastructure and cellular composition of the outer posterior pole. This technique should be applicable for morphological evaluation of mouse models, in which detailed visualization of subtle ocular structural changes is needed or in cases where post-processing methods introduce unacceptable artifacts.
Collapse
Affiliation(s)
- Ebraheim N Ismail
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Jeffrey W Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, United States; Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, MA, United States.
| | - Goldis Malek
- Departments of Ophthalmology and Pathology, Duke University School of Medicine, Durham, NC, United States.
| |
Collapse
|
11
|
Lawal AO. Air particulate matter induced oxidative stress and inflammation in cardiovascular disease and atherosclerosis: The role of Nrf2 and AhR-mediated pathways. Toxicol Lett 2017; 270:88-95. [PMID: 28189649 DOI: 10.1016/j.toxlet.2017.01.017] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/28/2017] [Accepted: 01/31/2017] [Indexed: 12/31/2022]
Abstract
Air particulate matter (PM) is an important component of air pollution, which has been reported to play important role in the adverse health effects of the latter. Extensive experimental data and epidemiological studies have shown that the increased cardiovascular morbidity and mortality and atherosclerosis caused by air pollution are mainly due to the PM component. Implicated in these adverse health effects of PM, is their ability to induce oxidative stress and pro-inflammatory events in the vascular system. The association between the cardiovascular ischemic events and atherosclerosis induced by PM has been linked to the ultrafine and fine components. These particles have a high content of redox cyclic chemicals. This, together with their ability to combine with proatherogenic molecules enhanced tissue oxidative stress. Studies have shown that the oxidative stress induced by PM could up-regulates the expression of phase I and phase II metabolize enzymes. This up-regulation occurs by the activation of transcription factors (such as nuclear factor (erythroid-derived 2) -like 2-related factor (Nrf2) and aryl hydrocarbon receptor (AhR)). This review will focus on data supporting the role of oxidative stress and inflammation in PM-induced cardiovascular diseases and atherosclerosis and the importance of Nrf2-and AhR- dependent regulatory pathways in the PM-induced cardiovascular events and atherosclerosis.
Collapse
Affiliation(s)
- Akeem O Lawal
- Department of Biochemistry, School of Sciences, Federal University of Technology, Akure P.M.B. 704, Akure, Ondo-State, Nigeria.
| |
Collapse
|
12
|
Low-density lipoprotein transport through an arterial wall under hypertension - A model with time and pressure dependent fraction of leaky junction consistent with experiments. J Theor Biol 2016; 411:81-91. [PMID: 27686595 DOI: 10.1016/j.jtbi.2016.09.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/19/2016] [Accepted: 09/23/2016] [Indexed: 12/11/2022]
Abstract
The influence of hypertension on low-density lipoproteins intake into the arterial wall is an important factor for understanding mechanisms of atherosclerosis. It has been experimentally observed that the increased pressure leads to the higher level of the LDL inside the wall. In this paper we attempt to construct a model of the LDL transport which reproduces quantitatively experimental outcomes. We supplement the well-known four-layer arterial wall model to include two pressure induced effects: the compression of the intima tissue and the increase of the fraction of leaky junctions. We demonstrate that such model can reach the very good agreement with experimental data.
Collapse
|
13
|
Bairaktari ET, Seferiadis KI, Elisaf MS. Evaluation of Methods for the Measurement of Low-Density Lipoprotein Cholesterol. J Cardiovasc Pharmacol Ther 2016; 10:45-54. [PMID: 15821838 DOI: 10.1177/107424840501000106] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A high concentration of low-density lipoprotein cholesterol (LDL-C) in plasma is one of the strongest risk factors for atherosclerotic cardiovascular disease and mortality. The most common approach to determining LDL-C in the clinical laboratory is the Friedewald calculation. There is an increased interest to improve the accuracy of LDL-C estimated by this equation. The expert panel convened by National Cholesterol Education Program has recommended the development of accurate direct methods to measure LDL-C. Several homogeneous and fully automated methods have been introduced in recent years that show improved precision and accuracy over earlier methods, especially the Friedewald calculation. Each of the atherogenic particles in plasma—very-low, intermediate-, and low-density lipoprotein—as well as lipoprotein (a), contain one molecule of apolipoprotein B (apoB) and thus, plasma total concentration of apoB reflects the number of atherogenic particles. Several studies suggested that the measurement of apoB could improve the prediction of risk of coronary artery disease. Thus, in addition to the newly developed direct assays, alternative calculation procedures have been proposed that also take into consideration total serum apoB concentration for the estimation of LDL-C and the presence of small, dense LDL particles. The new generation of homogenous methods for the measurement of LDL-C and the use of serum apoB concentration for the estimation of LDL-C can contribute to the accurate LDL-C determination.
Collapse
Affiliation(s)
- Eleni T Bairaktari
- Laboratory of Clinical Chemistry, Medical School, University of Ioannina, Ioannina, Greece
| | | | | |
Collapse
|
14
|
Zheng J, Zhou H, Zhao Y, Lun Q, Liu B, Tu P. Triterpenoid-enriched extract of Ilex kudingcha inhibits aggregated LDL-induced lipid deposition in macrophages by downregulating low density lipoprotein receptor-related protein 1 (LRP1). J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.08.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
15
|
Joshi S, Jan KM, Rumschitzki DS. Aquaporin-1 shifts the critical transmural pressure to compress the aortic intima and change transmural flow: theory and implications. Am J Physiol Heart Circ Physiol 2015; 309:H1974-86. [PMID: 26342066 DOI: 10.1152/ajpheart.00316.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/22/2015] [Indexed: 11/22/2022]
Abstract
Transmural-pressure (ΔP)-driven plasma advection carries macromolecules into the vessel wall, the earliest prelesion atherosclerotic event. The wall's hydraulic conductivity, LP, the water flux-to-ΔP ratio, is high at low pressures, rapidly decreases, and remains flat to high pressures (Baldwin AL, Wilson LM. Am J Physiol Heart Circ Physiol 264: H26-H32, 1993; Nguyen T, Toussaint, Xue JD, Raval Y, Cancel CB, Russell LM, Shou S, Sedes Y, Sun O, Yakobov Y, Tarbell JM, Jan KM, Rumschitzki DS. Am J Physiol Heart Circ Physiol 308: H1051-H1064, 2015; Tedgui A, Lever MJ. Am J Physiol Heart Circ Physiol. 247: H784-H791, 1984. Shou Y, Jan KM, Rumschitzki DS. Am J Physiol Heart Circ Physiol 291: H2758-H2771, 2006) due to pressure-induced subendothelial intima (SI) compression that causes endothelial cells to partially block internal elastic laminar fenestrae. Nguyen et al. showed that rat and bovine aortic endothelial cells express the membrane protein aquaporin-1 (AQP1) and transmural water transport is both transcellular and paracellular. They found that LP lowering by AQP1 blocking was perplexingly ΔP dependent. We hypothesize that AQP1 blocking lowers average SI pressure; therefore, a lower ΔP achieves the critical force/area on the endothelium to partially block fenestrae. To test this hypothesis, we improve the approximate model of Huang et al. (Huang Y, Rumschitzki D, Chien S, Weinbaum SS. Am J Physiol Heart Circ Physiol 272: H2023-H2039, 1997) and extend it by including transcellular AQP1 water flow. Results confirm the observation by Nguyen et al.: wall LP and water transport decrease with AQP1 disabling. The model predicts 1) low-pressure LP experiments correctly; 2) AQP1s contribute 30-40% to both the phenomenological endothelial + SI and intrinsic endothelial LP; 3) the force on the endothelium for partial SI decompression with functioning AQP1s at 60 mmHg equals that on the endothelium at ∼43 mmHg with inactive AQP1s; and 4) increasing endothelial AQP1 expression increases wall LP and shifts the ΔP regime where LP drops to significantly higher ΔP than in Huang et al. Thus AQP1 upregulation (elevated wall LP) might dilute and slow low-density lipoprotein binding to SI extracellular matrix, which may be beneficial for early atherogenesis.
Collapse
Affiliation(s)
- Shripad Joshi
- Department of Chemical Engineering, City College of the City University of New York, New York; and
| | - Kung-Ming Jan
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York
| | - David S Rumschitzki
- Department of Chemical Engineering, City College of the City University of New York, New York; and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York
| |
Collapse
|
16
|
Lu M, Gursky O. Aggregation and fusion of low-density lipoproteins in vivo and in vitro. Biomol Concepts 2015; 4:501-18. [PMID: 25197325 DOI: 10.1515/bmc-2013-0016] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Low-density lipoproteins (LDLs, also known as 'bad cholesterol') are the major carriers of circulating cholesterol and the main causative risk factor of atherosclerosis. Plasma LDLs are 20- to 25-nm nanoparticles containing a core of cholesterol esters surrounded by a phospholipid monolayer and a single copy of apolipoprotein B (550 kDa). An early sign of atherosclerosis is the accumulation of LDL-derived lipid droplets in the arterial wall. According to the widely accepted 'response-to-retention hypothesis', LDL binding to the extracellular matrix proteoglycans in the arterial intima induces hydrolytic and oxidative modifications that promote LDL aggregation and fusion. This enhances LDL uptake by the arterial macrophages and triggers a cascade of pathogenic responses that culminate in the development of atherosclerotic lesions. Hence, LDL aggregation, fusion, and lipid droplet formation are important early steps in atherogenesis. In vitro, a variety of enzymatic and nonenzymatic modifications of LDL can induce these reactions and thereby provide useful models for their detailed analysis. Here, we summarize current knowledge of the in vivo and in vitro modifications of LDLs leading to their aggregation, fusion, and lipid droplet formation; outline the techniques used to study these reactions; and propose a molecular mechanism that underlies these pro-atherogenic processes. Such knowledge is essential in identifying endogenous and exogenous factors that can promote or prevent LDL aggregation and fusion in vivo and to help establish new potential therapeutic targets to decelerate or even block these pathogenic reactions.
