1
|
Anchouche K, Baass A, Thanassoulis G. Lp(a): A Clinical Review. Clin Biochem 2025; 137:110929. [PMID: 40258460 DOI: 10.1016/j.clinbiochem.2025.110929] [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: 01/06/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
Elevated lipoprotein(a) (Lp[a]) is a genetically determined cardiovascular risk factor, linked to both atherosclerotic cardiovascular disease and aortic stenosis. Elevated Lp(a) is widely prevalent, and consequently, several cardiovascular societies now recommend performing Lp(a) screening at least once in all adults. While there are presently no approved drugs specifically aimed at lowering Lp(a), several promising candidates are currently in the drug development pipeline, and many of these are now undergoing late phase clinical trials. In this comprehensive review, we outline Lp(a) biology and genetics, describe Lp(a)'s relationship to various cardiovascular clinical phenotypes of interest, highlight novel Lp(a)-lowering therapies, and outline what role these may have in future clinical practice.
Collapse
Affiliation(s)
- Khalil Anchouche
- McGill University Health Centre and Research Institute, Montreal, QC, Canada; McGill University, Montreal, QC, Canada
| | - Alexis Baass
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montreal, QC, Canada; Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Montreal, QC, Canada
| | - George Thanassoulis
- McGill University Health Centre and Research Institute, Montreal, QC, Canada; McGill University, Montreal, QC, Canada.
| |
Collapse
|
2
|
Goldfarb JW, Wang L, Chen L, Khan JM, Ali ZA, Khalique OK. Anatomically indexed aortic valve calcium score more accurately predicts transaortic peak velocities and gradients compared to radiomics features in patients with severe aortic stenosis. J Cardiovasc Comput Tomogr 2025:S1934-5925(25)00093-0. [PMID: 40414767 DOI: 10.1016/j.jcct.2025.05.005] [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: 03/15/2025] [Revised: 05/07/2025] [Accepted: 05/11/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND This study examines the use of computed tomography (CT)-derived calcified and non-calcified aortic valve (AV) features, including radiomics-based quantitative imaging biomarkers, for predicting aortic stenosis (AS) severity and evaluating sex-specific differences. METHODS In this retrospective, single-center study, 270 patients (50 % female) with severe AS and preserved left ventricular ejection fraction were assessed in the primary-cohort using both echocardiography and CT angiography. Correlation-based feature selection and Lasso regression were employed to refine the most predictive features. Logistic regression models were developed for the overall-, male-, and female-cohorts, evaluating the predictive power of calcified and non-calcified AV features for identification of peak aortic valve jet velocity (PAV) ≥ 4 m/s and mean pressure gradient (MPG) ≥ 40 mmHg. RESULTS Statistical methods reduced the initial 44 CT variables to 13 in overall-cohort models, 10 in male-cohort models, and 12 in female-cohort models. The inclusion of these additional features significantly improved model performance compared to using the AV calcium score (AVCS) alone or its indexed variants. Indexing the AVCS to anatomical features resulted in modest improvements, with ROC-AUC values increasing from 0.71 (non-indexed Agatston) to 0.78 (indexed to sinus-of-Valsalva (SOV) volume) for PAV prediction in overall-cohort models. However, incorporating the full set of selected features further enhanced predictive accuracy, raising the ROC-AUC to 0.80. Similar trends were observed for MPG, with the best-performing models achieving a ROC-AUC of 0.84 compared to 0.73 using the non-indexed AVCS score alone. The male- and female-cohort models demonstrated similar improvements, with sex-specific feature sets significantly enhancing performance beyond indexed AVCSs. CONCLUSIONS Indexing the AVCS to SOV volume and aortic annulus area enhances the predictive power of AS severity models, though incorporating a broader set of calcified and non-calcified CT features provides the greatest improvement. These findings underscore the importance of considering anatomical and sex-specific differences in the assessment of AS.
Collapse
Affiliation(s)
- James W Goldfarb
- Division of Cardiovascular Imaging, St Francis Hospital and Heart Center, Roslyn, NY, USA.
| | - Lin Wang
- Division of Cardiovascular Imaging, St Francis Hospital and Heart Center, Roslyn, NY, USA
| | - Lu Chen
- Division of Cardiovascular Imaging, St Francis Hospital and Heart Center, Roslyn, NY, USA
| | - Jaffar M Khan
- Department of Cardiology, St Francis Hospital and Heart Center, Roslyn, NY, USA; New York Institute of Technology, Old Westbury, NY, USA
| | - Ziad A Ali
- Department of Cardiology, St Francis Hospital and Heart Center, Roslyn, NY, USA; New York Institute of Technology, Old Westbury, NY, USA
| | - Omar K Khalique
- Division of Cardiovascular Imaging, St Francis Hospital and Heart Center, Roslyn, NY, USA; New York Institute of Technology, Old Westbury, NY, USA
| |
Collapse
|
3
|
Zamani P, Houessou U, Manikpurage HD, Li Z, Dahmene M, Gaudreault N, Dagenais F, Clavel MA, Pibarot P, Arsenault BJ, Mathieu P, Bossé Y, Thériault S. Aortic valve-specific genes dysregulated in calcific aortic valve stenosis as potential biomarkers and therapeutic targets. HGG ADVANCES 2025; 6:100448. [PMID: 40329539 DOI: 10.1016/j.xhgg.2025.100448] [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: 11/01/2024] [Revised: 04/30/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
Calcific aortic valve stenosis (CAVS) is the most frequent heart valve disease. Elucidating specific gene expression patterns in the aortic valve could provide new insights for understanding disease pathophysiology. We used local RNA sequencing data from 500 explanted human aortic valves to identify aortic valve-specific genes and compared their expression according to disease status and CAVS severity. We identified 100 specific protein-coding genes in the aortic valve compared to 45 other tissues from the Genotype-Tissue Expression (GTEx) project. Among them, 38 were differentially expressed in CAVS. Ten had a gradient of expression between severity levels and were central in a protein-protein interaction network, most of which were involved in extracellular matrix regulation or inflammation. Among the aortic valve-specific genes, four of the corresponding proteins had a significantly different plasma level in individuals with CAVS. These findings represent a robust foundation for the development of specific biomarkers and therapies for CAVS.
Collapse
Affiliation(s)
- Pardis Zamani
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada
| | - Ursula Houessou
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada
| | - Hasanga D Manikpurage
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada
| | - Zhonglin Li
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada
| | - Manel Dahmene
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada
| | - Nathalie Gaudreault
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada
| | - François Dagenais
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada; Department of Surgery, Université Laval, Quebec City, QC, Canada
| | - Marie-Annick Clavel
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada; Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Philippe Pibarot
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada; Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Benoit J Arsenault
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada; Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Patrick Mathieu
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada; Department of Surgery, Université Laval, Quebec City, QC, Canada
| | - Yohan Bossé
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada; Department of Molecular Medicine, Université Laval, Quebec City, QC, Canada
| | - Sébastien Thériault
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, QC, Canada; Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
4
|
D’Alonzo M, Massimo B, Fiore A, Capussela J, Abrami G, Folliguet T, Muneretto C. The role of the Prognostic Nutritional Index in predicting survival and rehospitalization after surgical aortic valve replacement. Indian J Thorac Cardiovasc Surg 2025; 41:532-540. [PMID: 40247965 PMCID: PMC12000483 DOI: 10.1007/s12055-024-01891-7] [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: 08/11/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 04/19/2025] Open
Abstract
Purpose The Prognostic Nutritional Index (PNI), calculated using serum albumin levels and blood lymphocyte count, reflects a patient's nutritional and immune status. It is commonly used as a prognostic tool following oncological surgery and in certain cardiovascular conditions. This study aims to assess whether the PNI can also serve as a prognostic indicator in patients undergoing surgical aortic valve replacement (SAVR). Methods A total of 471 low-risk patients with EuroSCORE II (European System for Cardiac Operative Risk Evaluation II) of ≤4%, who underwent isolated SAVR, were retrospectively analysed. Patients were divided into two groups based on their PNI values (cut-off, 46.75). Outcomes such as length of hospital stay, 30-day mortality, 1-year survival, and rehospitalization rates were compared between the groups. Results The Low PNI group consisted of 116 patients, while the High PNI group included 355 patients. The latter were younger, but both groups had comparable comorbidities. All patients underwent SAVR with a bioprosthesis. There was no significant difference in 30-day mortality between the groups (Low PNI, 2.6% vs. High PNI, 0.9%, p = 0.162). However, the 1-year survival rate was significantly lower in the Low PNI group (Low PNI, 5.2 ± 4.1% vs. High PNI, 1.7 ± 1.3%, p = 0.039). Additionally, the 1-year rehospitalization rate was significantly higher in the Low PNI group (Low PNI, 13.8 ± 6.3% vs. High PNI, 7.7 ± 2.7%, p = 0.040). Multivariate analysis identified PNI as a protective factor, while mechanical ventilation was associated with increased risk of death or rehospitalization at 1-year after SAVR. Conclusions The PNI is an inexpensive, accessible, and reliable tool that can be easily integrated into existing risk stratification scores for SAVR. Graphical Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s12055-024-01891-7.
Collapse
Affiliation(s)
- Michele D’Alonzo
- Cardiac Surgery Department, “Spedali Civili” Hospital, University of Brescia, Brescia, Italy
- Cardiac Surgery Department, “Henri Mondor” Hospital, University of Paris East, Creteil, France
- Department of Cardiac and Thoracic Surgery, 11 Stage, CHU Henri Mondor, 51 Avenue du Maréchal de Lattre de Tassigny, 94000 Créteil, France
| | - Baudo Massimo
- Department of Cardiac Surgery Research, Lankenau Institute for Medical Research, Main Line Health, Wynnewood, PA USA
| | - Antonio Fiore
- Cardiac Surgery Department, “Henri Mondor” Hospital, University of Paris East, Creteil, France
| | - Jacopo Capussela
- Cardiac Surgery Department, “Spedali Civili” Hospital, University of Brescia, Brescia, Italy
| | - Gianluca Abrami
- Cardiac Surgery Department, “Spedali Civili” Hospital, University of Brescia, Brescia, Italy
| | - Thierry Folliguet
- Cardiac Surgery Department, “Henri Mondor” Hospital, University of Paris East, Creteil, France
| | - Claudio Muneretto
- Cardiac Surgery Department, “Spedali Civili” Hospital, University of Brescia, Brescia, Italy
| |
Collapse
|
5
|
Xian G, Huang R, Hu D, Xu M, Chen Y, Ren H, Xu D, Zeng Q. Interleukin-37 attenuates aortic valve lesions by inhibiting N6-methyladenosine-mediated interleukin-1 receptor-associated kinase M degradation. Cardiovasc Res 2025; 121:492-506. [PMID: 39913240 DOI: 10.1093/cvr/cvaf012] [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: 03/05/2024] [Revised: 09/11/2024] [Accepted: 11/12/2024] [Indexed: 02/26/2025] Open
Abstract
AIMS Calcific aortic valve disease (CAVD) has become an increasingly important global medical problem without effective pharmacological intervention. Accumulating evidence indicates that aortic valve calcification is driven by inflammation. Interleukin-1 receptor-associated kinase M (IRAK-M) is a well-known negative regulator of inflammation, but its role in CAVD remains unclear. METHODS AND RESULTS Here, we stimulated aortic valve interstitial cells (AVICs) with low-dose lipopolysaccharide (LPS) to mimic the inflammatory response in aortic valve calcification and observed the expression pattern of IRAK-M. Furthermore, we generated IRAK-M-/- mice to explore the effect of IRAK-M deficiency on the aortic valve in vivo. Additionally, overexpression and knockdown experiments were performed to verify the role of IRAK-M in AVICs. Methylated RNA immunoprecipitation-quantitative polymerase chain reaction was used to detect the N6-methyladenosine (m6A) level of IRAK-M, and recombinant interleukin (IL)-37-treated AVICs were used to determine the regulatory relationship between IL-37 and IRAK-M. We found that IRAK-M expression was upregulated in the early stages of inflammation as part of a negative feedback mechanism to modulate the immune response. However, persistent inflammation increased overall m6A levels, ultimately leading to reduced IRAK-M expression. In vivo, IRAK-M-/- mice exhibited a propensity for aortic valve thickening and calcification. Overexpression and knockdown experiments showed that IRAK-M inhibited inflammation and osteogenic responses in AVICs. In addition, IL-37 restored IRAK-M expression by inhibiting m6A-mediated IRAK-M degradation to suppress inflammation and aortic valve calcification. CONCLUSION Our findings confirm that inflammation and epigenetic modifications synergistically regulate IRAK-M expression. Moreover, IRAK-M represents a potential target for mitigating aortic valve calcification. Meanwhile, IL-37 exhibited inhibitory effects on CAVD development both in vivo and in vitro, giving us hope that CAVD can be treated with drugs rather than surgery.
Collapse
Affiliation(s)
- Gaopeng Xian
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Rong Huang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Dongtu Hu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Minhui Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Yangchao Chen
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Hao Ren
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| |
Collapse
|
6
|
Liu F, Cai H. Diabetes and calcific aortic valve disease: implications of glucose-lowering medication as potential therapy. Front Pharmacol 2025; 16:1583267. [PMID: 40356984 PMCID: PMC12066769 DOI: 10.3389/fphar.2025.1583267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Calcific aortic valve disease (CAVD) is a progressive disease, of which the 2-year mortality is >50% for symptomatic disease. However, there are currently no pharmacotherapies to prevent the progression of CAVD unless transcatheter or surgical aortic valve replacement is performed. The prevalence of diabetes among CAVD has increased rapidly in recent decades, especially among those undergoing aortic valve replacement. Diabetes and its comorbidities, such as hypertension, hyperlipidemia, chronic kidney disease and ageing, participated jointly in the initiation and progression of CAVD, which increased the management complexity in patients with CAVD. Except from hyperglycemia, the molecular links between diabetes and CAVD included inflammation, oxidative stress and endothelial dysfunction. Traditional cardiovascular drugs like lipid-lowering agents and renin-angiotensin system blocking drugs have proven to be unsuccessful in retarding the progression of CAVD in clinical trials. In recent years, almost all kinds of glucose-lowering medications have been specifically assessed for decelerating the development of CAVD. Based on the efficacy for atherosclerotic cardiovascular disease and CAVD, this review summarized current knowledge about glucose-lowering medications as promising treatment options with the potential to retard CAVD.
