1
|
Prabutzki P, Schiller J, Engel KM. Phospholipid-derived lysophospholipids in (patho)physiology. Atherosclerosis 2024:118569. [PMID: 39227208 DOI: 10.1016/j.atherosclerosis.2024.118569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/17/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024]
Abstract
Phospholipids (PL) are major components of cellular membranes and changes in PL metabolism have been associated with the pathogenesis of numerous diseases. Lysophosphatidylcholine (LPC) in particular, is a comparably abundant component of oxidatively damaged tissues. LPC originates from the cleavage of phosphatidylcholine (PC) by phospholipase A2 or the reaction of lipids with reactive oxygen species (ROS) such as HOCl. Another explanation of increased LPC concentration is the decreased re-acylation of LPC into PC. While there are also several other lysophospholipids, LPC is the most abundant lysophospholipid in mammals and will therefore be the focus of this review. LPC is involved in many physiological processes. It induces the migration of lymphocytes, fostering the production of pro-inflammatory compounds by inducing oxidative stress. LPC also "signals" via G protein-coupled and Toll-like receptors and has been implicated in the development of different diseases. However, LPCs are not purely "bad": this is reflected by the fact that the concentration and fatty acyl composition of LPC varies under different conditions, in plasma of healthy and diseased individuals, in tissues and different tumors. Targeting LPC and lipid metabolism and restoring homeostasis might be a potential therapeutic method for inflammation-related diseases.
Collapse
Affiliation(s)
- Patricia Prabutzki
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Härtelstr. 16-18, D 04107 Leipzig, Germany
| | - Jürgen Schiller
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Härtelstr. 16-18, D 04107 Leipzig, Germany
| | - Kathrin M Engel
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Härtelstr. 16-18, D 04107 Leipzig, Germany.
| |
Collapse
|
2
|
Mutithu DW, Aremu OO, Mokaila D, Bana T, Familusi M, Taylor L, Martin LJ, Heathfield LJ, Kirwan JA, Wiesner L, Adeola HA, Lumngwena EN, Manganyi R, Skatulla S, Naidoo R, Ntusi NAB. A study protocol to characterise pathophysiological and molecular markers of rheumatic heart disease and degenerative aortic stenosis using multiparametric cardiovascular imaging and multiomics techniques. PLoS One 2024; 19:e0303496. [PMID: 38739622 PMCID: PMC11090351 DOI: 10.1371/journal.pone.0303496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/26/2024] [Indexed: 05/16/2024] Open
Abstract
INTRODUCTION Rheumatic heart disease (RHD), degenerative aortic stenosis (AS), and congenital valve diseases are prevalent in sub-Saharan Africa. Many knowledge gaps remain in understanding disease mechanisms, stratifying phenotypes, and prognostication. Therefore, we aimed to characterise patients through clinical profiling, imaging, histology, and molecular biomarkers to improve our understanding of the pathophysiology, diagnosis, and prognosis of RHD and AS. METHODS In this cross-sectional, case-controlled study, we plan to recruit RHD and AS patients and compare them to matched controls. Living participants will undergo clinical assessment, echocardiography, CMR and blood sampling for circulatory biomarker analyses. Tissue samples will be obtained from patients undergoing valve replacement, while healthy tissues will be obtained from cadavers. Immunohistology, proteomics, metabolomics, and transcriptome analyses will be used to analyse circulatory- and tissue-specific biomarkers. Univariate and multivariate statistical analyses will be used for hypothesis testing and identification of important biomarkers. In summary, this study aims to delineate the pathophysiology of RHD and degenerative AS using multiparametric CMR imaging. In addition to discover novel biomarkers and explore the pathomechanisms associated with RHD and AS through high-throughput profiling of the tissue and blood proteome and metabolome and provide a proof of concept of the suitability of using cadaveric tissues as controls for cardiovascular disease studies.
Collapse
Affiliation(s)
- Daniel W. Mutithu
- Department of Medicine, Cape Heart Institute, University of Cape Town, Cape Town, South Africa
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
- Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, South African Medical Research Council, Cape Town, South Africa
| | - Olukayode O. Aremu
- Department of Medicine, Cape Heart Institute, University of Cape Town, Cape Town, South Africa
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
- Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, South African Medical Research Council, Cape Town, South Africa
| | - Dipolelo Mokaila
- Department of Medicine, Cape Heart Institute, University of Cape Town, Cape Town, South Africa
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
- Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, South African Medical Research Council, Cape Town, South Africa
| | - Tasnim Bana
- Department of Medicine, Cape Heart Institute, University of Cape Town, Cape Town, South Africa
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
- Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, South African Medical Research Council, Cape Town, South Africa
| | - Mary Familusi
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
- Department of Civil Engineering, University of Cape Town, Cape Town, South Africa
| | - Laura Taylor
- Division of Forensic Medicine and Toxicology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Lorna J. Martin
- Division of Forensic Medicine and Toxicology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Laura J. Heathfield
- Division of Forensic Medicine and Toxicology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Jennifer A. Kirwan
- Metabolomics Platform, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center (MDC) for Molecular Medicine, Helmholtz Association, Berlin, Germany
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Henry A. Adeola
- Division of Dermatology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Evelyn N. Lumngwena
- Department of Medicine, Cape Heart Institute, University of Cape Town, Cape Town, South Africa
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
- Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, South African Medical Research Council, Cape Town, South Africa
- School of Clinical Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Rodgers Manganyi
- Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Sebastian Skatulla
- Department of Civil Engineering, University of Cape Town, Cape Town, South Africa
| | - Richard Naidoo
- Division of Anatomical Pathology, Department of Pathology, University of Cape Town and National Health Laboratory Service, Cape Town, South Africa
| | - Ntobeko A. B. Ntusi
- Department of Medicine, Cape Heart Institute, University of Cape Town, Cape Town, South Africa
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
- Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, South African Medical Research Council, Cape Town, South Africa
- Cape Universities Body Imaging Centre, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Disease Research, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
3
|
Yoon D, Choi B, Kim JE, Kim EY, Chung SH, Min HJ, Sung Y, Chang EJ, Song JK. Autotaxin inhibition attenuates the aortic valve calcification by suppressing inflammation-driven fibro-calcific remodeling of valvular interstitial cells. BMC Med 2024; 22:122. [PMID: 38486246 PMCID: PMC10941471 DOI: 10.1186/s12916-024-03342-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Patients with fibro-calcific aortic valve disease (FCAVD) have lipid depositions in their aortic valve that engender a proinflammatory impetus toward fibrosis and calcification and ultimately valve leaflet stenosis. Although the lipoprotein(a)-autotaxin (ATX)-lysophosphatidic acid axis has been suggested as a potential therapeutic target to prevent the development of FCAVD, supportive evidence using ATX inhibitors is lacking. We here evaluated the therapeutic potency of an ATX inhibitor to attenuate valvular calcification in the FCAVD animal models. METHODS ATX level and activity in healthy participants and patients with FCAVD were analyzed using a bioinformatics approach using the Gene Expression Omnibus datasets, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, and western blotting. To evaluate the efficacy of ATX inhibitor, interleukin-1 receptor antagonist-deficient (Il1rn-/-) mice and cholesterol-enriched diet-induced rabbits were used as the FCAVD models, and primary human valvular interstitial cells (VICs) from patients with calcification were employed. RESULTS The global gene expression profiles of the aortic valve tissue of patients with severe FCAVD demonstrated that ATX gene expression was significantly upregulated and correlated with lipid retention (r = 0.96) or fibro-calcific remodeling-related genes (r = 0.77) in comparison to age-matched non-FCAVD controls. Orally available ATX inhibitor, BBT-877, markedly ameliorated the osteogenic differentiation and further mineralization of primary human VICs in vitro. Additionally, ATX inhibition significantly attenuated fibrosis-related factors' production, with a detectable reduction of osteogenesis-related factors, in human VICs. Mechanistically, ATX inhibitor prohibited fibrotic changes in human VICs via both canonical and non-canonical TGF-β signaling, and subsequent induction of CTGF, a key factor in tissue fibrosis. In the in vivo FCAVD model system, ATX inhibitor exposure markedly reduced calcific lesion formation in interleukin-1 receptor antagonist-deficient mice (Il1rn-/-, P = 0.0210). This inhibition ameliorated the rate of change in the aortic valve area (P = 0.0287) and mean pressure gradient (P = 0.0249) in the FCAVD rabbit model. Moreover, transaortic maximal velocity (Vmax) was diminished with ATX inhibitor administration (mean Vmax = 1.082) compared to vehicle control (mean Vmax = 1.508, P = 0.0221). Importantly, ATX inhibitor administration suppressed the effects of a high-cholesterol diet and vitamin D2-driven fibrosis, in association with a reduction in macrophage infiltration and calcific deposition, in the aortic valves of this rabbit model. CONCLUSIONS ATX inhibition attenuates the development of FCAVD while protecting against fibrosis and calcification in VICs, suggesting the potential of using ATX inhibitors to treat FCAVD.
Collapse
Affiliation(s)
- Dohee Yoon
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Bongkun Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Ji-Eun Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Eun-Young Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Soo-Hyun Chung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hyo-Jin Min
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Yoolim Sung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Jae-Kwan Song
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
4
|
Abstract
Prolonged or excessive exposure to oxidized phospholipids (OxPLs) generates chronic inflammation. OxPLs are present in atherosclerotic lesions and can be detected in plasma on apolipoprotein B (apoB)-containing lipoproteins. When initially conceptualized, OxPL-apoB measurement in plasma was expected to reflect the concentration of minimally oxidized LDL, but, surprisingly, it correlated more strongly with plasma lipoprotein(a) (Lp(a)) levels. Indeed, experimental and clinical studies show that Lp(a) particles carry the largest fraction of OxPLs among apoB-containing lipoproteins. Plasma OxPL-apoB levels provide diagnostic information on the presence and extent of atherosclerosis and improve the prognostication of peripheral artery disease and first and recurrent myocardial infarction and stroke. The addition of OxPL-apoB measurements to traditional cardiovascular risk factors improves risk reclassification, particularly in patients in intermediate risk categories, for whom improving decision-making is most impactful. Moreover, plasma OxPL-apoB levels predict cardiovascular events with similar or greater accuracy than plasma Lp(a) levels, probably because this measurement reflects both the genetics of elevated Lp(a) levels and the generalized or localized oxidation that modifies apoB-containing lipoproteins and leads to inflammation. Plasma OxPL-apoB levels are reduced by Lp(a)-lowering therapy with antisense oligonucleotides and by lipoprotein apheresis, niacin therapy and bariatric surgery. In this Review, we discuss the role of role OxPLs in the pathophysiology of atherosclerosis and Lp(a) atherogenicity, and the use of OxPL-apoB measurement for improving prognosis, risk reclassification and therapeutic interventions.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Joseph L Witztum
- Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
5
|
Dzobo KE, Cupido AJ, Mol BM, Stiekema LC, Versloot M, Winkelmeijer M, Peter J, Pennekamp AM, Havik SR, Vaz FM, van Weeghel M, Prange KH, Levels JH, de Winther MP, Tsimikas S, Groen AK, Stroes ES, de Kleijn DP, Kroon J. Diacylglycerols and Lysophosphatidic Acid, Enriched on Lipoprotein(a), Contribute to Monocyte Inflammation. Arterioscler Thromb Vasc Biol 2024; 44:720-740. [PMID: 38269588 PMCID: PMC10880937 DOI: 10.1161/atvbaha.123.319937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Oxidized phospholipids play a key role in the atherogenic potential of lipoprotein(a) (Lp[a]); however, Lp(a) is a complex particle that warrants research into additional proinflammatory mediators. We hypothesized that additional Lp(a)-associated lipids contribute to the atherogenicity of Lp(a). METHODS Untargeted lipidomics was performed on plasma and isolated lipoprotein fractions. The atherogenicity of the observed Lp(a)-associated lipids was tested ex vivo in primary human monocytes by RNA sequencing, ELISA, Western blot, and transendothelial migratory assays. Using immunofluorescence staining and single-cell RNA sequencing, the phenotype of macrophages was investigated in human atherosclerotic lesions. RESULTS Compared with healthy individuals with low/normal Lp(a) levels (median, 7 mg/dL [18 nmol/L]; n=13), individuals with elevated Lp(a) levels (median, 87 mg/dL [218 nmol/L]; n=12) demonstrated an increase in lipid species, particularly diacylglycerols (DGs) and lysophosphatidic acid (LPA). DG and the LPA precursor lysophosphatidylcholine were enriched in the Lp(a) fraction. Ex vivo stimulation with DG(40:6) demonstrated a significant upregulation in proinflammatory pathways related to leukocyte migration, chemotaxis, NF-κB (nuclear factor kappa B) signaling, and cytokine production. Functional assessment showed a dose-dependent increase in the secretion of IL (interleukin)-6, IL-8, and IL-1β after DG(40:6) and DG(38:4) stimulation, which was, in part, mediated via the NLRP3 (NOD [nucleotide-binding oligomerization domain]-like receptor family pyrin domain containing 3) inflammasome. Conversely, LPA-stimulated monocytes did not exhibit an inflammatory phenotype. Furthermore, activation of monocytes by DGs and LPA increased their transendothelial migratory capacity. Human atherosclerotic plaques from patients with high Lp(a) levels demonstrated colocalization of Lp(a) with M1 macrophages, and an enrichment of CD68+IL-18+TLR4+ (toll-like receptor) TREM2+ (triggering receptor expressed on myeloid cells) resident macrophages and CD68+CASP1+ (caspase) IL-1B+SELL+ (selectin L) inflammatory macrophages compared with patients with low Lp(a). Finally, potent Lp(a)-lowering treatment (pelacarsen) resulted in a reduction in specific circulating DG lipid subspecies in patients with cardiovascular disease with elevated Lp(a) levels (median, 82 mg/dL [205 nmol/L]). CONCLUSIONS Lp(a)-associated DGs and LPA have a potential role in Lp(a)-induced monocyte inflammation by increasing cytokine secretion and monocyte transendothelial migration. This DG-induced inflammation is, in part, NLRP3 inflammasome dependent.
