1
|
Wang X, Li Y, Cao Z, Li Y, Cao J, Wang Y, Li M, Zheng J, Peng S, Shi W, Wu Q, Yang J, Fang Y, Zhang A, Zhang X, Wang B. Development and external validation of a machine learning model for cardiac valve calcification early screening in dialysis patients: a multicenter study. Ren Fail 2025; 47:2491656. [PMID: 40275572 PMCID: PMC12035951 DOI: 10.1080/0886022x.2025.2491656] [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: 10/22/2024] [Revised: 03/22/2025] [Accepted: 04/06/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Cardiac valve calcification (CVC) is common in dialysis patients and associated with increased cardiovascular risk. However, early screening has been limited by cost concerns. This study aimed to develop and validate a machine learning model to enhance early detection of CVC. METHODS Data were collected at four centers between 2020 and 2023, including 852 dialysis patients in the development dataset and 661 in the external validation dataset. Predictive factors were selected using LASSO regression combined with univariate and multivariate analyses. Machine learning models including CatBoost, XGBoost, decision tree, support vector machine, random forest, and logistic regression were used to develop the CVC risk model. Model performance was evaluated in both validation sets. Risk thresholds were defined using the Youden index and validated in the external dataset. RESULTS In the development dataset, 32.9% of patients were diagnosed with CVC. Age, dialysis duration, alkaline phosphatase, apolipoprotein A1, and intact parathyroid hormone were selected to construct the CVC risk prediction model. CatBoost exhibited the best performance in the training dataset. The logistic regression model demonstrated the best predictive performance in both internal and external validation sets, with AUROCs of 0.806 (95% CI 0.750-0.863) and 0.757 (95% CI 0.720-0.793), respectively. Calibration curves and decision curves confirmed its predictive accuracy and clinical applicability. The logistic regression model was selected as the optimal model and achieved excellent risk stratification in CVC risk prediction. CONCLUSION The predictive model effectively identifies CVC risk in dialysis patients and offers a robust tool for early detection and improved management.
Collapse
Affiliation(s)
- Xiaoxu Wang
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
| | - Yinfang Li
- Department of Pediatric, The Second Affiliated Hospital of Nanjing Medical University, School of Pediatric, Nanjing Medical University, Nanjing, P.R. China
| | - Zixin Cao
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
| | - Yunuo Li
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
| | - Jingyuan Cao
- Department of Nephrology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, P.R. China
| | - Yao Wang
- Department of Nephrology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, P.R. China
| | - Min Li
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Soochow University, Changzhou, P.R. China
| | - Jing Zheng
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
- Department of Gerontology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
| | - Siqi Peng
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
| | - Wen Shi
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
| | - Qianqian Wu
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
| | - Junlan Yang
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
| | - Yaping Fang
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
| | - Aiqing Zhang
- Department of Pediatric, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, P.R. China
| | - Xiaoliang Zhang
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
| | - Bin Wang
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, P.R. China
| |
Collapse
|
2
|
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
|
3
|
Kajuluri LP, Guo YY, Lee S, Christof M, Malhotra R. Epigenetic Regulation of Human Vascular Calcification. Genes (Basel) 2025; 16:506. [PMID: 40428328 PMCID: PMC12111397 DOI: 10.3390/genes16050506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Vascular diseases present a significant threat to human health worldwide. Atherosclerosis is the most prevalent vascular disease, accounting for the majority of morbidity and mortality globally. Vascular calcification is a dynamic pathological process underlying the development of atherosclerotic plaques and involves the phenotypic transformation of vascular smooth muscle cells (VSMCs) into osteogenic cells. Specifically, the phenotypic switch in VSMCs often involves modifications in gene expression due to epigenetic changes, including DNA methylation, histone modification, and non-coding RNAs. Understanding the role of these epigenetic changes in regulating the pathophysiology of vascular calcification, along with the proteins and pathways that mediate these changes, will aid in identifying new therapeutic candidates to enhance vascular health. This review discusses a comprehensive range of epigenetic modifications and their implications for vascular health and the development of vascular calcification.
Collapse
Affiliation(s)
- Lova Prasadareddy Kajuluri
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| | - Yugene Young Guo
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| | - Sujin Lee
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| | - Michael Christof
- School of Arts and Sciences, University of Rochester, Rochester, NY 14627, USA;
| | - Rajeev Malhotra
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| |
Collapse
|
4
|
Otto CM, Newby DE, Hillis GS. Calcific Aortic Stenosis: A Review. JAMA 2024; 332:2014-2026. [PMID: 39527048 DOI: 10.1001/jama.2024.16477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Importance Calcific aortic stenosis (AS) restricts the aortic valve opening during systole due to calcification and fibrosis of either a congenital bicuspid or a normal trileaflet aortic valve. In the US, AS affects 1% to 2% of adults older than 65 years and approximately 12% of adults older than 75 years. Worldwide, AS leads to more than 100 000 deaths annually. Observations Calcific AS is characterized by aortic valve leaflet lipid infiltration and inflammation with subsequent fibrosis and calcification. Symptoms due to severe AS, such as exercise intolerance, exertional dyspnea, and syncope, are associated with a 1-year mortality rate of up to 50% without aortic valve replacement. Echocardiography can detect AS and measure the severity of aortic valve dysfunction. Although progression rates vary, once aortic velocity is higher than 2 m/s, progression to severe AS occurs typically within 10 years. Severe AS is defined by an aortic velocity 4 m/s or higher, a mean gradient 40 mm Hg or higher, or a valve area less than or equal to 1.0 cm2. Management of mild to moderate AS and asymptomatic severe AS consists of patient education about the typical progression of disease; clinical and echocardiographic surveillance at intervals of 3 to 5 years for mild AS, 1 to 2 years for moderate AS, and 6 to 12 months for severe AS; and treatment of hypertension, hyperlipidemia, and cigarette smoking as indicated. When a patient with severe AS develops symptoms, surgical aortic valve replacement (SAVR) or transcatheter aortic valve implantation (TAVI) is recommended, which restores an average life expectancy; in patients aged older than 70 years with a low surgical risk, 10-year all-cause mortality was 62.7% with TAVI and 64.0% with SAVR. TAVI is associated with decreased length of hospitalization, more rapid return to normal activities, and less pain compared with SAVR. However, evidence supporting TAVI for patients aged younger than 65 years and long-term outcomes of TAVI are less well defined than for SAVR. For patients with symptomatic severe AS, the 2020 American College of Cardiology/American Heart Association guideline recommends SAVR for individuals aged 65 years and younger, SAVR or TAVI for those aged 66 to 79 years, and TAVI for individuals aged 80 years and older or those with an estimated surgical mortality of 8% or higher. Conclusions Calcific AS is a common chronic progressive condition among older adults and is diagnosed via echocardiography. Symptomatic patients with severe AS have a mortality rate of up to 50% after 1 year, but treatment with SAVR or TAVI reduces mortality to that of age-matched control patients. The type and timing of valve replacement should be built on evidence-based guidelines, shared decision-making, and involvement of a multidisciplinary heart valve team.
