1
|
Blaser MC, Bäck M, Lüscher TF, Aikawa E. Calcific aortic stenosis: omics-based target discovery and therapy development. Eur Heart J 2024:ehae829. [PMID: 39656785 DOI: 10.1093/eurheartj/ehae829] [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: 07/19/2024] [Revised: 10/01/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Calcific aortic valve disease (CAVD) resulting in aortic stenosis (AS) is the most common form of valvular heart disease, affecting 2% of those over age 65. Those who develop symptomatic severe AS have an average further lifespan of <2 years without valve replacement, and three-quarters of these patients will develop heart failure, undergo valve replacement, or die within 5 years. There are no approved pharmaceutical therapies for AS, due primarily to a limited understanding of the molecular mechanisms that direct CAVD progression in the complex haemodynamic environment. Here, advances in efforts to understand the pathogenesis of CAVD and to identify putative drug targets derived from recent multi-omics studies [including (epi)genomics, transcriptomics, proteomics, and metabolomics] of blood and valvular tissues are reviewed. The recent explosion of single-cell omics-based studies in CAVD and the pathobiological and potential drug discovery insights gained from the application of omics to this disease area are a primary focus. Lastly, the translation of knowledge gained in valvular pathobiology into clinical therapies is addressed, with a particular emphasis on treatment regimens that consider sex-specific, renal, and lipid-mediated contributors to CAVD, and ongoing Phase I/II/III trials aimed at the prevention/treatment of AS are described.
Collapse
Affiliation(s)
- Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, 17th Floor, Boston, MA 02115, USA
| | - Magnus Bäck
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Valvular and Coronary Disease, Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, 17th Floor, Boston, MA 02115, USA
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB 741, Boston, MA 02115, USA
| |
Collapse
|
2
|
Gu Y, Chen R, Song J, Shi Z, Wu J, Chang H, Yuan C, Shi W, Zhang Y. MiR-21-5p Promotes Osteogenic Differentiation and Calcification of Valvular Interstitial Cells by Targeting TGFBI in Calcific Aortic Valve Disease. IRANIAN JOURNAL OF PUBLIC HEALTH 2024; 53:2260-2270. [PMID: 39544858 PMCID: PMC11557769 DOI: 10.18502/ijph.v53i10.16703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 04/12/2024] [Indexed: 11/17/2024]
Abstract
Background Calcific aortic valve disease (CAVD) is the most common heart relating disease with high morbidity and mortality, especially in elderly population. While extensive investigations have been devoted to the study of mechanistic pathways related to CAVD, the key factors and mechanisms mediating valve mineralization remain unclear. The aim of this study is to investigate the role of mirnas and their downstream targets in CAVD disease progression. A previous recent multi-omics study suggested a novel CAVD molecular interaction network contained miR-21-5p. Methods CAV and their pair-matched adjacent normal tissues were obtained from 15 patients pathologically diagnosed as CAVD and admitted in Yancheng Third People's Hospital (The Sixth Affiliated Hospital of Nantong University) from 2019-2021. RT-qPCR was utilized for detection of miR-21-5p and related protein expression levels to confirm the related factors in CAVD progression. Western blotting was applied to strengthen the results of RT-qPCR and confirm osteogenic differentiation of VICs via biomarker detection. The staining of alkaline phosphatase (ALP) and alizarin red was performed to assess the degree of VIC mineralization. Results We found that miR-21-5p was remarkably increased (P<0.0001) in calcified aortic valves (AVs) whereas TGFBI was diminished (P<0.01) in CAVD samples compared to the paired normal tissues from CAVD patients. Additionally, TGFBI was targeted by miR-21-5p. Furthermore, overexpressing TGFBI could block VIC osteogenic differentiation mediated by miR-21-5p. To sum up, miR-21-5p promotes VIC osteogenic differentiation and calcification via TGFBI in CAVD progression. Conclusion Our work might bring a sight on underlying mechanisms of CAVD progression and provide a possible therapeutic target for diagnosis and treatment.