Collapse
Affiliation(s)
- Kim E. Dzobo
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, the Netherlands (K.E.D., M.V., J.K.)
| | - Arjen J. Cupido
- Vascular Medicine (A.J.C., L.C.A.S., E.S.G.S.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Barend M. Mol
- Department of Vascular Surgery, University Medical Centre Utrecht, the Netherlands (B.M.M., D.P.V.d.K.)
| | - Lotte C.A. Stiekema
- Vascular Medicine (A.J.C., L.C.A.S., E.S.G.S.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Miranda Versloot
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, the Netherlands (K.E.D., M.V., J.K.)
| | - Maaike Winkelmeijer
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Jorge Peter
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Anne-Marije Pennekamp
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Stefan R. Havik
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Frédéric M. Vaz
- Core Facility Metabolomics (F.M.V., M.v.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Michel van Weeghel
- Core Facility Metabolomics (F.M.V., M.v.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Koen H.M. Prange
- Department of Medical Biochemistry, Amsterdam Infection and Immunity (K.H.M.P., M.P.J.d.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Johannes H.M. Levels
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Amsterdam Infection and Immunity (K.H.M.P., M.P.J.d.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla (S.T.)
| | - Albert K. Groen
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Erik S.G. Stroes
- Vascular Medicine (A.J.C., L.C.A.S., E.S.G.S.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Dominique P.V. de Kleijn
- Department of Vascular Surgery, University Medical Centre Utrecht, the Netherlands (B.M.M., D.P.V.d.K.)
| | - Jeffrey Kroon
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, the Netherlands (K.E.D., M.V., J.K.)
- Laboratory of Angiogenesis and Vascular Metabolism, Flanders Institute for Biotechnology (VIB)-KU Leuven Center for Cancer Biology, VIB, Belgium (J.K.)
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven and Leuven Cancer Institute, Belgium (J.K.)
| |
Collapse
|
6
|
Notenboom ML, Van Hoof L, Schuermans A, Takkenberg JJM, Rega FR, Taverne YJHJ. Aortic Valve Embryology, Mechanobiology, and Second Messenger Pathways: Implications for Clinical Practice. J Cardiovasc Dev Dis 2024; 11:49. [PMID: 38392263 PMCID: PMC10888685 DOI: 10.3390/jcdd11020049] [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: 12/24/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
During the Renaissance, Leonardo Da Vinci was the first person to successfully detail the anatomy of the aortic root and its adjacent structures. Ever since, novel insights into morphology, function, and their interplay have accumulated, resulting in advanced knowledge on the complex functional characteristics of the aortic valve (AV) and root. This has shifted our vision from the AV as being a static structure towards that of a dynamic interconnected apparatus within the aortic root as a functional unit, exhibiting a complex interplay with adjacent structures via both humoral and mechanical stimuli. This paradigm shift has stimulated surgical treatment strategies of valvular disease that seek to recapitulate healthy AV function, whereby AV disease can no longer be seen as an isolated morphological pathology which needs to be replaced. As prostheses still cannot reproduce the complexity of human nature, treatment of diseased AVs, whether stenotic or insufficient, has tremendously evolved, with a similar shift towards treatments options that are more hemodynamically centered, such as the Ross procedure and valve-conserving surgery. Native AV and root components allow for an efficient Venturi effect over the valve to allow for optimal opening during the cardiac cycle, while also alleviating the left ventricle. Next to that, several receptors are present on native AV leaflets, enabling messenger pathways based on their interaction with blood and other shear-stress-related stimuli. Many of these physiological and hemodynamical processes are under-acknowledged but may hold important clues for innovative treatment strategies, or as potential novel targets for therapeutic agents that halt or reverse the process of valve degeneration. A structured overview of these pathways and their implications for cardiothoracic surgeons and cardiologists is lacking. As such, we provide an overview on embryology, hemodynamics, and messenger pathways of the healthy and diseased AV and its implications for clinical practice, by relating this knowledge to current treatment alternatives and clinical decision making.
Collapse
Affiliation(s)
- Maximiliaan L. Notenboom
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| | - Lucas Van Hoof
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Art Schuermans
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Johanna J. M. Takkenberg
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| | - Filip R. Rega
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Yannick J. H. J. Taverne
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| |
Collapse
|
7
|
Chen J, Lyu L, Shen J, Pan Y, Jing J, Wang YJ, Wei T. Epidemiological study of calcified aortic valve stenosis in a Chinese community population. Postgrad Med J 2023; 99:868-874. [PMID: 37117153 DOI: 10.1136/pmj-2022-141721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/06/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND AIMS Due to the ageing global population, calcified aortic valve disease is currently the most common cardiac valve disorder. This study aimed to investigate the prevalence and the risk factors for calcified aortic valve stenosis (CAVS), and develop a prediction model for predicting CAVS risk. METHODS AND RESULTS This study was derived from the cross-sectional baseline survey of the PRECISE study (NCT03178448). The demographic, clinical and laboratory information of each participant was obtained. Univariable and multivariable logistic regression models were used to determine CAVS risk factors. A prediction model for predicting CAVS risk based on risk factors was developed and the result was performed by nomogram. The discrimination of the prediction model was assessed by receiver operating characteristic curve analysis. The degree of fitting for the prediction model was assessed by calibration curve analysis. A total of 3067 participants (1427 men and 1640 women) were included. The prevalence of CAVS among those aged below 60 years old, 60-70 years old and over 70 years old was 4.1%, 10.3% and 21.9%, respectively. Multivariable regression analysis revealed that age (OR: 1.099; 95% CI: 1.076 to 1.123, p<0.001), pulse pressure (OR: 1.020; 95% CI: 1.009 to 1.031, p<0.001), uric acid (OR: 1.003; 95% CI: 1.001 to 1.004, p<0.001), glycosylated haemoglobin (HbA1c) (OR: 1.152; 95% CI: 1.028 to 1.292, p=0.015) and lipoprotein(a) (OR: 1.002; 95% CI: 1.001 to 1.002, p<0.001) were independent risk factors for CAVS. High-density lipoprotein cholesterol (HDL-C) was a protective factor for CAVS (OR: 0.539; 95% CI: 0.349 to 0.831, p=0.005). The prediction model including the above risk factors showed a risk prediction of CAVS with good discrimination. The area under the curve value was found to be 0.743 (95% CI: 0.711 to 0.775). CONCLUSION CAVS is currently prevalent in the elderly Chinese population. Age, pulse pressure, HbA1c, lower-level HDL-C, lipoprotein(a) and uric acid are the independent risk factors for CAVS.
Collapse
Affiliation(s)
- Jun Chen
- Department of Cardiology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, Zhejiang, China
- Department of Cardiology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lingchun Lyu
- Department of Cardiology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, Zhejiang, China
| | - Jiayi Shen
- Department of Cardiology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, Zhejiang, China
| | - Yuesong Pan
- Department of Neurology, Beijing Tiantan Hospital, Beijing, China
| | - Jing Jing
- Department of Neurology, Beijing Tiantan Hospital, Beijing, China
| | - Yong-Jun Wang
- Department of Neurology, Beijing Tiantan Hospital, Beijing, China
| | - Tiemin Wei
- Department of Cardiology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, Zhejiang, China
| |
Collapse
|
8
|
Krzesińska A, Nowak M, Mickiewicz A, Chyła-Danił G, Ćwiklińska A, Koper-Lenkiewicz OM, Kamińska J, Matowicka-Karna J, Gruchała M, Jankowski M, Fijałkowski M, Kuchta A. Lipoprotein(a) As a Potential Predictive Factor for Earlier Aortic Valve Replacement in Patients with Bicuspid Aortic Valve. Biomedicines 2023; 11:1823. [PMID: 37509461 PMCID: PMC10376971 DOI: 10.3390/biomedicines11071823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/16/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Bicuspid aortic valve (BAV) affects 0.5-2% of the general population and constitutes the major cause of severe aortic valve stenosis (AVS) in individuals ≤70 years. The aim of the present study was to evaluate the parameters that may provide information about the risk of AVS developing in BAV patients, with particular emphasis on lipoprotein(a) (Lp(a)), which is a well-recognized risk factor for stenosis in the general population. We also analyzed the impact of autotaxin (ATX) and interleukin-6 (IL-6) as parameters potentially related to the pathomechanism of Lp(a) action. We found that high Lp(a) levels (>50 mg/dL) occurred significantly more frequently in patients with AVS than in patients without AVS, both in the group below and above 45 years of age (p = 0.036 and p = 0.033, respectively). Elevated Lp(a) levels were also strictly associated with the need for aortic valve replacement (AVR) at a younger age (p = 0.016). However, the Lp(a) concentration did not differ significantly between patients with and without AVS. Similarly, we observed no differences in ATX between the analyzed patient groups, and both ATX activity and concentration correlated significantly with Lp(a) level (R = 0.465, p < 0.001 and R = 0.599, p < 0.001, respectively). We revealed a significantly higher concentration of IL-6 in young patients with AVS. However, this observation was not confirmed in the group of patients over 45 years of age. We also did not observe a significant correlation between IL-6 and Lp(a) or between CRP and Lp(a) in any of the analyzed groups of BAV patients. Our results demonstrate that a high level of Lp(a), greater than 50 mg/dL, may be a significant predictive factor for earlier AVR. Lp(a)-related parameters, such as ATX and IL-6, may be valuable in providing information about the additional cardiovascular risks associated with developing AVS.
Collapse
Affiliation(s)
- Aleksandra Krzesińska
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Maria Nowak
- 1st Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Agnieszka Mickiewicz
- 1st Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Gabriela Chyła-Danił
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Agnieszka Ćwiklińska
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Olga M Koper-Lenkiewicz
- Department of Clinical Laboratory Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland
| | - Joanna Kamińska
- Department of Clinical Laboratory Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland
| | - Joanna Matowicka-Karna
- Department of Clinical Laboratory Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland
| | - Marcin Gruchała
- 1st Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Maciej Jankowski
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Marcin Fijałkowski
- 1st Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Agnieszka Kuchta
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| |
Collapse
|
9
|
Farina JM, Pereyra M, Mahmoud AK, Chao CJ, Barry T, Halli Demeter SM, Ayoub C, Arsanjani R. Current Management and Future Perspectives in the Treatment of Lp(a) with a Focus on the Prevention of Cardiovascular Diseases. Pharmaceuticals (Basel) 2023; 16:919. [PMID: 37513831 PMCID: PMC10385436 DOI: 10.3390/ph16070919] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Lipoprotein(a) [Lp(a)] is a lipid molecule with atherogenic, inflammatory, thrombotic, and antifibrinolytic effects, whose concentrations are predominantly genetically determined. The association between Lp(a) and cardiovascular diseases (CVDs) has been well-established in numerous studies, and the ability to measure Lp(a) levels is widely available in the community. As such, there has been increasing interest in Lp(a) as a therapeutic target for the prevention of CVD. The impact of the currently available lipid-modifying agents on Lp(a) is modest and heterogeneous, except for the monoclonal antibody proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), which demonstrated a significant reduction in Lp(a) levels. However, the absolute reduction in Lp(a) to significantly decrease CVD outcomes has not been definitely established, and the magnitude of the effect of PCSK9i seems insufficient to directly reduce the Lp(a)-related CVD risk. Therefore, emerging therapies are being developed that specifically aim to lower Lp(a) levels and the risk of CVD, including RNA interference (RNAi) agents, which have the capacity for temporary and reversible downregulation of gene expression. This review article aims to summarize the effects of Lp(a) on CVD and to evaluate the available evidence on established and emerging therapies targeting Lp(a) levels, focusing on the potential reduction of CVD risk attributable to Lp(a) concentrations.
Collapse
Affiliation(s)
- Juan M Farina
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Milagros Pereyra
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Ahmed K Mahmoud
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Chieh-Ju Chao
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Timothy Barry
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Susan M Halli Demeter
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| |
Collapse
|
10
|
Sud K, Narula N, Aikawa E, Arbustini E, Pibarot P, Merlini G, Rosenson RS, Seshan SV, Argulian E, Ahmadi A, Zhou F, Moreira AL, Côté N, Tsimikas S, Fuster V, Gandy S, Bonow RO, Gursky O, Narula J. The contribution of amyloid deposition in the aortic valve to calcification and aortic stenosis. Nat Rev Cardiol 2023; 20:418-428. [PMID: 36624274 PMCID: PMC10199673 DOI: 10.1038/s41569-022-00818-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/17/2022] [Indexed: 01/11/2023]
Abstract
Calcific aortic valve disease (CAVD) and stenosis have a complex pathogenesis, and no therapies are available that can halt or slow their progression. Several studies have shown the presence of apolipoprotein-related amyloid deposits in close proximity to calcified areas in diseased aortic valves. In this Perspective, we explore a possible relationship between amyloid deposits, calcification and the development of aortic valve stenosis. These amyloid deposits might contribute to the amplification of the inflammatory cycle in the aortic valve, including extracellular matrix remodelling and myofibroblast and osteoblast-like cell proliferation. Further investigation in this area is needed to characterize the amyloid deposits associated with CAVD, which could allow the use of antisense oligonucleotides and/or isotype gene therapies for the prevention and/or treatment of CAVD.