Collapse
Affiliation(s)
- Mengxiao Lu
- Department of Physiology and Biophysics, Boston University School of Medicine, W321, 700 Albany Street, Boston, MA 02118, USA.
| | | |
Collapse
|
17
|
Nguyen T, Toussaint J, Xue Y, Raval C, Cancel L, Russell S, Shou Y, Sedes O, Sun Y, Yakobov R, Tarbell JM, Jan KM, Rumschitzki DS. Aquaporin-1 facilitates pressure-driven water flow across the aortic endothelium. Am J Physiol Heart Circ Physiol 2015; 308:H1051-64. [PMID: 25659484 PMCID: PMC4551120 DOI: 10.1152/ajpheart.00499.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 01/23/2015] [Indexed: 01/23/2023]
Abstract
Aquaporin-1, a ubiquitous water channel membrane protein, is a major contributor to cell membrane osmotic water permeability. Arteries are the physiological system where hydrostatic dominates osmotic pressure differences. In the present study, we show that the walls of large conduit arteries constitute the first example where hydrostatic pressure drives aquaporin-1-mediated transcellular/transendothelial flow. We studied cultured aortic endothelial cell monolayers and excised whole aortas of male Sprague-Dawley rats with intact and inhibited aquaporin-1 activity and with normal and knocked down aquaporin-1 expression. We subjected these systems to transmural hydrostatic pressure differences at zero osmotic pressure differences. Impaired aquaporin-1 endothelia consistently showed reduced engineering flow metrics (transendothelial water flux and hydraulic conductivity). In vitro experiments with tracers that only cross the endothelium paracellularly showed that changes in junctional transport cannot explain these reductions. Percent reductions in whole aortic wall hydraulic conductivity with either chemical blocking or knockdown of aquaporin-1 differed at low and high transmural pressures. This observation highlights how aquaporin-1 expression likely directly influences aortic wall mechanics by changing the critical transmural pressure at which its sparse subendothelial intima compresses. Such compression increases transwall flow resistance. Our endothelial and historic erythrocyte membrane aquaporin density estimates were consistent. In conclusion, aquaporin-1 significantly contributes to hydrostatic pressure-driven water transport across aortic endothelial monolayers, both in culture and in whole rat aortas. This transport, and parallel junctional flow, can dilute solutes that entered the wall paracellularly or through endothelial monolayer disruptions. Lower atherogenic precursor solute concentrations may slow their intimal entrainment kinetics.
Collapse
Affiliation(s)
- Tieuvi Nguyen
- Department of Biomedical Engineering, City College of the City University of New York, New York, New York
| | - Jimmy Toussaint
- Department of Chemical Engineering, City College of the City University of New York, New York, New York
| | - Yan Xue
- Department of Chemical Engineering, City College of the City University of New York, New York, New York; Biology Department, City College and GSUC of The City College of New York, New York, New York; and
| | - Chirag Raval
- Department of Biomedical Engineering, City College of the City University of New York, New York, New York
| | - Limary Cancel
- Department of Biomedical Engineering, City College of the City University of New York, New York, New York
| | - Stewart Russell
- Department of Biomedical Engineering, City College of the City University of New York, New York, New York
| | - Yixin Shou
- Department of Chemical Engineering, City College of the City University of New York, New York, New York
| | - Omer Sedes
- Department of Chemical Engineering, City College of the City University of New York, New York, New York
| | - Yu Sun
- Department of Chemical Engineering, City College of the City University of New York, New York, New York
| | - Roman Yakobov
- Department of Chemical Engineering, City College of the City University of New York, New York, New York
| | - John M Tarbell
- Department of Biomedical Engineering, City College of the City University of New York, New York, New York
| | - Kung-ming Jan
- Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, New York
| | - David S Rumschitzki
- Department of Chemical Engineering, City College of the City University of New York, New York, New York; Biology Department, City College and GSUC of The City College of New York, New York, New York; and Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, New York
| |
Collapse
|
18
|
Reducing macrophage proteoglycan sulfation increases atherosclerosis and obesity through enhanced type I interferon signaling. Cell Metab 2014; 20:813-826. [PMID: 25440058 PMCID: PMC4254584 DOI: 10.1016/j.cmet.2014.09.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 08/30/2014] [Accepted: 09/26/2014] [Indexed: 01/05/2023]
Abstract
Heparan sulfate proteoglycans (HSPGs) are an important constituent of the macrophage glycocalyx and extracellular microenvironment. To examine their role in atherogenesis, we inactivated the biosynthetic gene N-acetylglucosamine N-deacetylase-N-sulfotransferase 1 (Ndst1) in macrophages and crossbred the strain to Ldlr(-/-) mice. When placed on an atherogenic diet, Ldlr(-/-)Ndst1(f/f)LysMCre(+) mice had increased atherosclerotic plaque area and volume compared to Ldlr(-/-) mice. Diminished sulfation of heparan sulfate resulted in enhanced chemokine expression; increased macrophages in plaques; increased expression of ACAT2, a key enzyme in cholesterol ester storage; and increased foam cell conversion. Motif analysis of promoters of upregulated genes suggested increased type I interferon signaling, which was confirmed by elevation of STAT1 phosphorylation induced by IFN-β. The proinflammatory macrophages derived from Ndst1(f/f)LysMCre(+) mice also sensitized the animals to diet-induced obesity. We propose that macrophage HSPGs control basal activation of macrophages by maintaining type I interferon reception in a quiescent state through sequestration of IFN-β.
Collapse
|
19
|
Chung S, Vafai K. Low-density lipoprotein transport within a multi-layered arterial wall—Effect of the atherosclerotic plaque/stenosis. J Biomech 2013; 46:574-85. [DOI: 10.1016/j.jbiomech.2012.09.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 09/05/2012] [Accepted: 09/15/2012] [Indexed: 11/27/2022]
|
20
|
Zeng Z, Jan KM, Rumschitzki DS. A theory for water and macromolecular transport in the pulmonary artery wall with a detailed comparison to the aorta. Am J Physiol Heart Circ Physiol 2011; 302:H1683-99. [PMID: 22198178 DOI: 10.1152/ajpheart.00447.2011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The pulmonary artery (PA) wall, which has much higher hydraulic conductivity and albumin void space and approximately one-sixth the normal transmural pressure of systemic arteries (e.g, aorta, carotid arteries), is rarely atherosclerotic, except under pulmonary hypertension. This study constructs a detailed, two-dimensional, wall-structure-based filtration and macromolecular transport model for the PA to investigate differences in prelesion transport processes between the disease-susceptible aorta and the relatively resistant PA. The PA and aorta models are similar in wall structure, but very different in parameter values, many of which have been measured (and therefore modified) since the original aorta model of Huang et al. (23). Both PA and aortic model simulations fit experimental data on transwall LDL concentration profiles and on the growth of isolated endothelial (horseradish peroxidase) tracer spots with circulation time very well. They reveal that lipid entering the aorta attains a much higher intima than media concentration but distributes better between these regions in the PA than aorta and that tracer in both regions contributes to observed tracer spots. Solutions show why both the overall transmural water flow and spot growth rates are similar in these vessels despite very different material transport parameters. Since early lipid accumulation occurs in the subendothelial intima and since (matrix binding) reaction kinetics depend on reactant concentrations, the lower intima lipid concentrations in the PA vs. aorta likely lead to slower accumulation of bound lipid in the PA. These findings may be relevant to understanding the different atherosusceptibilities of these vessels.
Collapse
Affiliation(s)
- Zhongqing Zeng
- Department of Chemical Engineering, City College of City University of New York, New York, New York 10031, USA
| | | | | |
Collapse
|
21
|
Hypoxia Stimulates Low-Density Lipoprotein Receptor–Related Protein-1 Expression Through Hypoxia-Inducible Factor-1α in Human Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2011; 31:1411-20. [DOI: 10.1161/atvbaha.111.225490] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective—
Hypoxia is considered a key factor in the progression of atherosclerotic lesions. Low-density lipoprotein receptor–related protein (LRP1) plays a pivotal role in the vasculature. The aim of this study was to investigate the effect of hypoxia on LRP1 expression and function in vascular smooth muscle cells (VSMC) and the role of hypoxia-inducible factor-α (HIF-1α).