Collapse
Affiliation(s)
| | - Haipeng Cai
- Department of Cardiology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| |
Collapse
|
7
|
Chen J, Ma C, Li J, Niu X, Fan Y. Collagen-mediated cardiovascular calcification. Int J Biol Macromol 2025; 301:140225. [PMID: 39864707 DOI: 10.1016/j.ijbiomac.2025.140225] [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: 11/14/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
Cardiovascular calcification is a pathological process commonly observed in the elderly. Based on the location of the calcification, cardiovascular calcification can be classified into two main types: vascular calcification and valvular calcification. Collagen plays a critical role in the development of cardiovascular calcification lesions. The content and type of collagen are the result of a dynamic balance between synthesis and degradation. Unregulated processes can lead to adverse outcomes. During cardiovascular calcification, collagen not only serves as a scaffold for ectopic mineral deposition but also acts as a signal transduction pathway that mediates calcification by guiding the aggregation and nucleation of matrix vesicles and promoting the proliferation, migration and phenotypic changes of cells involved in the lesion. This review provides an overview of collagen subtypes in the cardiovascular system under physiological conditions and discusses their distribution. Additionally, we introduce pathological changes and mechanisms of collagen in blood vessels and heart valves. Then, the formation process and characteristic stages of cardiovascular calcification are described. Finally, we highlight the role of collagen in cardiovascular calcification, explore strategied for mediating calcification, and suggest potential directions for future research.
Collapse
Affiliation(s)
- Junlin Chen
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Chunyang Ma
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Jinyu Li
- Department of Orthopedic, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, China.
| | - Xufeng Niu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China.
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; School of Engineering Medicine, Beihang University, Beijing 100083, China.
| |
Collapse
|
8
|
Yu YL, Jiang Q. Advances in Pathophysiological Mechanisms of Degenerative Aortic Valve Disease. Cardiol Res 2025; 16:86-101. [PMID: 40051666 PMCID: PMC11882237 DOI: 10.14740/cr2012] [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: 11/22/2024] [Accepted: 02/06/2025] [Indexed: 03/09/2025] Open
Abstract
Degenerative aortic valve disease (DAVD) represents the most prevalent valvular ailment among the elderly population, which significantly impacts their physical well-being and potentially poses a lethal risk. Currently, the underlying mechanisms of DAVD remain incompletely understood. While the progression of this disease has traditionally been attributed to degenerative processes associated with aging, numerous recent studies have revealed that heart valve calcification may represent a response of valve tissue to a specific initiating factor, involving the interaction of various genes and signaling pathways. This calcification process is further influenced by a range of factors, including genetic predispositions, environmental exposures, metabolic factors, and hemodynamic considerations. Based on the identification of its biomarkers, potential innovative therapeutic targets are proposed for the treatment of this complex condition. The present article primarily delves into the underlying pathophysiological mechanisms and advancements in diagnostic and therapeutic modalities pertaining to this malady.
Collapse
Affiliation(s)
- Ya Lu Yu
- School of Medicine, University of Electronic Science and Technology of China, 610072 Chengdu, Sichuan, China
| | - Qin Jiang
- School of Medicine, University of Electronic Science and Technology of China, 610072 Chengdu, Sichuan, China
- Department of Cardiac Surgery, Sichuan Provincial People’s Hospital, Affiliated Hospital of University of Electronic Science and Technology, 610072 Chengdu, Sichuan, China
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, 610072 Chengdu, Sichuan, China
| |
Collapse
|
9
|
Sádaba A, Garaikoetxea M, Tiraplegui C, San-Ildefonso-García S, Goñi-Olóriz M, Fernández-Celis A, Martín-Núñez E, Castillo P, Álvarez V, Sádaba R, Jover E, Navarro A, López-Andrés N. The Presence of Adipose Tissue in Aortic Valves Influences Inflammation and Extracellular Matrix Composition in Chronic Aortic Regurgitation. Int J Mol Sci 2025; 26:3128. [PMID: 40243913 PMCID: PMC11989201 DOI: 10.3390/ijms26073128] [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: 01/16/2025] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Adipose tissue is present in aortic valves (AVs). Valve interstitial cells (VICs) could differentiate into adipogenic lineages. We here characterize whether the presence of adipose tissue in the AV influences inflammation and extracellular matrix (ECM) composition in patients with aortic regurgitation (AR). A total of 144 AVs were analyzed by histological and molecular techniques. We performed discovery studies using Olink Proteomics® technology in 40 AVs (N = 16 without and N = 24 with adipose tissue). In vitro, human white adipocytes (HWAs) or VICs were cultured with adipogenic media and co-cultured with control VICs. Of Avs, 67% presented white-like adipocytes within the spongiosa. Discovery studies revealed increased levels of inflammatory and ECM molecules in AVs containing adipocytes. Interestingly, the presence of adipocytes was associated with greater AV thickness, higher inflammation, and ECM remodeling, which was characterized by increased proinflammatory molecules, collagen, fibronectin, proteoglycans, and metalloproteinases. AV thickness positively correlated with markers of adipose tissue, inflammation, and ECM. In vitro, adipocyte-like VICs expressed higher levels of adipocyte markers, increased cytokines, fibronectin, decorin, and MMP-13. Analyses of supernatants from co-cultured control VICs with HWA or adipocyte-like VICs showed higher expression of inflammatory mediators, collagen type I, proteoglycans, and metalloproteinases. AVs presenting adipocytes were thicker and exhibited changes characterized by increased inflammation accompanied by aberrant expression of collagen, proteoglycans, and metalloproteinases. VICs could differentiate into adipogenic pathway, affect neighbor VICs, and contribute to inflammation, collagen and proteoglycan accumulation, as well as to metalloproteinases secretion. In summary, the presence of adipose tissue in AV could modify its composition, favoring inflammation and remodeling with an impact on AV thickness.
Collapse
Affiliation(s)
- Alba Sádaba
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Mattie Garaikoetxea
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Carolina Tiraplegui
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Susana San-Ildefonso-García
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Miriam Goñi-Olóriz
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Ernesto Martín-Núñez
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Paula Castillo
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Virginia Álvarez
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Rafael Sádaba
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Eva Jover
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Adela Navarro
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (A.S.); (M.G.); (C.T.); (S.S.-I.-G.); (M.G.-O.); (A.F.-C.); (E.M.-N.); (P.C.); (V.Á.); (R.S.); (E.J.)
- French-Clinical Research Infrastructure Network (F-CRIN) Cardiovascular and Renal Clinical Trialists (INI-CRCT), 54500 Nancy, France
| |
Collapse
|
10
|
He Y, Yang YJ, Wang ZJ, Tang L. Bibliometric analysis of treatment modalities in calcific aortic valve stenosis. Front Pharmacol 2025; 16:1431311. [PMID: 40183078 PMCID: PMC11966050 DOI: 10.3389/fphar.2025.1431311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 02/04/2025] [Indexed: 04/05/2025] Open
Abstract
Background Calcific aortic valve stenosis (CAVS) is a common cardiovascular condition associated with significant adverse events and high mortality rates. Unfortunately, there are currently no effective pharmacological treatments to halt or prevent its progression. Through our analysis of global trends and treatment strategies, we have identified valuable insights and promising therapeutic possibilities. Additionally, by utilizing bibliometric and visualization techniques, we provide a comprehensive overview of the current research landscape in this field. Method According to our design idea, we used the Web of Science database to select publications on aortic stenosis and related treatments. Through our VOSviewer and CiteSpace analysis, a total of 787 articles have been analyzed by September 2024. We also summarize and explore the most prolific authors, the most prolific countries, and the journals and institutions that publish the most articles. Results A visual analysis of the collected articles reveals that Canada and the United States have the highest publication volumes in this field. Among institutions, Harvard University in the U.S. leads in publication count, followed by Laval University in Canada and the University of California in the U.S. The top three research hotspots are stenosis, calcification, and progression. The journal with the highest number of publications in this area is Frontiers in Cardiovascular Medicine, followed by Catheterization and Cardiovascular Interventions and Arteriosclerosis, Thrombosis and Vascular Biology. Furthermore, research on CAVS treatment spans various directions and focuses, including therapeutic approaches, pathogenesis, and diagnostic methods. Conclusion Research into CAVS treatment has advanced significantly over the years. While interventional and surgical valve replacement remains the mainstay treatments for aortic stenosis, they are insufficient to fully meet the needs of the patient. Emerging priorities now focus on improving diagnostics, exploring innovative therapies, uncovering disease mechanisms, and developing novel drugs. These findings highlight the evolving demands in this field and underscore the need for continued research to address these challenges.
Collapse
Affiliation(s)
| | | | | | - Liang Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Hénaut L, Candellier A, Huish S, Issa N, Sinha S, Massy ZA. Valvular calcification in chronic kidney disease: new insights from recent clinical and preclinical studies. Clin Kidney J 2025; 18:i27-i45. [PMID: 40083956 PMCID: PMC11903095 DOI: 10.1093/ckj/sfae421] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Indexed: 03/16/2025] Open
Abstract
Valvular calcification, developing either in the mitral or the aortic valve, is highly prevalent in patients suffering from chronic kidney disease (CKD), in whom their presence correlates with higher cardiovascular and all-cause mortality risk. To date, the exact mechanisms that promote heart valve calcification remain unclear, and none of the treatments tested so far have shown efficacy in preventing valvular fibrocalcific remodelling. It is therefore essential to improve our understanding of the mechanisms involved in the pathological process if we are to find new, effective therapies. The purpose of this review is to (i) summarize our current knowledge of the mechanisms by which CKD and related therapies affect valvular cell activity, (ii) present the latest therapeutic targets identified in preclinical studies, and (iii) discuss the most recent clinical trials evaluating the efficacy of therapies aimed at preventing valvular calcification in CKD.
Collapse
Affiliation(s)
- Lucie Hénaut
- MP3CV Laboratory, UR UPJV 7517, CURS, University of Picardie Jules Verne, Amiens, France
| | - Alexandre Candellier
- MP3CV Laboratory, UR UPJV 7517, CURS, University of Picardie Jules Verne, Amiens, France
| | - Sharon Huish
- Department of Nephrology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
- Donal O'Donoghue Renal Research Centre, Northern Care Alliance NHS Foundation Trust, Salford, UK
| | - Nervana Issa
- MP3CV Laboratory, UR UPJV 7517, CURS, University of Picardie Jules Verne, Amiens, France
| | - Smeeta Sinha
- Donal O'Donoghue Renal Research Centre, Northern Care Alliance NHS Foundation Trust, Salford, UK
- Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Ziad A Massy
- INSERM Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Saclay University and Versailles Saint-Quentin-en-Yvelines University (UVSQ), Villejuif, France
- Association pour l'Utilisation du Rein Artificiel dans la région parisienne (AURA), Paris, Paris, France
- Ambroise Paré University Hospital, APHP, Department of Nephrology Boulogne-Billancourt/Paris, Boulogne-Billancourt/Paris, France
| |
Collapse
|
12
|
Claes M, Pollari F, Mamdooh H, Fischlein T. Baseline CT-Based Risk Factors for Atrioventricular Block after Surgical AVR. Thorac Cardiovasc Surg 2025; 73:117-125. [PMID: 36914161 DOI: 10.1055/a-2052-8848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
BACKGROUND We aimed to evaluate the impact of membranous interventricular septum (MIS) length and calcifications of the native aortic valve (AV), via preoperative multidetector computed tomography (MDCT) scan, on postoperative atrioventricular block III (AVB/AVB III) and permanent pacemaker implantation in surgical aortic valve replacement (SAVR). METHODS We retrospectively analyzed preoperative contrast-enhanced MDCT scans and procedural outcomes of patients affected by AV stenosis who underwent SAVR at our center (June 2016-December 2019). The study population was divided into two groups (AVB and non-AVB), and variables were compared with a Mann-Whitney's U-test or chi-square test. Data were further analyzed using point biserial correlation and logistic regression. RESULTS A total of 155 (38% female) patients (mean age of 71.2 ± 6 years) were enrolled in our study: conventional stented bioprosthesis (N = 99) and sutureless prosthesis (N = 56) were implanted. A postoperative AVB III was observed in 11 patients (7.1%). AVB patients had significant greater calcifications in left coronary cusp (LCC) -AV (non-AVB = 181.0 mm3 [82.7-316.9] vs. AVB = 424.8 mm3 [115.9-563.2], p = 0.044), LCC left ventricular outflow tract (LVOT) (non-AVB = 2.1 mm3 [0-20.1] vs. AVB = 26.0 mm3 [0.1-138.0], p = 0.048), right coronary cusp (RCC) -LVOT (non-AVB = 0 mm3 [0-3.5] vs. AVB = 2.8 mm3 [0-29.0], p = 0.039), and consequently in total LVOT (non-AVB = 2.1 mm3 [0-20.1] vs. AVB = 26.0 mm3 [0.1-138.0], p = 0.02), while their MIS was significantly shorter than in non-AVB patients (non-AVB = 11.3 mm [9.9-13.4] vs. AVB = 9.44 mm [6.98-10.5]; p=0.014)). Partially, these group differences correlated positively (LCC -AV, r = 0.201, p = 0.012; RCC -LVOT, r = 0.283, p ≤ 0.001) or negatively (MIS length, r = -0.202, p = 0.008) with new-onset AVB III. CONCLUSION We recommend including an MDCT in preoperative diagnostic testing for all patients undergoing surgical AVR for further risk stratification.
Collapse
Affiliation(s)
- Marie Claes
- Department of Cardiac Surgery, Klinikum Nürnberg, Paracelsus Medical University, Nuremberg, Germany
| | - Francesco Pollari
- Department of Cardiac Surgery, Klinikum Nürnberg, Paracelsus Medical University, Nuremberg, Germany
| | - Hazem Mamdooh
- Department of Cardiac Surgery, Klinikum Nürnberg, Paracelsus Medical University, Nuremberg, Germany
| | - Theodor Fischlein
- Department of Cardiac Surgery, Klinikum Nürnberg, Paracelsus Medical University, Nuremberg, Germany
| |
Collapse
|
13
|
Allgayer R, Kabir RF, Bergeron A, Demers P, Mantovani D, Cerruti M. A collagen-based laboratory model to mimic sex-specific features of calcific aortic valve disease. Acta Biomater 2025; 194:204-218. [PMID: 39864641 DOI: 10.1016/j.actbio.2025.01.034] [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: 09/30/2024] [Revised: 01/08/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
Calcific aortic valve disease (CAVD) shows in the deposition of calcium phosphates in the collagen-rich layer of the valve leaflets. This stiffens the leaflets and eventually leads to heart failure. Recent research suggests that CAVD follows sex-specific pathways: at the same severity of the disease, women tend to have fewer and less crystalline calcifications, and the phases of their calcifications are decidedly different than those of men; namely, dicalcium phosphate dihydrate (DCPD) - one of the mineral phases in CAVD - occurs almost exclusively in females. Furthermore, the morphologies of heart valve calcifications might be sex-specific, but the sex dependence of the morphologies has not been systematically investigated. Herein, we first show that male CAVD patients have more compact and less fibrous calcifications than females, establishing sex-dependent morphological features of heart valve calcification. We then build a model that recapitulates the sex differences of the calcifications in CAVD, which is based on a collagen gel that we calcify in simulated body fluid with varying fetuin A concentrations. With increasing fetuin A concentration, the calcifications become less crystalline and more fibrous, and more DCPD deposits in the collagen matrix, resembling the physicochemical characteristics of the calcifications in female valves. Lower fetuin A concentrations give rise to a model that replicates male-specific mineral characteristics. The models could be used to develop sex-specific detection and treatment methods for CAVD. STATEMENT OF SIGNIFICANCE: Although calcific aortic valve disease (CAVD) affects ∼10 million people globally, researchers have only discovered recently that the disease follows sex-specific pathways, and many of its sex-specific features remain unknown. To further our understanding of sex differences in CAVD and to develop better detection and treatment methods, there is an urgent need to establish models for CAVD that account for its sex-specific manifestations. In this study, we first show that CAVD calcifications in men and women take on different morphologies. Second, we present a model that can replicate physicochemical calcification characteristics of male or female valves, including morphology, and that can help to develop sex-specific detection and treatment methods for CAVD.