Collapse
Affiliation(s)
- Karan Sud
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Navneet Narula
- New York University Grossman School of Medicine, New York, NY, USA.
| | - Elena Aikawa
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Philippe Pibarot
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
| | | | | | | | - Edgar Argulian
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amir Ahmadi
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fang Zhou
- New York University Grossman School of Medicine, New York, NY, USA
| | - Andre L Moreira
- New York University Grossman School of Medicine, New York, NY, USA
| | - Nancy Côté
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
| | | | | | - Sam Gandy
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert O Bonow
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Olga Gursky
- Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Jagat Narula
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
11
|
de Mello Barros Pimentel MV, Bertolami A, Fernandes LP, Barroso LP, Castro IA. Could a lipid oxidative biomarker be applied to improve risk stratification in the prevention of cardiovascular disease? Biomed Pharmacother 2023; 160:114345. [PMID: 36753953 DOI: 10.1016/j.biopha.2023.114345] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/08/2023] Open
Abstract
There is significant evidence demonstrating the influence of oxidative stress on atherosclerosis and cardiovascular diseases (CVD). However, oxidative biomarkers have not been applied to follow patients under primary or secondary prevention. Many factors can explain this paradox: the higher complexity of the methods applied to quantify oxidative markers, the high variability observed among the studies, the lack of reference values, and the weak correlation with clinical endpoints. This review presents the role of the major reactive oxygen species (ROS) involved in cardiovascular pathophysiology and how they can be neutralized by endogenous and exogenous antioxidants based on classical and recent studies, highlighting the importance of the secondary products of fatty acid oxidation as potential biomarkers. Furthermore, we discuss the great variability of oxidative stress biomarkers, using as an example data obtained from 55 studies. Among the molecules directly formed from lipid oxidation, such as malondialdehyde (MDA), oxidized LDL (oxLDL), and isoprostanes (F2-IsoP), and those associated with general oxidative conditions (ferric-reducing antioxidant power (FRAP), superoxide dismutase (SOD), glutathione (GSH)), MDA was the most lipid biomarker evaluated in the treatments and proved to be an independent factor compared with traditional markers used in the algorithms to stratify the patient's risk. Finally, this review suggests four steps to follow, aiming to include MDA in the algorithms applied to estimate CVD risk.
Collapse
Affiliation(s)
| | - Adriana Bertolami
- Dyslipidemia Medical Section, Dante Pazzanese Institute of Cardiology, São Paulo, Brazil
| | - Lígia Prestes Fernandes
- LADAF, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Lúcia Pereira Barroso
- Department of Statistics, Institute of Mathematics and Statistics, University of São Paulo, São Paulo, Brazil
| | - Inar Alves Castro
- LADAF, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; Food Research Center (FoRC), CEPID-FAPESP, Research Innovation and Dissemination Centers São Paulo Research Foundation, São Paulo 05468-140, Brazil.
| |
Collapse
|
12
|
Neels JG, Leftheriotis G, Chinetti G. Atherosclerosis Calcification: Focus on Lipoproteins. Metabolites 2023; 13:metabo13030457. [PMID: 36984897 PMCID: PMC10056669 DOI: 10.3390/metabo13030457] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipids in the vessel wall, leading to the formation of an atheroma and eventually to the development of vascular calcification (VC). Lipoproteins play a central role in the development of atherosclerosis and VC. Both low- and very low-density lipoproteins (LDL and VLDL) and lipoprotein (a) (Lp(a)) stimulate, while high-density lipoproteins (HDL) reduce VC. Apolipoproteins, the protein component of lipoproteins, influence the development of VC in multiple ways. Apolipoprotein AI (apoAI), the main protein component of HDL, has anti-calcific properties, while apoB and apoCIII, the main protein components of LDL and VLDL, respectively, promote VC. The role of lipoproteins in VC is also related to their metabolism and modifications. Oxidized LDL (OxLDL) are more pro-calcific than native LDL. Oxidation also converts HDL from anti- to pro-calcific. Additionally, enzymes such as autotaxin (ATX) and proprotein convertase subtilisin/kexin type 9 (PCSK9), involved in lipoprotein metabolism, have a stimulatory role in VC. In summary, a better understanding of the mechanisms by which lipoproteins and apolipoproteins contribute to VC will be crucial in the development of effective preventive and therapeutic strategies for VC and its associated cardiovascular disease.
Collapse
Affiliation(s)
- Jaap G Neels
- Université Côte d'Azur, INSERM, C3M, 06200 Nice, France
| | | | - Giulia Chinetti
- Université Côte d'Azur, CHU, INSERM, C3M, 06200 Nice, France
| |
Collapse
|
13
|
Calcific aortic valve disease: mechanisms, prevention and treatment. Nat Rev Cardiol 2023:10.1038/s41569-023-00845-7. [PMID: 36829083 DOI: 10.1038/s41569-023-00845-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/01/2023] [Indexed: 02/26/2023]
Abstract
Calcific aortic valve disease (CAVD) is the most common disorder affecting heart valves and is characterized by thickening, fibrosis and mineralization of the aortic valve leaflets. Analyses of surgically explanted aortic valve leaflets have shown that dystrophic mineralization and osteogenic transition of valve interstitial cells co-occur with neovascularization, microhaemorrhage and abnormal production of extracellular matrix. Age and congenital bicuspid aortic valve morphology are important and unalterable risk factors for CAVD, whereas additional risk is conferred by elevated blood pressure and plasma lipoprotein(a) levels and the presence of obesity and diabetes mellitus, which are modifiable factors. Genetic and molecular studies have identified that the NOTCH, WNT-β-catenin and myocardin signalling pathways are involved in the control and commitment of valvular cells to a fibrocalcific lineage. Complex interactions between valve endothelial and interstitial cells and immune cells promote the remodelling of aortic valve leaflets and the development of CAVD. Although no medical therapy is effective for reducing or preventing the progression of CAVD, studies have started to identify actionable targets.
Collapse
|
14
|
Ruder S, Mansfield B, Immelman AR, Varki N, Miu P, Raal F, Tsimikas S. Lp(a), oxidized phospholipids and oxidation-specific epitopes are increased in subjects with keloid formation. Lipids Health Dis 2022; 21:113. [PMID: 36320028 PMCID: PMC9623907 DOI: 10.1186/s12944-022-01720-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Keloid formation following trauma or surgery is common among darkly pigmented individuals. Since lipoprotein(a) [Lp(a)] has been postulated to have a putative role in wound healing, and also mediates atherosclerotic cardiovascular disease, it was assessed whether Lp(a), its associated oxidized phospholipids and other oxidation-specific biomarkers were associated with keloid formation. METHODS This case-control study included darkly pigmented individuals of African ancestry, 100 with keloid scarring and 100 non-keloid controls. The lipid panel, hsCRP, Lp(a), oxidized phospholipids on apolipoprotein B-100 (OxPL-apoB), IgG and IgM apoB-immune complexes and IgG and IgM autoantibodies to a malondialdehyde mimotope (MDA-mimotope) were measured. Immunohistochemistry of keloid specimens was performed for both Lp(a) and OxPL staining. RESULTS Cases and controls were well matched for age, sex and lipid profile. Mean Lp(a) (57.8 vs. 44.2 mg/dL; P = 0.01, OxPL-apoB 17.4 vs. 15.7 nmol/L; P = 0.009) and IgG and IgM apoB-immune complexes and IgG and IgM MDA-mimotope levels were significantly higher in keloid cases. Keloid tissue stained strongly for OxPL. CONCLUSION Darkly pigmented individuals of African ancestry with keloids have higher plasma levels of Lp(a), OxPL-apoB and oxidation-specific epitopes. The commonality of excessive wound healing in keloids and chronic complications from coronary revascularization suggests avenues of investigation to define a common mechanism driven by Lp(a) and the innate response to oxidized lipids.
Collapse
Affiliation(s)
- Sundeep Ruder
- Carbohydrate & Lipid Metabolism Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Brett Mansfield
- Carbohydrate & Lipid Metabolism Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Andrew Ronald Immelman
- Carbohydrate & Lipid Metabolism Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nissi Varki
- Department of Pathology, University of California, San Diego, USA
| | - Phuong Miu
- Sulpizio Cardiovascular Center, Division of Cardiovascular Medicine, University of California, 9500 Gilman Drive, 92093- 0682 San Diego, USA
| | - Frederick Raal
- Carbohydrate & Lipid Metabolism Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sotirios Tsimikas
- Sulpizio Cardiovascular Center, Division of Cardiovascular Medicine, University of California, 9500 Gilman Drive, 92093- 0682 San Diego, USA
| |
Collapse
|
15
|
Fu B, Wang J, Wang L, Wang Q, Guo Z, Xu M, Jiang N. Integrated proteomic and metabolomic profile analyses of cardiac valves revealed molecular mechanisms and targets in calcific aortic valve disease. Front Cardiovasc Med 2022; 9:944521. [PMID: 36312243 PMCID: PMC9606238 DOI: 10.3389/fcvm.2022.944521] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 09/13/2022] [Indexed: 12/02/2022] Open
Abstract
Background This study aimed to define changes in the metabolic and protein profiles of patients with calcific aortic valve disease (CAVD). Methods and results We analyzed cardiac valve samples of patients with and without (control) CAVD (n = 24 per group) using untargeted metabolomics and tandem mass tag-based quantitative proteomics. Significantly different metabolites and proteins between the CAVD and control groups were screened; then, functional enrichment was analyzed. We analyzed co-expressed differential metabolites and proteins, and constructed a metabolite-protein-pathway network. The expression of key proteins was validated using western blotting. Differential analysis identified 229 metabolites in CAVD among which, 2-aminophenol, hydroxykynurenine, erythritol, carnosine, and choline were the top five. Proteomic analysis identified 549 differentially expressed proteins in CAVD, most of which were localized in the nuclear, cytoplasmic, extracellular, and plasma membranes. Levels of selenium binding protein 1 (SELENBP1) positively correlated with multiple metabolites. Adenosine triphosphate-binding cassette transporters, starch and sucrose metabolism, hypoxia-inducible factor 1 (HIF-1) signaling, and purine metabolism were key pathways in the network. Ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), calcium2+/calmodulin-dependent protein kinase II delta (CAMK2D), and ATP binding cassette subfamily a member 8 (ABCA8) were identified as hub proteins in the metabolite-protein-pathway network as they interacted with ADP, glucose 6-phosphate, choline, and other proteins. Western blotting confirmed that ENPP1 was upregulated, whereas ABCA8 and CAMK2D were downregulated in CAVD samples. Conclusion The metabolic and protein profiles of cardiac valves from patients with CAVD significantly changed. The present findings provide a holistic view of the molecular mechanisms underlying CAVD that may lead to the development of novel diagnostic biomarkers and therapeutic targets to treat CAVD.
Collapse
Affiliation(s)
- Bo Fu
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin, China,Postdoctoral Mobile Station, Tianjin Medical University, Tianjin, China
| | - Jing Wang
- Department of Pathology, Tianjin Chest Hospital, Tianjin, China
| | - Lianqun Wang
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin, China
| | - Qiang Wang
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin, China
| | - Zhigang Guo
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin, China,Zhigang Guo,
| | - Meilin Xu
- Department of Pathology, Tianjin Chest Hospital, Tianjin, China
| | - Nan Jiang
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin, China,*Correspondence: Nan Jiang,
| |
Collapse
|
16
|
Bhatia HS, Wilkinson MJ. Lipoprotein(a): Evidence for Role as a Causal Risk Factor in Cardiovascular Disease and Emerging Therapies. J Clin Med 2022; 11:6040. [PMID: 36294361 PMCID: PMC9604626 DOI: 10.3390/jcm11206040] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/20/2022] [Accepted: 10/07/2022] [Indexed: 08/03/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) is an established risk factor for multiple cardiovascular diseases. Several lines of evidence including mechanistic, epidemiologic, and genetic studies support the role of Lp(a) as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and aortic stenosis/calcific aortic valve disease (AS/CAVD). Limited therapies currently exist for the management of risk associated with elevated Lp(a), but several targeted therapies are currently in various stages of clinical development. In this review, we detail evidence supporting Lp(a) as a causal risk factor for ASCVD and AS/CAVD, and discuss approaches to managing Lp(a)-associated risk.
Collapse
|
17
|
Koschinsky ML, Boffa MB. Oxidized phospholipid modification of lipoprotein(a): Epidemiology, biochemistry and pathophysiology. Atherosclerosis 2022; 349:92-100. [DOI: 10.1016/j.atherosclerosis.2022.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/22/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023]
|
18
|
DeVito LM, Dennis EA, Kahn BB, Shulman GI, Witztum JL, Sadhu S, Nickels J, Spite M, Smyth S, Spiegel S. Bioactive lipids and metabolic syndrome-a symposium report. Ann N Y Acad Sci 2022; 1511:87-106. [PMID: 35218041 DOI: 10.1111/nyas.14752] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 11/27/2022]
Abstract
Recent research has shed light on the cellular and molecular functions of bioactive lipids that go far beyond what was known about their role as dietary lipids. Bioactive lipids regulate inflammation and its resolution as signaling molecules. Genetic studies have identified key factors that can increase the risk of cardiovascular diseases and metabolic syndrome through their effects on lipogenesis. Lipid scientists have explored how these signaling pathways affect lipid metabolism in the liver, adipose tissue, and macrophages by utilizing a variety of techniques in both humans and animal models, including novel lipidomics approaches and molecular dynamics models. Dissecting out these lipid pathways can help identify mechanisms that can be targeted to prevent or treat cardiometabolic conditions. Continued investigation of the multitude of functions mediated by bioactive lipids may reveal additional components of these pathways that can provide a greater understanding of metabolic homeostasis.
Collapse
Affiliation(s)
| | | | - Barbara B Kahn
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | | | | | | | - Joseph Nickels
- Genesis Biotechnology Group, Hamilton Township, New Jersey
| | - Matthew Spite
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Susan Smyth
- University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Sarah Spiegel
- Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
19
|
Hu J, Lei H, Liu L, Xu D. Lipoprotein(a), a Lethal Player in Calcific Aortic Valve Disease. Front Cell Dev Biol 2022; 10:812368. [PMID: 35155427 PMCID: PMC8830536 DOI: 10.3389/fcell.2022.812368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/07/2022] [Indexed: 12/11/2022] Open
Abstract
Calcified aortic valve disease (CAVD) is the most common valvular cardiovascular disease with increasing incidence and mortality. The primary treatment for CAVD is surgical or transcatheter aortic valve replacement and there remains a lack of effective drug treatment. Recently, lipoprotein (a) (Lp(a)) has been considered to play a crucial role in CAVD pathophysiology. Multiple studies have shown that Lp(a) represents an independent risk factor for CAVD. Moreover, Lp(a) mediates the occurrence and development of CAVD by affecting aortic valve endothelial dysfunction, indirectly promoting foam cell formation through oxidized phospholipids (OxPL), inflammation, oxidative stress, and directly promotes valve calcification. However, there is a lack of clinical trials with Lp(a) reduction as a primary endpoint. This review aims to explore the relationship and mechanism between Lp(a) and CAVD, and focuses on the current drugs that can be used as potential therapeutic targets for CAVD.