Collapse
Affiliation(s)
- Catherine M Otto
- Division of Cardiology, University of Washington School of Medicine, Seattle
| | - David E Newby
- University of Edinburgh, British Heart Foundation Centre of Research Excellence, Royal Infirmary, Edinburgh, United Kingdom
| | - Graham S Hillis
- Department of Cardiology, Royal Perth Hospital and Medical School, University of Western Australia, Perth
| |
Collapse
|
5
|
Vassiliou VS, Johnson N, Langlands K, Tsampasian V. Genetics of Calcific Aortic Stenosis: A Systematic Review. Genes (Basel) 2024; 15:1309. [PMID: 39457433 PMCID: PMC11508093 DOI: 10.3390/genes15101309] [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: 08/30/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Calcific aortic stenosis is the most prevalent valvular abnormality in the Western world. Factors commonly associated with calcific aortic stenosis include advanced age, male sex, hypertension, diabetes and impaired renal function. This review synthesises the existing literature on genetic associations with calcific aortic stenosis. Methods: A systematic search was conducted in the PubMed, Ovid and Cochrane libraries from inception to 21 July 2024 to identify human studies investigating the genetic factors involved in calcific aortic stenosis. From an initial pool of 1392 articles, 78 were selected for full-text review and 31 were included in the final qualitative synthesis. The risk of bias in these studies was assessed using the Newcastle Ottawa Scale. Results: Multiple genes have been associated with calcific aortic stenosis. These genes are involved in different biological pathways, including the lipid metabolism pathway (PLA, LDL, APO, PCSK9, Lp-PLA2, PONS1), the inflammatory pathway (IL-6, IL-10), the calcification pathway (PALMD, TEX41) and the endocrine pathway (PTH, VIT D, RUNX2, CACNA1C, ALPL). Additional genes such as NOTCH1, NAV1 and FADS1/2 influence different pathways. Mechanistically, these genes may promote a pro-inflammatory and pro-calcific environment in the aortic valve itself, leading to increased osteoblastic activity and subsequent calcific degeneration of the valve. Conclusions: Numerous genetic associations contribute to calcific aortic stenosis. Recognition of these associations can enhance risk stratification for individuals and their first-degree relatives, facilitate family screening, and importantly, pave the way for targeted therapeutic interventions focusing on the identified genetic factors. Understanding these genetic factors can also lead to gene therapy to prevent calcific aortic stenosis in the future.
Collapse
Affiliation(s)
- Vassilios S. Vassiliou
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK; (N.J.); (V.T.)
- Fitzwilliam College, University of Cambridge, Cambridge CB3 0DG, UK
| | - Nicholas Johnson
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK; (N.J.); (V.T.)
| | - Kenneth Langlands
- Institute of Continuing Education, University of Cambridge, Cambridge CB23 8AQ, UK;
| | - Vasiliki Tsampasian
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK; (N.J.); (V.T.)
| |
Collapse
|
6
|
Chen M, Liu S. Atorvastatin reduces calcification in valve interstitial cells via the NF-κB signalling pathway by promoting Atg5-mediated autophagy. Eur J Histochem 2024; 68:3983. [PMID: 38619020 PMCID: PMC11110720 DOI: 10.4081/ejh.2024.3983] [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: 01/31/2024] [Accepted: 03/09/2024] [Indexed: 04/16/2024] Open
Abstract
Aortic valve calcification (AVC) is a common cardiovascular disease and a risk factor for sudden death. However, the potential mechanisms and effective therapeutic drugs need to be explored. Atorvastatin is a statin that can effectively prevent cardiovascular events by lowering cholesterol levels. However, whether atorvastatin can inhibit AVC by reducing low-density lipoprotein (LDL) and its possible mechanism of action require further exploration. In the current study, we constructed an in vitro AVC model by inducing calcification of the valve interstitial cells. We found that atorvastatin significantly inhibited osteogenic differentiation, reduced the deposition of calcium nodules in valve interstitial cells, and enhanced autophagy in calcified valve interstitial cells, manifested by increased expression levels of the autophagy proteins Atg5 and LC3B-II/I and the formation of smooth autophagic flow. Atorvastatin inhibited the NF-κB signalling pathway and the expression of inflammatory factors mediated by NF-κB in calcified valve interstitial cells. The activation of the NF-κB signalling pathway led to the reversal of atorvastatin's effect on enhancing autophagy and alleviating valve interstitial cell calcification. In conclusion, atorvastatin inhibited the NF-κB signalling pathway by upregulating autophagy, thereby alleviating valve interstitial cell calcification, which was conducive to improving AVC.
Collapse
Affiliation(s)
- Menghui Chen
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei; Department of Cardiothoracic Surgery, The Third Hospital of Shijiazhuang, Hebei.
| | - Su Liu
- Department of Surgery, Hebei Medical University, Shijiazhuang; Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei.
| |
Collapse
|
7
|
Notenboom ML, Van Hoof L, Schuermans A, Takkenberg JJM, Rega FR, Taverne YJHJ. Aortic Valve Embryology, Mechanobiology, and Second Messenger Pathways: Implications for Clinical Practice. J Cardiovasc Dev Dis 2024; 11:49. [PMID: 38392263 PMCID: PMC10888685 DOI: 10.3390/jcdd11020049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
During the Renaissance, Leonardo Da Vinci was the first person to successfully detail the anatomy of the aortic root and its adjacent structures. Ever since, novel insights into morphology, function, and their interplay have accumulated, resulting in advanced knowledge on the complex functional characteristics of the aortic valve (AV) and root. This has shifted our vision from the AV as being a static structure towards that of a dynamic interconnected apparatus within the aortic root as a functional unit, exhibiting a complex interplay with adjacent structures via both humoral and mechanical stimuli. This paradigm shift has stimulated surgical treatment strategies of valvular disease that seek to recapitulate healthy AV function, whereby AV disease can no longer be seen as an isolated morphological pathology which needs to be replaced. As prostheses still cannot reproduce the complexity of human nature, treatment of diseased AVs, whether stenotic or insufficient, has tremendously evolved, with a similar shift towards treatments options that are more hemodynamically centered, such as the Ross procedure and valve-conserving surgery. Native AV and root components allow for an efficient Venturi effect over the valve to allow for optimal opening during the cardiac cycle, while also alleviating the left ventricle. Next to that, several receptors are present on native AV leaflets, enabling messenger pathways based on their interaction with blood and other shear-stress-related stimuli. Many of these physiological and hemodynamical processes are under-acknowledged but may hold important clues for innovative treatment strategies, or as potential novel targets for therapeutic agents that halt or reverse the process of valve degeneration. A structured overview of these pathways and their implications for cardiothoracic surgeons and cardiologists is lacking. As such, we provide an overview on embryology, hemodynamics, and messenger pathways of the healthy and diseased AV and its implications for clinical practice, by relating this knowledge to current treatment alternatives and clinical decision making.
Collapse
Affiliation(s)
- Maximiliaan L. Notenboom
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| | - Lucas Van Hoof
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Art Schuermans
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Johanna J. M. Takkenberg
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| | - Filip R. Rega
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Yannick J. H. J. Taverne
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| |
Collapse
|
8
|
Elzomor H, Elkoumy A, Hothi SS, Soliman O. Considering alternatives to transcatheter heart valves for managing patients with severe aortic valve stenosis. Expert Rev Med Devices 2024; 21:109-120. [PMID: 38166517 DOI: 10.1080/17434440.2023.2298716] [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: 08/05/2023] [Accepted: 12/20/2023] [Indexed: 01/04/2024]
Abstract
INTRODUCTION Transcatheter aortic valve implantation (TAVI) is becoming the standard of care for severe symptomatic aortic stenosis (AS). Yet, some patients with AS are not indicated/eligible for TAVI. Several noninvasive, catheter-based or surgical alternatives exist, and other therapeutic options are emerging. AREAS COVERED This review provides an overview of non-TAVI options for severe AS. Non-invasive, transcatheter, and alternative surgical strategies are discussed, emphasizing their backgrounds, techniques, and outcomes. EXPERT OPINION Alternative therapies to TAVI, whether device-based or non-device-based, continue to evolve or emerge and provide either alternative treatments or a bridge to TAVI, for patients not meeting indications for, or having contraindications to TAVI.Although TAVI and SAVR are the current dominant therapies, there are still some patients that could benefit in the future from other alternatives.Data on alternative options for such patients are scarce. Many advantages and disadvantages arise when selecting a specific treatment strategy for individual patients.Head-to-head comparison studies could guide physicians toward better patient selection and procedural planning. Awareness of therapeutic options, indications, techniques, and outcomes should enable heart teams to achieve optimized patient selection. Furthermore, it can increase the use of these alternatives to optimize the management of AS among different patient populations.