Collapse
Affiliation(s)
- Yan Gu
- Department of Cardiothoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, Jiangsu 224000, China
| | - Rongjin Chen
- Department of Cardiothoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, Jiangsu 224000, China
| | - Jianxiang Song
- Department of Cardiothoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, Jiangsu 224000, China
| | - Zhan Shi
- Department of Cardiothoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, Jiangsu 224000, China
| | - Jixiang Wu
- Department of Cardiothoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, Jiangsu 224000, China
| | - Huiwen Chang
- Department of Cardiothoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, Jiangsu 224000, China
| | - Conghu Yuan
- Department of Anesthesiology, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yan-cheng, Jiangsu 224000, China
| | - Woda Shi
- Department of Cardiothoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, Jiangsu 224000, China
| | - Yajun Zhang
- Department of Cardiothoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, Jiangsu 224000, China
| |
Collapse
|
3
|
Marrero N, Jha K, Hughes TM, Razavi AC, Grant JK, Boakye E, Anchouche K, Dzaye O, Budoff MJ, Rotter JI, Guo X, Yao J, Wood AC, Blumenthal RS, Michos ED, Thanassoulis G, Post WS, Blaha MJ, Ibeh C, Whelton SP. Association of aortic valve calcium with dementia and stroke: The Multi-Ethnic Study of Atherosclerosis. Atherosclerosis 2024; 397:117596. [PMID: 38890039 DOI: 10.1016/j.atherosclerosis.2024.117596] [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: 12/21/2023] [Revised: 05/01/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND AIMS Calcific aortic valve disease is associated with increased thrombin formation, platelet activation, decreased fibrinolysis, and subclinical brain infarcts. We examined the long-term association of aortic valve calcification (AVC) with newly diagnosed dementia and incident stroke in the Multi-Ethnic Study of Atherosclerosis (MESA). METHODS AVC was measured using non-contrast cardiac CT at Visit 1. We examined AVC as a continuous (log-transformed) and categorical variable (0, 1-99, 100-299, ≥300). Newly diagnosed dementia was adjudicated using International Classification of Disease codes. Stroke was adjudicated from medical records. We calculated absolute event rates (per 1000 person-years) and multivariable adjusted Cox proportional hazards ratios (HR). RESULTS Overall, 6812 participants had AVC quantified with a mean age of 62.1 years old, 52.9 % were women, and the median 10-year estimated atherosclerotic cardiovascular disease (ASCVD) risk was 13.5 %. Participants with AVC >0 were older and less likely to be women compared to those with AVC=0. Over a median 16-year follow-up, there were 535 cases of dementia and 376 cases of stroke. The absolute risk of newly diagnosed dementia increased in a stepwise pattern with higher AVC scores, and stroke increased in a logarithmic pattern. In multivariable analyses, AVC was significantly associated with newly diagnosed dementia as a log-transformed continuous variable (HR 1.09; 95 % CI 1.04-1.14) and persons with AVC ≥300 had nearly a two-fold higher risk (HR 1.77; 95 % CI 1.14-2.76) compared to those with AVC=0. AVC was associated with an increased risk of stroke after adjustment for age, sex, and race/ethnicity, but not after adjustment for ASCVD risk factors. CONCLUSIONS After multivariable adjustment, AVC >0 was significantly associated with an increased risk of newly diagnosed dementia, but not incident stroke. This suggests that AVC may be an important risk factor for the long-term risk of dementia beyond traditional ASCVD risk factors.
Collapse
Affiliation(s)
- Natalie Marrero
- University of Miami/Jackson Memorial Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kunal Jha
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA; University of Louisville, Division of Cardiology, Louisville, KY, USA
| | - Timothy M Hughes
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Alexander C Razavi
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA; Center for Heart Disease Prevention, Emory School of Medicine, Atlanta, GA, USA
| | - Jelani K Grant
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ellen Boakye
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Khalil Anchouche
- Preventive and Genomic Cardiology, Department of Medicine, McGill University, and the McGill University Health Center Research Institute, Montréal, Québec, Canada
| | - Omar Dzaye
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Matthew J Budoff
- Department of Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - 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, CA, USA
| | - 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, CA, USA
| | - 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, CA, USA
| | | | - Roger S Blumenthal
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Erin D Michos
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - George Thanassoulis
- Preventive and Genomic Cardiology, Department of Medicine, McGill University, and the McGill University Health Center Research Institute, Montréal, Québec, Canada
| | - Wendy S Post
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Michael J Blaha
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Chinwe Ibeh
- Columbia University, Department of Neurology, New York, NY, USA
| | - Seamus P Whelton
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Yao Q, The E, Nedumaran B, Zhai Y, Ao L, Fullerton DA, Meng X. Cathepsin D elevates the fibrocalcific activity in human aortic valve cells through the ERK1/2-Sox9 pathway. Front Cardiovasc Med 2024; 11:1410862. [PMID: 39380629 PMCID: PMC11458440 DOI: 10.3389/fcvm.2024.1410862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