Methods and Results—
Real-time polymerase chain reaction and Western blot analysis demonstrated that hypoxia (1% O
2
) time-dependently induced LRP1 mRNA (maximum levels at 1 to 2 hours) and protein expression (maximum levels at 12 to 24 hours). The delayed hypoxic upregulation of LRP1 protein versus mRNA may be explained by the long half-life of LRP1 protein. Luciferase assays demonstrated that hypoxia and HIF-1α overaccumulation induced LRP1 promoter activity and that 2 consensus hypoxia response element sites located at −1072/−1069 and −695/−692 participate in the induction. Chromatin immunoprecipitation showed the in vivo binding of HIF-1α to LRP1 promoter in hypoxic VSMC. Hypoxia effects on LRP1 protein expression were functionally translated into an increased cholesteryl ester (CE) accumulation from aggregated low-density lipoprotein (agLDL) uptake. The blockade of HIF-1α expression inhibited the upregulatory effect of hypoxia on LRP1 expression and agLDL-derived intracellular CE overaccumulation, suggesting that both LRP1 overexpression and CE overaccumulation in hypoxic vascular cells are dependent on HIF-1α. Immunohistochemical analysis showed the colocalization of LRP1 and HIF-1α in vascular cells of human advanced atherosclerotic plaques.
Conclusion—
Hypoxia upregulates LRP1 expression and agLDL-derived intracellular CE accumulation in human VSMC through HIF-1α induction.
Collapse
|
22
|
Borrell-Pagès M, Romero JC, Juan-Babot O, Badimon L. Wnt pathway activation, cell migration, and lipid uptake is regulated by low-density lipoprotein receptor-related protein 5 in human macrophages. Eur Heart J 2011; 32:2841-50. [PMID: 21398644 DOI: 10.1093/eurheartj/ehr062] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AIMS Atherosclerosis plaque development includes infiltration of inflammatory cells, accumulation of lipids and fibrous cap formation. Low-density lipoprotein receptor-related protein 1 (LRP1) is expressed on atherosclerotic lesions associated with macrophages and vascular smooth muscle cells. The aim of this work is to analyse the role in atherosclerosis lesion progression of another member of the LDL receptor protein family, low-density lipoprotein receptor-related protein 5 (LRP5), a co-receptor with Frizzled known to activate the Wnt signalling pathway in several cell types. METHODS AND RESULTS LRP5 is expressed in human vascular and innate inflammatory cells. LRP5 is transcriptionally regulated by aggregated LDL (agLDL), participating in the lipid uptake and transformation of macrophages into foam cells, a critical step in atherosclerosis progression. AgLDL-treated macrophages show up-regulated expression of β-catenin, LEF1, c-jun, cyclinD1, bone morphogenetic protein 2 (BMP2), and osteopontin (OPN), proteins and targets of the Wnt signalling pathway, whereas LRP5-silenced macrophages show a significant down-regulation of OPN and BMP2 expression. Furthermore, LRP5-deficient macrophages exhibit an impaired migration both in wound-repair and modified Boyden chambers models. CONCLUSION These results demonstrate the involvement of LRP5 in the innate inflammatory reaction to lipid infiltration in atherosclerosis.
Collapse
Affiliation(s)
- Maria Borrell-Pagès
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, UAB, Barcelona, Spain
| | | | | | | |
Collapse
|
23
|
Shiomi M, Ito T. The Watanabe heritable hyperlipidemic (WHHL) rabbit, its characteristics and history of development: A tribute to the late Dr. Yoshio Watanabe. Atherosclerosis 2009; 207:1-7. [DOI: 10.1016/j.atherosclerosis.2009.03.024] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2009] [Revised: 03/13/2009] [Accepted: 03/17/2009] [Indexed: 11/28/2022]
|
24
|
Curcio CA, Johnson M, Huang JD, Rudolf M. Aging, age-related macular degeneration, and the response-to-retention of apolipoprotein B-containing lipoproteins. Prog Retin Eye Res 2009; 28:393-422. [PMID: 19698799 PMCID: PMC4319375 DOI: 10.1016/j.preteyeres.2009.08.001] [Citation(s) in RCA: 189] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The largest risk factor for age-related macular degeneration (ARMD) is advanced age. A prominent age-related change in the human retina is the accumulation of histochemically detectable neutral lipid in normal Bruch's membrane (BrM) throughout adulthood. This change has the potential to have a major impact on physiology of the retinal pigment epithelium (RPE). It occurs in the same compartment as drusen and basal linear deposit, the pathognomonic extracellular, lipid-containing lesions of ARMD. Here we present evidence from light microscopic histochemistry, ultrastructure, lipid profiling of tissues and isolated lipoproteins, and gene expression analysis that this deposition can be accounted for by esterified cholesterol-rich, apolipoprotein B-containing lipoprotein particles constitutively produced by the RPE. This work collectively allows ARMD lesion formation and its aftermath to be conceptualized as a response to the retention of a sub-endothelial apolipoprotein B lipoprotein, similar to a widely accepted model of atherosclerotic coronary artery disease (CAD) (Tabas et al., 2007). This approach provides a wide knowledge base and sophisticated clinical armamentarium that can be readily exploited for the development of new model systems and the future benefit of ARMD patients.
Collapse
Affiliation(s)
- Christine A Curcio
- Department of Ophthalmology, University of Alabama School of Medicine, Birmingham, AL 35294-0009, USA.
| | | | | | | |
Collapse
|
25
|
Curcio CA, Johnson M, Huang JD, Rudolf M. Apolipoprotein B-containing lipoproteins in retinal aging and age-related macular degeneration. J Lipid Res 2009; 51:451-67. [PMID: 19797256 DOI: 10.1194/jlr.r002238] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The largest risk factor for age-related macular degeneration (ARMD) is advanced age. With aging, there is a striking accumulation of neutral lipids in Bruch's membrane (BrM) of normal eye that continues through adulthood. This accumulation has the potential to significantly impact the physiology of the retinal pigment epithelium (RPE). It also ultimately leads to the creation of a lipid wall at the same locations where drusen and basal linear deposit, the pathognomonic extracellular, lipid-containing lesions of ARMD, subsequently form. Here, we summarize evidence obtained from light microscopy, ultrastructural studies, lipid histochemistry, assay of isolated lipoproteins, and gene expression analysis. These studies suggest that lipid deposition in BrM is at least partially due to accumulation of esterified cholesterol-rich, apolipoprotein B-containing lipoprotein particles produced by the RPE. Furthermore, we suggest that the formation of ARMD lesions and their aftermath may be a pathological response to the retention of a sub-endothelial apolipoprotein B lipoprotein, similar to a widely accepted model of atherosclerotic coronary artery disease (Tabas, I., K. J. Williams, and J. Borén. 2007. Subendothelial lipoprotein retention as the initiating process in atherosclerosis: update and therapeutic implications. Circulation. 116:1832-1844). This view provides a conceptual basis for the development of novel treatments that may benefit ARMD patients in the future.
Collapse
Affiliation(s)
- Christine A Curcio
- Department of Ophthalmology, University of Alabama School of Medicine, Birmingham, AL, USA.
| | | | | | | |
Collapse
|
26
|
Lu Y, Guo J, Di Y, Zong Y, Qu S, Tian J. Proteomic analysis of the triglyceride-rich lipoprotein-laden foam cells. Mol Cells 2009; 28:175-81. [PMID: 19756395 DOI: 10.1007/s10059-009-0120-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 07/01/2009] [Accepted: 07/22/2009] [Indexed: 11/29/2022] Open
Abstract
In hypertriglyceridaemic individuals, atherosclerogenesis is associated with the increased concentrations of very low density lipoprotein (VLDL) and VLDL-associated remnant particles. In vitro studies have suggested that VLDL induces foam cells formation. To reveal the changes of the proteins expression in the process of foam cells formation induced by VLDL, we performed a proteomic analysis of the foam cells based on the stimulation of differentiated THP-1 cells with VLDL. Using two-dimensional gel electrophoresis (2-DE) and matrix-assisted laser-desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) analysis, 14 differentially expressed proteins, containing 8 up-regulated proteins and 6 down-regulated proteins were identified. The proteins are involved in energy metabolism, oxidative stress, cell growth, differentiation and apoptosis, such as adipose differentiation-related protein (ADRP), enolase, S100A11, heat shock protein 27 and so on. In addition, the expression of some selected proteins was confirmed by Western blot and RT-PCR analysis. The results suggest that VLDL not only induces lipid accumulation, but also brings about foam cells diverse characteristics by altering the expression of various proteins.