Collapse
Affiliation(s)
- Hesham Elzomor
- Department of Cardiology, Saolta Group, Galway University Hospital, Galway, Ireland
- CORRIB Core Lab, University of Galway, Galway, Ireland
- Islamic Center of Cardiology, Al-Azhar University, Nasr City, Cairo, Egypt
- CÚRAM, SFI Research Centre for Medical Devices, Galway, Ireland
- Department of Cardiology, Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Ahmed Elkoumy
- Department of Cardiology, Saolta Group, Galway University Hospital, Galway, Ireland
- CORRIB Core Lab, University of Galway, Galway, Ireland
- Islamic Center of Cardiology, Al-Azhar University, Nasr City, Cairo, Egypt
- CÚRAM, SFI Research Centre for Medical Devices, Galway, Ireland
| | - Sandeep S Hothi
- Department of Cardiology, Royal Wolverhampton NHS Trust, Wolverhampton, UK
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, UK
| | - Osama Soliman
- Department of Cardiology, Saolta Group, Galway University Hospital, Galway, Ireland
- CORRIB Core Lab, University of Galway, Galway, Ireland
- Islamic Center of Cardiology, Al-Azhar University, Nasr City, Cairo, Egypt
- Department of Cardiology, Royal Wolverhampton NHS Trust, Wolverhampton, UK
- Euro Heart Foundation, Rotterdam, The Netherlands
| |
Collapse
|
9
|
Murali S, Smith ER, Tiong MK, Tan S, Toussaint ND. Interventions to Attenuate Cardiovascular Calcification Progression: A Systematic Review of Randomized Clinical Trials. J Am Heart Assoc 2023; 12:e031676. [PMID: 38014685 PMCID: PMC10727339 DOI: 10.1161/jaha.123.031676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/23/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Cardiovascular calcification, characterized by deposition of calcium phosphate in the arterial wall and heart valves, is associated with cardiovascular morbidity and mortality and is commonly seen in aging, diabetes, and chronic kidney disease. Whether evidence-based interventions could significantly attenuate cardiovascular calcification progression remains uncertain. METHODS AND RESULTS We conducted a systematic review of randomized controlled trials involving interventions, compared with placebo, another comparator, or standard of care, to attenuate cardiovascular calcification. Included clinical trials involved participants without chronic kidney disease, and the outcome was cardiovascular calcification measured using radiological methods. Quality of evidence was determined by the Cochrane risk of bias and Grading of Recommendations, Assessment, Development, and Evaluations assessment. Forty-nine randomized controlled trials involving 9901 participants (median participants 104, median duration 12 months) were eligible for inclusion. Trials involving aged garlic extract (n=6 studies) consistently showed attenuation of cardiovascular calcification. Trials involving 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (n=14), other lipid-lowering agents (n=2), hormone replacement therapies (n=3), vitamin K (n=5), lifestyle measures (n=4), and omega-3 fatty acids (n=2) consistently showed no attenuation of cardiovascular calcification with these therapies. Trials involving antiresorptive (n=2), antihypertensive (n=2), antithrombotic (n=4), and hypoglycemic agents (n=3) showed mixed results. Singleton studies involving salsalate, folate with vitamin B6 and 12, and dalcetrapib showed no attenuation of cardiovascular calcification. Overall, Cochrane risk of bias was moderate, and the Grading of Recommendations, Assessment, Development, and Evaluations assessment for a majority of analyses was moderate certainty of evidence. CONCLUSIONS Currently, there are insufficient or conflicting data for interventions evaluated in clinical trials for mitigation of cardiovascular calcification. Therapy involving aged garlic extract appears most promising, but evaluable studies were small and of short duration.
Collapse
Affiliation(s)
- Shashank Murali
- Department of NephrologyThe Royal Melbourne HospitalParkvilleVictoriaAustralia
| | - Edward R. Smith
- Department of NephrologyThe Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of Medicine (RMH)University of MelbourneParkvilleVictoriaAustralia
| | - Mark K. Tiong
- Department of NephrologyThe Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of Medicine (RMH)University of MelbourneParkvilleVictoriaAustralia
| | - Sven‐Jean Tan
- Department of NephrologyThe Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of Medicine (RMH)University of MelbourneParkvilleVictoriaAustralia
| | - Nigel D. Toussaint
- Department of NephrologyThe Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of Medicine (RMH)University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
10
|
Koljonen E, Lappalainen L, Kotiranta S, Turpeinen A, Vepsäläinen V, Kärkkäinen S, Romppanen J, Selander T, Hartikainen J, Rysä J. Plasma N-terminal pro-B-type natriuretic peptide in the detection of aortic valve stenosis. Scand J Clin Lab Invest 2023; 83:489-494. [PMID: 37933910 DOI: 10.1080/00365513.2023.2275290] [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: 06/16/2023] [Accepted: 10/22/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Systolic murmur suggestive of aortic valve origin is a common accidental finding, particularly in the elderly. Usually, it is due to aortic stenosis (AS) or aortic sclerosis (ASc). Currently, echocardiography is used to differentiate AS from ASc. Plasma N-terminal (NT)-prohormone BNP (NT-proBNP) is known to correlate with the severity of AS. We assessed whether NT-proBNP separates AS from ASc. METHODS The study population consisted of three groups: AS (n = 87, age 77 ± 7 years), ASc (n = 76, age 72 ± 10 years), and healthy controls (n = 101, age 55 ± 10 years). All subjects underwent transthoracic echocardiography and measurement of plasma NT-proBNP. Patients with diseases known to increase NT-proBNP were excluded. RESULTS The crude plasma NT-proBNP (median; IQR) in AS patients (413; 165-1055 ng/l) was significantly higher compared to ASc patients (96; 53-237 ng/l, p < 0.001) and healthy controls (50; 29-76 ng/l, p < 0.001). After adjusting for the confounding factors (age, coronary artery disease, renal function and diastolic blood pressure), plasma NT-proBNP remained significantly higher in AS patients as compared to ASc (p < 0.002) and controls (p < 0.0001). In the receiver-operating characteristic curve for NT-proBNP to identify AS from ASc and controls, the area under the curve was 0.878 with optimal cutoff of 115 ng/l. In addition, using 115 ng/l to separate AS from ASc yielded sensitivity of 0.885, and negative predictive value of 0.808. CONCLUSIONS NT-proBNP was sensitive to identify AS and useful to rule out AS in patients with systolic murmur in the left ventricular outflow tract provided the patient does not have coexisting disease known to impact NT-proBNP.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jarkko Romppanen
- Eastern Finland Laboratory Centre, Kuopio University Hospital, Finland
| | | | | | - Jaana Rysä
- School of Pharmacy, University of Eastern Finland, Finland
| |
Collapse
|
11
|
Shahraki MN, Jouabadi SM, Bos D, Stricker BH, Ahmadizar F. Statin Use and Coronary Artery Calcification: a Systematic Review and Meta-analysis of Observational Studies and Randomized Controlled Trials. Curr Atheroscler Rep 2023; 25:769-784. [PMID: 37796384 PMCID: PMC10618336 DOI: 10.1007/s11883-023-01151-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2023] [Indexed: 10/06/2023]
Abstract
PURPOSE OF REVIEW This review aimed to determine the association between statin use and coronary artery calcification (CAC), as detected by computed tomography in the general population, in previously published observational studies (OSs) and randomized controlled trials (RCTs). RECENT FINDINGS A systematic search until February 2022 identified 41 relevant studies, comprising 29 OSs and 12 RCTs. We employed six meta-analysis models, stratifying studies based on design and effect metrics. For cohort studies, the pooled β of the association with CAC quantified by the Agatston score was 0.11 (95% CI = 0.05; 0.16), with an average follow-up time per person (AFTP) of 3.68 years. Cross-sectional studies indicated a pooled odds ratio of 2.11 (95% CI = 1.61; 2.78) for the presence of CAC. In RCTs, the pooled standardized mean differences (SMDs) for CAC, quantified by Agatston score or volume, over and AFTP of 1.25 years were not statistically significant (SMD = - 0.06, 95% CI = - 0.19; 0.06 and SMD = 0.26, 95% CI = - 0.66; 1.19), but significantly different (p-value = 0.04). Meta-regression and subgroup analyses did not show any significant differences in pooled estimates across covariates. The effect of statins on CAC differs across study designs. OSs demonstrate associations between statin use and higher CAC scores and presence while being prone to confounding by indication. Effects from RCTs do not reach statistical significance and vary depending on the quantification method, hampering drawing conclusions. Further investigations are required to address the limitations inherent in each approach.