Collapse
Affiliation(s)
- Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Hao Lei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Leiling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
- *Correspondence: Danyan Xu,
| |
Collapse
|
20
|
Trends in testing and prevalence of elevated Lp(a) among patients with aortic valve stenosis. Atherosclerosis 2022; 349:144-150. [PMID: 35144769 PMCID: PMC9674369 DOI: 10.1016/j.atherosclerosis.2022.01.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/10/2022] [Accepted: 01/27/2022] [Indexed: 11/25/2022]
Abstract
Background and aims: Lipoprotein(a) [Lp(a)] is causally associated with aortic valve stenosis (AS) but Lp(a) testing among AS patients is not broadly incorporated into clinical practice. We evaluated trends in Lp(a) testing in an academic medical center. Methods: Educational efforts and adding Lp(a) to the lipid panel on the electronic medical record (EMR) and preprocedure order sets were used to increase awareness of Lp(a) as a risk factor in AS. Medical records at University of California San Diego Health (UCSDH) were analyzed from 2010 to 2020 to define the yearly frequency of first time Lp(a) testing in patients with diagnosis codes for AS or undergoing transcatheter aortic valve replacement (TAVR). Results: Lp(a) testing for any indication increased over 5-fold from 2010 to 2020. A total of 3808 patients had a diagnosis of AS and 417 patients had TAVR. Lp(a) levels >30 mg/dL were present in 37% of AS and 35% of TAVR patients. The rates of Lp(a) testing in AS and TAVR were 14.0% and 65.7%, respectively. In AS, Lp(a) testing increased over time from 8.5% in 2010, peaking at 24.2% in 2017, and declining to 13.9% in 2020 (p < 0.001 for trend). Following implementation of EMR order-sets in 2016, Lp(a) testing in TAVR cases increased to a peak of 88.5% in 2018. Conclusions: Elevated Lp(a) is prevalent in AS and TAVR patients. Implementation of educational efforts and practice pathways resulted in increased Lp(a) testing in patients with AS. This study represents a paradigm that may allow increased global awareness of Lp(a) as a risk factor for AS.
Collapse
|
21
|
Bourgeois R, Bourgault J, Despres AA, Perrot N, Guertin J, Girard A, Mitchell PL, Gotti C, Bourassa S, Scipione CA, Gaudreault N, Boffa MB, Koschinsky ML, Pibarot P, Droit A, Thériault S, Mathieu P, Bossé Y, Arsenault BJ. Lipoprotein Proteomics and Aortic Valve Transcriptomics Identify Biological Pathways Linking Lipoprotein(a) Levels to Aortic Stenosis. Metabolites 2021; 11:metabo11070459. [PMID: 34357353 PMCID: PMC8307014 DOI: 10.3390/metabo11070459] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/17/2022] Open
Abstract
Lipoprotein(a) (Lp(a)) is one of the most important risk factors for the development of calcific aortic valve stenosis (CAVS). However, the mechanisms through which Lp(a) causes CAVS are currently unknown. Our objectives were to characterize the Lp(a) proteome and to identify proteins that may be differentially associated with Lp(a) in patients with versus without CAVS. Our second objective was to identify genes that may be differentially regulated by exposure to high versus low Lp(a) levels in explanted aortic valves from patients with CAVS. We isolated Lp(a) from the blood of 21 patients with CAVS and 22 volunteers and performed untargeted label-free analysis of the Lp(a) proteome. We also investigated the transcriptomic signature of calcified aortic valves from patients who underwent aortic valve replacement with high versus low Lp(a) levels (n = 118). Proteins involved in the protein activation cascade, platelet degranulation, leukocyte migration, and response to wounding may be associated with Lp(a) depending on CAVS status. The transcriptomic analysis identified genes involved in cardiac aging, chondrocyte development, and inflammation as potentially influenced by Lp(a). Our multi-omic analyses identified biological pathways through which Lp(a) may cause CAVS, as well as key molecular events that could be triggered by Lp(a) in CAVS development.
Collapse
Affiliation(s)
- Raphaëlle Bourgeois
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Jérôme Bourgault
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Audrey-Anne Despres
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Nicolas Perrot
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Jakie Guertin
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Arnaud Girard
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Patricia L. Mitchell
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
| | - Clarisse Gotti
- Proteomics Platform of the CHU de Québec, QC G1V 4G2, Canada; (C.G.); (S.B.); (A.D.)
| | - Sylvie Bourassa
- Proteomics Platform of the CHU de Québec, QC G1V 4G2, Canada; (C.G.); (S.B.); (A.D.)
| | - Corey A. Scipione
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada;
| | - Nathalie Gaudreault
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
| | - Michael B. Boffa
- Robarts Research Institute, London, ON N6A 5B7, Canada; (M.B.B.); (M.L.K.)
| | | | - Philippe Pibarot
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Arnaud Droit
- Proteomics Platform of the CHU de Québec, QC G1V 4G2, Canada; (C.G.); (S.B.); (A.D.)
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada
| | - Sébastien Thériault
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Patrick Mathieu
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Yohan Bossé
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Benoit J. Arsenault
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (R.B.); (J.B.); (A.-A.D.); (N.P.); (J.G.); (A.G.); (P.L.M.); (N.G.); (P.P.); (S.T.); (P.M.); (Y.B.)
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: ; Tel.: +1-418-656-8711 (ext. 3498)
| |
Collapse
|
22
|
Afshar M, Yazdan-Ashoori S, Engert JC, Thanassoulis G. Drugs for Prevention and Treatment of Aortic Stenosis: How Close Are We? Can J Cardiol 2021; 37:1016-1026. [DOI: 10.1016/j.cjca.2021.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 12/25/2022] Open
|
23
|
Natorska J, Kopytek M, Undas A. Aortic valvular stenosis: Novel therapeutic strategies. Eur J Clin Invest 2021; 51:e13527. [PMID: 33621361 DOI: 10.1111/eci.13527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Aortic stenosis (AS) prevalence is estimated to reach 4.5 million cases worldwide by the year 2030. AS is a progressive disease without a pharmacological treatment. In the current review, we aimed to investigate novel therapeutic approaches for non-surgical AS treatment, at least in patients with mild-to-moderate AS. MATERIALS AND METHODS The most recent and relevant papers concerned with novel molecular pathways that have potential as therapeutic targets in AS were selected from searches of PubMed and Web of Science up to February 2021. RESULTS Growing evidence indicates that therapies using proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, simvastatin/ezetimibe combination, cholesteryl ester transfer protein inhibitors or antisense oligonucleotides targeting apolipoprotein(a) reduce the risk of AS progression. It has been shown that enhanced valvular lipid oxidation may drive AS development by leading to the activation of valvular interstitial cells (VICs), the most abundant valvular cells having a major contribution to valve calcification. Since VICs are able to release pro-inflammatory cytokines, clotting factors and proteins involved in calcification, strategies targeting these cell activations seem promising as therapeutic interventions. Recently, non-vitamin K antagonist oral anticoagulants (NOACs) have been shown to inhibit activation of VICs. CONCLUSION Several novel molecular pathways of AS development have been identified over the past few years. Therapies using PCSK9 inhibitors, simvastatin/ezetimibe combination, lipoprotein(a)-lowering therapy are highly promising candidates as therapeutics in the prevention of mild AS progression, while preclinical studies show that NOACs may inhibit valvular inflammation and coagulation activation and slower the rate of AS progression.
Collapse
Affiliation(s)
- Joanna Natorska
- John Paul II Hospital, Kraków, Poland.,Institute of Cardiology, Jagiellonian University Medical College, Kraków, Poland
| | - Magdalena Kopytek
- John Paul II Hospital, Kraków, Poland.,Institute of Cardiology, Jagiellonian University Medical College, Kraków, Poland
| | - Anetta Undas
- John Paul II Hospital, Kraków, Poland.,Institute of Cardiology, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
24
|
Donis N, Jiang Z, D'Emal C, Dulgheru R, Giera M, Blomberg N, Delvenne P, Nchimi A, Lancellotti P, Oury C. Regular Dietary Intake of Palmitate Causes Vascular and Valvular Calcification in a Rabbit Model. Front Cardiovasc Med 2021; 8:692184. [PMID: 34250045 PMCID: PMC8261064 DOI: 10.3389/fcvm.2021.692184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/24/2021] [Indexed: 01/23/2023] Open
Abstract
Aims: Palmitic acid (PA) and oleic acid (OA) are two main dietary fatty acids. Dietary intake of PA has been associated with cardiovascular disease risk, and the effect of OA remains uncertain. Our study aimed to assess the effect of a short-term intake of lard, as source of PA and OA, on aorta and aortic valve. Methods and Results: Rabbits were fed with two lard-enriched diets, containing either elevated levels of PA or of both PA and OA as compared to chow diet. After 16 weeks of each diet, calcification was observed in the aortic intima and in the aortic valve. The extent of calcification did not differ between the two diets. In contrast, rabbits fed chow diet did not develop any calcification. In blood, PA enrichment resulted in decreased lymphocyte and monocyte counts and increased levels of hemoglobin and haematocrit. Levels of the calcification inhibitor fetuin-A were also diminished, whereas creatinine levels were raised. Of note, none of the diets changed cholesterol levels in LDL or HDL. Comprehensive quantitative lipidomics analysis identified diet-related changes in plasma lipids. Dietary PA enrichment led to a drop of polyunsaturated fatty acids (PUFA), in particular of linoleic acid in cholesteryl esters, triglycerides and diacylglycerols (DAG). Ratios of PA to 18-carbon PUFA in DAG were positively correlated with the extent of aortic valve calcification, and inversely with monocyte counts. PA content in blood correlated with aorta calcification. Conclusions: Regular dietary PA intake induces vascular and valvular calcification independently of traditional risk factors. Our findings raise awareness about PA-rich food consumption and its potential deleterious effect on cardiovascular health.
Collapse
Affiliation(s)
- Nathalie Donis
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, CHU Sart Tilman, University of Liège, Liège, Belgium
| | - Zheshen Jiang
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, CHU Sart Tilman, University of Liège, Liège, Belgium
| | - Céline D'Emal
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, CHU Sart Tilman, University of Liège, Liège, Belgium
| | - Raluca Dulgheru
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, CHU Sart Tilman, University of Liège, Liège, Belgium
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Niek Blomberg
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Philippe Delvenne
- Department of Pathology, Centre Hospitalier Universitaire (CHU) University Hospital, Liège University, Liège, Belgium.,Laboratory of Experimental Pathology, Groupe Interdisciplinaire de Géno-protéomique Appliquée (GIGA) Institute, Liege University, Liège, Belgium
| | - Alain Nchimi
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, CHU Sart Tilman, University of Liège, Liège, Belgium
| | - Patrizio Lancellotti
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, CHU Sart Tilman, University of Liège, Liège, Belgium.,Gruppo Villa Maria Care and Research, Maria Cecilia Hospital, Cotignola, Italy.,Anthea Hospital, Bari, Italy
| | - Cécile Oury
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, CHU Sart Tilman, University of Liège, Liège, Belgium
| |
Collapse
|
25
|
Beyond Lipoprotein(a) plasma measurements: Lipoprotein(a) and inflammation. Pharmacol Res 2021; 169:105689. [PMID: 34033878 PMCID: PMC9247870 DOI: 10.1016/j.phrs.2021.105689] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022]
Abstract
Genome wide association, epidemiological, and clinical studies have established high lipoprotein(a) [Lp(a)] as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD). Lp(a) is an apoB100 containing lipoprotein covalently bound to apolipoprotein(a) [apo(a)], a glycoprotein. Plasma Lp(a) levels are to a large extent determined by genetics. Its link to cardiovascular disease (CVD) may be driven by its pro-inflammatory effects, of which its association with oxidized phospholipids (oxPL) bound to Lp(a) is the most studied. Various inflammatory conditions, such as rheumatoid arthritis (RA), systemic lupus erythematosus, acquired immunodeficiency syndrome, and chronic renal failure are associated with high Lp(a) levels. In cases of RA, high Lp(a) levels are reversed by interleukin-6 receptor (IL-6R) blockade by tocilizumab, suggesting a potential role for IL-6 in regulating Lp(a) plasma levels. Elevated levels of IL-6 and IL-6R polymorphisms are associated with CVD. Therapies aimed at lowering apo(a) and thereby reducing plasma Lp(a) levels are in clinical trials. Their results will determine if reductions in apo(a) and Lp(a) decrease cardiovascular outcomes. As we enter this new arena of available treatments, there is a need to improve our understanding of mechanisms. This review will focus on the role of Lp(a) in inflammation and CVD.
Collapse
|
26
|
Hoekstra M, Chen HY, Rong J, Dufresne L, Yao J, Guo X, Tsai MY, Tsimikas S, Post WS, Vasan RS, Rotter JI, Larson MG, Thanassoulis G, Engert JC. Genome-Wide Association Study Highlights APOH as a Novel Locus for Lipoprotein(a) Levels-Brief Report. Arterioscler Thromb Vasc Biol 2021; 41:458-464. [PMID: 33115273 PMCID: PMC7769958 DOI: 10.1161/atvbaha.120.314965] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Lp(a) (lipoprotein[a]) is an independent risk factor for cardiovascular diseases and plasma levels are primarily determined by variation at the LPA locus. We performed a genome-wide association study in the UK Biobank to determine whether additional loci influence Lp(a) levels. Approach and Results: We included 293 274 White British individuals in the discovery analysis. Approximately 93 095 623 variants were tested for association with natural log-transformed Lp(a) levels using linear regression models adjusted for age, sex, genotype batch, and 20 principal components of genetic ancestry. After quality control, 131 independent variants were associated at genome-wide significance (P≤5×10-8). In addition to validating previous associations at LPA, APOE, and CETP, we identified a novel variant at the APOH locus, encoding β2GPI (beta2-glycoprotein I). The APOH variant rs8178824 was associated with increased Lp(a) levels (β [95% CI] [ln nmol/L], 0.064 [0.047-0.081]; P=2.8×10-13) and demonstrated a stronger effect after adjustment for variation at the LPA locus (β [95% CI] [ln nmol/L], 0.089 [0.076-0.10]; P=3.8×10-42). This association was replicated in a meta-analysis of 5465 European-ancestry individuals from the Framingham Offspring Study and Multi-Ethnic Study of Atherosclerosis (β [95% CI] [ln mg/dL], 0.16 [0.044-0.28]; P=0.0071). CONCLUSIONS In a large-scale genome-wide association study of Lp(a) levels, we identified APOH as a novel locus for Lp(a) in individuals of European ancestry. Additional studies are needed to determine the precise role of β2GPI in influencing Lp(a) levels as well as its potential as a therapeutic target.