Collapse
Affiliation(s)
- Raphaela Allgayer
- Department of Mining and Materials Engineering, McGill University, 3610 Rue University, Montreal, QC H3A 0C5, Canada
| | - Reefah Fahmida Kabir
- Department of Bioengineering, McGill University, 3480 Rue University, Montreal, QC H2A 0E9, Canada
| | - Alexandre Bergeron
- Department of Surgery, Montreal Heart Institute, 5000 Rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Philippe Demers
- Department of Surgery, Montreal Heart Institute, 5000 Rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, CRC-I, Department of Min-Met-Materials Engineering & Research Center of CHU de Quebec, Division of Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Marta Cerruti
- Department of Mining and Materials Engineering, McGill University, 3610 Rue University, Montreal, QC H3A 0C5, Canada.
| |
Collapse
|
14
|
Mo B, Ding Y, Ji Q. NLRP3 inflammasome in cardiovascular diseases: an update. Front Immunol 2025; 16:1550226. [PMID: 40079000 PMCID: PMC11896874 DOI: 10.3389/fimmu.2025.1550226] [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] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Cardiovascular disease (CVD) continues to be the leading cause of mortality worldwide. The nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome is involved in numerous types of CVD. As part of innate immunity, the NLRP3 inflammasome plays a vital role, requiring priming and activation signals to trigger inflammation. The NLRP3 inflammasome leads both to the release of IL-1 family cytokines and to a distinct form of programmed cell death called pyroptosis. Inflammation related to CVD has been extensively investigated in relation to the NLRP3 inflammasome. In this review, we describe the pathways triggering NLRP3 priming and activation and discuss its pathogenic effects on CVD. This study also provides an overview of potential therapeutic approaches targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Binhai Mo
- People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yudi Ding
- First People’s Hospital of Nanning, Nanning, Guangxi, China
| | - Qingwei Ji
- People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
15
|
Blaser MC, Bäck M, Lüscher TF, Aikawa E. Calcific aortic stenosis: omics-based target discovery and therapy development. Eur Heart J 2025; 46:620-634. [PMID: 39656785 PMCID: PMC11825147 DOI: 10.1093/eurheartj/ehae829] [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: 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
|
16
|
Blaser MC, Aikawa E. Disease Drivers in Aortic Stenosis vs Atherosclerosis. JAMA Cardiol 2025; 10:109-111. [PMID: 39504008 DOI: 10.1001/jamacardio.2024.3749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Affiliation(s)
- Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
17
|
Hafiane A, Pisaturo A, Favari E, Bortnick AE. Atherosclerosis, calcific aortic valve disease and mitral annular calcification: same or different? Int J Cardiol 2025; 420:132741. [PMID: 39557087 DOI: 10.1016/j.ijcard.2024.132741] [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: 07/09/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
There are similarities in the pathophysiologic mechanisms of atherosclerosis, calcific aortic valve disease (CAVD) and mitral annular calcification (MAC), however, medical treatment to slow or stop the progression of CAVD or MAC has been more elusive as compared to atherosclerosis. Atherosclerosis and CAVD share common demographic, clinical, protein, and genetic factors even more so than with MAC, which supports the possibility of shared medical therapies, though abrogating calcific extracellular vesicle shedding could be a common target for all three conditions. Herein, we summarize the overlapping and distinct pathways for further investigation, as well as key areas where additional research is needed.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Institute of the McGill University Health Centre, McGill University, Montreal, Canada.
| | | | - Elda Favari
- Department of Food and Drug, University of Parma, Parma, Italy.
| | - Anna E Bortnick
- Department of Medicine, Divisions of Cardiology and Geriatrics, and Department of Obstetrics & Gynecology and Women's Health, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, United States of America.
| |
Collapse
|
18
|
Selig JI, Sugimura Y, Katahira S, Polidori M, Jacobi LA, Medovoj O, Betke S, Barth M, Lichtenberg A, Akhyari P, Minol JP. The Focal Induction of Reactive Oxygen Species in Rats as a Trigger of Aortic Valve Degeneration. Antioxidants (Basel) 2024; 13:1570. [PMID: 39765897 PMCID: PMC11673780 DOI: 10.3390/antiox13121570] [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: 10/30/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Degenerative aortic valve disease (DAVD) is a multifactorial process. We developed an animal model to analyze the isolated, local effect of reactive oxygen species (ROS) on its pathophysiology. METHODS We utilized a photodynamic reaction (PDR) as a source of ROS in the aortic valve by aiming a laser at the aortic valve for 60 min after the administration of a photosensitizer 24 h prior. ROS, laser, and sham groups (n = 7 each) for every observation period (t = 0; t = 8 d; t = 84 d; t = 168 d) were established. The amount of ROS generation; morphological changes; inflammatory, immune, and apoptotic reactions; and hemodynamic changes in the aortic valves were assessed using appropriate histological, immunohistological, immunohistochemical, and echocardiographic methods. RESULTS The ROS group displayed an increased amount of ROS (p < 0.01) and increased inflammatory activation of the endothelium (p < 0.05) at t = 0. In the ROS group, aortic valves were calcified (p < 0.05) and the transvalvular gradient was increased (p < 0.01) at t = 168 d. CONCLUSION The small animal model employed here may serve as a platform for analyzing ROS's isolated role in the DAVD context.
Collapse
Affiliation(s)
- Jessica Isabel Selig
- Department of Cardiac Surgery, University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany; (J.I.S.); (Y.S.); (S.K.); (M.P.); (L.A.J.); (O.M.); (S.B.); (M.B.); (A.L.)
| | - Yukiharu Sugimura
- Department of Cardiac Surgery, University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany; (J.I.S.); (Y.S.); (S.K.); (M.P.); (L.A.J.); (O.M.); (S.B.); (M.B.); (A.L.)
- Department of Cardiac Surgery, University of Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Shintaro Katahira
- Department of Cardiac Surgery, University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany; (J.I.S.); (Y.S.); (S.K.); (M.P.); (L.A.J.); (O.M.); (S.B.); (M.B.); (A.L.)
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryomachi, Aoba-ku, Sendai 980-8574, Japan
| | - Marco Polidori
- Department of Cardiac Surgery, University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany; (J.I.S.); (Y.S.); (S.K.); (M.P.); (L.A.J.); (O.M.); (S.B.); (M.B.); (A.L.)
| | - Laura Alida Jacobi
- Department of Cardiac Surgery, University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany; (J.I.S.); (Y.S.); (S.K.); (M.P.); (L.A.J.); (O.M.); (S.B.); (M.B.); (A.L.)
| | - Olga Medovoj
- Department of Cardiac Surgery, University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany; (J.I.S.); (Y.S.); (S.K.); (M.P.); (L.A.J.); (O.M.); (S.B.); (M.B.); (A.L.)
| | - Sarah Betke
- Department of Cardiac Surgery, University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany; (J.I.S.); (Y.S.); (S.K.); (M.P.); (L.A.J.); (O.M.); (S.B.); (M.B.); (A.L.)
| | - Mareike Barth
- Department of Cardiac Surgery, University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany; (J.I.S.); (Y.S.); (S.K.); (M.P.); (L.A.J.); (O.M.); (S.B.); (M.B.); (A.L.)
- Department of Cardiac Surgery, University of Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Artur Lichtenberg
- Department of Cardiac Surgery, University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany; (J.I.S.); (Y.S.); (S.K.); (M.P.); (L.A.J.); (O.M.); (S.B.); (M.B.); (A.L.)
- Cardiovascular Research Institute Dusseldorf (CARID), University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany
| | - Payam Akhyari
- Department of Cardiac Surgery, University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany; (J.I.S.); (Y.S.); (S.K.); (M.P.); (L.A.J.); (O.M.); (S.B.); (M.B.); (A.L.)
- Department of Cardiac Surgery, University of Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Jan-Philipp Minol
- Department of Cardiac Surgery, University of Dusseldorf, Moorenstrasse 5, 40225 Dusseldorf, Germany; (J.I.S.); (Y.S.); (S.K.); (M.P.); (L.A.J.); (O.M.); (S.B.); (M.B.); (A.L.)
| |
Collapse
|
19
|
Klauzen P, Basovich L, Shishkova D, Markova V, Malashicheva A. Macrophages in Calcific Aortic Valve Disease: Paracrine and Juxtacrine Disease Drivers. Biomolecules 2024; 14:1547. [PMID: 39766254 PMCID: PMC11673549 DOI: 10.3390/biom14121547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
A significant role in the pathogenesis of CAVD is played by innate immunity cells, such as macrophages. In stenotic valves, macrophages have enhanced inflammatory activity, and the population's balance is shifted toward pro-inflammatory ones. Pro-inflammatory macrophages release cytokines, chemokines, and microRNA, which can directly affect the resident valvular cells and cause valve calcification. In CAVD patients, macrophages may have more pronounced pro-inflammatory properties, enhanced not only by paracrine signals but also by juxtacrine Notch signaling and epigenetic factors, which influence the maturation of macrophages' progenitors. In this review, we observe the accumulated data on the involvement of macrophages in CAVD development via paracrine and juxtacrine interactions.
Collapse
Affiliation(s)
- Polina Klauzen
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| | - Liubov Basovich
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| | - Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo 650002, Russia; (D.S.); (V.M.)
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo 650002, Russia; (D.S.); (V.M.)
| | - Anna Malashicheva
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| |
Collapse
|
20
|
Subrahmanian S, Varshney R, Subramani K, Murphy B, Woolington S, Ahamed J. N-Acetylcysteine Inhibits Aortic Stenosis Progression in a Murine Model by Blocking Shear-Induced Activation of Platelet Latent Transforming Growth Factor Beta 1. Antioxid Redox Signal 2024; 41:e1187-e1196. [PMID: 34619980 PMCID: PMC11693965 DOI: 10.1089/ars.2021.0037] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 01/08/2023]
Abstract
Objective: Aortic stenosis (AS) is characterized by narrowing of the aortic valve opening, resulting in peak blood flow velocity that induces high wall shear stress (WSS) across the valve. Severe AS leads to heart failure and death. There is no treatment available for AS other than valve replacement. Platelet-derived transforming growth factor beta 1 (TGF-β1) partially contributes to AS progression in mice, and WSS is a potent activator of latent TGF-β1. N-acetylcysteine (NAC) inhibits WSS-induced TGF-β1 activation in vitro. We hypothesize that NAC will inhibit AS progression by inhibiting WSS-induced TGF-β1 activation. Approach: We treated a cohort of Ldlr(-/-)Apob(100/100) low density lipoprotein receptor (LDLR) mice fed a high-fat diet with NAC (2% in drinking water) at different stages of disease progression and measured its effect on AS progression and TGF-β1 activation. Results: Short-term NAC treatment inhibited AS progression in mice with moderate and severe AS relative to controls, but not in LDLR mice lacking platelet-derived TGF-β1 (TGF-β1platlet-KO-LDLR). NAC treatment reduced TGF-β signaling, p-Smad2 and collagen levels, and mesenchymal transition from isolectin B4 and CD45-positive cells in LDLR mice. Mechanistically, NAC treatment resulted in plasma NAC concentrations ranging from 75.5 to 449.2 ng/mL, which were sufficient to block free thiol labeling of plasma proteins and reduce active TGF-β1 levels without substantially affecting reactive oxygen species-modified products in valvular cells. Conclusions: Short-term treatment with NAC inhibits AS progression by inhibiting WSS-induced TGF-β1 activation in the LDLR mouse model of AS, motivating a clinical trial of NAC and/or other thiol-reactive agent(s) as a potential therapy for AS.
Collapse
Affiliation(s)
- Sandeep Subrahmanian
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Rohan Varshney
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Kumar Subramani
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Brennah Murphy
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Sean Woolington
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Jasimuddin Ahamed
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
21
|
Sohlman M, Jauhiainen R, Vangipurapu J, Laakso A, Ala-Korpela M, Kuulasmaa T, Kuusisto J. Biomarkers reflecting insulin resistance increase the risk of aortic stenosis in a population-based study of 10,144 Finnish men. Ann Med 2024; 56:2419996. [PMID: 39593205 DOI: 10.1080/07853890.2024.2419996] [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: 07/08/2024] [Revised: 07/08/2024] [Accepted: 07/23/2024] [Indexed: 11/28/2024] Open
Abstract
AIMS To investigate a comprehensive panel of biomarkers and risk of aortic stenosis (AS) in a prospective population-based study. METHODS Anthropometric, metabolic, and inflammatory biomarkers were measured in the Metabolic Syndrome in the Men Study of 10,144 Finnish men without AS at baseline. Cases of AS were identified from the medical records. Cox regression analysis was used to identify variables predicting AS over a follow-up time of 10.8 years. Principal component (PC) analysis was applied to the biomarkers that predicted AS. Cox regression analysis was used to investigate the resulting PCs as AS predictors. RESULTS AS was diagnosed in 116 men (1.1%), with a median age of 62 years. In Cox regression analyses, fasting, 30 min, and 120 min plasma insulin, and proinsulin, with hazard ratios (HR) ranging from 1.38 (1.12-1.69, p = 2.1E-3) to 1.44 (1.23-1.68, p = 4.0E-6), Matsuda index [HR 0.68 (0.56-0.82, p = 6.9E-5)], and serum C-peptide [HR 1.47 (1.22-1.77, p = 5.0E-5)] were associated with incident AS, in addition to age, systolic blood pressure, BMI, waist circumference, waist/hip ratio, height, body fat mass, fat-free mass, and hs-CRP, and remained significant after adjustments, or if diabetic subjects were excluded. PC 1, consisting of fasting plasma insulin, C-peptide, Matsuda index, waist/hip ratio, and urine albumin excretion, and PC 2, consisting of age, body fat mass, and systolic blood pressure, were significantly associated with AS [HRs 1.37(1.09-1.73) and 1.77 (1.45-2.17), respectively]. CONCLUSION Biomarkers reflecting insulin resistance are risk factors for AS, a novel finding indicating that insulin resistance is important in the pathogenesis of AS.