Collapse
Affiliation(s)
- Mitra Nekouei Shahraki
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Soroush Mohammadi Jouabadi
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bruno H Stricker
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Fariba Ahmadizar
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands.
- Department of Data Science and Biostatistics, Julius Global Health, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
12
|
Zhou Y, Yuan Z, Wang M, Zhang Z, Tan C, Yu J, Bi Y, Liao X, Zhou X, Ali Sheikh MS, Yang D. Liraglutide Attenuates Aortic Valve Calcification in a High-Cholesterol-Diet-Induced Experimental Calcific Aortic Valve Disease Model in Apolipoprotein E-Deficient Mice. J Cardiovasc Dev Dis 2023; 10:386. [PMID: 37754815 PMCID: PMC10531705 DOI: 10.3390/jcdd10090386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is a significant cause of morbidity and mortality among elderly people. However, no effective medications have been approved to slow or prevent the progression of CAVD. Here, we examined the effect of liraglutide on aortic valve stenosis. METHODS Male Apoe-/- mice were fed with a high-cholesterol diet for 24 weeks to generate an experimental CAVD model and randomly assigned to a liraglutide treatment group or control group. Echocardiography and immunohistological analyses were performed to examine the aortic valve function and morphology, fibrosis, and calcium deposition. Plasma Glucagon-like peptide-1 (GLP-1) levels and inflammatory contents were measured via ELISA, FACS, and immunofluorescence. RNA sequencing (RNA-seq) was used to identify liraglutide-affected pathways and processes. RESULTS Plasma GLP-1 levels were reduced in the CAVD model, and liraglutide treatment significantly improved aortic valve calcification and functions and attenuated inflammation. RNA-seq showed that liraglutide affects multiple myofibroblastic and osteogenic differentiations or inflammation-associated biological states or processes in the aortic valve. Those liraglutide-mediated beneficial effects were associated with increased GLP-1 receptor (GLP-1R) expression. CONCLUSIONS Liraglutide blocks aortic valve calcification and may serve as a potential therapeutic drug for CAVD treatment.
Collapse
Affiliation(s)
- Yangzhao Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.Z.); (Z.Y.); (M.W.); (Z.Z.); (C.T.); (J.Y.); (Y.B.); (X.L.); (X.Z.)
| | - Zhaoshun Yuan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.Z.); (Z.Y.); (M.W.); (Z.Z.); (C.T.); (J.Y.); (Y.B.); (X.L.); (X.Z.)
| | - Min Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.Z.); (Z.Y.); (M.W.); (Z.Z.); (C.T.); (J.Y.); (Y.B.); (X.L.); (X.Z.)
| | - Zhiyuan Zhang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.Z.); (Z.Y.); (M.W.); (Z.Z.); (C.T.); (J.Y.); (Y.B.); (X.L.); (X.Z.)
| | - Changming Tan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.Z.); (Z.Y.); (M.W.); (Z.Z.); (C.T.); (J.Y.); (Y.B.); (X.L.); (X.Z.)
| | - Jiaolian Yu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.Z.); (Z.Y.); (M.W.); (Z.Z.); (C.T.); (J.Y.); (Y.B.); (X.L.); (X.Z.)
| | - Yanfeng Bi
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.Z.); (Z.Y.); (M.W.); (Z.Z.); (C.T.); (J.Y.); (Y.B.); (X.L.); (X.Z.)
| | - Xiaobo Liao
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.Z.); (Z.Y.); (M.W.); (Z.Z.); (C.T.); (J.Y.); (Y.B.); (X.L.); (X.Z.)
| | - Xinmin Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.Z.); (Z.Y.); (M.W.); (Z.Z.); (C.T.); (J.Y.); (Y.B.); (X.L.); (X.Z.)
| | - Md Sayed Ali Sheikh
- Department of Internal Medicine, Cardiology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Dafeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.Z.); (Z.Y.); (M.W.); (Z.Z.); (C.T.); (J.Y.); (Y.B.); (X.L.); (X.Z.)
| |
Collapse
|
13
|
Cavalcante PN, Kanhouche G, Rosa VEE, Campos CM, Lopes MP, Lopes MAAADM, Sampaio RO, de Brito Júnior FS, Tarasoutchi F, Abizaid AAC. B-type natriuretic peptide and N-terminal Pro-B-type natriuretic peptide in severe aortic stenosis: a comprehensive literature review. Front Cardiovasc Med 2023; 10:1182530. [PMID: 37727304 PMCID: PMC10506406 DOI: 10.3389/fcvm.2023.1182530] [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: 03/08/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023] Open
Abstract
B-type natriuretic peptide (BNP) and N-terminal pro-BNP (NT-pro BNP) are cardiac biomarkers that are released in response to increased ventricular and atrial wall stress. Aortic stenosis (AS) leads to hemodynamic changes and left ventricular hypertrophy and may be associated with natriuretic peptide levels. Several studies have shown that increased natriuretic peptide levels are correlated with AS severity and can predict the need for intervention. It can be useful in risk stratification, monitoring follow-up, and predicting cardiovascular outcomes of patients with severe AS. This paper aims to summarize the evidence of the role of BNP and NT-pro BNP in AS, before and after intervention.