Collapse
Affiliation(s)
- Mary Hoekstra
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - Hao Yu Chen
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - Jian Rong
- Boston University’s and NHLBI’s Framingham Heart Study, Boston, Massachusetts
| | - Line Dufresne
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, California
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Martin G. Larson
- Boston University’s and NHLBI’s Framingham Heart Study, Boston, Massachusetts
| | - George Thanassoulis
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - James C. Engert
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
- Department of Human Genetics, McGill University, Montreal, Quebec
| |
Collapse
|
27
|
Metabolomic Signature of Human Aortic Valve Stenosis. JACC Basic Transl Sci 2020; 5:1163-1177. [PMID: 33426374 PMCID: PMC7775961 DOI: 10.1016/j.jacbts.2020.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 12/17/2022]
Abstract
This study is the first step towards the creation of a metabolomic map of calcified human aortic valves. The study highlights an independent association of LysoPA with CAVS severity. The study demonstrates that LysoPA levels are associated with faster CAVS progression rate.
This study outlines the first step toward creating the metabolite atlas of human calcified aortic valves by identifying the expression of metabolites and metabolic pathways involved at various stages of calcific aortic valve stenosis progression. Untargeted analysis identified 72 metabolites and lipids that were significantly altered (p < 0.01) across different stages of disease progression. Of these metabolites and lipids, the levels of lysophosphatidic acid were shown to correlate with faster hemodynamic progression and could select patients at risk for faster progression rate.
Collapse
Key Words
- AS, aortic stenosis
- ATX, autotaxin
- AV, aortic valve
- AVA, aortic valve area
- BAV, bicuspid aortic valve
- CAVS, calcific aortic valve stenosis
- CV, correlation of variation
- Lp(a), lipoprotein(a)
- LysoPA, lysophosphatidic acid
- LysoPC, lysophosphatidylcholine
- LysoPE, lysophosphatidylethanolamine
- MG, monoglyceride
- MPG, mean pressure gradient
- PC, phosphatidylcholine
- QC, quality control
- TAV, tricuspid aortic valve
- Vmax, peak aortic jet velocity
- aortic stenosis
- calcific aortic valve stenosis
- lysophosphatidic acids
- nontargeted metabolomics
- targeted lipidomics
- valvular calcification
Collapse
|
28
|
Bourgeois R, Girard A, Perrot N, Guertin J, Mitchell PL, Couture C, Gotti C, Bourassa S, Poggio P, Mass E, Capoulade R, Scipione CA, Després AA, Couture P, Droit A, Pibarot P, Boffa MB, Thériault S, Koschinsky ML, Mathieu P, Arsenault BJ. A Comparative Analysis of the Lipoprotein(a) and Low-Density Lipoprotein Proteomic Profiles Combining Mass Spectrometry and Mendelian Randomization. CJC Open 2020; 3:450-459. [PMID: 34027348 PMCID: PMC8129481 DOI: 10.1016/j.cjco.2020.11.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 11/28/2020] [Indexed: 12/30/2022] Open
Abstract
Background Lipoprotein(a) (Lp[a]), which consists of a low-density lipoprotein (LDL) bound to apolipoprotein(a), is one of the strongest genetic risk factors for atherosclerotic cardiovascular diseases. Few studies have performed hypothesis-free direct comparisons of the Lp(a) and the LDL proteomes. Our objectives were to compare the Lp(a) and the LDL proteomic profiles and to evaluate the effect of lifelong exposure to elevated Lp(a) or LDL cholesterol levels on the plasma proteomic profile. Methods We performed a label-free analysis of the Lp(a) and LDL proteomic profiles of healthy volunteers in a discovery (n = 6) and a replication (n = 9) phase. We performed inverse variance weighted Mendelian randomization to document the effect of lifelong exposure to elevated Lp(a) or LDL cholesterol levels on the plasma proteomic profile of participants of the INTERVAL study. Results We identified 15 proteins that were more abundant on Lp(a) compared with LDL (serping1, pi16, itih1, itih2, itih3, pon1, podxl, cd44, cp, ptprg, vtn, pcsk9, igfals, vcam1, and ttr). We found no proteins that were more abundant on LDL compared with Lp(a). After correction for multiple testing, lifelong exposure to elevated LDL cholesterol levels was associated with the variation of 18 plasma proteins whereas Lp(a) did not appear to influence the plasma proteome. Conclusions Results of this study highlight marked differences in the proteome of Lp(a) and LDL as well as in the effect of lifelong exposure to elevated LDL cholesterol or Lp(a) on the plasma proteomic profile.
Collapse
Affiliation(s)
- Raphaëlle Bourgeois
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Arnaud Girard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Nicolas Perrot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Jakie Guertin
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Patricia L Mitchell
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada
| | - Christian Couture
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada
| | - Clarisse Gotti
- Proteomics platform of the CHU de Québec, Quebec, Canada
| | | | | | - Elvira Mass
- University of Bonn, Developmental Biology of the Immune System, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Romain Capoulade
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Corey A Scipione
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Audrey-Anne Després
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Patrick Couture
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada.,Centre de recherche du CHU de Québec, Quebec, Canada
| | - Arnaud Droit
- Proteomics platform of the CHU de Québec, Quebec, Canada.,Centre de recherche du CHU de Québec, Quebec, Canada
| | - Philippe Pibarot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Michael B Boffa
- Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Sébastien Thériault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Patrick Mathieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Surgery, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| |
Collapse
|
29
|
Chignon A, Bon-Baret V, Boulanger MC, Bossé Y, Mathieu P. Oxyphospholipids in Cardiovascular Calcification. Arterioscler Thromb Vasc Biol 2020; 41:11-19. [PMID: 33232199 DOI: 10.1161/atvbaha.120.313790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mineralization of cardiovascular structures including blood vessels and heart valves is a common feature. We postulate that ectopic mineralization is a response-to-injury in which signals delivered to cells trigger a chain of events to restore and repair tissues. Maladaptive response to external or internal signals promote the expression of danger-associated molecular patterns, which, in turn, promote, when expressed chronically, a procalcifying gene program. Growing evidence suggest that danger-associated molecular patterns such as oxyphospholipids and small lipid mediators, generated by enzyme activity, are involved in the transition of vascular smooth muscle cells and valve interstitial cells to an osteoblast-like phenotype. Understanding the regulation and the molecular processes underpinning the mineralization of atherosclerotic plaques and cardiac valves are providing valuable mechanistic insights, which could lead to the development of novel therapies. Herein, we provide a focus account on the role oxyphospholipids and their mediators in the development of mineralization in plaques and calcific aortic valve disease.
Collapse
Affiliation(s)
- Arnaud Chignon
- Department of Surgery, Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center (A.C., V.B.-B., M.-C.B., P.M.), Laval University, Canada
| | - Valentin Bon-Baret
- Department of Surgery, Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center (A.C., V.B.-B., M.-C.B., P.M.), Laval University, Canada
| | - Marie-Chloé Boulanger
- Department of Surgery, Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center (A.C., V.B.-B., M.-C.B., P.M.), Laval University, Canada
| | - Yohan Bossé
- Department of Molecular Medicine (Y.B.), Laval University, Canada
| | - Patrick Mathieu
- Department of Surgery, Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center (A.C., V.B.-B., M.-C.B., P.M.), Laval University, Canada
| |
Collapse
|
30
|
Donato M, Ferri N, Lupo MG, Faggin E, Rattazzi M. Current Evidence and Future Perspectives on Pharmacological Treatment of Calcific Aortic Valve Stenosis. Int J Mol Sci 2020; 21:ijms21218263. [PMID: 33158204 PMCID: PMC7663524 DOI: 10.3390/ijms21218263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Calcific aortic valve stenosis (CAVS), the most common heart valve disease, is characterized by the slow progressive fibro-calcific remodeling of the valve leaflets, leading to progressive obstruction to the blood flow. CAVS is an increasing health care burden and the development of an effective medical treatment is a major medical need. To date, no effective pharmacological therapies have proven to halt or delay its progression to the severe symptomatic stage and aortic valve replacement represents the only available option to improve clinical outcomes and to increase survival. In the present report, the current knowledge and latest advances in the medical management of patients with CAVS are summarized, placing emphasis on lipid-lowering agents, vasoactive drugs, and anti-calcific treatments. In addition, novel potential therapeutic targets recently identified and currently under investigation are reported.
Collapse
Affiliation(s)
- Maristella Donato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Maria Giovanna Lupo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Elisabetta Faggin
- Department of Medicine—DIMED, University of Padova, 35122 Padova, Italy;
| | - Marcello Rattazzi
- Department of Medicine—DIMED, University of Padova, 35122 Padova, Italy;
- Correspondence: ; Tel.: +39-0498-211-867 or +39-0422-322-207
| |
Collapse
|
31
|
Makshood M, Joshi PH, Kanaya AM, Ayers C, Budoff M, Tsai MY, Blaha M, Michos ED, Post WS. Lipoprotein (a) and aortic valve calcium in South Asians compared to other race/ethnic groups. Atherosclerosis 2020; 313:14-19. [PMID: 33002750 PMCID: PMC8247632 DOI: 10.1016/j.atherosclerosis.2020.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/04/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS South Asians are at increased risk for cardiovascular disease (CVD). Aortic valve calcium (AVC) is associated with CVD risk and aortic stenosis. Elevated Lp(a) is a heritable risk factor for CVD and AVC. AVC prevalence and its association with Lp(a) have not been studied in South Asians. METHODS Among participants in the Mediators of Atherosclerosis in South Asians Living in America (MASALA) study (n = 695), AVC prevalence and extent were compared to four race/ethnic groups in the Multi-Ethnic Study of Atherosclerosis (MESA) (n = 4671). Multivariable regression was performed to evaluate associations between Lp(a) and AVC stratified by race/ethnic groups, adjusting for cardiovascular risk factors. RESULTS After age and sex adjustment, South Asians had higher median Lp(a) (17.0 mg/dL) compared to Whites (12.9 mg/dL), Hispanics (13.1 mg/dL) and Chinese Americans (12.9 mg/dL), and Blacks had highest Lp(a) levels (35.1 mg/dL). There were no differences in the odds of AVC in South Asians compared with Whites or Hispanics, after age and sex adjustment (p = 0.64 and 0.63, respectively). Odds of AVC was lower in Chinese (OR 0.35; 95%CI 0.23-0.54) and somewhat lower in Blacks compared with South Asians (OR 0.76; 0.56-1.04). There were no associations between Lp(a) and AVC presence or extent in South Asians. Lp(a) was associated with AVC only among Blacks and Whites. CONCLUSIONS Although present in Whites and Blacks, there were no associations between Lp(a) and AVC in South Asians. These differences may be due to statistic power or race specific modifying factors that influences the effect of Lp(a) particles on AVC pathogenesis.
Collapse
Affiliation(s)
- Minhal Makshood
- Johns Hopkins University, School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA.
| | - Parag H Joshi
- Department of Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Alka M Kanaya
- University of California San Francisco, 1545 Divisadero Street, Suite 311, San Francisco, CA, 94117, USA
| | - Colby Ayers
- Department of Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Matthew Budoff
- Lundquist Institute, California, 1000 W Carston St, Torrance, CA, 90502, USA
| | - Michael Y Tsai
- University of Minnesota, 420 Delaware St SE, Minneapolis, MN, 55455, USA
| | - Michael Blaha
- Johns Hopkins University, School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA; Ciccarone Center for the Prevention of Cardiovascular Disease Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD, 21287, USA
| | - Erin D Michos
- Johns Hopkins University, School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA; Ciccarone Center for the Prevention of Cardiovascular Disease Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD, 21287, USA
| | - Wendy S Post
- Johns Hopkins University, School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA; Ciccarone Center for the Prevention of Cardiovascular Disease Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD, 21287, USA
| |
Collapse
|
32
|
Nie J, Yang J, Wei Y, Wei X. The role of oxidized phospholipids in the development of disease. Mol Aspects Med 2020; 76:100909. [PMID: 33023753 DOI: 10.1016/j.mam.2020.100909] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/29/2020] [Accepted: 09/21/2020] [Indexed: 02/05/2023]
Abstract
Oxidized phospholipids (OxPLs), complex mixtures of phospholipid oxidation products generated during normal or pathological processes, are increasingly recognized to show bioactive effects on many cellular signalling pathways. There is a growing body of evidence showing that OxPLs play an important role in many diseases, so it is essential to define the specific role of OxPLs in different diseases for the design of disease therapies. In vastly diverse pathological processes, OxPLs act as pro-inflammatory agents and contribute to the progression of many diseases; in addition, they play a role in anti-inflammatory processes, promoting the dissipation of inflammation and inhibiting the progression of some diseases. In addition to participating in the regulation of inflammatory responses, OxPLs affect the occurrence and development of diseases through other pathways, such as apoptosis promotion. In this review, the different and even opposite effects of different OxPL molecular species are discussed. Furthermore, the specific effects of OxPLs in various diseases, as well as the receptor and cellular mechanisms involved, are summarized.