Background Human Aortic valve interstitial cells (AVICs) from calcific aortic valve disease (CAVD)-affected valves exhibit elevated fibrocalcific activity although the underlying mechanism remains incompletely understood. This study aimed to identify endogenous factors that promote aortic valve fibrocalcification. Methods and results Proteomic analysis found increased cathepsin D levels in AVICs from CAVD-affected valves compared to AVICs from normal valves, and this finding was validated by immunoblotting. ELISA assay identified exacerbated release of cathepsin D by AVICs of diseased valves. Recombinant human cathepsin D upregulated the expression of runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP), collagen I and collagen IV in human AVICs, resulting in the deposition of calcium and collagen. Blocking of the ERK1/2-Sox9 signaling pathway markedly reduced the pro-fibrocalcific effect of cathepsin D. Moreover, normal AVICs express and release greater levels of cathepsin D when exposed to soluble matrilin 2. Knockdown of cathepsin D attenuated the fibrocalcific response induced by soluble matrilin 2. Conclusion AVICs of diseased aortic valves produce and release greater levels of cathepsin D that exerts a pro-fibrocalcific effect on AVICs through the ERK1/2-Sox9 pathway. Soluble matrilin 2 up-regulates cathepsin D to elevate AVIC fibrocalcific activity. Over-expression of cathepsin D in the aortic valve may enhance the pathobiological activities in AVICs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xianzhong Meng
- Department of Surgery, University of Colorado, Denver, CO, United States
| |
Collapse
|
5
|
Annabi M, Carter‐Storch R, Zaroui A, Galat A, Oghina S, Kharoubi M, Bezard M, Derumeaux G, Fanen P, Lemonnier F, Poullot E, Itti E, Gallet R, Teiger E, Pibarot P, Damy T, Clavel M. Prevalence, Characteristics, and Impact on Prognosis of Aortic Stenosis in Patients With Cardiac Amyloidosis. J Am Heart Assoc 2024; 13:e034723. [PMID: 38904242 PMCID: PMC11255711 DOI: 10.1161/jaha.124.034723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Cardiac amyloidosis (CA) is frequently found in older patients with aortic stenosis (AS). However, the prevalence of AS among patients with CA is unknown. The objective was to study the prevalence and prognostic impact of AS among patients with CA. METHODS AND RESULTS We conducted a retrospective analysis of a prospective registry comprising 976 patients with native aortic valves who were confirmed with wild type transthyretin amyloid (ATTRwt), hereditary variant transthyretin amyloid (ATTRv), or immunoglobulin light-chain (AL) CA. CA patients' echocardiograms were re-analyzed focusing on the aortic valve. Multivariable Cox regression analysis was performed to assess the mortality risk associated with moderate or greater AS in ATTRwt CA. The crude prevalence of AS among patients with CA was 26% in ATTRwt, 8% in ATTRv, and 5% in AL. Compared with population-based controls, all types of CA had higher age- and sex-standardized rate ratios (SRRs) of having any degree of AS (AL: SRR, 2.62; 95% Confidence Interval (CI) [1.09-3.64]; ATTRv: SRR, 3.41; 95%CI [1.64-4.60]; ATTRwt: SRR, 10.8; 95%CI [5.25-14.53]). Compared with hospital controls, only ATTRwt had a higher SRR of having any degree of AS (AL: SRR, 0.97, 95%CI [0.56-1.14]; ATTRv: SRR, 1.27; 95%CI [0.85-1.44]; ATTRwt: SRR, 4.01; 95%CI [2.71-4.54]). Among patients with ATTRwt, moderate or greater AS was not associated with increased all-cause death after multivariable adjustment (hazard ratio, 0.71; 95%CI [0.42-1.19]; P=0.19). CONCLUSIONS Among patients with CA, ATTRwt but not ATTRv or AL is associated with a higher prevalence of patients with AS compared with hospital controls without CA, even after adjusting for age and sex. In our population, having moderate or greater AS was not associated with a worse outcome in patients with ATTRwt.
Collapse
Affiliation(s)
- Mohamed‐Salah Annabi
- Institut Universitaire de Cardiologie et de PneumologieUniversité LavalQuébecCanada
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
- INSERM U955, Team «Senescence, Metabolism and Cardiovascular Diseases»Paris‐Est Créteil University (UPEC)CréteilFrance
| | - Rasmus Carter‐Storch
- Institut Universitaire de Cardiologie et de PneumologieUniversité LavalQuébecCanada
- Department of CardiologyOdense University HospitalOdenseDenmark
| | - Amira Zaroui
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
- INSERM Unit U955Clinical Epidemiology and Ageing (CEpiA, Paris‐Est Créteil University, Val‐de‐Marne)CréteilFrance
| | - Arnault Galat
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
- INSERM Unit U955Clinical Epidemiology and Ageing (CEpiA, Paris‐Est Créteil University, Val‐de‐Marne)CréteilFrance
| | - Silvia Oghina
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
| | - Mounira Kharoubi
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
- INSERM Unit U955Clinical Epidemiology and Ageing (CEpiA, Paris‐Est Créteil University, Val‐de‐Marne)CréteilFrance
| | - Mélanie Bezard
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
- INSERM Unit U955Clinical Epidemiology and Ageing (CEpiA, Paris‐Est Créteil University, Val‐de‐Marne)CréteilFrance
| | - Geneviève Derumeaux
- INSERM U955, Team «Senescence, Metabolism and Cardiovascular Diseases»Paris‐Est Créteil University (UPEC)CréteilFrance
- AP‐HP, Department of PhysiologyHenri Mondor Hospital, FHU‐SENECCréteilFrance
| | - Pascale Fanen
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
- INSERM Unit U955Clinical Epidemiology and Ageing (CEpiA, Paris‐Est Créteil University, Val‐de‐Marne)CréteilFrance
| | - François Lemonnier
- AP‐HP, Henri Mondor HospitalLymphoid Malignancies UnitCréteilFrance
- Univ Paris Est Créteil, INSERM, IMRBCréteilFrance
| | - Elsa Poullot
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
- Univ Paris Est Créteil, INSERM, IMRBCréteilFrance
- Department of PathologyAP‐HP Henri Mondor HospitalCréteilFrance
| | - Emmanuel Itti
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
- INSERM Unit U955Clinical Epidemiology and Ageing (CEpiA, Paris‐Est Créteil University, Val‐de‐Marne)CréteilFrance
| | - Romain Gallet
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
- INSERM Unit U955Clinical Epidemiology and Ageing (CEpiA, Paris‐Est Créteil University, Val‐de‐Marne)CréteilFrance
| | - Emmanuel Teiger
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