Collapse
Affiliation(s)
- Yanjun Lu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | | | | | | | | | | |
Collapse
|
27
|
Otero-Viñas M, Llorente-Cortés V, Peña E, Padró T, Badimon L. Aggregated low density lipoproteins decrease metalloproteinase-9 expression and activity in human coronary smooth muscle cells. Atherosclerosis 2007; 194:326-33. [PMID: 17134708 DOI: 10.1016/j.atherosclerosis.2006.10.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2006] [Revised: 10/10/2006] [Accepted: 10/23/2006] [Indexed: 11/22/2022]
Abstract
Plaque stability largely depends on vascular smooth muscle cell (VSMC) function. VSMC secrete metalloproteinases (MMPs), matrix degrading endopeptidases, that regulate VSMC migration and function. Among them, gelatinase B or MMP-9 seems to have a protective effect by promoting a stable plaque phenotype. In macrophage foam cells oxidized LDL (oxLDL) uptake regulates MMP-9 expression. However, it is unknown whether VSMC-lipid loading by aggregated LDL (agLDL) internalization produces any effect on MMP-9 production by human resident vascular cells. In the present study, we analyzed the effect of lipid-internalization in MMP-9 and MMP-2 expression and activity and its consequences in VSMC migration. Our results show that agLDL-internalization down-regulates MMP-9 activity in a time-dependent manner up to 42% at 48h and in a dose-dependent manner up to 87% at 300 microg/mL. nLDL induced similar but not sustained decrease on MMP-9 activity. However, neither agLDL nor nLDL exerted any significant effect on MMP-2 and TIMP-1. VSMC regrowth after a scratch injury was significantly reduced by exposure to agLDL. We conclude that agLDL-lipid loading reduces MMP-9 activity and this effect is associated to inhibition of VSMC migration. Thus, agLDL internalization may have consequences on vascular remodeling after injury, and the stability of lipid-rich atherosclerotic plaques.
Collapse
|
28
|
Llorente-Cortés V, Royo T, Juan-Babot O, Badimon L. Adipocyte differentiation-related protein is induced by LRP1-mediated aggregated LDL internalization in human vascular smooth muscle cells and macrophages. J Lipid Res 2007; 48:2133-40. [PMID: 17620659 DOI: 10.1194/jlr.m700039-jlr200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Aggregated LDL (agLDL) is internalized by LDL receptor-related protein (LRP1) in vascular smooth muscle cells (VSMCs) and human monocyte-derived macrophages (HMDMs). AgLDL is, therefore, a potent inducer of massive intracellular cholesteryl ester accumulation in lipid droplets. The adipocyte differentiation-related protein (ADRP) has been found on the surface of lipid droplets. The objectives of this work were to analyze whether agLDL uptake modulates ADRP expression levels and whether the effect of agLDL internalization on ADRP expression depends on LRP1 in human VSMCs and HMDMs. AgLDL strongly upregulates ADRP mRNA (real-time PCR) and protein expression (Western blot) in human VSMCs (mRNA: by 3.06-fold; protein: 8.58-fold) and HMDMs (mRNA: by 3.5-fold; protein: by 3.71-fold). Treatment of VSMCs and HMDMs with small anti-LRP1-interfering RNA (siRNA-LRP1) leads to specific inhibition of LRP1 expression. siRNA-LRP1 treatment significantly reduced agLDL-induced ADRP overexpression in HMDMs (by 69%) and in VSMCs (by 53%). Immunohystochemical studies evidence a colocolocalization between ADRP/macrophages and ADRP/VSMCs in advanced lipid-enriched atherosclerotic plaques. These results demonstrate that agLDL-LRP1 engagement induces ADRP overexpression in both HMDMs and human VSMCs and that ADRP is highly expressed in advanced lipid-enriched human atherosclerotic plaques. Therefore, LRP1-mediated agLDL uptake might play a pivotal role in vascular foam cell formation.
Collapse
Affiliation(s)
- V Llorente-Cortés
- Cardiovascular Research Center, Consejo Superior de Investigaciones Científicas-Instituto Catalán de Ciencias Cardiovasculares, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | | |
Collapse
|
29
|
Shou Y, Jan KM, Rumschitzki DS. Transport in rat vessel walls. II. Macromolecular leakage and focal spot size growth in rat arteries and veins. Am J Physiol Heart Circ Physiol 2007; 292:H2881-90. [PMID: 17209003 DOI: 10.1152/ajpheart.00575.2006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Transendothelial lipid transport into and spread in the subendothelial intima of large arteries, and subsequent lipid accumulation, appear to start plaque formation. We experimentally examine transendothelial horseradish peroxidase (HRP) transport in vessels that are usually, e.g., pulmonary artery (PA), or almost always, e.g., inferior vena cava (IVC), atherosclerosis resistant vs. disease prone, e.g., aorta, vessels. In these vessels, HRP traverses the endothelium at isolated, focal spots, rather than uniformly, for short circulation times. For femoral vein HRP introduction, PA spots have 30-s radii [∼53.2 μm (SD10.4); compare aorta: 54.6 μm (SD8.75)] and grow quickly from 30 s to 1 min (40%, P < 0.05) and more slowly afterward ( P > 0.05). This trend resembles the aorta, suggesting the PA has a similarly sparse intima. With carotid artery (CA) HRP introduction, the 30-s spot (132.86 ± 37.32 μm) is far larger than the PAs, grows little (∼28%, P < 0.05) from 30 to 60 s, and is much flatter than the artery curves. Transverse electron microscopic sections after ∼10 min HRP circulation show thin, intense staining immediately beneath both vessels’ endothelia with an almost step change to diffuse staining beyond. This indicates the existence of a sparse, subendothelial intima, even when there is no internal elastic lamina (IVC). This motivates a simple model that translates growth rates into lower bounds for the flow through focal leaks. The model results and our earlier wall and medial hydraulic conductivity data explain these spot growth curves and point to differences in transport patterns that might be relevant in understanding the immunity of IVC to disease initiation.
Collapse
Affiliation(s)
- Yixin Shou
- Department of Chemical Engineering, City College of the City University of New York, Convent Ave. at 140th St., New York, NY 10031, USA
| | | | | |
Collapse
|
30
|
Zeng Z, Yin Y, Jan KM, Rumschitzki DS. Macromolecular transport in heart valves. II. Theoretical models. Am J Physiol Heart Circ Physiol 2007; 292:H2671-86. [PMID: 17220189 DOI: 10.1152/ajpheart.00608.2006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This paper proposes a new, two-dimensional convection-diffusion model for macromolecular transport in heart valves based on horseradish peroxidase (HRP) experiments on rats presented in the first of the papers in this series (Part I; Zeng Z, Yin Y, Huang AL, Jan KM, Rumschitzki DS. Am J Physiol Heart Circ Physiol 292: H2664–H2670, 2007). Experiments require two valvular intimae, one underneath each endothelium. Tompkins et al. (Tompkins RG, Schnitzer JJ, Yarmush ML. Circ Res 64: 1213–1223, 1989) found large variations in shape and magnitude in transvalvular125I-labeled low-density lipoprotein (LDL) profiles from identical experiments on four squirrel monkeys. Their one-dimensional, uniform-medium diffusion-only model fit three parameters independently for each profile; data variability resulted in large parameter spreads. Our theory aims to explain their data with one parameter set. It uses measured parameters and some aortic values but fits the endothelial mass transfer coefficient ( ka= kv= 1.63 × 10−8cm/s, where subscripts a and v indicate aortic aspect and ventricular aspect, respectively) and middle layer permeability ( K[Formula: see text]= 2.28 × 10−16cm2) and LDL diffusion coefficient [ D2(LDL) = 5.93 × 10−9cm2/s], using one of Tompkins et al.'s profiles, and fixes them throughout. It accurately predicts Part I's rapid localized HRP leakage spot growth rate in rat leaflets that results from the intima's much sparser structure, dictating its far larger transport parameters [ K[Formula: see text]= 1.10 × 10−12cm2, D1(LDL/HRP) = 1.02/4.09 × 10−7cm2/s] than the middle layer. This contrasts with large arteries with similarly large HRP spots, since the valve has no internal elastic lamina. The model quantitatively explains all of Tompkins et al.'s monkey profiles with these same parameters. Different numbers and locations of isolated macromolecular leaks on both aspects and different section-leak(s) distances yield all profiles.
Collapse
Affiliation(s)
- Zhongqing Zeng
- Department of Chemical Engineering, City College of the City University of New York, NY 10031, USA
| | | | | | | |
Collapse
|
31
|
Zeng Z, Nievelstein-Post P, Yin Y, Jan KM, Frank JS, Rumschitzki DS. Macromolecular transport in heart valves. III. Experiment and theory for the size distribution of extracellular liposomes in hyperlipidemic rabbits. Am J Physiol Heart Circ Physiol 2007; 292:H2687-97. [PMID: 17237250 DOI: 10.1152/ajpheart.00606.2006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The heart valve leaflets of 29-day cholesterol-fed rabbits were examined by ultrarapid freezing without conventional chemical fixation/processing, followed by rotary shadow freeze-etching. This procedure images the leaflets' subendothelial extracellular matrix in extraordinary detail, and extracellular lipid liposomes, from 23 to 220 nm in diameter, clearly appear there. These liposomes are linked to matrix filaments and appear in clusters. Their size distribution shows 60.7% with diameters 23–69 nm, 31.7% between 70 and 119 nm, 7.3% between 120 and 169 nm, and 0.3% between 170 and 220 nm (superlarge) and suggests that smaller liposomes can fuse into larger ones. We couple our model from Part II of this series (Zeng Z, Yin Y, Jan KM, Rumschitzki DS. Am J Physiol Heart Circ Physiol 292: H2671–H2686, 2007) for lipid transport into the leaflet to the nucleation-polymerization model hierarchy for liposome formation proposed originally for aortic liposomes to predict liposome formation/growth in heart valves. Simulations show that the simplest such model cannot account for the observed size distribution. However, modifying this model by including liposome fusing/merging, using parameters determined from aortic liposomes, leads to predicted size distributions in excellent agreement with our valve data. Evolutions of both the liposome size distribution and total liposome mass suggest that fusing becomes significant only after 2 wk of high lumen cholesterol. Inclusion of phagocytosis by macrophages limits the otherwise monotonically increasing total liposome mass, while keeping the excellent fit of the liposome size distribution to the data.