Collapse
Affiliation(s)
- Pâmela Nogueira Cavalcante
- Instituto do Coracao (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Gabriel Kanhouche
- Instituto do Coracao (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Vitor Emer Egypto Rosa
- Instituto do Coracao (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Carlos M. Campos
- Instituto do Coracao (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
- Departament of Hemodynamic, Instituto Prevent Senior, Sao Paulo, Brazil
| | - Mariana Pezzute Lopes
- Instituto do Coracao (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Roney Orismar Sampaio
- Instituto do Coracao (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Fábio Sândoli de Brito Júnior
- Instituto do Coracao (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Flavio Tarasoutchi
- Instituto do Coracao (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Alexandre Antonio Cunha Abizaid
- Instituto do Coracao (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
14
|
Calcific aortic valve disease: mechanisms, prevention and treatment. Nat Rev Cardiol 2023:10.1038/s41569-023-00845-7. [PMID: 36829083 DOI: 10.1038/s41569-023-00845-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/01/2023] [Indexed: 02/26/2023]
Abstract
Calcific aortic valve disease (CAVD) is the most common disorder affecting heart valves and is characterized by thickening, fibrosis and mineralization of the aortic valve leaflets. Analyses of surgically explanted aortic valve leaflets have shown that dystrophic mineralization and osteogenic transition of valve interstitial cells co-occur with neovascularization, microhaemorrhage and abnormal production of extracellular matrix. Age and congenital bicuspid aortic valve morphology are important and unalterable risk factors for CAVD, whereas additional risk is conferred by elevated blood pressure and plasma lipoprotein(a) levels and the presence of obesity and diabetes mellitus, which are modifiable factors. Genetic and molecular studies have identified that the NOTCH, WNT-β-catenin and myocardin signalling pathways are involved in the control and commitment of valvular cells to a fibrocalcific lineage. Complex interactions between valve endothelial and interstitial cells and immune cells promote the remodelling of aortic valve leaflets and the development of CAVD. Although no medical therapy is effective for reducing or preventing the progression of CAVD, studies have started to identify actionable targets.
Collapse
|
15
|
Xia C, Lei W, Hu Y, Yang H, Zeng X, Chen M. Association of serum levels of calcium, phosphate, and vitamin D with risk of developing aortic stenosis: the UK Biobank cohort. Eur J Prev Cardiol 2022; 29:1520-1528. [PMID: 35104862 DOI: 10.1093/eurjpc/zwac016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/04/2022] [Accepted: 01/26/2022] [Indexed: 11/13/2022]
Abstract
AIMS This study was aimed to investigate the associations of serum calcium, phosphate, and vitamin D levels with the risk of developing aortic stenosis (AS). METHODS AND RESULTS We included 296 415 participants who were free of prior diagnosis of any valvular heart disease from the UK Biobank. Serum levels of phosphate, calcium, and vitamin D were measured. Incidental AS was determined by the records of hospital data. Cox regression was used to examine the association of serum mineral levels with incidental AS after adjustment for potential confounders. The mean age was 56.4 years (SD 8.14) and 53.3% of participants were women. During an average follow-up of 8.1 years, 1232 individuals developed AS. After adjustment, each 0.5-unit increase in serum phosphate level was associated with a 50% increase of AS risk (hazard ratio 1.50, 95% confidence interval 1.26-1.80). We observed no association of serum calcium and vitamin D levels with AS. CONCLUSION Increased serum phosphate level, but not calcium or vitamin D, was associated with a higher risk of incident AS, this association did not differed substantially between patients with and without decreased kidney function. This finding implied that phosphate may be a potential interventional target for AS.
Collapse
Affiliation(s)
- Congying Xia
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan Province 610041, China
| | - Wenhua Lei
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan Province 610041, China
| | - Yao Hu
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan Province 610041, China
- Med-X Center for Informatics, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan Province 610041, China
| | - Huazhen Yang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan Province 610041, China
- Med-X Center for Informatics, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan Province 610041, China
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan Province 610041, China
- Division of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan Province 610041, China
| | - Mao Chen
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan Province 610041, China
| |
Collapse
|
16
|
Phua K, Chew NWS, Kong WKF, Tan RS, Ye L, Poh KK. The mechanistic pathways of oxidative stress in aortic stenosis and clinical implications. Theranostics 2022; 12:5189-5203. [PMID: 35836811 PMCID: PMC9274751 DOI: 10.7150/thno.71813] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/03/2022] [Indexed: 02/06/2023] Open
Abstract
Despite the elucidation of the pathways behind the development of aortic stenosis (AS), there remains no effective medical treatment to slow or reverse its progress. Instead, the gold standard of care in severe or symptomatic AS is replacement of the aortic valve. Oxidative stress is implicated, both directly as well as indirectly, in lipid infiltration, inflammation and fibro-calcification, all of which are key processes underlying the pathophysiology of degenerative AS. This culminates in the breakdown of the extracellular matrix, differentiation of the valvular interstitial cells into an osteogenic phenotype, and finally, calcium deposition as well as thickening of the aortic valve. Oxidative stress is thus a promising and potential therapeutic target for the treatment of AS. Several studies focusing on the mitigation of oxidative stress in the context of AS have shown some success in animal and in vitro models, however similar benefits have yet to be seen in clinical trials. Statin therapy, once thought to be the key to the treatment of AS, has yielded disappointing results, however newer lipid lowering therapies may hold some promise. Other potential therapies, such as manipulation of microRNAs, blockade of the renin-angiotensin-aldosterone system and the use of dipeptidylpeptidase-4 inhibitors will also be reviewed.
Collapse
Affiliation(s)
- Kailun Phua
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore
| | - Nicholas WS Chew
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore,✉ Corresponding authors: A/Prof Kian-Keong Poh, . Dr Nicholas Chew, MBChB, MMED (Singapore), MRCP (UK) . Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore. 1E Kent Ridge Rd, NUHS Tower Block, Level 9, Singapore 119228. Fax: (65) 68722998 Telephone: (65) 67722476
| | - William KF Kong
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore
| | - Ru-San Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, 169609, Singapore
| | - Lei Ye
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Singapore
| | - Kian-Keong Poh
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore,Yong Loo Lin School of Medicine, National University of Singapore, Singapore,✉ Corresponding authors: A/Prof Kian-Keong Poh, . Dr Nicholas Chew, MBChB, MMED (Singapore), MRCP (UK) . Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore. 1E Kent Ridge Rd, NUHS Tower Block, Level 9, Singapore 119228. Fax: (65) 68722998 Telephone: (65) 67722476
| |
Collapse
|
17
|
The Haemodynamic and Pathophysiological Mechanisms of Calcific Aortic Valve Disease. Biomedicines 2022; 10:biomedicines10061317. [PMID: 35740339 PMCID: PMC9220142 DOI: 10.3390/biomedicines10061317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 11/17/2022] Open
|
18
|
Greenberg HZE, Zhao G, Shah AM, Zhang M. Role of oxidative stress in calcific aortic valve disease and its therapeutic implications. Cardiovasc Res 2022; 118:1433-1451. [PMID: 33881501 PMCID: PMC9074995 DOI: 10.1093/cvr/cvab142] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the end result of active cellular processes that lead to the progressive fibrosis and calcification of aortic valve leaflets. In western populations, CAVD is a significant cause of cardiovascular morbidity and mortality, and in the absence of effective drugs, it will likely represent an increasing disease burden as populations age. As there are currently no pharmacological therapies available for preventing, treating, or slowing the development of CAVD, understanding the mechanisms underlying the initiation and progression of the disease is important for identifying novel therapeutic targets. Recent evidence has emerged of an important causative role for reactive oxygen species (ROS)-mediated oxidative stress in the pathophysiology of CAVD, inducing the differentiation of valve interstitial cells into myofibroblasts and then osteoblasts. In this review, we focus on the roles and sources of ROS driving CAVD and consider their potential as novel therapeutic targets for this debilitating condition.