- INSERM Unit U955Clinical Epidemiology and Ageing (CEpiA, Paris‐Est Créteil University, Val‐de‐Marne)CréteilFrance
| | - Philippe Pibarot
- Institut Universitaire de Cardiologie et de PneumologieUniversité LavalQuébecCanada
| | - Thibaud Damy
- Referral Center for Cardiac Amyloidosis, Mondor Amyloidosis Network, GRC Amyloid Research Institute and Cardiology DepartmentAPHP Henri Mondor HospitalCréteilFrance
- INSERM Unit U955Clinical Epidemiology and Ageing (CEpiA, Paris‐Est Créteil University, Val‐de‐Marne)CréteilFrance
| | - Marie‐Annick Clavel
- Institut Universitaire de Cardiologie et de PneumologieUniversité LavalQuébecCanada
| |
Collapse
|
6
|
Perez KA, Deppe DW, Filas A, Singh SA, Aikawa E. Multimodal Analytical Tools to Enhance Mechanistic Understanding of Aortic Valve Calcification. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:539-550. [PMID: 37517686 PMCID: PMC10988764 DOI: 10.1016/j.ajpath.2023.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 08/01/2023]
Abstract
This review focuses on technologies at the core of calcific aortic valve disease (CAVD) and drug target research advancement, including transcriptomics, proteomics, and molecular imaging. We examine how bulk RNA sequencing and single-cell RNA sequencing have engendered organismal genomes and transcriptomes, promoting the analysis of tissue gene expression profiles and cell subpopulations, respectively. We bring into focus how the field is also largely influenced by increasingly accessible proteome profiling techniques. In unison, global transcriptional and protein expression analyses allow for increased understanding of cellular behavior and pathogenic pathways under pathologic stimuli including stress, inflammation, low-density lipoprotein accumulation, increased calcium and phosphate levels, and vascular injury. We also look at how direct investigation of protein signatures paves the way for identification of targetable pathways for pharmacologic intervention. Here, we note that imaging techniques, once a clinical diagnostic tool for late-stage CAVD, have since been refined to address a clinical need to identify microcalcifications using positron emission tomography/computed tomography and even detect in vivo cellular events indicative of early stage CAVD and map the expression of identified proteins in animal models. Together, these techniques generate a holistic approach to CAVD investigation, with the potential to identify additional novel regulatory pathways.
Collapse
Affiliation(s)
- Katelyn A Perez
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel W Deppe
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aidan Filas
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
7
|
Blaser MC, Buffolo F, Halu A, Turner ME, Schlotter F, Higashi H, Pantano L, Clift CL, Saddic LA, Atkins SK, Rogers MA, Pham T, Vromman A, Shvartz E, Sukhova GK, Monticone S, Camussi G, Robson SC, Body SC, Muehlschlegel JD, Singh SA, Aikawa M, Aikawa E. Multiomics of Tissue Extracellular Vesicles Identifies Unique Modulators of Atherosclerosis and Calcific Aortic Valve Stenosis. Circulation 2023; 148:661-678. [PMID: 37427430 PMCID: PMC10527599 DOI: 10.1161/circulationaha.122.063402] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 06/02/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Fewer than 50% of patients who develop aortic valve calcification have concomitant atherosclerosis, implying differential pathogenesis. Although circulating extracellular vesicles (EVs) act as biomarkers of cardiovascular diseases, tissue-entrapped EVs are associated with early mineralization, but their cargoes, functions, and contributions to disease remain unknown. METHODS Disease stage-specific proteomics was performed on human carotid endarterectomy specimens (n=16) and stenotic aortic valves (n=18). Tissue EVs were isolated from human carotid arteries (normal, n=6; diseased, n=4) and aortic valves (normal, n=6; diseased, n=4) by enzymatic digestion, (ultra)centrifugation, and a 15-fraction density gradient validated by proteomics, CD63-immunogold electron microscopy, and nanoparticle tracking analysis. Vesiculomics, comprising vesicular proteomics and small RNA-sequencing, was conducted on tissue EVs. TargetScan identified microRNA targets. Pathway network analyses prioritized genes for validation in primary human carotid artery smooth muscle cells and aortic valvular interstitial cells. RESULTS Disease progression drove significant convergence (P<0.0001) of carotid artery plaque and calcified aortic valve proteomes (2318 proteins). Each tissue also retained a unique subset of differentially enriched proteins (381 in plaques; 226 in valves; q<0.05). Vesicular gene ontology terms increased 2.9-fold (P<0.0001) among proteins modulated by disease in both tissues. Proteomics identified 22 EV markers in tissue digest fractions. Networks of proteins and microRNA targets changed by disease progression in both artery and valve EVs revealed shared involvement in intracellular signaling and cell cycle regulation. Vesiculomics identified 773 proteins and 80 microRNAs differentially enriched by disease exclusively in artery or valve EVs (q<0.05); multiomics integration found tissue-specific EV cargoes associated with procalcific Notch and Wnt signaling in carotid arteries and aortic valves, respectively. Knockdown of tissue-specific EV-derived molecules FGFR2, PPP2CA, and ADAM17 in human carotid artery smooth muscle cells and WNT5A, APP, and APC in human aortic valvular interstitial cells significantly modulated calcification. CONCLUSIONS The first comparative proteomics study of human carotid artery plaques and calcified aortic valves identifies unique drivers of atherosclerosis versus aortic valve stenosis and implicates EVs in advanced cardiovascular calcification. We delineate a vesiculomics strategy to isolate, purify, and study protein and RNA cargoes from EVs entrapped in fibrocalcific tissues. Integration of vesicular proteomics and transcriptomics by network approaches revealed novel roles for tissue EVs in modulating cardiovascular disease.