Collapse
Affiliation(s)
- Zhongqing Zeng
- Department of Chemical Engineering, City College of the City University of New York, NY 10031, USA
| | | | | | | | | | | |
Collapse
|
32
|
Zeng Z, Yin Y, Huang AL, Jan KM, Rumschitzki DS. Macromolecular transport in heart valves. I. Studies of rat valves with horseradish peroxidase. Am J Physiol Heart Circ Physiol 2007; 292:H2664-70. [PMID: 17277015 DOI: 10.1152/ajpheart.01419.2006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study aims to experimentally elucidate subtle structural features of the rat valve leaflet and the related nature of macromolecular transport across its endothelium and in its subendothelial space, information necessary to construct a rational theoretical model that can explain observation. After intravenous injection of horseradish peroxidase (HRP), we perfusion-fixed the aortic valve of normal Sprague-Dawley rats and found under light microscopy that HRP leaked through the leaflet's endothelium at very few localized brown spots, rather than uniformly. These spots grew nearly as rapidly with HRP circulation time before euthanasia as aortic spots, particularly when the time axis only included the time the valve was closed. These results suggest that macromolecular transport in heart valves depends not only on the direction normal to, but also parallel to, the endothelial surface and that convection, as well as molecular diffusion, plays an important role in macromolecular transport in heart valves. Transmission electron microscopy of traverse leaflet sections after 4-min HRP circulation showed a very thin (∼150 nm), sparse layer immediately beneath the endothelium where the HRP concentration was much higher than that in the matrix below it. Nievelstein-Post et al.'s (Nievelstein-Post P, Mottino G, Fogelman A, Frank J. Arterioscler Thromb 14: 1151–1161, 1994) ultrarapid freezing/rotary shadow etching of the normal rabbit valve's subendothelial space supports the existence of this very thin, very sparse “valvular subendothelial intima,” in analogy to the vascular subendothelial intima.
Collapse
Affiliation(s)
- Zhongqing Zeng
- Department of Chemical Engineering, City College of the City University of New York, NY 10031, USA
| | | | | | | | | |
Collapse
|
33
|
Huang JD, Presley JB, Chimento MF, Curcio CA, Johnson M. Age-related changes in human macular Bruch's membrane as seen by quick-freeze/deep-etch. Exp Eye Res 2007; 85:202-18. [PMID: 17586493 PMCID: PMC2352152 DOI: 10.1016/j.exer.2007.03.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Revised: 03/22/2007] [Accepted: 03/29/2007] [Indexed: 10/23/2022]
Abstract
Lipid-containing inclusions have been observed in human Bruch's membrane (BrM) and are postulated to be associated with age-related maculopathy (ARM), a major cause of legal blindness in developed countries. The dehydration associated with specimen preparation for thin-section transmission electron microscopy causes loss of these inclusions. Better preservation of the ultrastructure of the inclusions and tissue is achieved by using a quick-freeze/deep-etch preparation. We use this technique to examine normal human macular BrM in order to characterize the deposition of the lipid-rich inclusions and their age-related accumulation within different layers of the tissue. We find that various inclusions mentioned in other studies can be formed by combinations of three basic structures: lipoprotein-like particles (LLPs), small granules (SGs) and membrane-like structures. These inclusions are associated with collagen and elastic fibrils by fine filaments. In younger eyes, these inclusions are found mostly in the elastic (EL) and outer collageneous layer (OCL) and occupy a small fraction of the interfibrillar spacing. As age increases, LLPs and SGs gradually fill the interfibrillar spacing of the EL and inner collageneous layer (ICL) of the tissue, and later form a new sublayer, the lipid wall, within the boundary region between the basal lamina of retinal pigment epithelium (RPE) and ICL. Because the formation of the lipid wall only occurs after these inclusions fill the ICL, and it seems unlikely that the LLPs can pass through the packed layer, this result suggests a possible RPE origin of the LLPs that make up the lipid wall.
Collapse
Affiliation(s)
- Jiahn-Dar Huang
- Department of Biomedical Engineering, Northwestern University, Evanston IL USA
| | - J. Brett Presley
- Department of Ophthalmology, University of Alabama School of Medicine, Birmingham AL USA
| | - Melissa F. Chimento
- Department of Ophthalmology, University of Alabama School of Medicine, Birmingham AL USA
| | - Christine A. Curcio
- Department of Ophthalmology, University of Alabama School of Medicine, Birmingham AL USA
| | - Mark Johnson
- Department of Biomedical Engineering, Northwestern University, Evanston IL USA
| |
Collapse
|
34
|
Xu S, Yu JJ. Beneath the minerals, a layer of round lipid particles was identified to mediate collagen calcification in compact bone formation. Biophys J 2006; 91:4221-9. [PMID: 16980361 PMCID: PMC1635673 DOI: 10.1529/biophysj.105.075804] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2005] [Accepted: 07/25/2006] [Indexed: 11/18/2022] Open
Abstract
Astronauts lose 1-2% of their bone minerals per month during space flights. A systematic search for a countermeasure relies on a good understanding of the mechanism of bone formation at the molecular level. How collagen fibers, the dominant matrix protein in bones, are mineralized remains mysterious. Atomic force microscopy was carried out, in combination with immunostaining and Western blotting, on bovine tibia to identify unrecognized building blocks involved in bone formation and for an elucidation of the process of collagen calcification in bone formation. Before demineralization, tiles of hydroxyapatite crystals were found stacked along bundles of collagen fibers. These tiles were homogeneous in size and shape with dimensions 0.69 x 0.77 x 0.2 micro m(3). Demineralization dissolved these tiles and revealed small spheres with an apparent diameter around 145 nm. These spheres appeared to be lipid particles since organic solvents dissolved them. The parallel collagen bundles had widths mostly <2 micro m. Composition analysis of compact bones indicated a high content of apolar lipids, including triglycerides and cholesterol esters. Apolar lipids are known to form lipid droplets or lipoproteins, and these spheres are unlikely to be matrix vesicles as reported for collagen calcification in epiphyseal cartilages. Results from this study suggest that the layer of round lipid particles on collagen fibers mediates the mineral deposition onto the fibers. The homogeneous size of these lipid particles and the presence of apolipoprotein in demineralized bone tissue suggest the possibility that these particles might be of lipoprotein origin. More studies are needed to verify the last claim and to exclude the possibility that they are secreted lipid droplets.
Collapse
Affiliation(s)
- Shaohua Xu
- Florida Space Research Institute, Space Life Sciences Laboratory, Kennedy Space Center, Cape Canaveral, FL 32899, USA.
| | | |
Collapse
|
35
|
Simionescu M. Implications of early structural-functional changes in the endothelium for vascular disease. Arterioscler Thromb Vasc Biol 2006; 27:266-74. [PMID: 17138941 DOI: 10.1161/01.atv.0000253884.13901.e4] [Citation(s) in RCA: 174] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
By location, between the blood and tissues and the multiple functions, the endothelial cells (ECs) play a major role in securing body homeostasis. The ECs sense all variations occurring in the plasma and interstitial fluid, and respond (function of intensity), initially by modulation of their constitutive functions, then by dysfunction, expressed by temporarily altered functions and a phenotypic shift, and ultimately by injury/death. In dyslipidemia/hyperglycemia, the initial response of EC is the modulation of 2 constitutive functions: permeability and biosynthesis. Increased transcytosis of plasma beta-lipoproteins leads to their accumulation within the hyperplasic basal lamina, interaction with matrix proteins, and conversion to modified and reassembled lipoproteins (MRL). This generates a multipart inflammatory process and EC dysfunction characterized by expression of new cell adhesion molecules and MCP-1 that trigger T-lymphocytes and monocyte recruitment, diapedesis, and homing within the subendothelium where activated macrophages become foam cells. The latter, together with the subendothelial accrual of MRL, growth factors, cytokines, and chemokines, and accretion of smooth muscle cells of various sources lead to atheroma formation; in advanced disease, the EC overlaying atheroma take up lipids, become EC-derived foam cells, and the cytotoxic ambient ultimately conducts to EC apoptosis. Understanding the mechanisms of EC dysfunction is a prerequisite for EC-targeted therapy to reduce the incidence of cardiovascular diseases.
Collapse
Affiliation(s)
- Maya Simionescu
- Institute of Cellular Biology and Pathology Nicolae Simionescu, 8, B. P. Hasdeu Street, Bucharest, Romania.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Binding of apolipoprotein B-100-containing lipoproteins (VLDL, IDL, and LDL) to proteoglycans and modifications of the lipoproteins, whether bound or unbound, are key processes in atherogenesis. The complex interplay between binding and modification has been studied at neutral pH conditions. It has been demonstrated that during atherogenesis the extracellular pH of the lesions decreases. We summarize findings suggesting that lipoprotein binding and modification are enhanced at acidic pH. RECENT FINDINGS Many enzymes found in the arterial intima, such as secretory sphingomyelinase and cathepsins, are able to hydrolyze lipoproteins in vitro. These enzymes function optimally at slightly acidic pH (pH 5.5-6.5), and are likely to act on lipoproteins optimally in the acidic plaque areas. Also, the ability of human aortic proteoglycans to bind native VLDL, IDL, and LDL is dramatically increased at acidic pH; this binding can be further increased if these apolipoprotein B-100-containing particles are hydrolytically modified. SUMMARY Recent in-vitro findings suggest that in areas of atherosclerotic arterial intima where the extracellular pH is decreased, binding of apolipoprotein B-100-containing lipoproteins to proteoglycans and modification of the lipoproteins by acidic enzymes are enhanced. The pH-induced amplification of these processes will lead to enhanced extracellular accumulation of lipoproteins and accelerated progression of the disease.