Collapse
Affiliation(s)
- Maija Sohlman
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Raimo Jauhiainen
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jagadish Vangipurapu
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Annamaria Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Center for Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Mika Ala-Korpela
- Systems Epidemiology, University of Oulu, Oulu, Finland
- Research Unit of Population Health, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Teemu Kuulasmaa
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Center for Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
22
|
Muzafarova T, Motovska Z. The role of pre-existing left-sided valvular heart disease in the prognosis of patients with acute myocardial infarction. Front Cardiovasc Med 2024; 11:1465723. [PMID: 39628551 PMCID: PMC11612903 DOI: 10.3389/fcvm.2024.1465723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/16/2024] [Indexed: 12/06/2024] Open
Abstract
Acute myocardial infarction (AMI) and valvular heart disease (VHD) are the leading causes of cardiovascular morbidity and mortality. The epidemiology of VHD has changed in recent decades with an aging population, increasing risk factors for cardiovascular disease and migration, all of which have a significant implifications for healthcare systems. Due to common pathophysiological mechanisms and risk factors, AMI and VHD often coexist. These patients have more complicated clinical characteristics, in-hospital course and outcomes, and are less likely to receive guideline-directed therapy. Because of the reciprocal negative pathophysiological influence, these patients need to be referred to VHD specialists and further discussed within the Heart team to assess the need for earlier intervention. Since the results of the number of studies show that one third of the patients are referred to the heart teams either too early or too late, there is a need to better define the communication networks between the treating physicians, including internists, general practitioners, outpatient cardiologists and heart teams, after the discharge of patients with pre-existing VHD and AMI.
Collapse
Affiliation(s)
| | - Zuzana Motovska
- Cardiocenter, Third Faculty of Medicine, Charles University and University Hospital Vinohrady, Prague, Czechia
| |
Collapse
|
23
|
Chong T, Lan NSR, Courtney W, He A, Strange G, Playford D, Dwivedi G, Hillis GS, Ihdayhid AR. Medical Therapy to Prevent or Slow Progression of Aortic Stenosis: Current Evidence and Future Directions. Cardiol Rev 2024; 32:473-482. [PMID: 36961371 DOI: 10.1097/crd.0000000000000528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
Degenerative aortic stenosis is a growing clinical problem owing to the high incidence in an aging population and its significant morbidity and mortality. Currently, aortic valve replacement remains the only treatment. Despite promising observational data, pharmacological management to slow or halt progression of aortic stenosis has remained elusive. Nevertheless, with a greater understanding of the mechanisms which underpin aortic stenosis, research has begun to explore novel treatment strategies. This review will explore the historical agents used to manage aortic stenosis and the emerging agents that are currently under investigation.
Collapse
Affiliation(s)
- Travis Chong
- From the Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Harry Perkins Institute of Medical Research, Perth, Australia
| | - Nick S R Lan
- From the Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Harry Perkins Institute of Medical Research, Perth, Australia
- Internal Medicine, Medical School, The University of Western Australia, Perth, Australia
| | - William Courtney
- Internal Medicine, Medical School, The University of Western Australia, Perth, Australia
- Department of Cardiology, Royal Perth Hospital, Perth, Australia
| | - Albert He
- From the Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Harry Perkins Institute of Medical Research, Perth, Australia
| | - Geoff Strange
- School of Medicine, University of Notre Dame, Fremantle, Australia
| | - David Playford
- School of Medicine, University of Notre Dame, Fremantle, Australia
| | - Girish Dwivedi
- From the Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Harry Perkins Institute of Medical Research, Perth, Australia
- Internal Medicine, Medical School, The University of Western Australia, Perth, Australia
| | - Graham S Hillis
- Internal Medicine, Medical School, The University of Western Australia, Perth, Australia
- Department of Cardiology, Royal Perth Hospital, Perth, Australia
| | - Abdul Rahman Ihdayhid
- From the Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Harry Perkins Institute of Medical Research, Perth, Australia
- Curtin Medical School, Curtin University, Perth, Australia
| |
Collapse
|
24
|
Prabhakar AP, Lopez-Candales A. Calcific aortic valve disease and cardiometabolic triggers: an explanation behind progression of aortic valvular disease and failure of medical therapy interventions. Postgrad Med 2024; 136:810-818. [PMID: 39297302 DOI: 10.1080/00325481.2024.2406740] [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: 02/20/2024] [Accepted: 09/17/2024] [Indexed: 09/25/2024]
Abstract
Calcific aortic valve disease (CAVD), a nonrheumatic stenosis of the trileaflet aortic valve, is a complex, multifaceted cardiovascular condition involving a widespread inflammatory process and an analogous atheromatous process affecting the arteries. It is currently the most encountered valvular abnormality in cardiology. Although distinctive abnormal mechanical forces are at the core propelling a responsive mechanosensitive feedback cascade, implicated in both initiation and perpetuation of CAVD; we propose a conundrum of metabolic abnormalities including hypertension, elevated fasting blood sugar, decreased high-density lipoprotein, hypertriglyceridemia, and abdominal obesity as perpetuators of this process. Furthermore, we suggest CAVD as a cardio metabolic disorder. New perspectives as well as which pathways we believe are critically involved and ideas for early intervention are discussed.
Collapse
Affiliation(s)
- Akruti Patel Prabhakar
- Advanced Cardiac Imaging Research Fellow, Department of Cardiovascular Medicine, University of Louisville, Louisville, KY, USA
| | - Angel Lopez-Candales
- Cardiology Section, Dayton VA Medical Center, Wright State Boonshoft School of Medicine, Dayton, OH, USA
| |
Collapse
|
25
|
Aasim M, Aziz R, Mohsin AU, Khan R, Aziz G, Ikram J. Comparison of the Outcomes of Aortic Valve Replacements (AVRs) Performed via Conventional Full Sternotomy and Upper Mini Sternotomy: Our Experience at Hayatabad Medical Complex, Pakistan. Cureus 2024; 16:e73278. [PMID: 39651028 PMCID: PMC11625441 DOI: 10.7759/cureus.73278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction Aortic valve replacement (AVR) for severe symptomatic aortic stenosis is a commonly performed procedure, yielding excellent long-term outcomes. Comparing a mini sternotomy with a conventional sternotomy is essential to evaluate less invasive options that can improve patient recovery and reduce postoperative complications. This insight supports surgical decision-making for better AVR patient outcomes. Methodology This retrospective comparative study aims to compare clinical outcomes between mini sternotomy for aortic valve replacement (mini-AVR) and conventional full sternotomy for aortic valve replacement (FS-AVR). Patient records of isolated AVR from January 2021 to July 2023 were reviewed, excluding those with comorbidities or requiring concomitant procedures. Outcomes measured included sternal wound infections, operative time, length of ICU stay, cardiopulmonary bypass (CPB) time, and aortic cross-clamp time. Results The study included 65 patients (47 males and 18 females). Among the participants, 30 patients underwent AVR using full sternotomy, while 35 patients had the procedure performed via upper mini-sternotomy. The mini-AVR group experienced significantly less bleeding and a reduced need for blood transfusions compared to the FS-AVR group. Additionally, patients in the FS-AVR group had longer ICU stays and prolonged ventilation times. Notably, in contrast to findings from other studies, our research revealed that CPB time and aortic cross-clamp time were shorter in the mini-AVR group. Conclusion Mini-sternotomy has proven to be a safe and effective approach for AVR, with the mini-AVR group experiencing fewer complications, such as reduced bleeding and decreased need for blood transfusions. Additionally, patients benefit from shorter ICU stays, reduced ventilation time, and quicker overall recovery.
Collapse
Affiliation(s)
- Muhammad Aasim
- Department of Cardiac Surgery, Hayatabad Medical Complex, Peshawar, PAK
- Department of Cardiac Surgery, Khyber Girls Medical College (KGMC), Peshawar, PAK
| | - Raheela Aziz
- Department of Cardiac Surgery, Hayatabad Medical Complex, Peshawar, PAK
| | - Atta Ul Mohsin
- Department of Cardiac Surgery, Hayatabad Medical Complex, Peshawar, PAK
| | - Raheel Khan
- Department of Cardiac Surgery, Hayatabad Medical Complex, Peshawar, PAK
| | - Gulshad Aziz
- Department of Anatomy, Khyber Medical University (KMU) Institute of Dental Sciences, Kohat, PAK
| | - Jibran Ikram
- Department of Cardiovascular Medicine, Hayatabad Medical Complex, Peshawar, PAK
| |
Collapse
|
26
|
Brette JB, Colombat M, Fournier P, Moninhas M, Marcheix B, Lairez O, Cariou E. Descriptive study of the clinical and myocardial status of a population with anatomopathological aortic valve amyloidosis. Cardiovasc Pathol 2024; 73:107674. [PMID: 39025343 DOI: 10.1016/j.carpath.2024.107674] [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: 01/16/2024] [Revised: 07/06/2024] [Accepted: 07/07/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Aortic stenosis (AS) and transthyretin (ATTR) cardiac amyloidosis (CA) share the same clinical profiles and cardiac phenotype. Amyloid deposits have been frequently reported in aortic valves of patients with severe AS referred for surgical aortic valve replacement (SAVR). The aim of this study was to determine the clinical and myocardial status of patients with aortic valve amyloidosis after aortic valve surgery. METHODS AND RESULTS We performed a retrospective descriptive study of 46 patients who underwent SAVR for severe AS with amyloid deposits upon histological analysis. All patients were screened for cardiac involvement. Amyloid deposits typing was successful in 35 (76%) patients and 28 (80%) were ATTR. Two (4%) had positive bone scintigraphy and among the 5 myocardial biopsies performed during surgery, 80% were positive for ATTR deposits. CONCLUSION ATTR is the predominant type in the presence of amyloid deposits on the aortic valve after surgery for severe AS but is only rarely accompanied by cardiac uptake on bone scintigraphy. Early stages of myocardial involvement are frequent and myocardial biopsy is more sensitive for detection of mild amyloid deposits than bone scintigraphy.
Collapse
Affiliation(s)
- Jean-Baptiste Brette
- Department of Cardiology, Toulouse University Hospital, France; Cardiac Imaging Center, Toulouse University Hospital, France
| | - Magali Colombat
- Medical School, Toulouse III Paul Sabatier University, Toulouse, France; Department of Pathology, IUCT Oncopôle, Toulouse France
| | - Pauline Fournier
- Department of Cardiology, Toulouse University Hospital, France; Cardiac Imaging Center, Toulouse University Hospital, France
| | - Maxime Moninhas
- Department of Cardiology, Toulouse University Hospital, France; Cardiac Imaging Center, Toulouse University Hospital, France
| | - Bertrand Marcheix
- Medical School, Toulouse III Paul Sabatier University, Toulouse, France; Department of Cardiac Surgery, Toulouse University Hospital, France
| | - Olivier Lairez
- Department of Cardiology, Toulouse University Hospital, France; Cardiac Imaging Center, Toulouse University Hospital, France; Department of Nuclear Medicine, Toulouse University Hospital, France; Medical School, Toulouse III Paul Sabatier University, Toulouse, France.
| | - Eve Cariou
- Department of Cardiology, Toulouse University Hospital, France; Cardiac Imaging Center, Toulouse University Hospital, France
| |
Collapse
|
27
|
Kontogeorgos S, Rosengren A, Sandström TZ, Fu M, Lindgren M, Basic C, Svanvik M, Djekic D, Thunström E. Association Between Body Mass Index and Risk of Aortic Stenosis in Women in the Swedish Medical Birth Registry. J Am Heart Assoc 2024; 13:e034891. [PMID: 39392145 PMCID: PMC11935566 DOI: 10.1161/jaha.123.034891] [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: 03/21/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Overweight and obesity are increasing globally with aging, as are life expectancy and aging-associated disorders, including calcific aortic stenosis (AS). Studies investigating the correlation between high body mass index (BMI) and AS are contradictory and inconclusive. This study examines a potential association between BMI and AS in women. METHODS AND RESULTS By linking the Swedish Medical Birth Register and the Swedish National Patient Register, we included women aged 18 to 55 years with a first childbirth from 1981 to 2020. Diagnosis of AS and comorbidities were defined according to the International Classification of Diseases (ICD) codes. The women were divided into groups on the basis of BMI. Cox proportional hazards regression models were used to investigate the difference in the risk of being diagnosed with AS, with reference BMI 20 to <22.5 kg/m2. Among the 1 722 625 included women, the mean age was 28 years, and mean BMI was 24 kg/m2, with 21% being overweight (BMI 25 to <30 kg/m2) and 8.5% obese (BMI ≥30 kg/m2). During median follow-up of 19.5 years, 2488 women (0.14%) were diagnosed with AS. The age-adjusted risk of being diagnosed with AS increased with higher BMI to 2.82 (95% CI, 2.44-3.25) times higher in women with BMI 30 to <35 kg/m2, and to 3.72 (95% CI, 2.95-4.70) times higher in those with BMI ≥35 kg/m2. Similar results were found after excluding AS of rheumatic pathogenesis. CONCLUSIONS An increase in BMI from its upper normal range was consistently and independently associated with the risk of developing AS in women.
Collapse
Affiliation(s)
- Silvana Kontogeorgos
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of MedicineGeriatrics and Emergency MedicineRegion Västra GötalandSahlgrenska University Hospital/ÖstraGothenburgSweden
- Department of Clinical PhysiologySahlgrenska University HospitalGothenburgSweden
| | - Annika Rosengren
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of MedicineGeriatrics and Emergency MedicineRegion Västra GötalandSahlgrenska University Hospital/ÖstraGothenburgSweden
| | - Tatiana Zverkova Sandström
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of MedicineGeriatrics and Emergency MedicineRegion Västra GötalandSahlgrenska University Hospital/ÖstraGothenburgSweden
| | - Michael Fu
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of MedicineGeriatrics and Emergency MedicineRegion Västra GötalandSahlgrenska University Hospital/ÖstraGothenburgSweden
| | - Martin Lindgren
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of MedicineGeriatrics and Emergency MedicineRegion Västra GötalandSahlgrenska University Hospital/ÖstraGothenburgSweden
| | - Carmen Basic
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of MedicineGeriatrics and Emergency MedicineRegion Västra GötalandSahlgrenska University Hospital/ÖstraGothenburgSweden
| | - Maria‐Teresia Svanvik
- Department of Obstetrics and GynecologyInstitute of Clinical SciencesSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Demir Djekic
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of MedicineGeriatrics and Emergency MedicineRegion Västra GötalandSahlgrenska University Hospital/ÖstraGothenburgSweden
| | - Erik Thunström
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of MedicineGeriatrics and Emergency MedicineRegion Västra GötalandSahlgrenska University Hospital/ÖstraGothenburgSweden
| |
Collapse
|
28
|
Tóth A, Balogh E, Jeney V. In Vitro Models of Cardiovascular Calcification. Biomedicines 2024; 12:2155. [PMID: 39335668 PMCID: PMC11429067 DOI: 10.3390/biomedicines12092155] [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: 08/30/2024] [Revised: 09/15/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiovascular calcification, characterized by hydroxyapatite deposition in the arterial wall and heart valves, is associated with high cardiovascular morbidity and mortality. Cardiovascular calcification is a hallmark of aging but is frequently seen in association with chronic diseases, such as chronic kidney disease (CKD), diabetes, dyslipidemia, and hypertension in the younger population as well. Currently, there is no therapeutic approach to prevent or cure cardiovascular calcification. The pathophysiology of cardiovascular calcification is highly complex and involves osteogenic differentiation of various cell types of the cardiovascular system, such as vascular smooth muscle cells and valve interstitial cells. In vitro cellular and ex vivo tissue culture models are simple and useful tools in cardiovascular calcification research. These models contributed largely to the discoveries of the numerous calcification inducers, inhibitors, and molecular mechanisms. In this review, we provide an overview of the in vitro cell culture and the ex vivo tissue culture models applied in the research of cardiovascular calcification.