Collapse
Affiliation(s)
- Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Fabrizio Buffolo
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Arda Halu
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Mandy E. Turner
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Florian Schlotter
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Hideyuki Higashi
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Lorena Pantano
- T H Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Cassandra L. Clift
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Louis A. Saddic
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Boston University School of Medicine, Boston, MA, USA
| | - Samantha K. Atkins
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Maximillian A. Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Tan Pham
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Amélie Vromman
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Eugenia Shvartz
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Galina K Sukhova
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Silvia Monticone
- Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Simon C. Robson
- Center for Inflammation Research, Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Simon C. Body
- Boston University School of Medicine, Boston, MA, USA
| | - Jochen D. Muehlschlegel
- Center for Perioperative Genomics, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sasha A. Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Anousakis-Vlachochristou N, Athanasiadou D, Carneiro KM, Toutouzas K. Focusing on the Native Matrix Proteins in Calcific Aortic Valve Stenosis. JACC Basic Transl Sci 2023; 8:1028-1039. [PMID: 37719438 PMCID: PMC10504402 DOI: 10.1016/j.jacbts.2023.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 09/19/2023]
Abstract
Calcific aortic valve stenosis (CAVS) is a widespread valvular heart disease affecting people in aging societies, primarily characterized by fibrosis, inflammation, and progressive calcification, leading to valve orifice stenosis. Understanding the factors associated with CAVS onset and progression is crucial to develop effective future pharmaceutical therapies. In CAVS, native extracellular matrix proteins modifications, play a significant role in calcification in vitro and in vivo. This work aimed to review the evidence on the alterations of structural native extracellular matrix proteins involved in calcification development during CAVS and highlight its link to deregulated biomechanical function.
Collapse
Affiliation(s)
| | | | - Karina M.M. Carneiro
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Konstantinos Toutouzas
- National and Kapodistrian University of Athens, Medical School, First Department of Cardiology, Athens, Greece
| |
Collapse
|
9
|
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: 3.5] [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
|
10
|
Blaser MC, Kraler S, Lüscher TF, Aikawa E. Network-Guided Multiomic Mapping of Aortic Valve Calcification. Arterioscler Thromb Vasc Biol 2023; 43:417-426. [PMID: 36727519 PMCID: PMC9975082 DOI: 10.1161/atvbaha.122.318334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/18/2023] [Indexed: 02/03/2023]
Abstract
Despite devastating clinical sequelae of calcific aortic valve disease that range from left ventricular remodeling to arrhythmias, heart failure, and early death, the molecular insights into disease initiation and progression are limited and pharmacotherapies remain unavailable. The pathobiology of calcific aortic valve disease is complex and comprehensive studies are challenging valvular calcification is heterogeneous and occurs preferentially on the aortic surface, along a fibrocalcific spectrum. Here, we review efforts to study (epi-)genomic, transcriptomic, proteomic, and metabolomic aspects of aortic valve calcification in combination with network medicine-/systems biology-based strategies to integrate multilayered omics datasets and prioritize druggable targets for experimental validation studies. Ultimately, such holistic approach efforts may open therapeutic avenues that go beyond invasive and costly valve replacement therapy.
Collapse
Affiliation(s)
- Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, CH
| | - Thomas F. Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, CH
- Heart Division, Royal Brompton & Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Zenses AS, Leduc C, Béchard S, Forcillo J, El Haffaf Z, Do QB, Pibarot P, Tournoux F. Amyloid Deposits in a Functionally Unicuspid Stenotic Aortic Valve. CJC Open 2022; 4:1069-1073. [PMID: 36562019 PMCID: PMC9764129 DOI: 10.1016/j.cjco.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/16/2022] [Indexed: 12/25/2022] Open
Abstract
Amyloidosis concomitant to aortic stenosis usually occurs with myocardial infiltration by the transthyretin protein. To our knowledge, this is the first report of localized amyloidosis of indeterminate type in a severely calcified and functionally unicuspid aortic valve. Isolated dystrophic valvular amyloidosis is believed to be related to fibrocalcific valve disease. In light of the literature on this topic, the present case raises new hypotheses on pathophysiology and further supports the contributory role of unusual non-tricuspid valve morphology in the development of dystrophic amyloid, likely secondary to altered hemodynamic stress.