Collapse
Affiliation(s)
- Katariina Oörni
- Wihuri Research Institute, Kalliolinnantie 4, FIN-00140 Helsinki, Finland.
| | | |
Collapse
|
37
|
Shou Y, Jan KM, Rumschitzki DS. Transport in rat vessel walls. I. Hydraulic conductivities of the aorta, pulmonary artery, and inferior vena cava with intact and denuded endothelia. Am J Physiol Heart Circ Physiol 2006; 291:H2758-71. [PMID: 16731638 DOI: 10.1152/ajpheart.00610.2005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this study, filtration flows through the walls of the rat aorta, pulmonary artery (PA), and inferior vena cava (IVC), vessels with very different susceptibilities to atherosclerosis, were measured as a function of transmural pressure (DeltaP), with intact and denuded endothelium on the same vessel. Aortic hydraulic conductivity (L(p)) is high at 60 mmHg, drops approximately 40% by 100 mmHg, and is pressure independent to 140 mmHg. The trends are similar in the PA and IVC, dropping 42% from 10 to 40 mmHg and flat to 100 mmHg (PA) and dropping 33% from 10 to 20 mmHg and essentially flat to 60 mmHg (IVC). Removal of the endothelium renders L(p)(DeltaP) flat: it increases L(p) of the aorta by approximately 75%, doubles L(p) of the PA, and quadruples L(p) of the IVC. Specific resistance (1/L(p)) of the aortic endothelium is approximately 47% of total resistance; i.e., the endothelium accounts for approximately 47% of the DeltaP drop at 100 mmHg. The PA value is 55% at >40 mmHg, and the IVC value is 23% at 10 mmHg. L(p) of the intact aorta, PA, and IVC are order 10(-8), 10(-7), and 5 x 10(-7) cm.s(-1).mmHg(-1), and wall thicknesses are 145.8 microm (SD 9.3), 78.9 microm (SD 3.3), and 66.1 microm (SD 4.1), respectively. These data are consistent with the different wall structures of the three vessels. The rat aortic L(p) data are quantitatively consistent with rabbit L(p)(DeltaP) (Tedgui A and Lever MJ. Am J Physiol Heart Circ Physiol 247: H784-H791, 1984; Baldwin AL and Wilson LM. Am J Physiol Heart Circ Physiol 264: H26-H32, 1993), suggesting that intimal compression under pressure loading may also play a role in L(p)(DeltaP) in these other vessels. Despite very different driving DeltaP, nominal transmural water fluxes of these three vessels are very similar and, therefore, cannot alone account for their differences in disease susceptibility. The different fates of macromolecular tracers convected by these water fluxes into the walls of these vessels may account for this difference.
Collapse
Affiliation(s)
- Yixin Shou
- Dept. of Chemical Engineering, City University of New York, New York, NY 10031, USA
| | | | | |
Collapse
|
38
|
Nakajima K, Nakano T, Tanaka A. The oxidative modification hypothesis of atherosclerosis: The comparison of atherogenic effects on oxidized LDL and remnant lipoproteins in plasma. Clin Chim Acta 2006; 367:36-47. [PMID: 16448638 DOI: 10.1016/j.cca.2005.12.013] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2005] [Revised: 12/07/2005] [Accepted: 12/08/2005] [Indexed: 12/18/2022]
Abstract
A tremendous number of articles on oxidized LDL (Ox-LDL) and scavenger receptor in macrophage have been published since Steinberg proposed Ox-LDL hypothesis as the major cause of atherosclerosis. This hypothesis has provided strong support for the efficacy of LDL lowering drugs, indicating that lowering LDL means lowering Ox-LDL in vivo. This manuscript proposed a new oxidative modification hypothesis that remnant lipoproteins determined as remnant-like lipoprotein particles (RLP), not LDL are the major oxidized lipoproteins in plasma, resulting from the plasma concentration of these oxidized lipoproteins. Remnant lipoproteins may play a pivotal role for the initiation of atherosclerosis via lectin-like oxidized LDL receptor-1 (LOX-1) in endothelial cells. Isolated remnant lipoproteins were found to be oxidized or susceptible to be oxidized in plasma, not necessary to be further oxidized in vitro as Ox-LDL. High similarity of proatherogenic and proinflammatory properties of isolated Ox-LDL and remnant lipoporteins have been reported and predicted the presence of similar oxidized phospholipids in both lipoproteins as bioactive components. These results suggest the possibility that reducing plasma remnant lipoproteins rather than LDL should be the target for hyperlipidemic therapy especially in patients with metabolic syndrome for the prevention of endothelial dysfunction in the initiation of atherosclerosis.
Collapse
Affiliation(s)
- Katsuyuki Nakajima
- Japan Immunoresearch Laboratories, Co. Ltd., Takasaki, Gunma, and Department of Health and Nutrition, College of Human and Environmental Studies, Kanto-Gakuin University, Yokohama, Japan.
| | | | | |
Collapse
|
39
|
Miyoshi T, Tian J, Matsumoto AH, Shi W. Differential response of vascular smooth muscle cells to oxidized LDL in mouse strains with different atherosclerosis susceptibility. Atherosclerosis 2006; 189:99-105. [PMID: 16405896 DOI: 10.1016/j.atherosclerosis.2005.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2005] [Revised: 11/26/2005] [Accepted: 12/01/2005] [Indexed: 10/25/2022]
Abstract
Oxidized low-density lipoprotein (LDL) has numerous atherogenic properties, including induction of inflammatory genes, and vascular smooth muscle cells (VSMC) are involved in the development of atherosclerosis. In this study, we examined whether variations of VSMC in the capacity to oxidize LDL or in response to minimally modified LDL (MM-LDL) constitute a genetic component in atherosclerosis. VSMC were isolated from the aorta of two inbred mouse strains C57BL/6J (B6) and C3H, which differ markedly in susceptibility to atherosclerosis. LDL oxidation was assessed by measuring thiobarbituric acid-reactive substance (TBARS) production. Responses to MM-LDL were evaluated by examining the expression of inflammatory genes involved atherosclerosis, including monocyte chemotactic protein-1 (MCP-1) and vascular cell adhesion molecule-1 (VCAM-1), and an oxidant stress gene, heme oxygenase-1 (HO-1). VSMC from the two strains exhibited a comparable ability to transform native LDL to oxidized LDL, whereas their response to MM-LDL differed markedly. MM-LDL resulted in dramatic induction of MCP-1, VCAM-1, and HO-1 mRNAs in the cells from B6 mice but exerted little effect in cells from C3H mice. MCP-1 and soluble VCAM-1 protein levels in conditioned media were measured by ELISA. B6 cells produced significantly more MCP-1 and VCAM-1 proteins in response to MM-LDL than C3H cells. These data suggest that variation in the response of VSMC to oxidized LDL may contribute to the difference between B6 and C3H mice in atherosclerosis susceptibility.
Collapse
MESH Headings
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cells, Cultured
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Disease Models, Animal
- Enzyme-Linked Immunosorbent Assay
- Female
- Gene Expression
- Genetic Predisposition to Disease
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/metabolism
- Lipoproteins, LDL/genetics
- Lipoproteins, LDL/metabolism
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Oxidation-Reduction
- RNA, Messenger/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Thiobarbituric Acid Reactive Substances/metabolism
- Vascular Cell Adhesion Molecule-1/genetics
- Vascular Cell Adhesion Molecule-1/metabolism
Collapse
Affiliation(s)
- Toru Miyoshi
- Department of Radiology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
40
|
Sohn M, Tan Y, Wang B, Klein RL, Trojanowska M, Jaffa AA. Mechanisms of low-density lipoprotein-induced expression of connective tissue growth factor in human aortic endothelial cells. Am J Physiol Heart Circ Physiol 2005; 290:H1624-34. [PMID: 16272194 DOI: 10.1152/ajpheart.01233.2004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyperlipidemia is a recognized risk factor for atherosclerotic vascular disease. The underlying mechanisms that link lipoproteins and vascular disease are undefined. Connective tissue growth factor (CTGF) is emerging as a key determinant of progressive fibrotic diseases, and its expression is upregulated by diabetes. To define the mechanisms through which low-density lipoproteins (LDL) promote vascular injury, we evaluated whether LDL can modulate the expression of CTGF and collagen IV in human aortic endothelial cells (HAECs). Treatment of HAECs with LDL (50 microg/ml) for 24 h produced a significant increase in the mRNA and the protein levels of CTGF and collagen IV compared with unstimulated controls. To explore the mechanisms by which LDL regulates CTGF and collagen IV expression in HAECs, we determined first if CTGF and collagen IV are downstream targets for regulation by transforming growth factor-beta (TGF-beta). The results demonstrated that TGF-beta produced a concentration-dependent increase in the protein levels of CTGF. To assess whether the induction of CTGF in response to LDL is mediated via autocrine activation of TGF-beta, HAECs were treated with LDL for 24 h in the presence and absence of anti-TGF-beta neutralizing antibodies (anti-TGF-beta NA). The results demonstrated that the increase in CTGF induced by LDL was significantly inhibited by the anti-TGF-beta NA. To investigate the upstream mediators of TGF-beta on activity of CTGF in response to LDL, HAECs were treated with LDL for 24 h in the presence and absence of cell-permeable MAPK inhibitors. Inhibition of p38(mapk) activities did not affect LDL-induced TGF-beta1, CTGF, and collagen IV expression. On the other hand, SP-600125, a specific inhibitor of c-Jun NH(2)-terminal kinase, suppressed LDL-induced TGF-beta, CTGF, and collagen IV expression, and PD-98059, a selective inhibitor of p44/42(mapk), suppressed LDL-induced TGF-beta and CTGF expression. These findings are the first to implicate the MAPK pathway and TGF-beta as key players in LDL signaling, leading to CTGF and collagen IV expression in HAECs. The data also point to a potential mechanistic pathway through which lipoproteins may promote vascular injury.