Collapse
Affiliation(s)
- Andrea Tóth
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Enikő Balogh
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Viktória Jeney
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
29
|
Navani RV, Dayawansa NH, Nanayakkara S, Palmer S, Noaman S, Htun NM, Walton AS, Peter K, Stub D. Post-Procedure Monocyte Count Levels Predict Major Adverse Cardiovascular Events (MACE) Following Transcatheter Aortic Valve Implantation (TAVI) for Aortic Stenosis. Heart Lung Circ 2024; 33:1340-1347. [PMID: 38845242 DOI: 10.1016/j.hlc.2024.03.013] [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: 01/03/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Aortic stenosis has recently been characterised as having an inflammatory aetiology, beyond the traditional degenerative model. Recruitment of monocytes has been associated with inflammation contributing to progression of calcific aortic-valve disease. Prior research has demonstrated that pre-procedure inflammatory biomarkers do not consistently discriminate poorer outcomes in those with aortic stenosis. It remains, however, unclear if postprocedure inflammatory biomarkers, which are influenced by intraprocedural pro-inflammatory insults, can predict major adverse cardiovascular events (MACE) post transcatheter aortic valve implantation (TAVI). METHOD All patients with postprocedure monocyte levels undergoing transcatheter aortic valve implantation at The Alfred Hospital, Melbourne, Australia (2008-2019) were included. The highest monocyte count from postprocedure days 1 to 3 was used. Patients were divided into "high" or "low" postprocedure monocyte count groups using the Youden Index. The incidence of 30-day MACE a composite of stroke, acute myocardial infarction, and death) was then compared. RESULTS In total, 472 patients were included (54% men, median age 84 years). Fourteen (14) patients (3%) suffered a 30-day MACE. Those with high postprocedure monocyte count were more likely to: be hypertensive (p=0.049); have a higher Society of Thoracic Surgeons risk score (p=0.032); and, undergo non-transfemoral access (p=0.018). A high (≥0.975) postprocedure monocyte count was significantly associated with 30-day MACE (odds ratio [OR] 1.16 for each 0.1 increase in monocyte, p=0.025). This association remained present on multivariable analysis adjusted for age, sex, Society of Thoracic Surgeons risk score, and self-expanding valve prosthesis type (OR 1.17, p=0.028). CONCLUSIONS The association between postprocedure monocytosis and 30-day MACE suggests that minimising peri-procedural inflammatory insults may improve outcomes. This inexpensive and readily available biomarker may also aid in tailored risk stratification for patients.
Collapse
Affiliation(s)
- Rohan V Navani
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia. https://www.twitter.com/RohanNavani
| | - Nalin H Dayawansa
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia; Atherothrombosis and Vascular Biology Laboratory, Baker Heart & Diabetes Institute, Melbourne, Vic, Australia; School of Translational Medicine, Monash University, Melbourne, Vic, Australia
| | - Shane Nanayakkara
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia; Atherothrombosis and Vascular Biology Laboratory, Baker Heart & Diabetes Institute, Melbourne, Vic, Australia; School of Translational Medicine, Monash University, Melbourne, Vic, Australia
| | - Sonny Palmer
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia; Department of Medicine, The University of Melbourne, Melbourne, Vic, Australia
| | - Samer Noaman
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia
| | - Nay M Htun
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia
| | - Antony S Walton
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia; School of Translational Medicine, Monash University, Melbourne, Vic, Australia
| | - Karlheinz Peter
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia; Atherothrombosis and Vascular Biology Laboratory, Baker Heart & Diabetes Institute, Melbourne, Vic, Australia; School of Translational Medicine, Monash University, Melbourne, Vic, Australia
| | - Dion Stub
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia; School of Translational Medicine, Monash University, Melbourne, Vic, Australia.
| |
Collapse
|
30
|
Mueller KAL, Langnau C, Harm T, Sigle M, Mott K, Droppa M, Borst O, Rohlfing AK, Gekeler S, Günter M, Goebel N, Franke UF, Radwan M, Schlensak C, Janning H, Scheuermann S, Seitz CM, Rath D, Kreisselmeier KP, Castor T, Mueller II, Schulze H, Autenrieth SE, Gawaz MP. Macrophage Migration Inhibitory Factor Promotes Thromboinflammation and Predicts Fast Progression of Aortic Stenosis. Arterioscler Thromb Vasc Biol 2024; 44:2118-2135. [PMID: 38989580 PMCID: PMC11335082 DOI: 10.1161/atvbaha.124.321000] [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: 03/25/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Aortic stenosis (AS) is driven by progressive inflammatory and fibrocalcific processes regulated by circulating inflammatory and valve resident endothelial and interstitial cells. The impact of platelets, platelet-derived mediators, and platelet-monocyte interactions on the acceleration of local valvular inflammation and mineralization is presently unknown. METHODS We prospectively enrolled 475 consecutive patients with severe symptomatic AS undergoing aortic valve replacement. Clinical workup included repetitive echocardiography, analysis of platelets, monocytes, chemokine profiling, aortic valve tissue samples for immunohistochemistry, and gene expression analysis. RESULTS The patients were classified as fast-progressive AS by the median ∆Vmax of 0.45 m/s per year determined by echocardiography. Immunohistological aortic valve analysis revealed enhanced cellularity in fast-progressive AS (slow- versus fast-progressive AS; median [interquartile range], 247 [142.3-504] versus 717.5 [360.5-1234]; P<0.001) with less calcification (calcification area, mm2: 33.74 [27.82-41.86] versus 20.54 [13.52-33.41]; P<0.001). MIF (macrophage migration inhibitory factor)-associated gene expression was significantly enhanced in fast-progressive AS accompanied by significantly elevated MIF plasma levels (mean±SEM; 6877±379.1 versus 9959±749.1; P<0.001), increased platelet activation, and decreased intracellular MIF expression indicating enhanced MIF release upon platelet activation (CD62P, %: median [interquartile range], 16.8 [11.58-23.8] versus 20.55 [12.48-32.28], P=0.005; MIF, %: 4.85 [1.48-9.75] versus 2.3 [0.78-5.9], P<0.001). Regression analysis confirmed that MIF-associated biomarkers are strongly associated with an accelerated course of AS. CONCLUSIONS Our findings suggest a key role for platelet-derived MIF and its interplay with circulating and valve resident monocytes/macrophages in local and systemic thromboinflammation during accelerated AS. MIF-based biomarkers predict an accelerated course of AS and represent a novel pharmacological target to attenuate progression of AS.
Collapse
Affiliation(s)
- Karin Anne Lydia Mueller
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Carolin Langnau
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Tobias Harm
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Manuel Sigle
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Kristina Mott
- Institute for Experimental Biomedicine, Chair I University Hospital Würzburg, Germany (K.M., H.S.)
| | - Michal Droppa
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
- DFG Heisenberg Group Thrombocardiology (O.B.), University of Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Sarah Gekeler
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Manina Günter
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology (M.G., S.E.A.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
- Dendritic Cells in Infection and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany (M.G., S.E.A.)
| | - Nora Goebel
- Robert-Bosch Hospital, Department of Cardiovascular Surgery, Stuttgart, Germany (N.G., U.F.W.F.)
| | - Ulrich F.W. Franke
- Robert-Bosch Hospital, Department of Cardiovascular Surgery, Stuttgart, Germany (N.G., U.F.W.F.)
| | - Medhat Radwan
- Department of Thoracic and Cardiovascular Surgery (M.R., C.S.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Christian Schlensak
- Department of Thoracic and Cardiovascular Surgery (M.R., C.S.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Henrik Janning
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Sophia Scheuermann
- Cluster of Excellence iFIT (EXC 2180) Image-Guided and Functionally Instructed Tumor Therapies (S.S., C.M.S.), University of Tübingen, Germany
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Tuebingen, Germany (S.S., C.M.S.)
| | - Christian M. Seitz
- Cluster of Excellence iFIT (EXC 2180) Image-Guided and Functionally Instructed Tumor Therapies (S.S., C.M.S.), University of Tübingen, Germany
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Tuebingen, Germany (S.S., C.M.S.)
| | - Dominik Rath
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Klaus-Peter Kreisselmeier
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Iris Irmgard Mueller
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Harald Schulze
- Institute for Experimental Biomedicine, Chair I University Hospital Würzburg, Germany (K.M., H.S.)
| | - Stella E. Autenrieth
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology (M.G., S.E.A.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
- Dendritic Cells in Infection and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany (M.G., S.E.A.)
| | - Meinrad Paul Gawaz
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| |
Collapse
|
31
|
Fan L, Yao D, Fan Z, Zhang T, Shen Q, Tong F, Qian X, Xu L, Jiang C, Dong N. Beyond VICs: Shedding light on the overlooked VECs in calcific aortic valve disease. Biomed Pharmacother 2024; 178:117143. [PMID: 39024838 DOI: 10.1016/j.biopha.2024.117143] [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: 04/29/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024] Open
Abstract
Calcific aortic valve disease (CAVD) is prevalent in developed nations and has emerged as a pressing global public health concern due to population aging. The precise etiology of this disease remains uncertain, and recent research has primarily focused on examining the role of valvular interstitial cells (VICs) in the development of CAVD. The predominant treatment options currently available involve open surgery and minimally invasive interventional surgery, with no efficacious pharmacological treatment. This article seeks to provide a comprehensive understanding of valvular endothelial cells (VECs) from the aspects of valvular endothelium-derived nitric oxide (NO), valvular endothelial mechanotransduction, valvular endothelial injury, valvular endothelial-mesenchymal transition (EndMT), and valvular neovascularization, which have received less attention, and aims to establish their role and interaction with VICs in CAVD. The ultimate goal is to provide new perspectives for the investigation of non-invasive treatment options for this disease.
Collapse
Affiliation(s)
- Lin Fan
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dingyi Yao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengfeng Fan
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tailong Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Shen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuqiang Tong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingyu Qian
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chen Jiang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
32
|
Wada S, Iwanaga Y, Nakai M, Miyamoto Y, Noguchi T. Clinical impact of cardiovascular calcifications on stroke incidence in primary prevention: analysis in NADESICO study. Heart Vessels 2024; 39:754-762. [PMID: 38568474 DOI: 10.1007/s00380-024-02394-6] [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: 12/11/2023] [Accepted: 03/07/2024] [Indexed: 07/23/2024]
Abstract
The utility of assessment of cardiovascular calcifications for predicting stroke incidence remains unclear. This study assessed the relationship between cardiovascular calcifications including coronary artery calcification (CAC), aortic valve (AVC), and aortic root (ARC) assessed by coronary computed tomography (CT) and stroke incidence in patients with suspected CAD. In this multicenter prospective cohort study, 1187 patients suspected of CAD who underwent coronary CT were enrolled. Cardiovascular events including stroke were documented. Hazard ratio (HR) and confidence interval (CI) were assessed by Cox proportional hazard model adjusted for the Framingham risk score. C statistics for stroke incidence were also examined by models including cardiovascular calcifications. A total of 980 patients (mean age, 65 ± 7 years; females, 45.8%) were assessed by the CAC, AVC, and ARC Agatston scores. During a median follow-up of 4.0 years, 19 patients developed stroke. Cox proportional hazard model showed severe CAC (Agatston score ≥ 90th percentile [580.0 value]) and presence of AVC and ARC were associated with stroke incidence (HR; 10.33 [95% CI; 2.08-51.26], 3.08 [1.19-7.98], and 2.75 [1.03-7.30], respectively). C statistic in the model with CAC and AVC severity for predicting stroke incidence was 0.841 (95% CI; 0.761-0.920), which was superior to the model with CAC alone (0.762 [95% CI; 0.665-0.859], P < 0.01). CAC, AVC, and ARC were associated with stroke incidence in patients suspected of CAD. Assessment of both CAC and AVC may be useful for prediction of stroke incidence.
Collapse
Affiliation(s)
- Shinichi Wada
- Department of Medical and Health Information Management, National Cerebral and Cardiovascular Center, 6-1 kishibe-shimmachi, Suita, Osaka, 564-8565, Japan
- Department of Neurology, Kansai Electric Power Hospital, Osaka, Japan
| | - Yoshitaka Iwanaga
- Department of Medical and Health Information Management, National Cerebral and Cardiovascular Center, 6-1 kishibe-shimmachi, Suita, Osaka, 564-8565, Japan.
- Department of Cardiology, Sakurabashi-Watanabe Hospital, Osaka, Japan.
| | - Michikazu Nakai
- Department of Medical and Health Information Management, National Cerebral and Cardiovascular Center, 6-1 kishibe-shimmachi, Suita, Osaka, 564-8565, Japan
- Clinical Research Support Center, University of Miyazaki Hospital, Miyazaki, Japan
| | - Yoshihiro Miyamoto
- Department of Medical and Health Information Management, National Cerebral and Cardiovascular Center, 6-1 kishibe-shimmachi, Suita, Osaka, 564-8565, Japan
| | - Teruo Noguchi
- Department of Cardiology, National Cerebral and Cardiovascular Center, Suita, Japan
| |
Collapse
|
33
|
Shan W, Cui J, Song Y, Yan D, Feng L, Jian Y, Yi W, Sun Y. Itaconate as a key player in cardiovascular immunometabolism. Free Radic Biol Med 2024; 219:64-75. [PMID: 38604314 DOI: 10.1016/j.freeradbiomed.2024.04.218] [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: 01/13/2024] [Revised: 03/23/2024] [Accepted: 04/09/2024] [Indexed: 04/13/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally, resulting in a major health burden. Thus, an urgent need exists for exploring effective therapeutic targets to block progression of CVDs and improve patient prognoses. Immune and inflammatory responses are involved in the development of atherosclerosis, ischemic myocardial damage responses and repair, calcification, and stenosis of the aortic valve. These responses can involve both large and small blood vessels throughout the body, leading to increased blood pressure and end-organ damage. While exploring potential avenues for therapeutic intervention in CVDs, researchers have begun to focus on immune metabolism, where metabolic changes that occur in immune cells in response to exogenous or endogenous stimuli can influence immune cell effector responses and local immune signaling. Itaconate, an intermediate metabolite of the tricarboxylic acid (TCA) cycle, is related to pathophysiological processes, including cellular metabolism, oxidative stress, and inflammatory immune responses. The expression of immune response gene 1 (IRG1) is upregulated in activated macrophages, and this gene encodes an enzyme that catalyzes the production of itaconate from the TCA cycle intermediate, cis-aconitate. Itaconate and its derivatives have exerted cardioprotective effects through immune modulation in various disease models, such as ischemic heart disease, valvular heart disease, vascular disease, heart transplantation, and chemotherapy drug-induced cardiotoxicity, implying their therapeutic potential in CVDs. In this review, we delve into the associated signaling pathways through which itaconate exerts immunomodulatory effects, summarize its specific roles in CVDs, and explore emerging immunological therapeutic strategies for managing CVDs.