Collapse
Affiliation(s)
- Anne-Sophie Zenses
- Research Centre of the University of Montreal Hospital (CRCHUM), Montreal, Quebec, Canada,Corresponding author: Dr Anne-Sophie Zenses, Centre de recherche du CHUM (CRCHUM) - Pavillon R, 11e étage – 900, rue Saint-Denis, Montréal, Québec H2X 0A9, Canada. Tel.: +1-514-890-8000 ext. 13517.
| | - Charles Leduc
- Department of Pathology and Cell Biology, University of Montreal Hospital (CHUM), Montreal, Quebec, Canada
| | - Stéphanie Béchard
- Department of Cardiology, University of Montreal Hospital (CHUM), Montreal, Quebec, Canada
| | - Jessica Forcillo
- Department of Surgery, University of Montreal Hospital (CHUM), Montreal, Quebec, Canada
| | - Zaki El Haffaf
- Department of Genetic Medicine, University of Montreal Hospital (CHUM), Montreal, Quebec, Canada
| | - Quoc-Bao Do
- Department of Surgery, University of Montreal Hospital (CHUM), Montreal, Quebec, Canada
| | - Philippe Pibarot
- Research Center of the Quebec Heart and Lung Institute, Laval University, Quebec, Quebec, Canada
| | - François Tournoux
- Department of Cardiology, University of Montreal Hospital (CHUM), Montreal, Quebec, Canada
| |
Collapse
|
12
|
Heuschkel MA, Babler A, Heyn J, van der Vorst EPC, Steenman M, Gesper M, Kappel BA, Magne D, Gouëffic Y, Kramann R, Jahnen-Dechent W, Marx N, Quillard T, Goettsch C. Distinct role of mitochondrial function and protein kinase C in intimal and medial calcification in vitro. Front Cardiovasc Med 2022; 9:959457. [PMID: 36204585 PMCID: PMC9530266 DOI: 10.3389/fcvm.2022.959457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction Vascular calcification (VC) is a major risk factor for cardiovascular morbidity and mortality. Depending on the location of mineral deposition within the arterial wall, VC is classified as intimal and medial calcification. Using in vitro mineralization assays, we developed protocols triggering both types of calcification in vascular smooth muscle cells (SMCs) following diverging molecular pathways. Materials and methods and results Human coronary artery SMCs were cultured in osteogenic medium (OM) or high calcium phosphate medium (CaP) to induce a mineralized extracellular matrix. OM induces osteoblast-like differentiation of SMCs-a key process in intimal calcification during atherosclerotic plaque remodeling. CaP mimics hyperphosphatemia, associated with chronic kidney disease-a risk factor for medial calcification. Transcriptomic analysis revealed distinct gene expression profiles of OM and CaP-calcifying SMCs. OM and CaP-treated SMCs shared 107 differentially regulated genes related to SMC contraction and metabolism. Real-time extracellular efflux analysis demonstrated decreased mitochondrial respiration and glycolysis in CaP-treated SMCs compared to increased mitochondrial respiration without altered glycolysis in OM-treated SMCs. Subsequent kinome and in silico drug repurposing analysis (Connectivity Map) suggested a distinct role of protein kinase C (PKC). In vitro validation experiments demonstrated that the PKC activators prostratin and ingenol reduced calcification triggered by OM and promoted calcification triggered by CaP. Conclusion Our direct comparison results of two in vitro calcification models strengthen previous observations of distinct intracellular mechanisms that trigger OM and CaP-induced SMC calcification in vitro. We found a differential role of PKC in OM and CaP-calcified SMCs providing new potential cellular and molecular targets for pharmacological intervention in VC. Our data suggest that the field should limit the generalization of results found in in vitro studies using different calcification protocols.
Collapse
Affiliation(s)
- Marina A. Heuschkel
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Anne Babler
- Institute of Experimental Medicine and Systems Biology, University Hospital, RWTH Aachen, Aachen, Germany
| | - Jonas Heyn
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Emiel P. C. van der Vorst
- Interdisciplinary Center for Clinical Research, Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Marja Steenman
- L’institut Du Thorax, Inserm UMR 1087, CNRS, INSERM, France and Nantes Université, Nantes, France
| | - Maren Gesper
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Ben A. Kappel
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - David Magne
- ICBMS UMR CNRS 5246, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Yann Gouëffic
- Department of Vascular Surgery, Vascular Center, Groupe Hospitalier Paris Saint-Joseph, Paris, France
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems Biology, University Hospital, RWTH Aachen, Aachen, Germany
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | - Willi Jahnen-Dechent
- Biointerface Laboratory, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Nikolaus Marx
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Thibaut Quillard
- L’institut Du Thorax, Inserm UMR 1087, CNRS, INSERM, France and Nantes Université, Nantes, France
- PHY-OS Laboratory, INSERM UMR 1238, Nantes University of Medicine, Nantes, France
| | - Claudia Goettsch
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
13
|
Wu R, Zhou S, Liu M, An H, Wang Z, Liu T. Clinical Significance of miR-21-5p in Predicting Occurrence and Progression of Uremic Vascular Calcification in Patients with End-Stage Renal Disease. Yonsei Med J 2022; 63:252-258. [PMID: 35184427 PMCID: PMC8860934 DOI: 10.3349/ymj.2022.63.3.252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/29/2021] [Accepted: 11/12/2021] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Vascular calcification (VC) is a common complication of end-stage renal disease (ESRD). This study aimed to examine changes in the expression of miR-21-5p in ESRD patients with VC and to explore its clinical value in predicting the occurrence and progression of uremic VC. MATERIALS AND METHODS 120 ESRD patients were divided into patients without VC group (n=38) and patients with VC group (n=82). All patients were followed up for 2 years to evaluate VC progression. qRT-PCR was used to detect serum miR-21-5p levels. Receiver operating characteristic curves were constructed to assess diagnostic value. Kaplan-Meier and log-rank methods were utilized to calculate associations between VC progression and risk factors. RESULTS Serum miR-21-5p levels were significantly higher in ESRD patients with VC than in those without VC and increased progressively with increasing disease severity. Serum miR-21-5p levels were able to distinguish patients with VC from those without VC, with an area under the curve value of 0.883, a sensitivity of 81.7%, and a specificity of 84.2%. After 2 years of follow-up, miR-21-5p expression had increased in patients with worse VC severity, compared with those with stable VC severity. Patients with high miR-21-5p levels were more likely to develop more severe VC, indicating an association between miR-21-5p and VC progression (log-rank p=0.002). Multivariable Cox regression analysis suggested that serum miR-21-5p is an independent predictive factor of VC progression in ESRD patients (hazard ratio=2.064, 95% confidence interval=1.225-3.478, p=0.006). CONCLUSION miR-21-5p is overexpressed in the serum of ESRD patients with VC. Our results suggest that overexpression of miR-21-5p is closely associated with VC progression.