Collapse
Affiliation(s)
- Mimi Sohn
- Dept. of Medicine, Div. of Endocrinology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | | | |
Collapse
|
41
|
Steinberg D. Thematic review series: The Pathogenesis of Atherosclerosis. An interpretive history of the cholesterol controversy, part III: mechanistically defining the role of hyperlipidemia. J Lipid Res 2005; 46:2037-51. [PMID: 15995167 DOI: 10.1194/jlr.r500010-jlr200] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
In this third installment of the series, we point out that the absence of an explicit, detailed and plausible hypothesis linking hypercholesterolemia to the events in the artery wall was probably an important reason for continuing skepticism and for failure to treat elevated blood cholesterol levels. The rapid advances in understanding of lipoprotein metabolism in the 1950s and 1960s and the application of modern cellular biology in the 1970s provided the context for a modern consensus on pathogenetic mechanisms of atherogenesis.
Collapse
Affiliation(s)
- Daniel Steinberg
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
42
|
Navab M, Anantharamaiah GM, Reddy ST, Hama S, Hough G, Frank JS, Grijalva VR, Ganesh VK, Mishra VK, Palgunachari MN, Fogelman AM. Oral Small Peptides Render HDL Antiinflammatory in Mice and Monkeys and Reduce Atherosclerosis in ApoE Null Mice. Circ Res 2005; 97:524-32. [PMID: 16100046 DOI: 10.1161/01.res.0000181229.69508.2f] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A peptide containing only 4 amino acid residues (KRES) that is too small to form an amphipathic helix, reduced lipoprotein lipid hydroperoxides (LOOH), increased paraoxonase activity, increased plasma HDL-cholesterol levels, rendered HDL antiinflammatory, and reduced atherosclerosis in apoE null mice. KRES was orally effective when synthesized from either L or D-amino acids suggesting that peptide-protein interactions were not required. Remarkably, changing the order of 2 amino acids (from KRES to KERS) resulted in the loss of all biologic activity. Solubility in ethyl acetate and interaction with lipids, as determined by differential scanning calorimetry, indicated significant differences between KRES and KERS. Negative stain electron microscopy showed that KRES formed organized peptide-lipid structures whereas KERS did not. Another tetrapeptide FREL shared many of the physical-chemical properties of KRES and was biologically active in mice and monkeys when synthesized from either L- or D-amino acids. After oral administration KRES and FREL were found associated with HDL whereas KERS was not. We conclude that the ability of peptides to interact with lipids, remove LOOH and activate antioxidant enzymes associated with HDL determines their antiinflammatory and antiatherogenic properties regardless of their ability to form amphipathic helixes.
Collapse
Affiliation(s)
- Mohamad Navab
- David Geffen School of Medicine, UCLA, Los Angeles, CA 90095-1679, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Daugherty A, Webb NR, Rateri DL, King VL. Thematic review series: The Immune System and Atherogenesis. Cytokine regulation of macrophage functions in atherogenesis. J Lipid Res 2005; 46:1812-22. [PMID: 15995168 DOI: 10.1194/jlr.r500009-jlr200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This review will focus on the role of cytokines in the behavior of macrophages, a prominent cell type of atherosclerotic lesions. Once these macrophages have immigrated into the vessel wall, they propagate the development of atherosclerosis by modifying lipoproteins, accumulating intracellular lipids, remodeling the extracellular environment, and promoting local coagulation. The numerous cytokines that have been detected in atherosclerosis, combined with the expression of large numbers of cytokine receptors on macrophages, are consistent with this axis being an important contributor to lesion development. Given the vast literature on cytokine-macrophage interactions, this review will be selective, with an emphasis on the major cytokines that have been detected in atherosclerotic lesions and their effects on properties that are relevant to lesion formation and maturation. There will be an emphasis on the role of cytokines in regulating lipid metabolism by macrophages. We will provide an overview of the major findings in cell culture and then put these in the context of in vivo studies.
Collapse
Affiliation(s)
- Alan Daugherty
- Cardiovascular Research Center, Gill Heart Institute, University of Kentucky, Lexington, KY, USA.
| | | | | | | |
Collapse
|
44
|
Shibeshi SS, Everett J, Venable DD, Collins WE. Simulated Blood Transport of Low Density Lipoproteins in a Three-Dimensional and Permeable T-Junction. ASAIO J 2005; 51:269-74. [PMID: 15968958 DOI: 10.1097/01.mat.0000160579.11018.ae] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Previous studies indicate that blood flow and transport of macromolecules in the cardiovascular system and tissues are essential to understand the genesis and progression of arterial diseases and for the effective implementation of arterial grafts, as well as to devise efficient drug delivery mechanisms. In the present study, we use computational fluid dynamics to simulate the blood flow and transport of low-density lipoproteins (LDL) in a three-dimensional and permeable T junction. The Navier-Stokes equation, Darcy's Law, and the advective diffusion equations are the mathematical models used to simulate the flow and transport phenomena of the system. In the numeric model to implement the finite volume method, we used the computational fluid dynamics software Fluent 6.1. The simulation shows higher LDL concentration in the luminal surface at the junction under physiologic flow conditions. At 1 mm depth into the artery from the luminal surface, the LDL concentration is approximately 40% of the lumenal concentration, and at 2 mm depth, it reduces to 20%. Ultimately, the concentration drops further and reaches zero at the outer wall boundary.
Collapse
|
45
|
Abstract
This review focuses on the role of oxidative processes in atherosclerosis and its resultant cardiovascular events. There is now a consensus that atherosclerosis represents a state of heightened oxidative stress characterized by lipid and protein oxidation in the vascular wall. The oxidative modification hypothesis of atherosclerosis predicts that low-density lipoprotein (LDL) oxidation is an early event in atherosclerosis and that oxidized LDL contributes to atherogenesis. In support of this hypothesis, oxidized LDL can support foam cell formation in vitro, the lipid in human lesions is substantially oxidized, there is evidence for the presence of oxidized LDL in vivo, oxidized LDL has a number of potentially proatherogenic activities, and several structurally unrelated antioxidants inhibit atherosclerosis in animals. An emerging consensus also underscores the importance in vascular disease of oxidative events in addition to LDL oxidation. These include the production of reactive oxygen and nitrogen species by vascular cells, as well as oxidative modifications contributing to important clinical manifestations of coronary artery disease such as endothelial dysfunction and plaque disruption. Despite these abundant data however, fundamental problems remain with implicating oxidative modification as a (requisite) pathophysiologically important cause for atherosclerosis. These include the poor performance of antioxidant strategies in limiting either atherosclerosis or cardiovascular events from atherosclerosis, and observations in animals that suggest dissociation between atherosclerosis and lipoprotein oxidation. Indeed, it remains to be established that oxidative events are a cause rather than an injurious response to atherogenesis. In this context, inflammation needs to be considered as a primary process of atherosclerosis, and oxidative stress as a secondary event. To address this issue, we have proposed an "oxidative response to inflammation" model as a means of reconciling the response-to-injury and oxidative modification hypotheses of atherosclerosis.