Collapse
Affiliation(s)
- Wenju Shan
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jun Cui
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yujie Song
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Dongxu Yan
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Linqi Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yuhong Jian
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Yang Sun
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
34
|
Tao J, Wang J, Peng H, Yuan Y, Lai H, Gu P, Xier Z, Li G, Yang YN. Association between serum phosphate, magnesium, calcium and aortic valve sclerosis: a propensity score-matched case-control study. Acta Cardiol 2024; 79:522-529. [PMID: 39041382 DOI: 10.1080/00015385.2024.2379682] [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: 04/28/2024] [Revised: 06/14/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024]
Abstract
OBJECTIVES Aortic valve sclerosis has been proposed to signify greater cardiovascular risk; the correlation between serum trace elements and aortic valve sclerosis has been reported. Therefore, an in-depth exploration of the risk factors for aortic valve sclerosis and early intervention may reduce the risk of cardiovascular disease. METHODS In this study, Patients with aortic valve sclerosis and non-aortic valve sclerosis who underwent echocardiographic diagnosis in the People's Hospital of Xinjiang Uygur Autonomous Region during the period from 2019 to 2021 were selected for this study. The correlation between aortic valve sclerosis and serum phosphorus, calcium, and magnesium levels was explored using the propensity score matching technique by pairing the two groups of patients 1:1. RESULTS A total of 1,533 non-aortic valve sclerosis and 1,533 aortic valve sclerosis patients were included. Logistic regression analysis showed that serum magnesium [OR: 0.346; 95%CI: 0.227, 0.528] and serum calcium [OR: 7.022; 95%CI: 4.755, 10.369] were influential factors. Patients with low, intermediate, and high serum magnesium levels had a significantly lower risk of aortic valve sclerosis compared to patients with very low micronutrient levels (p < 0.05). Comparatively, patients with low or high serum calcium levels had an elevated risk of aortic valve sclerosis (p < 0.05). CONCLUSION Serum magnesium may have a protective role against aortic valve sclerosis, while both low and high levels of serum calcium could be risk factor for the condition. These serum micronutrients may be indications of cardiovascular disease risk prediction or prevention, and more research is required.
Collapse
Affiliation(s)
- Jing Tao
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, Xinjiang, China
- Department of Cardiovascular Medicine, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, China
| | - Juan Wang
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, Xinjiang, China
- Department of Cardiovascular Medicine, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, China
| | - Hui Peng
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, Xinjiang, China
- Department of Cardiovascular Medicine, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, China
| | - Yujuan Yuan
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, Xinjiang, China
- Department of Cardiovascular Medicine, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, China
| | - Hongmei Lai
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, Xinjiang, China
- Department of Cardiovascular Medicine, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, China
| | - Peipei Gu
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, Xinjiang, China
- Department of Cardiovascular Medicine, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, China
| | - Zulipiyemu Xier
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, Xinjiang, China
- Department of Cardiovascular Medicine, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, China
| | - Guoqing Li
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, Xinjiang, China
- Department of Cardiovascular Medicine, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, China
| | - Yi-Ning Yang
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, Xinjiang, China
- Department of Cardiovascular Medicine, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, China
| |
Collapse
|
35
|
Benfari G, Essayagh B, Michelena HI, Ye Z, Inojosa JM, Ribichini FL, Crestanello J, Messika-Zeitoun D, Prendergast B, Wong BF, Thapa P, Enriquez-Sarano M. Severe aortic stenosis: secular trends of incidence and outcomes. Eur Heart J 2024; 45:1877-1886. [PMID: 38190428 DOI: 10.1093/eurheartj/ehad887] [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: 06/25/2023] [Revised: 11/17/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND AND AIMS Severe aortic stenosis (AS) is the guideline-based indication for aortic valve replacement (AVR), which has markedly increased with transcatheter approaches, suggesting possible increasing AS incidence. However, reported secular trends of AS incidence remain contradictory and lack quantitative Doppler echocardiographic ascertainment. METHODS All adults residents in Olmsted County (MN, USA) diagnosed over 20 years (1997-2016) with incident severe AS (first diagnosis) based on quantitatively defined measures (aortic valve area ≤ 1 cm2, aortic valve area index ≤ 0.6 cm2/m2, mean gradient ≥ 40 mmHg, peak velocity ≥ 4 m/s, Doppler velocity index ≤ 0.25) were counted to define trends in incidence, presentation, treatment, and outcome. RESULTS Incident severe AS was diagnosed in 1069 community residents. The incidence rate was 52.5 [49.4-55.8] per 100 000 patient-year, slightly higher in males vs. females and was almost unchanged after age and sex adjustment for the US population 53.8 [50.6-57.0] per 100 000 residents/year. Over 20 years, severe AS incidence remained stable (P = .2) but absolute burden of incident cases markedly increased (P = .0004) due to population growth. Incidence trend differed by sex, stable in men (incidence rate ratio 0.99, P = .7) but declining in women (incidence rate ratio 0.93, P = .02). Over the study, AS clinical characteristics remained remarkably stable and AVR performance grew and was more prompt (from 1.3 [0.1-3.3] years in 1997-2000 to 0.5 [0.2-2.1] years in 2013-16, P = .001) but undertreatment remained prominent (>40%). Early AVR was associated with survival benefit (adjusted hazard ratio 0.55 [0.42-0.71], P < .0001). Despite these improvements, overall mortality (3-month 8% and 3-year 36%), was swift, considerable and unabated (all P ≥ .4) throughout the study. CONCLUSIONS Over 20 years, the population incidence of severe AS remained stable with increased absolute case burden related to population growth. Despite stable severe AS presentation, AVR performance grew notably, but while declining, undertreatment remained substantial and disease lethality did not yet decline. These population-based findings have important implications for improving AS management pathways.
Collapse
Affiliation(s)
- Giovanni Benfari
- Department ofCardiovascular Disease, Mayo Clinic, Rochester, MN, USA
- Section of Cardiology, University of Verona, Verona, Italy
| | - Benjamin Essayagh
- Department ofCardiovascular Disease, Mayo Clinic, Rochester, MN, USA
- Department of Echocardiography, Cardio X Clinic, Cannes, France
| | | | - Zi Ye
- Department ofCardiovascular Disease, Mayo Clinic, Rochester, MN, USA
| | | | | | - Juan Crestanello
- Department ofCardiovascular Disease, Mayo Clinic, Rochester, MN, USA
| | - David Messika-Zeitoun
- Department of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Bernard Prendergast
- Department of Cardiology, Cleveland Clinic and Saint Thomas' Hospitals, London, UK
| | | | - Prabin Thapa
- Department ofCardiovascular Disease, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
36
|
Marrero N, Jha K, Razavi AC, Boakye E, Anchouche K, Dzaye O, Budoff MJ, Tsai MY, Shah SJ, Rotter JI, Guo X, Yao J, Blumenthal RS, Thanassoulis G, Post WS, Blaha MJ, Whelton SP. Identifying People at High Risk for Severe Aortic Stenosis: Aortic Valve Calcium Versus Lipoprotein(a) and Low-Density Lipoprotein Cholesterol. Circ Cardiovasc Imaging 2024; 17:e016372. [PMID: 38889215 PMCID: PMC11423705 DOI: 10.1161/circimaging.123.016372] [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: 11/17/2023] [Accepted: 04/11/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Aortic valve calcification (AVC), Lp(a) [lipoprotein(a)], and low-density lipoprotein cholesterol (LDL-C) are associated with severe aortic stenosis (AS). We aimed to determine which of these risk factors were most strongly associated with the risk of incident severe AS. METHODS A total of 6792 participants from the MESA study (Multi-Ethnic Study of Atherosclerosis) had computed tomography-quantified AVC, Lp(a), and LDL-C values at MESA visit 1 (2000-2002). We calculated the absolute event rate of incident adjudicated severe AS per 1000 person-years and performed multivariable adjusted Cox proportional hazards regression. RESULTS The mean age was 62 years old, and 47% were women. Over a median 16.7-year follow-up, the rate of incident severe AS increased exponentially with higher AVC, regardless of Lp(a) or LDL-C values. Participants with AVC=0 had a very low rate of severe AS even with elevated Lp(a) ≥50 mg/dL (<0.1/1000 person-years) or LDL-C ≥130 mg/dL (0.1/1000 person-years). AVC >0 was strongly associated with severe AS when Lp(a) <50 mg/dL hazard ratio (HR) of 33.8 (95% CI, 16.4-70.0) or ≥50 mg/dL HR of 61.5 (95% CI, 7.7-494.2) and when LDL-C <130 mg/dL HR of 31.1 (95% CI, 14.4-67.1) or ≥130 mg/dL HR of 50.2 (95% CI, 13.2-191.9). CONCLUSIONS AVC better identifies people at high risk for severe AS compared with Lp(a) or LDL-C, and people with AVC=0 have a very low long-term rate of severe AS regardless of Lp(a) or LDL-C level. These results suggest AVC should be the preferred prognostic risk marker to identify patients at high risk for severe AS, which may help inform participant selection for future trials testing novel strategies to prevent severe AS.
Collapse
Affiliation(s)
| | - Kunal Jha
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
- University of Louisville, Division of Cardiology, KY (K.J.)
| | - Alexander C Razavi
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
- Center for Heart Disease Prevention, Emory School of Medicine, Atlanta, GA (A.C.R.)
| | - Ellen Boakye
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| | - Khalil Anchouche
- Preventive and Genomic Cardiology, Department of Medicine, McGill University, and the McGill University Health Center Research Institute, Montréal, Québec, Canada (K.A., G.T.)
| | - Omar Dzaye
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| | - Matthew J Budoff
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (M.Y.T.)
| | - Sanjiv J Shah
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.)
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences (J.I.R., X.G., J.Y.), The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Xiuqing Guo
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences (J.I.R., X.G., J.Y.), The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Jie Yao
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences (J.I.R., X.G., J.Y.), The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Roger S Blumenthal
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| | - George Thanassoulis
- Preventive and Genomic Cardiology, Department of Medicine, McGill University, and the McGill University Health Center Research Institute, Montréal, Québec, Canada (K.A., G.T.)
| | - Wendy S Post
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| | - Michael J Blaha
- Department of Medicine (M.J.B.), The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Seamus P Whelton
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD (K.J., A.C.R., E.B., O.D., R.S.B., W.S.P., M.J.B., S.P.W.)
| |
Collapse
|
37
|
Gong S, Xiang K, Chen L, Zhuang H, Song Y, Chen J. Integrated bioinformatics analysis identified leucine rich repeat containing 15 and secreted phosphoprotein 1 as hub genes for calcific aortic valve disease and osteoarthritis. IET Syst Biol 2024; 18:77-91. [PMID: 38566328 PMCID: PMC11179158 DOI: 10.1049/syb2.12091] [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: 06/05/2023] [Revised: 01/05/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
Calcific aortic valve disease (CAVD) and osteoarthritis (OA) are common diseases in the ageing population and share similar pathogenesis, especially in inflammation. This study aims to discover potential diagnostic and therapeutic targets in patients with CAVD and OA. Three CAVD datasets and one OA dataset were obtained from the Gene Expression Omnibus database. We used bioinformatics methods to search for key genes and immune infiltration, and established a ceRNA network. Immunohistochemical staining was performed to verify the expression of candidate genes in human and mice aortic valve tissues. Two key genes obtained, leucine rich repeat containing 15 (LRRC15) and secreted phosphoprotein 1 (SPP1), were further screened using machine learning and verified in human and mice aortic valve tissues. Compared to normal tissues, the infiltration of immune cells in CAVD tissues was significantly higher, and the expressions of LRRC15 and SPP1 were positively correlated with immune cells infiltration. Moreover, the ceRNA network showed extensive regulatory interactions based on LRRC15 and SPP1. The authors' findings identified LRRC15 and SPP1 as hub genes in immunological mechanisms during CAVD and OA initiation and progression, as well as potential targets for drug development.
Collapse
Affiliation(s)
- Shuji Gong
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Kun Xiang
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Le Chen
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Huanwei Zhuang
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Yaning Song
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Jinlan Chen
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| |
Collapse
|
38
|
Kilaru V, Patel S, Patel H, Ghasemzadeh N. An Unusual Case of Bicuspid Aortic Valve With Prolapse Masquerading as Cusp Perforation. Cureus 2024; 16:e60562. [PMID: 38887341 PMCID: PMC11181233 DOI: 10.7759/cureus.60562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2024] [Indexed: 06/20/2024] Open
Abstract
We present a unique clinical scenario of a 58-year-old male with a past medical history of hypertension who initially presented with chest pain and was ruled in for non-ST elevation myocardial infarction (NSTEMI) but rapidly developed respiratory failure secondary to aortic insufficiency complicated by cardiogenic shock (CS), attributed to aortic valve prolapse. Intriguingly, the patient had a normal ECG on presentation, underscoring the dynamic nature of valvular pathology. The development of CS highlights the importance of early recognition, prompt diagnosis, and interdisciplinary management in such complex cases.
Collapse
Affiliation(s)
- Vikas Kilaru
- Internal Medicine, Northeast Georgia Medical Center Gainesville, Gainesville, USA
| | - Sharvil Patel
- Internal Medicine, Northeast Georgia Medical Center Gainesville, Gainesville, USA
| | - Hely Patel
- Pediatrics, Emory University, Atlanta, USA
| | - Nima Ghasemzadeh
- Interventional Cardiology, Georgia Heart Institute, Gainesville, USA
| |
Collapse
|
39
|
Botezatu SB, Yu X, Meah MN, Williams MC, Dey D, Newby DE, Tzolos E, Dweck MR. Aortic valve perivascular adipose tissue computed tomography attenuation in patients with aortic stenosis. Heart 2024; 110:657-665. [PMID: 37918901 DOI: 10.1136/heartjnl-2023-323217] [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: 07/14/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVE Aortic stenosis (AS) shares pathophysiological similarities with atherosclerosis including active inflammation. CT attenuation of perivascular adipose tissue provides a measure of vascular inflammation that is linked to prognosis and has the potential to be applied to the aortic valve. We investigated perivascular adipose tissue attenuation around the aortic valve in patients with AS. METHODS CT attenuation was measured in the perivascular adipose tissue extending 3 mm radially and 10 mm longitudinally around the aortic valve in patients with and without AS. Associations between perivascular adipose tissue attenuation and AS disease severity, activity and progression were investigated. RESULTS Perivascular adipose tissue attenuation around the aortic valve demonstrated good intraobserver and interobserver repeatability (interobserver: intraclass correlation coefficient 0.977 (95% CI: 0.94, 0.99)) but was similar between patients with AS (n=120) and control subjects (n=80) (-62.4 (-68.7, -56.5) Hounsfield units (HU) vs -61.2 (-65.3, -55.6) HU, p=0.099). There were no differences between perivascular adipose tissue attenuation in patients with mild (-60.2 (-66.9, -55.1) HU), moderate (-62.8 (-69.6, -56.80) HU) or severe (-62.3 (-69.3, -55.4) HU) AS (all p>0.05), and perivascular adipose tissue attenuation did not demonstrate an association with AS severity as assessed by echocardiography or CT calcium scoring, nor with disease activity assessed by 18F-sodium fluoride positron emission tomography. Moreover, there was no association between baseline aortic valve perivascular adipose tissue attenuation and subsequent AS progression (annualised change in peak velocity: r=0.072, p=0.458). Similar results were found using five other image analysis methods. CONCLUSIONS CT-derived aortic valve perivascular adipose tissue attenuation is not associated with AS disease severity, activity or progression suggesting that it has no value in the investigation and management of patients with AS.