Collapse
Affiliation(s)
- Rong Wu
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Sen Zhou
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Minglong Liu
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Haiqian An
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhe Wang
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Tianxi Liu
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
14
|
Jiang C, Jiang W. AGTR1, PLTP, and SCG2 associated with immune genes and immune cell infiltration in calcific aortic valve stenosis: analysis from integrated bioinformatics and machine learning. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:3787-3802. [PMID: 35341274 DOI: 10.3934/mbe.2022174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Background: Calcific aortic valve stenosis (CAVS) is a crucial cardiovascular disease facing aging societies. Our research attempts to identify immune-related genes through bioinformatics and machine learning analysis. Two machine learning strategies include Least Absolute Shrinkage Selection Operator (LASSO) and Support Vector Machine Recursive Feature Elimination (SVM-RFE). In addition, we deeply explore the role of immune cell infiltration in CAVS, aiming to study the potential therapeutic targets of CAVS and explore possible drugs. Methods: Download three data sets related to CAVS from the Gene Expression Omnibus. Gene set variation analysis (GSVA) looks for potential mechanisms, determines differentially expressed immune-related genes (DEIRGs) by combining the ImmPort database with CAVS differential genes, and explores the functions and pathways of enrichment. Two machine learning methods, LASSO and SVM-RFE, screen key immune signals and validate them in external data sets. Single-sample GSEA (ssGSEA) and CIBERSORT analyze the subtypes of immune infiltrating cells and integrate the analysis with DEIRGs and key immune signals. Finally, the possible targeted drugs are analyzed through the Connectivity Map (CMap). Results: GSVA analysis of the gene set suggests that it is highly correlated with multiple immune pathways. 266 differential genes (DEGs) integrate with immune genes to obtain 71 DEIRGs. Enrichment analysis found that DEIRGs are related to oxidative stress, synaptic membrane components, receptor activity, and a variety of cardiovascular diseases and immune pathways. Angiotensin II Receptor Type 1(AGTR1), Phospholipid Transfer Protein (PLTP), Secretogranin II (SCG2) are identified as key immune signals of CAVS by machine learning. Immune infiltration found that B cells naï ve and Macrophages M2 are less in CAVS, while Macrophages M0 is more in CAVS. Simultaneously, AGTR1, PLTP, SCG2 are highly correlated with a variety of immune cell subtypes. CMap analysis found that isoliquiritigenin, parthenolide, and pyrrolidine-dithiocarbamate are the top three targeted drugs related to CAVS immunity. Conclusion: The key immune signals, immune infiltration and potential drugs obtained from the research play a vital role in the pathophysiological progress of CAVS.
Collapse
Affiliation(s)
- Chenyang Jiang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Weidong Jiang
- Department of Cardiology, Nantong Traditional Chinese Medicine Hospital, Nantong 226001, China
| |
Collapse
|
15
|
Biological knowledge-slanted random forest approach for the classification of calcified aortic valve stenosis. BioData Min 2021; 14:35. [PMID: 34301292 PMCID: PMC8305490 DOI: 10.1186/s13040-021-00269-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/18/2021] [Indexed: 11/29/2022] Open
Abstract
Background Calcific aortic valve stenosis (CAVS) is a fatal disease and there is no pharmacological treatment to prevent the progression of CAVS. This study aims to identify genes potentially implicated with CAVS in patients with congenital bicuspid aortic valve (BAV) and tricuspid aortic valve (TAV) in comparison with patients having normal valves, using a knowledge-slanted random forest (RF). Results This study implemented a knowledge-slanted random forest (RF) using information extracted from a protein-protein interactions network to rank genes in order to modify their selection probability to draw the candidate split-variables. A total of 15,191 genes were assessed in 19 valves with CAVS (BAV, n = 10; TAV, n = 9) and 8 normal valves. The performance of the model was evaluated using accuracy, sensitivity, and specificity to discriminate cases with CAVS. A comparison with conventional RF was also performed. The performance of this proposed approach reported improved accuracy in comparison with conventional RF to classify cases separately with BAV and TAV (Slanted RF: 59.3% versus 40.7%). When patients with BAV and TAV were grouped against patients with normal valves, the addition of prior biological information was not relevant with an accuracy of 92.6%. Conclusion The knowledge-slanted RF approach reflected prior biological knowledge, leading to better precision in distinguishing between cases with BAV, TAV, and normal valves. The results of this study suggest that the integration of biological knowledge can be useful during difficult classification tasks. Supplementary Information The online version contains supplementary material available at 10.1186/s13040-021-00269-4.