Collapse
Affiliation(s)
- Ji Nie
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Department of Respiration, First People's Hospital of Yunnan Province, Yunnan, 650032, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Jing Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
33
|
Bourgeois R, Devillers R, Perrot N, Després AA, Boulanger MC, Mitchell PL, Guertin J, Couture P, Boffa MB, Scipione CA, Pibarot P, Koschinsky ML, Mathieu P, Arsenault BJ. Interaction of Autotaxin With Lipoprotein(a) in Patients With Calcific Aortic Valve Stenosis. ACTA ACUST UNITED AC 2020; 5:888-897. [PMID: 33015412 PMCID: PMC7524777 DOI: 10.1016/j.jacbts.2020.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022]
Abstract
Our objectives were to determine whether autotaxin (ATX) is transported by lipoprotein(a) [Lp(a)] in human plasma and if could be used as a biomarker of calcific aortic valve stenosis (CAVS). We first found that ATX activity was higher in Lp(a) compared to low-density lipoprotein fractions in isolated fractions of 10 healthy participants. We developed a specific assay to measure ATX-Lp(a) in 88 patients with CAVS and 144 controls without CAVS. In a multivariable model corrected for CAVS risk factors, ATX-Lp(a) was associated with CAVS (p = 0.003). We concluded that ATX is preferentially transported by Lp(a) and might represent a novel biomarker for CAVS.
Collapse
Key Words
- ALR, adiponectin-to-leptin ratio
- ATX, autotaxin
- ATX-apo(a), ATX carried by Lp(a)
- ATX-apoB, ATX carried by apoB-containing lipoproteins
- BMI, body mass index
- CAD, coronary artery disease
- CAVS, calcific aortic valve stenosis
- HDL, high-density lipoprotein
- LDL, low-density lipoprotein
- Lp(a), lipoprotein(a)
- LysoPA, lysophosphatidic acid
- LysoPC, lysophosphatidylcholine
- OxPLs, oxidized phospholipids
- apo(a), apolipoprotein(a)
- apoB, apolipoprotein B
- autotaxin
- calcific aortic valve stenosis
- lipoprotein(a)
- low-density lipoproteins
- obesity
Collapse
Affiliation(s)
- Raphaëlle Bourgeois
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Romain Devillers
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada.,Department of Surgery, Faculty of Medicine, Université Laval, Québec, Canada
| | - Nicolas Perrot
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Audrey-Anne Després
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Marie-Chloé Boulanger
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada
| | - Patricia L Mitchell
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada
| | - Jakie Guertin
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Patrick Couture
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada.,Centre de Recherche du CHU de Québec, Quebec, Canada
| | - Michael B Boffa
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Corey A Scipione
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Philippe Pibarot
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Patrick Mathieu
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada.,Department of Surgery, Faculty of Medicine, Université Laval, Québec, Canada
| | - Benoit J Arsenault
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| |
Collapse
|
34
|
Roles for lysophosphatidic acid signaling in vascular development and disease. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158734. [PMID: 32376340 DOI: 10.1016/j.bbalip.2020.158734] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 01/28/2023]
Abstract
The bioactive lipid lysophosphatidic acid (LPA) is emerging as an important mediator of inflammation in cardiovascular diseases. Produced in large part by the secreted lysophospholipase D autotaxin (ATX), LPA acts on a series of G protein-coupled receptors and may have action on atypical receptors such as RAGE to exert potent effects on vascular cells, including the promotion of foam cell formation and phenotypic modulation of smooth muscle cells. The signaling effects of LPA can be terminated by integral membrane lipid phosphate phosphatases (LPP) that hydrolyze the lipid to receptor inactive products. Human genetic variants in PLPP3, that predict lower levels of LPP3, associate with risk for premature coronary artery disease, and reductions of LPP3 expression in mice promote the development of experimental atherosclerosis and enhance inflammation in the atherosclerotic lesions. Recent evidence also supports a role for ATX, and potentially LPP3, in calcific aortic stenosis. In summary, LPA may be a relevant inflammatory mediator in atherosclerotic cardiovascular disease and heightened LPA signaling may explain the cardiovascular disease risk effect of PLPP3 variants.
Collapse
|
35
|
Zheng KH, Tsimikas S, Pawade T, Kroon J, Jenkins WSA, Doris MK, White AC, Timmers NKLM, Hjortnaes J, Rogers MA, Aikawa E, Arsenault BJ, Witztum JL, Newby DE, Koschinsky ML, Fayad ZA, Stroes ESG, Boekholdt SM, Dweck MR. Lipoprotein(a) and Oxidized Phospholipids Promote Valve Calcification in Patients With Aortic Stenosis. J Am Coll Cardiol 2020; 73:2150-2162. [PMID: 31047003 PMCID: PMC6494952 DOI: 10.1016/j.jacc.2019.01.070] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/28/2019] [Accepted: 01/28/2019] [Indexed: 01/05/2023]
Abstract
Background Lipoprotein(a) [Lp(a)], a major carrier of oxidized phospholipids (OxPL), is associated with an increased incidence of aortic stenosis (AS). However, it remains unclear whether elevated Lp(a) and OxPL drive disease progression and are therefore targets for therapeutic intervention. Objectives This study investigated whether Lp(a) and OxPL on apolipoprotein B-100 (OxPL-apoB) levels are associated with disease activity, disease progression, and clinical events in AS patients, along with the mechanisms underlying any associations. Methods This study combined 2 prospective cohorts and measured Lp(a) and OxPL-apoB levels in patients with AS (Vmax >2.0 m/s), who underwent baseline 18F-sodium fluoride (18F-NaF) positron emission tomography (PET), repeat computed tomography calcium scoring, and repeat echocardiography. In vitro studies investigated the effects of Lp(a) and OxPL on valvular interstitial cells. Results Overall, 145 patients were studied (68% men; age 70.3 ± 9.9 years). On baseline positron emission tomography, patients in the top Lp(a) tertile had increased valve calcification activity compared with those in lower tertiles (n = 79; 18F-NaF tissue-to-background ratio of the most diseased segment: 2.16 vs. 1.97; p = 0.043). During follow-up, patients in the top Lp(a) tertile had increased progression of valvular computed tomography calcium score (n = 51; 309 AU/year [interquartile range: 142 to 483 AU/year] vs. 93 AU/year [interquartile range: 56 to 296 AU/year; p = 0.015), faster hemodynamic progression on echocardiography (n = 129; 0.23 ± 0.20 m/s/year vs. 0.14 ± 0.20 m/s/year] p = 0.019), and increased risk for aortic valve replacement and death (n = 145; hazard ratio: 1.87; 95% CI: 1.13 to 3.08; p = 0.014), compared with lower tertiles. Similar results were noted with OxPL-apoB. In vitro, Lp(a) induced osteogenic differentiation of valvular interstitial cells, mediated by OxPL and inhibited with the E06 monoclonal antibody against OxPL. Conclusions In patients with AS, Lp(a) and OxPL drive valve calcification and disease progression. These findings suggest lowering Lp(a) or inactivating OxPL may slow AS progression and provide a rationale for clinical trials to test this hypothesis.
Collapse
Affiliation(s)
- Kang H Zheng
- Department of Vascular Medicine, Academic Medical Center, Amsterdam UMC, Amsterdam, the Netherlands. https://twitter.com/Zheng_KH
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California, San Diego, La Jolla, California
| | - Tania Pawade
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Jeffrey Kroon
- Department of Vascular Medicine, Academic Medical Center, Amsterdam UMC, Amsterdam, the Netherlands
| | - William S A Jenkins
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Mhairi K Doris
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Audrey C White
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Nyanza K L M Timmers
- Department of Vascular Medicine, Academic Medical Center, Amsterdam UMC, Amsterdam, the Netherlands
| | - Jesper Hjortnaes
- Department of Cardiothoracic Surgery, UMC Utrecht, Utrecht, the Netherlands
| | - Maximillian A Rogers
- Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elena Aikawa
- Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Benoit J Arsenault
- Centre de recherche de l'Institut Universitaire de cardiologie et de pneumologie de Québec-Université Laval, Québec City, Québec, Canada
| | - Joseph L Witztum
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Marlys L Koschinsky
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Erik S G Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam UMC, Amsterdam, the Netherlands
| | - S Matthijs Boekholdt
- Department of Cardiology, Academic Medical Center, Amsterdam UMC, Amsterdam, the Netherlands
| | - Marc R Dweck
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
36
|
Association of serum lipoprotein(a) level with the severity and prognosis of calcific aortic valve stenosis: a Chinese cohort study. J Geriatr Cardiol 2020; 17:133-140. [PMID: 32280329 PMCID: PMC7118012 DOI: 10.11909/j.issn.1671-5411.2020.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND There was a causal relationship between elevated lipoprotein(a) [Lp(a)] levels and increased risk of calcific aortic valve stenosis (CAVS) in whites and blacks. The present study aimed to investigate whether Lp(a) levels were associated with aortic stenosis (AS) severity and clinical events in Chinese patients. METHODS Levels of serum Lp(a) were measured in 652 patients with CAVS, whom all underwent baseline echocardiographic examination. The clinical endpoint was defined as a composite of aortic valve replacement (AVR) and cardiac death. RESULTS Patients in the tertile 3 of Lp(a) had a higher percentage of severe AS compared with those in the tertile 1 and 2 of Lp(a) (46.2% vs. 33.9%, P = 0.005). Moreover, the top tertile of Lp(a) was an independent predictor of severe AS (OR = 1.78, 95% CI: 1.18-2.66, P = 0.006). However, there was no significant association between tertile 3 of Lp(a) and clinical events (hazard ratio: 0.73; 95% CI: 0.43-1.24; P = 0.239) in the multivariate Cox regression analysis during a mean follow-up time of 3.16 ± 2.74 years. CONCLUSIONS Elevated Lp(a) level was an independent predictor of severe AS by echocardiography in the Chinese population, but was not associated with the increased risk of AVR and cardiac death, suggesting that Lp(a) levels might be helpful in the risk stratification of patients with CAVS.
Collapse
|
37
|
Capoulade R, Torzewski M, Mayr M, Chan KL, Mathieu P, Bossé Y, Dumesnil JG, Tam J, Teo KK, Burnap SA, Schmid J, Gobel N, Franke UFW, Sanchez A, Witztum JL, Yang X, Yeang C, Arsenault B, Després JP, Pibarot P, Tsimikas S. ApoCIII-Lp(a) complexes in conjunction with Lp(a)-OxPL predict rapid progression of aortic stenosis. Heart 2020; 106:738-745. [PMID: 32054669 DOI: 10.1136/heartjnl-2019-315840] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE This study assessed whether apolipoprotein CIII-lipoprotein(a) complexes (ApoCIII-Lp(a)) associate with progression of calcific aortic valve stenosis (AS). METHODS Immunostaining for ApoC-III was performed in explanted aortic valve leaflets in 68 patients with leaflet pathological grades of 1-4. Assays measuring circulating levels of ApoCIII-Lp(a) complexes were measured in 218 patients with mild-moderate AS from the AS Progression Observation: Measuring Effects of Rosuvastatin (ASTRONOMER) trial. The progression rate of AS, measured as annualised changes in peak aortic jet velocity (Vpeak), and combined rates of aortic valve replacement (AVR) and cardiac death were determined. For further confirmation of the assay data, a proteomic analysis of purified Lp(a) was performed to confirm the presence of apoC-III on Lp(a). RESULTS Immunohistochemically detected ApoC-III was prominent in all grades of leaflet lesion severity. Significant interactions were present between ApoCIII-Lp(a) and Lp(a), oxidised phospholipids on apolipoprotein B-100 (OxPL-apoB) or on apolipoprotein (a) (OxPL-apo(a)) with annualised Vpeak (all p<0.05). After multivariable adjustment, patients in the top tertile of both apoCIII-Lp(a) and Lp(a) had significantly higher annualised Vpeak (p<0.001) and risk of AVR/cardiac death (p=0.03). Similar results were noted with OxPL-apoB and OxPL-apo(a). There was no association between autotaxin (ATX) on ApoB and ATX on Lp(a) with faster progression of AS. Proteomic analysis of purified Lp(a) showed that apoC-III was prominently present on Lp(a). CONCLUSION ApoC-III is present on Lp(a) and in aortic valve leaflets. Elevated levels of ApoCIII-Lp(a) complexes in conjunction with Lp(a), OxPL-apoB or OxPL-apo(a) identify patients with pre-existing mild-moderate AS who display rapid progression of AS and higher rates of AVR/cardiac death. TRIAL REGISTRATION NCT00800800.