Collapse
Affiliation(s)
- Simona B Botezatu
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
- Cardiology Department, Euroecolab, University of Medicine and Pharmacy 'Carol Davila', Bucharest, Romania
| | - Xinming Yu
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Mohammed N Meah
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Michelle C Williams
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Damini Dey
- Biomedical Imaging Research Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Evangelos Tzolos
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Marc R Dweck
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
40
|
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: 2] [Impact Index Per Article: 2.0] [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
|
41
|
Chyrchel M, Siłka W, Wylaź M, Wójcik W, Surdacki A. Electrocardiography versus Echocardiography in Severe Aortic Stenosis with the Consideration of Coexistent Coronary Artery Disease. J Clin Med 2024; 13:1013. [PMID: 38398326 PMCID: PMC10888567 DOI: 10.3390/jcm13041013] [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: 12/31/2023] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
(1) Background: Coexistent coronary artery disease (CAD) might influence the ability of electrocardiogram (ECG) to identify echocardiographic left ventricular hypertrophy (ECHO-LVH) in patients with aortic stenosis (AS). We aimed to assess the relation between ECG-LVH (by the Sokolov-Lyon or Cornell criteria) and ECHO-LVH considering coexistent CAD. (2) Methods: We retrospectively analyzed the medical records of 74 patients (36 males) with severe AS who were hospitalized in the University Hospital in Cracow from 2021 to 2022. (3) Results: ECHO-LVH was present in 49 (66%) patients, whereas 35 (47.3%) patients had ECG-LVH. There was no difference between the rate of ECG-LVH in patients with vs. without ECHO-LVH. Single-vessel and multi-vessel CAD were diagnosed by invasive coronary angiography in 18% and 11% of patients, respectively. The sensitivity of the classical ECG-LVH criteria with regard to ECHO-LVH was low, reaching at best 41% for the Sokolov-Lyon and Cornell criteria. The results were similar and lacked a pattern when considering patients without significant stenosis, with single- and multi-vessel disease separately. Correlations between the left ventricular mass index and ECG-derived parameters were weak and present solely for the Lewis index (r = 0.31), R wave's amplitude >1.1 mV in aVL (r = 0.36), as well as the Cornell (r = 0.32) and Sokolov-Lyon (r = 0.31) voltage criteria (p < 0.01). The presence, location of stenoses, and CAD extent were not associated with the presence of either ECHO-LVH or ECG-LVH, irrespective of individual ECG-LVH criteria. (4) Conclusions: The sensitivity of classical ECG criteria for echocardiographic LVH in severe AS is low, regardless of coexistent CAD or its angiographic extent.
Collapse
Affiliation(s)
- Michał Chyrchel
- Second Department of Cardiology, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, 30-688 Cracow, Poland; (M.C.); (A.S.)
- Department of Cardiology and Cardiovascular Interventions, University Hospital, 30-688 Cracow, Poland
| | - Wojciech Siłka
- Students’ Scientific Group, Second Department of Cardiology, Jagiellonian University Medical College, 30-688 Cracow, Poland; (M.W.); (W.W.)
| | - Mateusz Wylaź
- Students’ Scientific Group, Second Department of Cardiology, Jagiellonian University Medical College, 30-688 Cracow, Poland; (M.W.); (W.W.)
| | - Wiktor Wójcik
- Students’ Scientific Group, Second Department of Cardiology, Jagiellonian University Medical College, 30-688 Cracow, Poland; (M.W.); (W.W.)
| | - Andrzej Surdacki
- Second Department of Cardiology, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, 30-688 Cracow, Poland; (M.C.); (A.S.)
- Department of Cardiology and Cardiovascular Interventions, University Hospital, 30-688 Cracow, Poland
| |
Collapse
|
42
|
Al‐Kassou B, Al‐Kassou L, Mahn T, Lütjohann D, Shamekhi J, Willemsen N, Niepmann ST, Baldus S, Kelm M, Nickenig G, Latz E, Zimmer S. Cholesterol Crystal Dissolution Rate of Serum Predicts Outcomes in Patients With Aortic Stenosis Undergoing Transcatheter Aortic Valve Replacement. J Am Heart Assoc 2024; 13:e031997. [PMID: 38240198 PMCID: PMC11056150 DOI: 10.1161/jaha.123.031997] [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: 08/01/2023] [Accepted: 12/12/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND Aortic stenosis has pathophysiological similarities with atherosclerosis, including the deposition of cholesterol-containing lipoproteins. The resulting cholesterol crystals activate the NLRP3 (NOD-like receptor protein 3) inflammasome, leading to inflammation and cardiovascular diseases. We aimed to investigate the cholesterol crystal dissolution rate (CCDR) of serum in patients with aortic stenosis and to assess the prognostic value of this biomarker. METHODS AND RESULTS The study included 348 patients with aortic stenosis undergoing transcatheter aortic valve replacement. The CCDR was measured using flow cytometry to enumerate cholesterol crystals that were added to a serum solution, at baseline and after 2 hours of incubation. Based on the median CCDR, the cohort was stratified into high and low cholesterol crystal dissolvers. The incidence of the primary end point, a composite of 1-year all-cause mortality and major vascular complication, was significantly lower in the high CCDR group (7.3 per 100 person-years) compared with the low CCDR group (17.0 per 100 person-years, P=0.01). This was mainly driven by a lower 1-year mortality rate in patients with a high CCDR (7.3 versus 15.1 per 100 person-years, P=0.04). Unplanned endovascular interventions were significantly less frequent in high cholesterol crystal dissolvers (12.8 versus 22.6 per 100 person-years, P=0.04). Although low-density lipoprotein cholesterol levels were comparable in both groups (101.8±37.3 mg/dL versus 97.9±37.6 mg/dL, P=0.35), only patients with a low CCDR showed a benefit from statin treatment. In multivariate analysis, low CCDR (hazard ratio, 2.21 [95% CI, 0.99-4.92], P=0.04) was significantly associated with 1-year mortality. CONCLUSIONS The CCDR is a novel biomarker associated with outcome in patients with aortic stenosis undergoing transcatheter aortic valve replacement. It may provide new insights into patients' anti-inflammatory capacity and additional prognostic information beyond classic risk assessment.
Collapse
Affiliation(s)
- Baravan Al‐Kassou
- Heart Center, Department of Medicine IIUniversity Hospital BonnBonnGermany
| | - Lara Al‐Kassou
- Heart Center, Department of Medicine IIUniversity Hospital BonnBonnGermany
| | - Thorsten Mahn
- Heart Center, Department of Medicine IIUniversity Hospital BonnBonnGermany
| | - Dieter Lütjohann
- Institute of Clinical Chemistry und Clinical PharmacologyUniversity Hospital BonnBonnGermany
| | - Jasmin Shamekhi
- Heart Center, Department of Medicine IIUniversity Hospital BonnBonnGermany
| | - Nicola Willemsen
- Heart Center, Department of Medicine IIUniversity Hospital BonnBonnGermany
| | | | - Stephan Baldus
- Department of Cardiology, Heart CenterUniversity of CologneGermany
| | - Malte Kelm
- Division of CardiologyUniversity Hospital of DuesseldorfGermany
- CARID, Cardiovascular Research Institute DuesseldorfGermany
| | - Georg Nickenig
- Heart Center, Department of Medicine IIUniversity Hospital BonnBonnGermany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospitals BonnBonnGermany
- German Center of Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Infectious Diseases and ImmunologyUMass Medical SchoolWorcesterMA
| | - Sebastian Zimmer
- Heart Center, Department of Medicine IIUniversity Hospital BonnBonnGermany
| |
Collapse
|
43
|
Sastre-Oliva T, Corbacho-Alonso N, Rodriguez-Sanchez E, Mercado-García E, Perales-Sanchez I, Hernandez-Fernandez G, Juarez-Alia C, Tejerina T, López-Almodóvar LF, Padial LR, Sánchez PL, Martín-Núñez E, López-Andrés N, Ruiz-Hurtado G, Mourino-Alvarez L, Barderas MG. Albumin Redox Modifications Promote Cell Calcification Reflecting the Impact of Oxidative Status on Aortic Valve Disease and Atherosclerosis. Antioxidants (Basel) 2024; 13:108. [PMID: 38247532 PMCID: PMC10812654 DOI: 10.3390/antiox13010108] [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: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
Calcific aortic valve disease (CAVD) and coronary artery disease (CAD) are related cardiovascular diseases in which common mechanisms lead to tissue calcification. Oxidative stress plays a key role in these diseases and there is also evidence that the redox state of serum albumin exerts a significant influence on these conditions. To further explore this issue, we used multimarker scores (OxyScore and AntioxyScore) to assess the global oxidative status in patients with CAVD, with and without CAD, also evaluating their plasma thiol levels. In addition, valvular interstitial cells were treated with reduced, oxidized, and native albumin to study how this protein and its modifications affect cell calcification. The differences we found suggest that oxidative status is distinct in CAVD and CAD, with differences in redox markers and thiol levels. Importantly, the in vitro interstitial cell model revealed that modified albumin affects cell calcification, accelerating this process. Hence, we show here the importance of the redox system in the development of CAVD, emphasizing the relevance of multimarker scores, while also offering evidence of how the redox state of albumin influences vascular calcification. These data highlight the relevance of understanding the overall redox processes involved in these diseases, opening the door to new studies on antioxidants as potential therapies for these patients.
Collapse
Affiliation(s)
- Tamara Sastre-Oliva
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Servicio de Salud de Castilla-La Mancha (SESCAM), 45071 Toledo, Spain; (T.S.-O.); (N.C.-A.); (I.P.-S.); (G.H.-F.); (C.J.-A.); (L.M.-A.)
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Nerea Corbacho-Alonso
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Servicio de Salud de Castilla-La Mancha (SESCAM), 45071 Toledo, Spain; (T.S.-O.); (N.C.-A.); (I.P.-S.); (G.H.-F.); (C.J.-A.); (L.M.-A.)
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Elena Rodriguez-Sanchez
- Cardiorenal Translational Laboratory, Instituto de Investigación Imas12, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain (E.M.-G.); (G.R.-H.)
| | - Elisa Mercado-García
- Cardiorenal Translational Laboratory, Instituto de Investigación Imas12, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain (E.M.-G.); (G.R.-H.)
| | - Ines Perales-Sanchez
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Servicio de Salud de Castilla-La Mancha (SESCAM), 45071 Toledo, Spain; (T.S.-O.); (N.C.-A.); (I.P.-S.); (G.H.-F.); (C.J.-A.); (L.M.-A.)
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - German Hernandez-Fernandez
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Servicio de Salud de Castilla-La Mancha (SESCAM), 45071 Toledo, Spain; (T.S.-O.); (N.C.-A.); (I.P.-S.); (G.H.-F.); (C.J.-A.); (L.M.-A.)
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Cristina Juarez-Alia
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Servicio de Salud de Castilla-La Mancha (SESCAM), 45071 Toledo, Spain; (T.S.-O.); (N.C.-A.); (I.P.-S.); (G.H.-F.); (C.J.-A.); (L.M.-A.)
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Teresa Tejerina
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Luis F. López-Almodóvar
- Cardiac Surgery, Hospital General Universitario de Toledo, Servicio de Salud de Castilla-La Mancha (SESCAM), 45007 Toledo, Spain;
| | - Luis R. Padial
- Department of Cardiology, Hospital General Universitario de Toledo, Servicio de Salud de Castilla-La Mancha (SESCAM), 45007 Toledo, Spain;
| | - Pedro L. Sánchez
- Department of Cardiology, Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ernesto Martín-Núñez
- Cardiovascular Translational Research, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (E.M.-N.); (N.L.-A.)
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (E.M.-N.); (N.L.-A.)
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Instituto de Investigación Imas12, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain (E.M.-G.); (G.R.-H.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Laura Mourino-Alvarez
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Servicio de Salud de Castilla-La Mancha (SESCAM), 45071 Toledo, Spain; (T.S.-O.); (N.C.-A.); (I.P.-S.); (G.H.-F.); (C.J.-A.); (L.M.-A.)
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Maria G. Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Servicio de Salud de Castilla-La Mancha (SESCAM), 45071 Toledo, Spain; (T.S.-O.); (N.C.-A.); (I.P.-S.); (G.H.-F.); (C.J.-A.); (L.M.-A.)
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| |
Collapse
|
44
|
Dolmaci OB, Hilhorst TL, Malekzadeh A, Mertens BJ, Klautz RJ, Poelmann RE, Grewal N. The Prevalence of Coronary Artery Disease in Bicuspid Aortic Valve Patients: An Overview of the Literature. AORTA (STAMFORD, CONN.) 2023; 11:191-197. [PMID: 38698623 PMCID: PMC11219131 DOI: 10.1055/s-0044-1785190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/07/2024] [Indexed: 05/05/2024]
Abstract
The prevalence of coronary artery disease (CAD) in bicuspid aortic valve (BAV) patients is a debatable topic. Several studies have indicated that BAV patients have a lower prevalence of CAD compared with patients with a tricuspid aortic valve (TAV), but the effects of age and gender have not always been considered. This systematic review provides an overview of articles which report on CAD in BAV and TAV patients. Searches were executed in April 2021 and January 2022 according to the PRISMA (Preferred Reporting Items for Systematic reviews and Meta-analyses) guidelines in three online databases: Medline, Embase, and Scopus. Screening and data extraction was done by two investigators separately. Primary and secondary outcomes were compared between BAV and TAV patients; a fixed effects model was used for correcting on confounders. Literature search yielded 1,529 articles with 44 being eligible for inclusion. BAV patients were younger (56.4 ± 8.3 years) than TAV patients (64 ± 10.3 years, p < 0.001). All CAD risk factors and CAD were more prevalent in TAV patients. No significant difference remained after correcting for age and gender as confounders. BAV patients have a lower prevalence of CAD and CAD risk factors compared with TAV patients. However, when the age differences between both groups are considered in the analyses, a similar prevalence of both CAD and CAD risk factors is found.