Collapse
|
16
|
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: 3.0] [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
|
17
|
Wieczorek E, Ożyhar A. Transthyretin: From Structural Stability to Osteoarticular and Cardiovascular Diseases. Cells 2021; 10:1768. [PMID: 34359938 PMCID: PMC8307983 DOI: 10.3390/cells10071768] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/29/2021] [Accepted: 07/09/2021] [Indexed: 01/10/2023] Open
Abstract
Transthyretin (TTR) is a tetrameric protein transporting hormones in the plasma and brain, which has many other activities that have not been fully acknowledged. TTR is a positive indicator of nutrition status and is negatively correlated with inflammation. TTR is a neuroprotective and oxidative-stress-suppressing factor. The TTR structure is destabilized by mutations, oxidative modifications, aging, proteolysis, and metal cations, including Ca2+. Destabilized TTR molecules form amyloid deposits, resulting in senile and familial amyloidopathies. This review links structural stability of TTR with the environmental factors, particularly oxidative stress and Ca2+, and the processes involved in the pathogenesis of TTR-related diseases. The roles of TTR in biomineralization, calcification, and osteoarticular and cardiovascular diseases are broadly discussed. The association of TTR-related diseases and vascular and ligament tissue calcification with TTR levels and TTR structure is presented. It is indicated that unaggregated TTR and TTR amyloid are bound by vicious cycles, and that TTR may have an as yet undetermined role(s) at the crossroads of calcification, blood coagulation, and immune response.
Collapse
Affiliation(s)
- Elżbieta Wieczorek
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland;
| | | |
Collapse
|
18
|
Vakili D, Radenkovic D, Chawla S, Bhatt DL. Panomics: New Databases for Advancing Cardiology. Front Cardiovasc Med 2021; 8:587768. [PMID: 34041278 PMCID: PMC8142819 DOI: 10.3389/fcvm.2021.587768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 03/01/2021] [Indexed: 11/13/2022] Open
Abstract
The multifactorial nature of cardiology makes it challenging to separate noisy signals from confounders and real markers or drivers of disease. Panomics, the combination of various omic methods, provides the deepest insights into the underlying biological mechanisms to develop tools for personalized medicine under a systems biology approach. Questions remain about current findings and anticipated developments of omics. Here, we search for omic databases, investigate the types of data they provide, and give some examples of panomic applications in health care. We identified 104 omic databases, of which 72 met the inclusion criteria: genomic and clinical measurements on a subset of the database population plus one or more omic datasets. Of those, 65 were methylomic, 59 transcriptomic, 41 proteomic, 42 metabolomic, and 22 microbiomic databases. Larger database sample sizes and longer follow-up are often better suited for panomic analyses due to statistical power calculations. They are often more complete, which is important when dealing with large biological variability. Thus, the UK BioBank rises as the most comprehensive panomic resource, at present, but certain study designs may benefit from other databases.
Collapse
Affiliation(s)
- Dara Vakili
- Imperial College School of Medicine, Imperial College London, London, United Kingdom
| | | | - Shreya Chawla
- Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Deepak L Bhatt
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
19
|
Abstract
Calcific aortic valve disease sits at the confluence of multiple world-wide epidemics of aging, obesity, diabetes, and renal dysfunction, and its prevalence is expected to nearly triple over the next 3 decades. This is of particularly dire clinical relevance, as calcific aortic valve disease can progress rapidly to aortic stenosis, heart failure, and eventually premature death. Unlike in atherosclerosis, and despite the heavy clinical toll, to date, no pharmacotherapy has proven effective to halt calcific aortic valve disease progression, with invasive and costly aortic valve replacement representing the only treatment option currently available. This substantial gap in care is largely because of our still-limited understanding of both normal aortic valve biology and the key regulatory mechanisms that drive disease initiation and progression. Drug discovery is further hampered by the inherent intricacy of the valvular microenvironment: a unique anatomic structure, a complex mixture of dynamic biomechanical forces, and diverse and multipotent cell populations collectively contributing to this currently intractable problem. One promising and rapidly evolving tactic is the application of multiomics approaches to fully define disease pathogenesis. Herein, we summarize the application of (epi)genomics, transcriptomics, proteomics, and metabolomics to the study of valvular heart disease. We also discuss recent forays toward the omics-based characterization of valvular (patho)biology at single-cell resolution; these efforts promise to shed new light on cellular heterogeneity in healthy and diseased valvular tissues and represent the potential to efficaciously target and treat key cell subpopulations. Last, we discuss systems biology- and network medicine-based strategies to extract meaning, mechanisms, and prioritized drug targets from multiomics datasets.
Collapse
Affiliation(s)
- Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, CH
| | - Thomas F. Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, CH
- Heart Division, Royal Brompton & Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|