Collapse
Affiliation(s)
- Harry Z E Greenberg
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Guoan Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Henan, China
| | - Ajay M Shah
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Min Zhang
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
19
|
Liu Q, Yu Y, Xi R, Li J, Lai R, Wang T, Fan Y, Zhang Z, Xu H, Ju J. Association Between Lipoprotein(a) and Calcific Aortic Valve Disease: A Systematic Review and Meta-Analysis. Front Cardiovasc Med 2022; 9:877140. [PMID: 35548407 PMCID: PMC9082602 DOI: 10.3389/fcvm.2022.877140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/28/2022] [Indexed: 01/03/2023] Open
Abstract
Background Preliminary studies indicated that enhanced plasma levels of lipoprotein(a) [lp(a)] might link with the risk of calcific aortic valve disease (CAVD), but the clinical association between them remained inconclusive. This systematic review and meta-analysis were aimed to determine this association. Methods We comprehensively searched PubMed, Embase, Web of Science, and Scopus databases for studies reporting the incidence of CAVD and their plasma lp(a) concentrations. Pooled risk ratio (RR) and 95% confidence interval (95% CI) were calculated to evaluate the effect of lp(a) on CAVD using the random-effects model. Subgroup analyses by study types, countries, and the level of adjustment were also conducted. Funnel plots, Egger's test and Begg's test were conducted to evaluate the publication bias. Results Eight eligible studies with 52,931 participants were included in this systematic review and meta-analysis. Of these, four were cohort studies and four were case-control studies. Five studies were rated as high quality, three as moderate quality. The pooled results showed that plasma lp(a) levels ≥50 mg/dL were associated with a 1.76-fold increased risk of CAVD (RR, 1.76; 95% CI, 1.47–2.11), but lp(a) levels ≥30 mg/dL were not observed to be significantly related with CAVD (RR, 1.28; 95% CI, 0.98–1.68). We performed subgroup analyses by study type, the RRs of cohort studies revealed lp(a) levels ≥50 mg/dL and lp(a) levels ≥30 mg/dL have positive association with CAVD (RR, 1.70; 95% CI, 1.39–2.07; RR 1.38; 95% CI, 1.19–1.61). Conclusion High plasma lp(a) levels (≥50 mg/dL) are significantly associated with increased risk of CAVD.
Collapse
Affiliation(s)
- Qiyu Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yanqiao Yu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Ruixi Xi
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingen Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Runmin Lai
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tongxin Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yixuan Fan
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zihao Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Hao Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Hao Xu
| | - Jianqing Ju
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Jianqing Ju
| |
Collapse
|
20
|
Innate immune cells in the pathophysiology of calcific aortic valve disease: lessons to be learned from atherosclerotic cardiovascular disease? Basic Res Cardiol 2022; 117:28. [PMID: 35581364 PMCID: PMC9114076 DOI: 10.1007/s00395-022-00935-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 01/31/2023]
Abstract
Calcific aortic valve disease (CAVD) is the most common valvular disease in the developed world with currently no effective pharmacological treatment available. CAVD results from a complex, multifactorial process, in which valvular inflammation and fibro-calcific remodelling lead to valve thickening and cardiac outflow obstruction. The exact underlying pathophysiology of CAVD is still not fully understood, yet the development of CAVD shows many similarities with the pathophysiology of atherosclerotic cardiovascular disease (ASCVD), such as coronary artery disease. Innate immune cells play a crucial role in ASCVD and might also play a pivotal role in the development of CAVD. This review summarizes the current knowledge on the role of innate immune cells, both in the circulation and in the aortic valve, in the development of CAVD and the similarities and differences with ASCVD. Trained immunity and clonal haematopoiesis of indeterminate potential are proposed as novel immunological mechanisms that possibly contribute to the pathophysiology of CAVD and new possible treatment targets are discussed.
Collapse
|
21
|
Morselli F, McNally R, Nesti L, Liu B, Khan H, Thomson RJ, Stevenson A, Banerjee A, Ahmad M, Hanif M, Steeds R, Khan M. Pharmacological interventions for the treatment of aortic root and heart valve disease. Hippokratia 2021. [DOI: 10.1002/14651858.cd014767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Franca Morselli
- School of Cardiovascular Medicine and Sciences; King's College London; London UK
- Department of Cardiology; East Kent Hospitals Univestity NHS Foundation Trust; Kent UK
| | - Ryan McNally
- Department of Clinical Pharmacology; King's College London; London UK
| | - Lorenzo Nesti
- Department of Clinical and Experimental Medicine; University of Pisa; Pisa Italy
| | - Boyang Liu
- Department of Cardiology; University Hospitals Birmingham (Queen Elizabeth) NHS Foundation Trust; Birmingham UK
| | - Haris Khan
- Department of Renal Medicine; Guys and St Thomas NHS Foundation Trust; London UK
| | - Ross J Thomson
- William Harvey Research Institute; Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London UK
| | - Alex Stevenson
- Department of Acute Medicine; Royal Free NHS Foundation Trust; London UK
| | - Amitava Banerjee
- Institute of Health Informatics Research; University College London; London UK
| | - Mahmood Ahmad
- Department of Cardiology; Royal Free Hospital, Royal Free London NHS Foundation Trust; London UK
| | - Moghees Hanif
- William Harvey Research Institute; Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London UK
| | - Richard Steeds
- Department of Cardiology; University Hospitals Birmingham (Queen Elizabeth) NHS Foundation Trust; Birmingham UK
| | | |
Collapse
|
22
|
Nordquist EM, Dutta P, Kodigepalli KM, Mattern C, McDermott MR, Trask AJ, LaHaye S, Lindner V, Lincoln J. Tgfβ1-Cthrc1 Signaling Plays an Important Role in the Short-Term Reparative Response to Heart Valve Endothelial Injury. Arterioscler Thromb Vasc Biol 2021; 41:2923-2942. [PMID: 34645278 PMCID: PMC8612994 DOI: 10.1161/atvbaha.121.316450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/23/2021] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Aortic valve disease is a common worldwide health burden with limited treatment options. Studies have shown that the valve endothelium is critical for structure-function relationships, and disease is associated with its dysfunction, damage, or injury. Therefore, therapeutic targets to maintain a healthy endothelium or repair damaged endothelial cells could hold promise. In this current study, we utilize a surgical mouse model of heart valve endothelial cell injury to study the short-term response at molecular and cellular levels. The goal is to determine if the native heart valve exhibits a reparative response to injury and identify the mechanisms underlying this process. Approach and Results: Mild aortic valve endothelial injury and abrogated function was evoked by inserting a guidewire down the carotid artery of young (3 months) and aging (16-18 months) wild-type mice. Short-term cellular responses were examined at 6 hours, 48 hours, and 4 weeks following injury, whereas molecular profiles were determined after 48 hours by RNA-sequencing. Within 48 hours following endothelial injury, young wild-type mice restore endothelial barrier function in association with increased cell proliferation, and upregulation of transforming growth factor beta 1 (Tgfβ1) and the glycoprotein, collagen triple helix repeat containing 1 (Cthrc1). Interestingly, this beneficial response to injury was not observed in aging mice with known underlying endothelial dysfunction. CONCLUSIONS Data from this study suggests that the healthy valve has the capacity to respond to mild endothelial injury, which in short term has beneficial effects on restoring endothelial barrier function through acute activation of the Tgfβ1-Cthrc1 signaling axis and cell proliferation.