Collapse
Affiliation(s)
- Romain Capoulade
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec Heart and Lung Institut, Quebec city, Quebec, Canada.,Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Michael Torzewski
- Department of Cardiovascular Surgery, Robert Bosch Hospital, Stuttgart, Baden-Wurttemberg, Germany
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Kwan-Leung Chan
- Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Patrick Mathieu
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec Heart and Lung Institut, Quebec city, Quebec, Canada
| | - Yohan Bossé
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec Heart and Lung Institut, Quebec city, Quebec, Canada
| | - Jean G Dumesnil
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec Heart and Lung Institut, Quebec city, Quebec, Canada
| | - James Tam
- Department of Internal Medecine, St Boniface Hospital Research, Winnipeg, Manitoba, Canada
| | - Koon K Teo
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sean A Burnap
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Jens Schmid
- Department of Cardiovascular Surgery, Robert Bosch Hospital, Stuttgart, Baden-Wurttemberg, Germany
| | - Nora Gobel
- Department of Cardiovascular Surgery, Robert Bosch Hospital, Stuttgart, Baden-Wurttemberg, Germany
| | - Ulrich F W Franke
- Department of Cardiovascular Surgery, Robert Bosch Hospital, Stuttgart, Baden-Wurttemberg, Germany
| | - Amber Sanchez
- Department of Nephrology, University of California San Diego, La Jolla, California, USA
| | - Joseph L Witztum
- Department of Endocrinology and Metabolism, University of California San Diego, La Jolla, California, USA
| | - Xiaohong Yang
- Department of cardiology, University of California San Diego, La Jolla, California, USA
| | - Calvin Yeang
- Department of cardiology, University of California San Diego, La Jolla, California, USA
| | - Benoit Arsenault
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec Heart and Lung Institut, Quebec city, Quebec, Canada
| | - Jean-Pierre Després
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec Heart and Lung Institut, Quebec city, Quebec, Canada
| | - Philippe Pibarot
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec Heart and Lung Institut, Quebec city, Quebec, Canada
| | - Sotirios Tsimikas
- Department of cardiology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
38
|
Tsimikas S. Potential Causality and Emerging Medical Therapies for Lipoprotein(a) and Its Associated Oxidized Phospholipids in Calcific Aortic Valve Stenosis. Circ Res 2019; 124:405-415. [PMID: 30702993 DOI: 10.1161/circresaha.118.313864] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The prevalence of calcific aortic valve disease is increasing with aging of the population. Current treatment options for advanced or symptomatic aortic stenosis are limited to traditional surgical or percutaneous aortic valve replacement. Medical therapies that impact the progression of calcific aortic valve disease do not currently exist. New pathophysiological insights suggest that the processes leading to calcific aortic valve disease are metabolically active for many years before and during the clinical expression of disease. The identification of genetic and potentially causal mediators of calcific aortic valve disease allows opportunities for therapies that may slow progression to the point where aortic valve replacement can be avoided. Recent studies suggest that approximately one-third of aortic stenosis cases are associated with highly elevated lipoprotein(a) [Lp(a)] and pathways related to the metabolism of procalcifying oxidized phospholipids. Oxidized phospholipids can be carried by Lp(a) into valve leaflets but can also be formed in situ from cell membranes, lipoproteins, and apoptotic cells. This review will summarize the clinical data implicating the potential causality of Lp(a)/oxidized phospholipids, describe emerging therapeutic agents, and propose clinical trial designs to test the hypothesis that lowering Lp(a) will reduce progression aortic stenosis and the need for aortic valve replacement.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- From the Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla
| |
Collapse
|
39
|
Capoulade R, Yeang C, Chan KL, Pibarot P, Tsimikas S. Association of Mild to Moderate Aortic Valve Stenosis Progression With Higher Lipoprotein(a) and Oxidized Phospholipid Levels: Secondary Analysis of a Randomized Clinical Trial. JAMA Cardiol 2019; 3:1212-1217. [PMID: 30476957 DOI: 10.1001/jamacardio.2018.3798] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Importance Several studies have reported an association of levels of lipoprotein(a) (Lp[a]) and the content of oxidized phospholipids on apolipoprotein B (OxPL-apoB) and apolipoprotein(a) (OxPL-apo[a]) with faster calcific aortic valve stenosis (CAVS) progression. However, whether this association is threshold or linear remains unclear. Objective To determine whether the plasma levels of Lp(a), OxPL-apoB, and OxPL-apo(a) have a linear association with a faster rate of CAVS progression. Design, Setting, and Participants This secondary analysis of a randomized clinical trial tested the association of baseline plasma levels of Lp(a), OxPL-apoB, and OxPL-apo(a) with the rate of CAVS progression. Participants were included from the ASTRONOMER (Effects of Rosuvastatin on Aortic Stenosis Progression) trial, a multicenter study conducted in 23 Canadian sites designed to test the effect of statin therapy (median follow-up, 3.5 years [interquartile range, 2.9-4.5 years]). Patients with mild to moderate CAVS defined by peak aortic jet velocity ranging from 2.5 to 4.0 m/s were recruited; those with peak aortic jet velocity of less than 2.5 m/s or with an indication for statin therapy were excluded. Data were collected from January 1, 2002, through December 31, 2005, and underwent ad hoc analysis from April 1 through September 1, 2018. Interventions After the randomization process, patients were followed up by means of echocardiography for 3 to 5 years. Main Outcomes and Measures Progression rate of CAVS as assessed by annualized progression of peak aortic jet velocity. Results In this cohort of 220 patients (60.0% male; mean [SD] age, 58 [13] years), a linear association was found between plasma levels of Lp(a) (odds ratio [OR] per 10-mg/dL increase, 1.10; 95% CI, 1.03-1.19; P = .006), OxPL-apoB (OR per 1-nM increase, 1.06; 95% CI, 1.01-1.12; P = .02), and OxPL-apo(a) (OR per 10-nM increase, 1.16; 95% CI, 1.05-1.27; P = .002) and faster CAVS progression, which is marked in younger patients (OR for Lp[a] level per 10-mg/dL increase, 1.19 [95% CI, 1.07-1.33; P = .002]; OR for OxPL-apoB level per 1-nM increase, 1.06 [95% CI, 1.02-1.17; P = .01]; and OR for OxPL-apo[a] level per 10-nM increase, 1.26 [95% CI, 1.10-1.45; P = .001]) and remained statistically significant after comprehensive multivariable adjustment (β coefficient, ≥ 0.25; SE, ≤ 0.004 [P ≤ .005]; OR, ≥1.10 [P ≤ .007]). Conclusions and Relevance This study demonstrates that the association of Lp(a) levels and its content in OxPL with faster CAVS progression is linear, reinforcing the concept that Lp(a) levels should be measured in patients with mild to moderate CAVS to enhance management and risk stratification. Trial Registration ClinicalTrials.gov Identifier: NCT00800800.
Collapse
Affiliation(s)
- Romain Capoulade
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Québec City, Québec, Canada.,L'institut du thorax, Institut National de la Santé et de la Recherche Medicale, Centre National de la Recherche Scientifique, Centre Hospitalier Universitaire Nantes, University of Nantes, Nantes, France
| | - Calvin Yeang
- Vascular Medicine Program, University of California, San Diego, La Jolla
| | - Kwan L Chan
- Ottawa Heart Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Philippe Pibarot
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Québec City, Québec, Canada
| | - Sotirios Tsimikas
- Vascular Medicine Program, University of California, San Diego, La Jolla
| |
Collapse
|
40
|
Kraemer MP, Mao G, Hammill C, Yan B, Li Y, Onono F, Smyth SS, Morris AJ. Effects of diet and hyperlipidemia on levels and distribution of circulating lysophosphatidic acid. J Lipid Res 2019; 60:1818-1828. [PMID: 31484695 DOI: 10.1194/jlr.m093096] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 08/19/2019] [Indexed: 12/12/2022] Open
Abstract
Lysophosphatidic acids (LPAs) are bioactive radyl hydrocarbon-substituted derivatives of glycerol 3-phosphate. LPA metabolism and signaling are implicated in heritable risk of coronary artery disease. Genetic and pharmacological inhibition of these processes attenuate experimental atherosclerosis. LPA accumulates in atheromas, which may be a consequence of association with LDLs. The source, regulation, and biological activity of LDL-associated LPA are unknown. We examined the effects of experimental hyperlipidemia on the levels and distribution of circulating LPA in mice. The majority of plasma LPA was associated with albumin in plasma from wild-type mice fed normal chow. LDL-associated LPA was increased in plasma from high-fat Western diet-fed mice that are genetically prone to hyperlipidemia (LDL receptor knockout or activated proprotein convertase subtilisin/kexin type 9-overexpressing C57Bl6). Adipose-specific deficiency of the ENPP2 gene encoding the LPA-generating secreted lysophospholipase D, autotaxin (ATX), attenuated these Western diet-dependent increases in LPA. ATX-dependent increases in LDL-associated LPA were observed in isolated incubated plasma. ATX acted directly on LDL-associated lysophospholipid substrates in vitro. LDL from all human subjects examined contained LPA and was decreased by lipid-lowering drug therapies. Human and mouse plasma therefore contains a diet-sensitive LDL-associated LPA pool that might contribute to the cardiovascular disease-promoting effects of LPA.
Collapse
Affiliation(s)
- Maria P Kraemer
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY.,Lexington Veterans Affairs Medical Center, Lexington, KY
| | - Guogen Mao
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY.,Lexington Veterans Affairs Medical Center, Lexington, KY
| | - Courtney Hammill
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY.,Lexington Veterans Affairs Medical Center, Lexington, KY
| | - Baoxiang Yan
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY
| | - Yu Li
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY
| | - Fredrick Onono
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY
| | - Susan S Smyth
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY.,Lexington Veterans Affairs Medical Center, Lexington, KY
| | - Andrew J Morris
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY .,Lexington Veterans Affairs Medical Center, Lexington, KY
| |
Collapse
|
41
|
Tsimikas S, Fazio S, Ferdinand KC, Ginsberg HN, Koschinsky ML, Marcovina SM, Moriarty PM, Rader DJ, Remaley AT, Reyes-Soffer G, Santos RD, Thanassoulis G, Witztum JL, Danthi S, Olive M, Liu L. NHLBI Working Group Recommendations to Reduce Lipoprotein(a)-Mediated Risk of Cardiovascular Disease and Aortic Stenosis. J Am Coll Cardiol 2019; 71:177-192. [PMID: 29325642 DOI: 10.1016/j.jacc.2017.11.014] [Citation(s) in RCA: 326] [Impact Index Per Article: 65.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022]
Abstract
Pathophysiological, epidemiological, and genetic studies provide strong evidence that lipoprotein(a) [Lp(a)] is a causal mediator of cardiovascular disease (CVD) and calcific aortic valve disease (CAVD). Specific therapies to address Lp(a)-mediated CVD and CAVD are in clinical development. Due to knowledge gaps, the National Heart, Lung, and Blood Institute organized a working group that identified challenges in fully understanding the role of Lp(a) in CVD/CAVD. These included the lack of research funding, inadequate experimental models, lack of globally standardized Lp(a) assays, and inadequate understanding of the mechanisms underlying current drug therapies on Lp(a) levels. Specific recommendations were provided to facilitate basic, mechanistic, preclinical, and clinical research on Lp(a); foster collaborative research and resource sharing; leverage expertise of different groups and centers with complementary skills; and use existing National Heart, Lung, and Blood Institute resources. Concerted efforts to understand Lp(a) pathophysiology, together with diagnostic and therapeutic advances, are required to reduce Lp(a)-mediated risk of CVD and CAVD.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Vascular Medicine Program, Sulpizio Cardiovascular Center, Division of Cardiology, Department of Medicine, University of California San Diego, La Jolla, California.
| | - Sergio Fazio
- Oregon Health & Science University, Portland, Oregon
| | | | - Henry N Ginsberg
- College of Physicians and Surgeons, Columbia University, New York, New York
| | - Marlys L Koschinsky
- Robarts Research Institute and Department of Physiology & Pharmacology Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | | | | | - Daniel J Rader
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alan T Remaley
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Raul D Santos
- Heart Institute (InCor) University of Sao Paulo Medical School Hospital and Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | | | - Joseph L Witztum
- Division of Endocrinology, Department of Medicine, University of California San Diego, La Jolla, California
| | - Simhan Danthi
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Michelle Olive
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lijuan Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
42
|
Development of calcific aortic valve disease: Do we know enough for new clinical trials? J Mol Cell Cardiol 2019; 132:189-209. [PMID: 31136747 DOI: 10.1016/j.yjmcc.2019.05.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/11/2019] [Accepted: 05/19/2019] [Indexed: 12/19/2022]
Abstract
Calcific aortic valve disease (CAVD), previously thought to represent a passive degeneration of the valvular extracellular matrix (VECM), is now regarded as an intricate multistage disorder with sequential yet intertangled and interacting underlying processes. Endothelial dysfunction and injury, initiated by disturbed blood flow and metabolic disorders, lead to the deposition of low-density lipoprotein cholesterol in the VECM further provoking macrophage infiltration, oxidative stress, and release of pro-inflammatory cytokines. Such changes in the valvular homeostasis induce differentiation of normally quiescent valvular interstitial cells (VICs) into synthetically active myofibroblasts producing excessive quantities of the VECM and proteins responsible for its remodeling. As a result of constantly ongoing degradation and re-deposition, VECM becomes disorganised and rigid, additionally potentiating myofibroblastic differentiation of VICs and worsening adaptation of the valve to the blood flow. Moreover, disrupted and excessively vascularised VECM is susceptible to the dystrophic calcification caused by calcium and phosphate precipitating on damaged collagen fibers and concurrently accompanied by osteogenic differentiation of VICs. Being combined, passive calcification and biomineralisation synergistically induce ossification of the aortic valve ultimately resulting in its mechanical incompetence requiring surgical replacement. Unfortunately, multiple attempts have failed to find an efficient conservative treatment of CAVD; however, therapeutic regimens and clinical settings have also been far from the optimal. In this review, we focused on interactions and transitions between aforementioned mechanisms demarcating ascending stages of CAVD, suggesting a predisposing condition (bicuspid aortic valve) and drug combination (lipid-lowering drugs combined with angiotensin II antagonists and cytokine inhibitors) for the further testing in both preclinical and clinical trials.
Collapse
|
43
|
Després AA, Perrot N, Poulin A, Tastet L, Shen M, Chen HY, Bourgeois R, Trottier M, Tessier M, Guimond J, Nadeau M, Engert JC, Thériault S, Bossé Y, Witztum JL, Couture P, Mathieu P, Dweck MR, Tsimikas S, Thanassoulis G, Pibarot P, Clavel MA, Arsenault BJ. Lipoprotein(a), Oxidized Phospholipids, and Aortic Valve Microcalcification Assessed by 18F-Sodium Fluoride Positron Emission Tomography and Computed Tomography. CJC Open 2019; 1:131-140. [PMID: 32159096 PMCID: PMC7063623 DOI: 10.1016/j.cjco.2019.03.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 03/14/2019] [Indexed: 11/19/2022] Open
Abstract
Background Lipoprotein(a) (Lp[a]) is the preferential lipoprotein carrier of oxidized phospholipids (OxPLs) and a well-established genetic risk factor for calcific aortic valve stenosis (CAVS). Whether Lp(a) predicts aortic valve microcalcification in individuals without CAVS is unknown. Our objective was to estimate the prevalence of elevated Lp(a) and OxPL levels in patients with CAVS and to determine if individuals with elevated Lp(a) but without CAVS have higher aortic valve microcalcification. Methods We recruited 214 patients with CAVS from Montreal and 174 patients with CAVS and 108 controls from Québec City, Canada. In a second group of individuals with high (≥75 nmol/L, n = 27) or low (<75 nmol/L, n = 28) Lp(a) levels, 18F-sodium fluoride positron emission tomography/computed tomography was performed to determine the difference in mean tissue-to-background ratio (TBR) of the aortic valve. Results Patients with CAVS had 62.0% higher Lp(a) (median = 28.7, interquartile range [8.2-116.6] vs 10.9 [3.6-28.8] nmol/L, P < 0.0001), 50% higher OxPL-apolipoprotein-B (2.2 [1.3-6.0] vs 1.1 [0.7-2.6] nmol/L, P < 0.0001), and 69.9% higher OxPL-apolipoprotein(a) (7.3 [1.8-28.4] vs 2.2 [0.8-8.4] nmol/L, P < 0.0001) levels compared with individuals without CAVS (all P < 0.0001). Individuals without CAVS but elevated Lp(a) had 40% higher mean TBR compared with individuals with low Lp(a) levels (mean TBR = 1.25 ± 0.23 vs 1.15 ± 0.11, P = 0.02). Conclusions Elevated Lp(a) and OxPL levels are associated with prevalent CAVS in patients studied in an echocardiography laboratory setting. In individuals with elevated Lp(a), evidence of aortic valve microcalcification by 18F-sodium fluoride positron emission tomography/computed tomography is present before the development of clinically manifested CAVS.