Collapse
Affiliation(s)
- Roland Stocker
- Centre for Vascular Research, University of New South Wales, Sydney, New South Wales, Australia.
| | | |
Collapse
|
46
|
Zhao B, Huang W, Zhang WY, Ishii I, Kruth HS. Retention of aggregated LDL by cultured human coronary artery endothelial cells. Biochem Biophys Res Commun 2004; 321:728-35. [PMID: 15358167 DOI: 10.1016/j.bbrc.2004.07.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2004] [Indexed: 10/26/2022]
Abstract
Aggregated LDL (AgLDL) accumulates within the subendothelial space of developing atherosclerotic lesions. We were interested to learn whether endothelial cells can interact with AgLDL. Incubation of endothelial cells with AgLDL resulted in apparent cholesterol retention. Microscopic examination revealed that cholesterol retention resulted mainly from endothelial cell surface attachment of AgLDL. Little AgLDL entered endothelial cells consistent with the small amount of endothelial cell degradation of AgLDL. Although endothelial cell retention of AgLDL was inhibited by LDL, AgLDL retention was not blocked by lactoferrin, C7 anti-LDL receptor monoclonal antibody, or receptor-associated protein, suggesting that LDL receptor family members did not mediate this retention. Surface retention of AgLDL depended on microtubule function and could be regulated by the protein kinase C activator, PMA. Treatment of endothelial cells with PMA either before or during, but not after incubation with AgLDL, inhibited retention of AgLDL. Our findings show that endothelial cells can retain AgLDL but internalize and metabolize little of this AgLDL. Thus, it is unlikely that endothelial cells can transport AgLDL out of atherosclerotic lesions, but it is likely that retention of AgLDL affects endothelial function.
Collapse
Affiliation(s)
- Bin Zhao
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
47
|
Navab M, Anantharamaiah GM, Reddy ST, Hama S, Hough G, Grijalva VR, Wagner AC, Frank JS, Datta G, Garber D, Fogelman AM. Oral D-4F causes formation of pre-beta high-density lipoprotein and improves high-density lipoprotein-mediated cholesterol efflux and reverse cholesterol transport from macrophages in apolipoprotein E-null mice. Circulation 2004; 109:3215-20. [PMID: 15197147 DOI: 10.1161/01.cir.0000134275.90823.87] [Citation(s) in RCA: 276] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND These studies were designed to determine the mechanism of action of an oral apolipoprotein (apo) A-I mimetic peptide, D-4F, which previously was shown to dramatically reduce atherosclerosis in mice. METHODS AND RESULTS Twenty minutes after 500 microg of D-4F was given orally to apoE-null mice, small cholesterol-containing particles (CCPs) of 7 to 8 nm with pre-beta mobility and enriched in apoA-I and paraoxonase activity were found in plasma. Before D-4F, both mature HDL and the fast protein liquid chromatography fractions containing the CCPs were proinflammatory. Twenty minutes after oral D-4F, HDL and CCPs became antiinflammatory, and there was an increase in HDL-mediated cholesterol efflux from macrophages in vitro. Oral D-4F also promoted reverse cholesterol transport from intraperitoneally injected cholesterol-loaded macrophages in vivo. In addition, oral D-4F significantly reduced lipoprotein lipid hydroperoxides (LOOH), except for pre-beta HDL fractions, in which LOOH increased. CONCLUSIONS The mechanism of action of oral D-4F in apoE-null mice involves rapid formation of CCPs, with pre-beta mobility enriched in apoA-I and paraoxonase activity. As a result, lipoprotein LOOH are reduced, HDL becomes antiinflammatory, and HDL-mediated cholesterol efflux and reverse cholesterol transport from macrophages are stimulated.
Collapse
MESH Headings
- Administration, Oral
- Amino Acid Sequence
- Animals
- Apolipoprotein A-I/pharmacology
- Apolipoprotein A-I/therapeutic use
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Arteriosclerosis/blood
- Arteriosclerosis/genetics
- Aryldialkylphosphatase/blood
- Biological Transport/drug effects
- Cells, Cultured
- Chemotaxis/drug effects
- Cholesterol/metabolism
- Coculture Techniques
- Drug Evaluation, Preclinical
- Female
- High-Density Lipoproteins, Pre-beta
- Humans
- Hyperlipoproteinemia Type II/blood
- Hyperlipoproteinemia Type II/genetics
- Inflammation
- Lipid Peroxidation/drug effects
- Lipoproteins, HDL/biosynthesis
- Lipoproteins, HDL/blood
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Sequence Data
- Monocytes/cytology
- Monocytes/drug effects
- Monocytes/metabolism
Collapse
Affiliation(s)
- Mohamad Navab
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA 90095-1679, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Brown MD, Jin L, Jien ML, Matsumoto AH, Helm GA, Lusis AJ, Frank JS, Shi W. Lipid retention in the arterial wall of two mouse strains with different atherosclerosis susceptibility. J Lipid Res 2004; 45:1155-61. [PMID: 15105414 DOI: 10.1194/jlr.m400092-jlr200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
LDL deposition in the subendothelium of arterial walls is the initial event in the development of atherosclerosis. The deposited LDL undergoes oxidative modification by arterial wall cells to become oxidized LDL and consequently contributes to atherosclerotic formation. Using mouse strains C57BL/6J (B6) and C3H/HeJ (C3H), which differ markedly in susceptibility to atherosclerosis, we determined whether variation in subendothelial retention of apolipoprotein B (apoB)-containing lipoproteins constitutes a genetic component in atherosclerosis. Lipoprotein retention was quantitated by Western blot analysis to detect the presence of apoB in aortic walls before foam cells developed. In both dietary and apoE-deficient models, B6 mice exhibited up to a 2-fold increase of apoB in the aortic wall compared with C3H mice. This increase could not be attributed to differences in plasma lipid levels of the two strains. In vitro, endothelial cells from C3H mice took up more acetylated and oxidized LDL but not native LDL and converted more native LDL to oxidized LDL than did endothelial cells from B6 mice. C3H mice expressed more scavenger receptor A in their aortic wall than B6 mice. Thus, variation in the subendothelial retention of apoB-containing lipoproteins cannot explain the dramatic difference in atherosclerosis susceptibility between B6 and C3H mice, and endothelial cells may play a role in alleviating lipid accumulation in arterial walls.
Collapse
Affiliation(s)
- Morry D Brown
- Department of Radiology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Raiszadeh F, Solati M, Etemadi A, Azizi F. Serum paraoxonase activity before and after treatment of thyrotoxicosis. Clin Endocrinol (Oxf) 2004; 60:75-80. [PMID: 14678291 DOI: 10.1111/j.1365-2265.2004.01940.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Antioxidant effects of paraoxonase, a high density lipoprotein (HDL)-associated enzyme that inhibits low density lipoprotein cholesterol (LDL-C) oxidation in human serum, have been reported. Patients with thyroid dysfunction are more susceptible to oxidative stress, and may show enhanced LDL-C oxidation. The purpose of this study was to evaluate serum paraoxonase activity in patients with hyperthyroidism before and after treatment with methimazole (MMI). DESIGN AND PATIENTS Twenty-four hyperthyroid patients (15 women and nine men, aged 43.0 +/- 12.9 years) and 23 age- and sex-matched healthy controls were studied. Serum paraoxonase activity, lipid, lipoprotein and apolipoprotein levels were measured in fasting samples. Patients were treated with MMI 20-30 mg daily for the first month, and 5-10 mg daily thereafter, and re-evaluated after 6-9 months of treatment. RESULTS Significantly lower serum paraoxonase activity was present in hyperthyroid patients before treatment compared with the controls (43.4 +/- 21.9 vs. 72.6 +/- 41.2 U/ml, P < 0.005). After a mean follow-up of 7.3 months, 15 patients became euthyroid (treated) and nine were still hyperthyroid. After follow-up, serum paraoxonase activity had increased to 62.2 +/- 37.4 U/ml in those who became euthyroid (P < 0.05 compared with baseline). In patients who were still hyperthyroid serum paraoxonase was unchanged from baseline, at 43.2 +/- 23.2 U/ml. CONCLUSION Serum paraoxonase is reduced in patients with hyperthyroidism and reverts to normal after euthyroidism is attained. Reduced serum paraoxonase activity in thyrotoxicosis might predispose lipids to oxidation.
Collapse
Affiliation(s)
- Farbod Raiszadeh
- Endocrine Research Centre, Shaheed Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | |
Collapse
|
50
|
Talbot RM, del Rio JD, Weinberg PD. Effect of fluid mechanical stresses and plasma constituents on aggregation of LDL. J Lipid Res 2003; 44:837-45. [PMID: 12562846 DOI: 10.1194/jlr.m200477-jlr200] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
LDL aggregates when exposed to even moderate fluid mechanical stresses in the laboratory, yet its half-life in the circulation is 2-3 days, implying that little aggregation occurs. LDL may be protected from aggregation in vivo by components of plasma, or by a qualitative difference in flows. Previous studies have shown that HDL and albumin inhibit the aggregation induced by vortexing. Using a more reproducible method of inducing aggregation and assessing aggregation both spectrophotometrically and by sedimentation techniques, we showed that at physiological concentrations, albumin is the more effective inhibitor, and that aggregation is substantially but not completely inhibited in plasma. Heat denatured and fatty-acid-stripped albumin were more effective inhibitors than normal albumin, supporting the idea that hydrophobic interactions are involved. Aggregation of LDL in a model reproducing several aspects of flow in the circulation was 200-fold slower, but was still inhibited by HDL and albumin, suggesting similar mechanisms are involved. Within the sensitivity of our technique, LDL aggregation did not occur in plasma exposed to these flows. Thus, as a result of the characteristics of blood flow and the inhibitory effects of plasma components, particularly albumin, LDL aggregation is unlikely to occur within the circulation.
Collapse
Affiliation(s)
- Roy M Talbot
- School of Animal and Microbial Sciences, University of Reading, United Kingdom
| | | | | |
Collapse
|