Collapse
Affiliation(s)
- Emily M. Nordquist
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Pediatrics, Section of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI, USA
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Punashi Dutta
- Department of Pediatrics, Section of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI, USA
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Karthik M. Kodigepalli
- Department of Pediatrics, Section of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI, USA
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Carol Mattern
- Department of Pediatrics, Section of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI, USA
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Michael R. McDermott
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Aaron J. Trask
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Stephanie LaHaye
- The Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Volkhard Lindner
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Joy Lincoln
- Department of Pediatrics, Section of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI, USA
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
23
|
Kraler S, Blaser MC, Aikawa E, Camici GG, Lüscher TF. Calcific aortic valve disease: from molecular and cellular mechanisms to medical therapy. Eur Heart J 2021; 43:683-697. [PMID: 34849696 DOI: 10.1093/eurheartj/ehab757] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/12/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is a highly prevalent condition that comprises a disease continuum, ranging from microscopic changes to profound fibro-calcific leaflet remodelling, culminating in aortic stenosis, heart failure, and ultimately premature death. Traditional risk factors, such as hypercholesterolaemia and (systolic) hypertension, are shared among atherosclerotic cardiovascular disease and CAVD, yet the molecular and cellular mechanisms differ markedly. Statin-induced low-density lipoprotein cholesterol lowering, a remedy highly effective for secondary prevention of atherosclerotic cardiovascular disease, consistently failed to impact CAVD progression or to improve patient outcomes. However, recently completed phase II trials provide hope that pharmaceutical tactics directed at other targets implicated in CAVD pathogenesis offer an avenue to alter the course of the disease non-invasively. Herein, we delineate key players of CAVD pathobiology, outline mechanisms that entail compromised endothelial barrier function, and promote lipid homing, immune-cell infiltration, and deranged phospho-calcium metabolism that collectively perpetuate a pro-inflammatory/pro-osteogenic milieu in which valvular interstitial cells increasingly adopt myofibro-/osteoblast-like properties, thereby fostering fibro-calcific leaflet remodelling and eventually resulting in left ventricular outflow obstruction. We provide a glimpse into the most promising targets on the horizon, including lipoprotein(a), mineral-binding matrix Gla protein, soluble guanylate cyclase, dipeptidyl peptidase-4 as well as candidates involved in regulating phospho-calcium metabolism and valvular angiotensin II synthesis and ultimately discuss their potential for a future therapy of this insidious disease.
Collapse
Affiliation(s)
- Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland.,University Heart Center, Department of Cardiology, University Hospital, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, Boston, MA 02115, USA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, Boston, MA 02115, USA.,Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Ave Louis Pasteur, NRB7, Boston, MA 02115, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland.,University Heart Center, Department of Cardiology, University Hospital, Rämistrasse 100, 8091 Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland.,Heart Division, Royal Brompton & Harefield Hospitals, Sydney Street, London SW3 6NP, UK.,National Heart and Lung Institute, Imperial College, Guy Scadding Building, Dovehouse Street, London SW3 6LY, UK
| |
Collapse
|
24
|
Erkan G, Ozyaprak B, Kaya FA, Dursun İ, Korkmaz L. Comparison of anesthesia management in transcatheter aortic valve implantation: a retrospective cohort study. Braz J Anesthesiol 2021; 72:629-636. [PMID: 34252453 PMCID: PMC9515671 DOI: 10.1016/j.bjane.2021.06.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/20/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND AND OBJECTIVES We aimed to investigate the effects of two different anesthetic techniques in our patients who underwent transcatheter aortic valve implantation (TAVI). METHODS In this study, 303 patients who underwent TAVI procedure with a diagnosis of severe aortic stenosis between January 1, 2012 and December 31, 2018 were retrospectively evaluated. The patients were divided according to the type of anesthesia given during each procedure as; general anesthesia (GA), local anesthesia (LA). RESULTS LA was preferred in 245 (80.8%) of 303 patients who underwent TAVI, while GA was preferred in 58 patients (19.1%). Median ages of our patients who received LA and GA were 83 and 84, respectively. The procedure and anesthesia durations of the patients in the GA group were longer than the LA group (p< 0.00001, p < 0.00001, respectively). Demographic and pre-operative clinical data were similar in comparison between two groups (p > 0.05) except for peripheral artery disease. Hypertension was the most common comorbidity in both groups. While the number of inotrope use was significantly higher in patients who received GA (p < 0.00001), no significant differences were found between LA and GA patients in terms of major complications and mortality (p > 0.05). Intensive care and hospital stays were significantly shorter in the LA group (p = 0.001, p = 0.023, respectively). CONCLUSION The anesthetic technique of TAVI procedure did not have a significant effect on outcomes including; complications, mortality and success of the procedure. LA provides shorter duration of procedure and hospital stay.
Collapse
Affiliation(s)
- Gönül Erkan
- Health Sciences University, Ahi Evren Thoracic and Cardiovascular Surgery Training and Research Hospital, Department of Anesthesiology and Reanimation, Trabzon, Turkey
| | - Buket Ozyaprak
- Health Sciences University, Bursa Yüksek Ihtisas Training and Research Hospital, Department of Anesthesiology and Reanimation, Bursa, Turkey
| | - Ferdane Aydoğdu Kaya
- Health Sciences University, Ahi Evren Thoracic and Cardiovascular Surgery Training and Research Hospital, Department of Anesthesiology and Reanimation, Trabzon, Turkey
| | - İhsan Dursun
- Health Sciences University, Ahi Evren Thoracic and Cardiovascular Surgery Training and Research Hospital, Department of Cardiology, Trabzon, Turkey.
| | - Levent Korkmaz
- Health Sciences University, Ahi Evren Thoracic and Cardiovascular Surgery Training and Research Hospital, Department of Cardiology, Trabzon, Turkey
| |
Collapse
|
25
|
Afshar M, Yazdan-Ashoori S, Engert JC, Thanassoulis G. Drugs for Prevention and Treatment of Aortic Stenosis: How Close Are We? Can J Cardiol 2021; 37:1016-1026. [DOI: 10.1016/j.cjca.2021.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 12/25/2022] Open
|
26
|
Dutta P, Kodigepalli KM, LaHaye S, Thompson JW, Rains S, Nagel C, Thatcher K, Hinton RB, Lincoln J. KPT-330 Prevents Aortic Valve Calcification via a Novel C/EBPβ Signaling Pathway. Circ Res 2021; 128:1300-1316. [PMID: 33601919 PMCID: PMC8085092 DOI: 10.1161/circresaha.120.318503] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Punashi Dutta
- Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
- Pediatric Cardiology, The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Karthik M. Kodigepalli
- Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
- Pediatric Cardiology, The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Stephanie LaHaye
- The Institute for Genomic Medicine at Nationwide Children’s Hospital, Columbus, OH, USA
| | - J. Will Thompson
- Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Sarah Rains
- Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
- Duke Proteomics and Metabolomics Shared Resource, Durham, NC, USA
| | - Casey Nagel
- Ocean Ridge Biosciences, Deerfield Beach, Florida, USA
| | - Kaitlyn Thatcher
- Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
- Pediatric Cardiology, The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Robert B. Hinton
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Joy Lincoln
- Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
- Pediatric Cardiology, The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
27
|
Abstract
Aortic stenosis (AS) remains one of the most common forms of valve disease, with significant impact on patient survival. The disease is characterized by left ventricular outflow obstruction and encompasses a series of stenotic lesions starting from the left ventricular outflow tract to the descending aorta. Obstructions may be subvalvar, valvar, or supravalvar and can be present at birth (congenital) or acquired later in life. Bicuspid aortic valve, whereby the aortic valve forms with two instead of three cusps, is the most common cause of AS in younger patients due to primary anatomic narrowing of the valve. In addition, the secondary onset of premature calcification, likely induced by altered hemodynamics, further obstructs left ventricular outflow in bicuspid aortic valve patients. In adults, degenerative AS involves progressive calcification of an anatomically normal, tricuspid aortic valve and is attributed to lifelong exposure to multifactoral risk factors and physiological wear-and-tear that negatively impacts valve structure-function relationships. AS continues to be the most frequent valvular disease that requires intervention, and aortic valve replacement is the standard treatment for patients with severe or symptomatic AS. While the positive impacts of surgical interventions are well documented, the financial burden, the potential need for repeated procedures, and operative risks are substantial. In addition, the clinical management of asymptomatic patients remains controversial. Therefore, there is a critical need to develop alternative approaches to prevent the progression of left ventricular outflow obstruction, especially in valvar lesions. This review summarizes our current understandings of AS cause; beginning with developmental origins of congenital valve disease, and leading into the multifactorial nature of AS in the adult population.