Collapse
Affiliation(s)
- Audrey-Anne Després
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Nicolas Perrot
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Anthony Poulin
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
| | - Lionel Tastet
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Mylène Shen
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Hao Yu Chen
- McGill University Health Research Center, Montreal, Québec, Canada
| | - Raphaëlle Bourgeois
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Mikaël Trottier
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
| | - Michel Tessier
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
| | - Jean Guimond
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
| | - Maxime Nadeau
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
| | - James C. Engert
- McGill University Health Research Center, Montreal, Québec, Canada
| | - Sébastien Thériault
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Yohan Bossé
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Joseph L. Witztum
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Patrick Couture
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
- Centre de recherche du CHU de Québec, Québec, Québec, Canada
| | - Patrick Mathieu
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Marc R. Dweck
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Sotirios Tsimikas
- Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, California, USA
| | | | - Philippe Pibarot
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Marie-Annick Clavel
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Benoit J. Arsenault
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
- Corresponding author: Dr Benoit J. Arsenault, Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Y-3106, Pavillon Marguerite D’Youville, 2725 chemin Ste-Foy, Québec, Québec G1V 4G5, Canada. Tel.: +1-418-656-8711, ext. 3498.
| |
Collapse
|
44
|
Zhao Y, Hasse S, Zhao C, Bourgoin SG. Targeting the autotaxin - Lysophosphatidic acid receptor axis in cardiovascular diseases. Biochem Pharmacol 2019; 164:74-81. [PMID: 30928673 DOI: 10.1016/j.bcp.2019.03.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/26/2019] [Indexed: 02/06/2023]
Abstract
Lysophosphatidic acid (LPA) is a well-characterized bioactive lipid mediator, which is involved in development, physiology, and pathological processes of the cardiovascular system. LPA can be produced both inside cells and in biological fluids. The majority of extracellularLPAis produced locally by the secreted lysophospholipase D, autotaxin (ATX), through its binding to various β integrins or heparin sulfate on cell surface and hydrolyzing various lysophospholipids. LPA initiates cellular signalling pathways upon binding to and activation of its G protein-coupled receptors (LPA1-6). LPA has potent effects on various blood cells and vascular cells involved in the development of cardiovascular diseases such as atherosclerosis and aortic valve sclerosis. LPA signalling drives cell migration and proliferation, cytokine production, thrombosis, fibrosis, as well as angiogenesis. For instance, LPA promotes activation and aggregation of platelets through LPA5, increases expression of adhesion molecules in endothelial cells, and enhances expression of tissue factor in vascular smooth muscle cells. Furthermore, LPA induces differentiation of monocytes into macrophages and stimulates oxidized low-density lipoproteins (oxLDLs) uptake by macrophages to form foam cells during formation of atherosclerotic lesions through LPA1-3. This review summarizes recent findings of the roles played by ATX, LPA and LPA receptors (LPARs) in atherosclerosis and calcific aortic valve disease. Targeting the ATX-LPAR axis may have potential applications for treatment of patients suffering from various cardiovascular diseases.
Collapse
Affiliation(s)
- Yang Zhao
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V4G2, Canada
| | - Stephan Hasse
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V4G2, Canada
| | - Chenqi Zhao
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada
| | - Sylvain G Bourgoin
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V4G2, Canada.
| |
Collapse
|
45
|
Mathieu P, Boulanger MC. Autotaxin and Lipoprotein Metabolism in Calcific Aortic Valve Disease. Front Cardiovasc Med 2019; 6:18. [PMID: 30881959 PMCID: PMC6405425 DOI: 10.3389/fcvm.2019.00018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/12/2019] [Indexed: 02/06/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is a complex trait disorder characterized by calcific remodeling of leaflets. Genome-wide association (GWA) study and Mendelian randomization (MR) have highlighted that LPA, which encodes for apolipoprotein(a) [apo(a)], is causally associated with CAVD. Apo(a) is the protein component of Lp(a), a LDL-like particle, which transports oxidized phospholipids (OxPLs). Autotaxin (ATX), which is encoded by ENPP2, is a member of the ecto-nucleotidase family of enzymes, which is, however, a lysophospholipase. As such, ATX converts phospholipids into lysophosphatidic acid (LysoPA), a metabolite with potent and diverse biological properties. Studies have recently underlined that ATX is enriched in the Lp(a) lipid fraction. Functional experiments and data obtained in mouse models suggest that ATX mediates inflammation and mineralization of the aortic valve. Recent findings also indicate that epigenetically-driven processes lower the expression of phospholipid phosphatase 3 (PLPP3) and increased LysoPA signaling and inflammation in the aortic valve during CAVD. These recent data thus provide novel insights about how lipoproteins mediate the development of CAVD. Herein, we review the implication of lipoproteins in CAVD and examine the role of ATX in promoting the osteogenic transition of valve interstitial cells (VICs).
Collapse
Affiliation(s)
- Patrick Mathieu
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Research Center, Quebec Heart and Lung Institute, Laval University, Quebec, QC, Canada
| | - Marie-Chloé Boulanger
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Research Center, Quebec Heart and Lung Institute, Laval University, Quebec, QC, Canada
| |
Collapse
|
46
|
Vuorio A, Watts GF, Kovanen PT. Lipoprotein(a) as a risk factor for calcific aortic valvulopathy in heterozygous familial hypercholesterolemia. Atherosclerosis 2019; 281:25-30. [DOI: 10.1016/j.atherosclerosis.2018.11.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/17/2018] [Accepted: 11/28/2018] [Indexed: 12/24/2022]
|
47
|
Ma GS, Wilkinson MJ, Reeves RR, Yeang C, DeMaria AN, Cotter B, Patel M, Mahmud E, Tsimikas S. Lipoprotein(a) in Patients Undergoing Transcatheter Aortic Valve Replacement. Angiology 2019; 70:332-336. [DOI: 10.1177/0003319719826461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Lipoprotein(a) [Lp(a)] is a genetically determined risk factor for calcific aortic valve stenosis (CAVS) for which transcatheter aortic valve replacement (TAVR) is increasingly utilized as treatment. We evaluated the effect of a program to increase testing of and define the prevalence of elevated Lp(a) among patients undergoing TAVR. Educational efforts and incorporation of a “check-box” Lp(a) order to the preoperative TAVR order set were instituted. Retrospective chart review was performed in 229 patients requiring TAVR between May 2013 and September 2018. Of these patients, 57% had an Lp(a) level measured; testing rates increased from 0% in 2013 to 96% in 2018. Lipoprotein(a) testing occurred in 11% of patients before and in 80% of patients after the “check-box” order set ( P < .001). The prevalence of elevated Lp(a) (≥30 mg/dL) was 35%; these patients had a higher incidence of coronary artery disease requiring revascularization compared with patients with normal Lp(a) (65% vs 47%; P = .047). Patients with Lp(a) ≥30 mg/dL also had higher incidence of paravalvular leak compared with those with normal Lp(a) (13% vs 4%; P = .04). This study defines the prevalence of elevated Lp(a) in advanced stages of CAVS and provides a practice pathway to assess procedural complications and long-term outcomes of TAVR in patients with elevated Lp(a) levels.
Collapse
Affiliation(s)
- Gary S. Ma
- Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California San Diego, San Diego, CA, USA
| | - Michael J. Wilkinson
- Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California San Diego, San Diego, CA, USA
| | - Ryan R. Reeves
- Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California San Diego, San Diego, CA, USA
| | - Calvin Yeang
- Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California San Diego, San Diego, CA, USA
| | - Anthony N. DeMaria
- Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California San Diego, San Diego, CA, USA
| | - Bruno Cotter
- Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California San Diego, San Diego, CA, USA
| | - Mitul Patel
- Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California San Diego, San Diego, CA, USA
| | - Ehtisham Mahmud
- Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California San Diego, San Diego, CA, USA
| | - Sotirios Tsimikas
- Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
48
|
Boffa MB, Koschinsky ML. Oxidized phospholipids as a unifying theory for lipoprotein(a) and cardiovascular disease. Nat Rev Cardiol 2019; 16:305-318. [DOI: 10.1038/s41569-018-0153-2] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
49
|
McCormick SPA, Schneider WJ. Lipoprotein(a) catabolism: a case of multiple receptors. Pathology 2018; 51:155-164. [PMID: 30595508 DOI: 10.1016/j.pathol.2018.11.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 10/31/2018] [Accepted: 11/01/2018] [Indexed: 01/09/2023]
Abstract
Lipoprotein(a) [Lp(a)] is an apolipoprotein B (apoB)-containing plasma lipoprotein similar in structure to low-density lipoprotein (LDL). Lp(a) is more complex than LDL due to the presence of apolipoprotein(a) [apo(a)], a large glycoprotein sharing extensive homology with plasminogen, which confers some unique properties onto Lp(a) particles. ApoB and apo(a) are essential for the assembly and catabolism of Lp(a); however, other proteins associated with the particle may modify its metabolism. Lp(a) specifically carries a cargo of oxidised phospholipids (OxPL) bound to apo(a) which stimulates many proinflammatory pathways in cells of the arterial wall, a key property underlying its pathogenicity and association with cardiovascular disease (CVD). While the liver and kidney are the major tissues implicated in Lp(a) clearance, the pathways for Lp(a) uptake appear to be complex and are still under investigation. Biochemical studies have revealed an exceptional array of receptors that associate with Lp(a) either via its apoB, apo(a), or OxPL components. These receptors fall into five main categories, namely 'classical' lipoprotein receptors, toll-like and scavenger receptors, lectins, and plasminogen receptors. The roles of these receptors have largely been dissected by genetic manipulation in cells or mice, although their relative physiological importance for removal of Lp(a) from the circulation remains unclear. The LPA gene encoding apo(a) has an overwhelming effect on Lp(a) levels which precludes any clear associations between potential Lp(a) receptor genes and Lp(a) levels in population studies. Targeted approaches and the selection of unique Lp(a) phenotypes within populations has nevertheless allowed for some associations to be made. Few of the proposed Lp(a) receptors can specifically be manipulated with current drugs and, as such, it is not currently clear whether any of these receptors could provide relevant targets for therapeutic manipulation of Lp(a) levels. This review summarises the current status of knowledge about receptor-mediated pathways for Lp(a) catabolism.
Collapse
Affiliation(s)
- Sally P A McCormick
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.
| | - Wolfgang J Schneider
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
50
|
Koleini N, Nickel BE, Edel AL, Fandrich RR, Ravandi A, Kardami E. Non-mitogenic FGF2 protects cardiomyocytes from acute doxorubicin-induced toxicity independently of the protein kinase CK2/heme oxygenase-1 pathway. Cell Tissue Res 2018; 374:607-617. [PMID: 30159756 PMCID: PMC6267702 DOI: 10.1007/s00441-018-2905-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/25/2018] [Indexed: 12/01/2022]
Abstract
Doxorubicin (Dox)-induced cardiotoxicity, a limiting factor in the use of Dox to treat cancer, can be mitigated by the mitogenic factor FGF2 in vitro, via a heme oxygenase 1 (HO-1)-dependent pathway. HO-1 upregulation was reported to require protein kinase CK2 activity. We show that a mutant non-mitogenic FGF2 (S117A-FGF2), which does not activate CK2, is cardioprotective against acute cardiac ischemic injury. We now investigate the potential of S117A-FGF2 to protect cardiomyocytes against acute Dox injury and decrease Dox-induced upregulation of oxidized phospholipids. The roles of CK2 and HO-1 in cardiomyocyte protection are also addressed.Rat neonatal cardiomyocyte cultures were used as an established in vitro model of acute Dox toxicity. Pretreatment with S117A-FGF2 protected against Dox-induced: oxidative stress; upregulation of fragmented and non-fragmented oxidized phosphatidylcholine species, measured by LC/MS/MS; and cardiomyocyte injury and cell death measured by LDH release and a live-dead assay. CK2 inhibitors (TBB and Ellagic acid), did not affect protection by S117A-FGF2 but prevented protection by mitogenic FGF2. Furthermore, protection by S117A-FGF2, unlike that of FGF2, was not prevented by HO-1 inhibitors and S117A-FGF2 did not upregulate HO-1. Protection by S117A-FGF2 required the activity of FGF receptor 1 and ERK.We conclude that mitogenic and non-mitogenic FGF2 protect from acute Dox toxicity by common (FGFR1) and distinct, CK2/HO-1- dependent or CK2/HO-1-independent (respectively), pathways. Non-mitogenic FGF2 merits further consideration as a preventative treatment against Dox cardiotoxicity.
Collapse
Affiliation(s)
- Navid Koleini
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre., 351 Tache Ave, Winnipeg, Manitoba, R2H2A6, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Barbara E Nickel
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre., 351 Tache Ave, Winnipeg, Manitoba, R2H2A6, Canada
| | - Andrea L Edel
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre., 351 Tache Ave, Winnipeg, Manitoba, R2H2A6, Canada
| | - Robert R Fandrich
- Department of Human Anatomy and Cell Sciences, University of Manitoba, Winnipeg, Canada
| | - Amir Ravandi
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre., 351 Tache Ave, Winnipeg, Manitoba, R2H2A6, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
- Interventional Cardiology, Section of Cardiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Elissavet Kardami
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre., 351 Tache Ave, Winnipeg, Manitoba, R2H2A6, Canada.
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.
- Department of Human Anatomy and Cell Sciences, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|