Collapse
Affiliation(s)
- Onur B. Dolmaci
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Tijmen L. Hilhorst
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Arjan Malekzadeh
- Medical Library, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, Netherlands
| | - Bart J.A. Mertens
- Department of Statistics, Leiden University Medical Center, Leiden, Netherlands
| | - Robert J.M. Klautz
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Robert E. Poelmann
- Institute of Biology, Leiden University, Sylvius Laboratory, Leiden, Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nimrat Grewal
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
45
|
Hansén N, Ljungberg J, Bergdahl IA, Hultdin J, Näslund U, Johansson B, Söderberg S. Adipokines are possible risk markers for aortic stenosis requiring surgery. SCAND CARDIOVASC J 2023; 57:2247193. [PMID: 37592808 DOI: 10.1080/14017431.2023.2247193] [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: 02/06/2023] [Revised: 06/15/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023]
Abstract
OBJECTIVES Aortic stenosis (AS) is the most prevalent valvular heart disease among adults. The adipocyte-derived hormones, leptin and adiponectin, have profound metabolic actions. We examined whether these adipokines are independently associated with future aortic valve replacement (AVR). DESIGN In this longitudinal case-control study, we identified 336 cases who had undergone AVR due to AS, and who had previously participated in population-based health surveys. Two referents were matched to each case and leptin and adiponectin concentrations were analysed from stored baseline survey samples. Uni- and multivariable logistic regression analyses were used to estimate the risk of future AVR. An additional cohort was identified for validation including 106 cases with AVR and 212 matched referents. RESULTS Median age (interquartile range (IQR)) in years at survey was 59.9 (10.4) and at surgery 68.3 (12.7), and 48% were women. An elevated concentration of leptin was not associated with future AVR (odds ratio [95% confidence interval]) (1.10 [0.92-1.32]), although leptin was associated with a higher risk in patients with coronary artery disease (CAD) having more than 5 years between survey and AVR (1.41 [1.08-1.84]). Adiponectin was not associated with higher risk for future AVR (0.95 [0.82-1.11]), although after stratification for age, higher levels were associated with reduced risk for AVR in persons aged ≥60 years at surgery (0.79 [0.64-0.98]). In the validation study, leptin was associated with future AVR whereas adiponectin was not. None of the associations remained significant after adjustment for body mass index (BMI). CONCLUSIONS The adipokine leptin may promote the development of AS.
Collapse
Affiliation(s)
- Nike Hansén
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | - Johan Ljungberg
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | | | - Johan Hultdin
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Ulf Näslund
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | - Bengt Johansson
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | - Stefan Söderberg
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
46
|
Feldmann A, Nitschke Y, Linß F, Mulac D, Stücker S, Bertrand J, Buers I, Langer K, Rutsch F. Improved Reversion of Calcifications in Porcine Aortic Heart Valves Using Elastin-Targeted Nanoparticles. Int J Mol Sci 2023; 24:16471. [PMID: 38003660 PMCID: PMC10671589 DOI: 10.3390/ijms242216471] [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: 09/29/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Calcified aortic valve disease in its final stage leads to aortic valve stenosis, limiting cardiac function. To date, surgical intervention is the only option for treating calcific aortic valve stenosis. This study combined controlled drug delivery by nanoparticles (NPs) and active targeting by antibody conjugation. The chelating agent diethylenetriaminepentaacetic acid (DTPA) was covalently bound to human serum albumin (HSA)-based NP, and the NP surface was modified using conjugating antibodies (anti-elastin or isotype IgG control). Calcification was induced ex vivo in porcine aortic valves by preincubation in an osteogenic medium containing 2.5 mM sodium phosphate for five days. Valve calcifications mainly consisted of basic calcium phosphate crystals. Calcifications were effectively resolved by adding 1-5 mg DTPA/mL medium. Incubation with pure DTPA, however, was associated with a loss of cellular viability. Reversal of calcifications was also achieved with DTPA-coupled anti-elastin-targeted NPs containing 1 mg DTPA equivalent. The addition of these NPs to the conditioned media resulted in significant regression of the valve calcifications compared to that in the IgG-NP control without affecting cellular viability. These results represent a step further toward the development of targeted nanoparticular formulations to dissolve aortic valve calcifications.
Collapse
Affiliation(s)
- Anja Feldmann
- Department of General Pediatrics, Muenster University Children’s Hospital, D-48149 Muenster, Germany; (A.F.); (Y.N.); (I.B.)
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
| | - Yvonne Nitschke
- Department of General Pediatrics, Muenster University Children’s Hospital, D-48149 Muenster, Germany; (A.F.); (Y.N.); (I.B.)
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
| | - Franziska Linß
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, D-48149 Muenster, Germany; (D.M.); (K.L.)
| | - Dennis Mulac
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, D-48149 Muenster, Germany; (D.M.); (K.L.)
| | - Sina Stücker
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
- Department of Orthopaedic Surgery, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany
| | - Jessica Bertrand
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
- Department of Orthopaedic Surgery, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany
| | - Insa Buers
- Department of General Pediatrics, Muenster University Children’s Hospital, D-48149 Muenster, Germany; (A.F.); (Y.N.); (I.B.)
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
| | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, D-48149 Muenster, Germany; (D.M.); (K.L.)
| | - Frank Rutsch
- Department of General Pediatrics, Muenster University Children’s Hospital, D-48149 Muenster, Germany; (A.F.); (Y.N.); (I.B.)
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
| |
Collapse
|
47
|
Scalia IG, Farina JM, Padang R, Jokerst CE, Pereyra M, Mahmoud AK, Naqvi TZ, Chao CJ, Oh JK, Arsanjani R, Ayoub C. Aortic Valve Calcium Score by Computed Tomography as an Adjunct to Echocardiographic Assessment-A Review of Clinical Utility and Applications. J Imaging 2023; 9:250. [PMID: 37998097 PMCID: PMC10672559 DOI: 10.3390/jimaging9110250] [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: 09/01/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/25/2023] Open
Abstract
Aortic valve stenosis (AS) is increasing in prevalence due to the aging population, and severe AS is associated with significant morbidity and mortality. Echocardiography remains the mainstay for the initial detection and diagnosis of AS, as well as for grading of severity. However, there are important subgroups of patients, for example, patients with low-flow low-gradient or paradoxical low-gradient AS, where quantification of severity of AS is challenging by echocardiography and underestimation of severity may delay appropriate management and impart a worse prognosis. Aortic valve calcium score by computed tomography has emerged as a useful clinical diagnostic test that is complimentary to echocardiography, particularly in cases where there may be conflicting data or clinical uncertainty about the degree of AS. In these situations, aortic valve calcium scoring may help re-stratify grading of severity and, therefore, further direct clinical management. This review presents the evolution of aortic valve calcium score by computed tomography, its diagnostic and prognostic value, as well as its utility in clinical care.
Collapse
Affiliation(s)
- Isabel G. Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Juan M. Farina
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Ratnasari Padang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Milagros Pereyra
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Ahmed K. Mahmoud
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Tasneem Z. Naqvi
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Chieh-Ju Chao
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jae K. Oh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| |
Collapse
|
48
|
Tagzirt M, Rosa M, Corseaux D, Vincent F, Vincentelli A, Daoudi M, Jashari R, Staels B, Van Belle E, Susen S, Dupont A. Modulation of inflammatory M1-macrophages phenotype by valvular interstitial cells. J Thorac Cardiovasc Surg 2023; 166:e377-e389. [PMID: 36182586 DOI: 10.1016/j.jtcvs.2022.08.027] [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: 03/03/2022] [Revised: 08/01/2022] [Accepted: 08/25/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Aortic valve stenosis involves inflammation, excess deposition of a collagen-rich extracellular matrix, and calcification. Recent studies have shown that M1 or inflammatory macrophages derived from infiltrating monocytes promote calcification of valvular interstitial cells, the most prevalent cell type of the aortic valve. We hypothesized that valvular interstitial cells could modulate inflammatory macrophages phenotype. METHODS We first assessed macrophage phenotype in human aortic valve stenosis and control aortic valves from donors. Then, we examined profibrotic and inflammatory-related gene expression in valves and valvular interstitial cells. Finally, we investigated whether valvular interstitial cells can modify the phenotype of inflammatory macrophages. RESULTS Circulating monocytes and plasma transforming growth factor beta-1 levels of patients with aortic valve stenosis were significantly higher compared with patients without aortic valve stenosis. Histologic analysis of thickened spongiosa of the aortic valve from patients with aortic valve stenosis showed a high macrophage infiltration but a low matrix metalloproteinase-9 expression compared with control aortic valves. On the other hand, valvular interstitial cell culture of aortic valve stenosis exhibited a profibrotic phenotype with a high expression of transforming growth factor beta-1 and transforming growth factor beta-1/transforming growth factor beta-3 ratio but a decreased expression of the peroxisome proliferator-activated receptor gamma nuclear receptor. Valvular interstitial cell-conditioned media of aortic valve stenosis led to a decrease in enzymatic activity of matrix metalloproteinase-9 and an increase in production of collagen in inflammatory macrophages compared with valvular interstitial cell-conditioned media from control aortic valve donors. CONCLUSIONS These findings indicate that profibrotic valvular interstitial cells promote the imbalance of extracellular matrix remodeling by reducing matrix metalloproteinase-9 production on inflammatory macrophages that lead to excessive collagen deposition observed in aortic valve stenosis. Further investigation is needed to clarify the role of transforming growth factor beta-1/proliferator-activated receptor gamma nuclear receptor/matrix metalloproteinase-9 in aortic valve stenosis.
Collapse
Affiliation(s)
- Madjid Tagzirt
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France.
| | - Mickael Rosa
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Delphine Corseaux
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Flavien Vincent
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - André Vincentelli
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | | | - Ramadan Jashari
- European Homograft Bank, Clinic Saint Jean, Brussels, Belgium
| | - Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Eric Van Belle
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Sophie Susen
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Annabelle Dupont
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
49
|
De Biase N, Mazzola M, Del Punta L, Di Fiore V, De Carlo M, Giannini C, Costa G, Paneni F, Mengozzi A, Nesti L, Gargani L, Masi S, Pugliese NR. Haemodynamic and metabolic phenotyping of patients with aortic stenosis and preserved ejection fraction: A specific phenotype of heart failure with preserved ejection fraction? Eur J Heart Fail 2023; 25:1947-1958. [PMID: 37655676 DOI: 10.1002/ejhf.3018] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/22/2023] [Accepted: 08/26/2023] [Indexed: 09/02/2023] Open
Abstract
AIMS Degenerative aortic valve stenosis with preserved ejection fraction (ASpEF) and heart failure with preserved ejection fraction (HFpEF) display intriguing similarities. This study aimed to provide a non-invasive, comparative analysis of ASpEF versus HFpEF at rest and during exercise. METHODS AND RESULTS We prospectively enrolled 148 patients with HFpEF and 150 patients with degenerative moderate-to-severe ASpEF, together with 66 age- and sex-matched healthy controls. All subjects received a comprehensive evaluation at rest and 351/364 (96%) performed a combined cardiopulmonary exercise stress echocardiography test. Patients with ASpEF eligible for transcatheter aortic valve replacement (n = 125) also performed cardiac computed tomography (CT). HFpEF and ASpEF patients showed similar demographic distribution and biohumoral profiles. Most patients with ASpEF (134/150, 89%) had severe high-gradient aortic stenosis; 6/150 (4%) had normal-flow, low-gradient ASpEF, while 10/150 (7%) had low-flow, low-gradient ASpEF. Both patient groups displayed significantly lower peak oxygen consumption (VO2 ), peak cardiac output, and peak arteriovenous oxygen difference compared to controls (all p < 0.01). ASpEF patients showed several extravalvular abnormalities at rest and during exercise, similar to HFpEF (all p < 0.01 vs. controls). Epicardial adipose tissue (EAT) thickness was significantly greater in ASpEF than HFpEF and was inversely correlated with peak VO2 in all groups. In ASpEF, EAT was directly related to echocardiography-derived disease severity and CT-derived aortic valve calcium burden. CONCLUSION Functional capacity is similarly impaired in ASpEF and HFpEF due to both peripheral and central components. Further investigation is warranted to determine whether extravalvular alterations may affect disease progression and prognosis in ASpEF even after valve intervention, which could support the concept of ASpEF as a specific sub-phenotype of HFpEF.
Collapse
Affiliation(s)
- Nicolò De Biase
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Mazzola
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Lavinia Del Punta
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Valerio Di Fiore
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marco De Carlo
- Cardiac, Thoracic and Vascular Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Cristina Giannini
- Cardiac, Thoracic and Vascular Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Giulia Costa
- Cardiac, Thoracic and Vascular Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Lorenzo Nesti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luna Gargani
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | |
Collapse
|
50
|
Feistner L, Penk A, Böttner J, Büttner P, Thiele H, Huster D, Schlotter F. Nuclear magnetic resonance spectroscopy to quantify major extracellular matrix components in fibro-calcific aortic valve disease. Sci Rep 2023; 13:18823. [PMID: 37914797 PMCID: PMC10620231 DOI: 10.1038/s41598-023-46143-7] [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: 07/05/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023] Open
Abstract
Fibro-calcific aortic valve disease (FCAVD) is a pathological condition marked by overt fibrous and calcific extracellular matrix (ECM) accumulation that leads to valvular dysfunction and left ventricular outflow obstruction. Costly valve implantation is the only approved therapy. Multiple pharmacological interventions are under clinical investigation, however, none has proven clinically beneficial. This failure of translational approaches indicates incomplete understanding of the underlying pathomechanisms and may result from a limited toolbox of scientific methods to assess the cornerstones of FCAVD: lipid deposition, fibrous and calcific ECM accumulation. In this study, we evaluated magic-angle spinning (MAS) nuclear magnetic resonance (NMR) spectroscopy to both, qualitatively and quantitatively assess these key elements of FCAVD pathogenesis. NMR spectra showed collagen, elastin, triacylglycerols, and phospholipids in human control and FCAVD tissue samples (n = 5). Calcification, measured by the hydroxyapatite content, was detectable in FCAVD tissues and in valve interstitial cells under procalcifying media conditions. Hydroxyapatite was significantly higher in FCAVD tissues than in controls (p < 0.05) as measured by 31P MAS NMR. The relative collagen content was lower in FCAVD tissues vs. controls (p < 0.05). Overall, we demonstrate the versatility of NMR spectroscopy as a diagnostic tool in preclinical FCAVD assessment.
Collapse
Affiliation(s)
- Lukas Feistner
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Struempellstr. 39, 04289, Leipzig, Germany
| | - Anja Penk
- Institute for Medical Physics and Biophysics, Leipzig University, Leipzig, Germany
| | - Julia Böttner
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Struempellstr. 39, 04289, Leipzig, Germany
| | - Petra Büttner
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Struempellstr. 39, 04289, Leipzig, Germany
| | - Holger Thiele
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Struempellstr. 39, 04289, Leipzig, Germany
| | - Daniel Huster
- Institute for Medical Physics and Biophysics, Leipzig University, Leipzig, Germany
| | - Florian Schlotter
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Struempellstr. 39, 04289, Leipzig, Germany.
| |
Collapse
|