Collapse
Affiliation(s)
- Punashi Dutta
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Pediatrics, Medical College of Wisconsin, Milwaukee (P.D., J.F.J., J.L.)
| | - Jeanne F James
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Pediatrics, Medical College of Wisconsin, Milwaukee (P.D., J.F.J., J.L.)
| | - Hail Kazik
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Biomedical Engineering, Marquette University & Medical College of Wisconsin, Milwaukee (H.K.)
| | - Joy Lincoln
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Pediatrics, Medical College of Wisconsin, Milwaukee (P.D., J.F.J., J.L.)
| |
Collapse
|
28
|
Metabolomics in Severe Aortic Stenosis Reveals Intermediates of Nitric Oxide Synthesis as Most Distinctive Markers. Int J Mol Sci 2021; 22:ijms22073569. [PMID: 33808189 PMCID: PMC8037707 DOI: 10.3390/ijms22073569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is a rapidly growing global health problem with an estimated 12.6 million cases globally in 2017 and a 112% increase of deaths since 1990 due to aging and population growth. CAVD may develop into aortic stenosis (AS) by progressive narrowing of the aortic valve. AS is underdiagnosed, and if treatment by aortic valve replacement (AVR) is delayed, this leads to poor recovery of cardiac function, absence of symptomatic improvement and marked increase of mortality. Considering the current limitations to define the stage of AS-induced cardiac remodeling, there is need for a novel method to aid in the diagnosis of AS and timing of intervention, which may be found in metabolomics profiling of patients. METHODS Serum samples of nine healthy controls and 10 AS patients before and after AVR were analyzed by untargeted mass spectrometry. Multivariate modeling was performed to determine a metabolic profile of 30 serum metabolites which distinguishes AS patients from controls. Human cardiac microvascular endothelial cells (CMECs) were incubated with serum of the AS patients and then stained for ICAM-1 with Western Blot to analyze the effect of AS patient serum on endothelial cell activation. RESULTS The top 30 metabolic profile strongly distinguishes AS patients from healthy controls and includes 17 metabolites related to nitric oxide metabolism and 12 metabolites related to inflammation, in line with the known pathomechanism for calcific aortic valve disease. Nine metabolites correlate strongly with left ventricular mass, of which three show reversal back to control values after AVR. Western blot analysis of CMECs incubated with AS patient sera shows a significant reduction (14%) in ICAM-1 in AS samples taken after AVR compared to AS patient sera before AVR. CONCLUSION Our study defined a top 30 metabolic profile with biological and clinical relevance, which may be used as blood biomarker to identify AS patients in need of cardiac surgery. Future studies are warranted in patients with mild-to-moderate AS to determine if these metabolites reflect disease severity and can be used to identify AS patients in need of cardiac surgery.
Collapse
|
29
|
Wei C, Li Z, Xu C, Yin T, Zhao C. Timing of surgery for asymptomatic patients with severe aortic valve stenosis: An updated systematic review and meta-analysis. Hellenic J Cardiol 2021; 62:270-277. [PMID: 33549719 DOI: 10.1016/j.hjc.2021.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/18/2020] [Accepted: 01/25/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The research findings concerning the effect of early surgery on mortality among asymptomatic patients with severe aortic valve stenosis (AS) are controversial and contradictory. In this study, the total weight of evidence regarding early surgery versus conservative management in these population was examined. METHODS Embase, PubMed, CENTRAL, and CINAHL databases were thoroughly searched without language restriction until 29 February, 2020. We included all studies exploring the mortality in asymptomatic patients with severe AS comparing early surgery with conservative management and obtained data in a standard form. Pooled relative risks ratios (RRs) and 95% confidence intervals (CI) were pooled by using a random- or fixed-effects model and by using intention-to-treat principle. Randomized controlled trials (RCTs) and observational studies (OSs) were analyzed separately. RESULTS Finally, 8 studies involving 2462 patients were enrolled, including 1 RCT and 7 OSs. In the OSs, early surgery was linked with significant reduction among all-cause mortality (RR & 95% CI, I2: 0.32 (0.18-0.57), 84.9%) and cardiac death (0.28 (0.18-0.45), 62.5%) in asymptomatic patients with severe AS. The superiority of early surgery over conservative management in reducing all-cause mortality in these patients is also intensified in the RCT. CONCLUSIONS The existing RCT and OSs indicate that earlier surgery is associated with better outcomes for asymptomatic patients with severe AS. Nevertheless, more well-designed and large-sized RCTs are needed to find an individual approach focusing on individual risk stratification and staging.
Collapse
Affiliation(s)
- Changmin Wei
- Department of Cardiology, Shengli Oilfield Central Hospital, Dongying, Shandong, PR China
| | - Zhen Li
- Department of Cardiology, Shengli Oilfield Central Hospital, Dongying, Shandong, PR China
| | - Congjian Xu
- Department of Cardiology, Shengli Oilfield Central Hospital, Dongying, Shandong, PR China.
| | - Ting Yin
- Nursing College of Beihua University, Jilin City, Jilin Province, PR China
| | - Chunshan Zhao
- Nursing College of Beihua University, Jilin City, Jilin Province, PR China
| |
Collapse
|
30
|
Treating Moderate Aortic Stenosis: Too Early or Too Late? CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2021. [DOI: 10.1007/s11936-020-00884-0] [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: 10/22/2022]
|
31
|
PCSK9: Associated with cardiac diseases and their risk factors? Arch Biochem Biophys 2020; 704:108717. [PMID: 33307067 DOI: 10.1016/j.abb.2020.108717] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 12/28/2022]
Abstract
PCSK9 plays a critical role in cholesterol metabolism via the PCSK9-LDLR axis. Liver-derived, circulating PCSK9 has become a novel drug target in lipid-lowering therapy. Accumulative evidence supports the possible association between PCSK9 and cardiac diseases and their risk factors. PCSK9 exerts various effects in the heart independently of LDL-cholesterol regulation. Acute myocardial infarction (AMI) induces local and systemic inflammation and reactive oxygen species generation, resulting in increased PCSK9 expression in hepatocytes and cardiomyocytes. PCSK9 upregulation promotes excessive autophagy and apoptosis in cardiomyocytes, thereby contributing to cardiac insufficiency. PCSK9 might also participate in the pathophysiology of heart failure by regulating fatty acid metabolism and cardiomyocyte contractility. It also promotes platelet activation and coagulation in patients with atrial fibrillation. PCSK9 is an independent predictor of aortic valve calcification and accelerates calcific aortic valve disease by regulating lipoprotein(a) catabolism. Accordingly, the use of PCSK9 inhibitors significantly reduced infarct sizes and arrhythmia and improves cardiac contractile function in a rat model of AMI. Circulating PCSK9 levels are positively correlated with age, diabetes mellitus, obesity, and hypertension. Here, we reviewed recent clinical and experimental studies exploring the association between PCSK9, cardiac diseases, and their related risk factors and aiming to identify possible underlying mechanisms.
Collapse
|
32
|
Medical management of symptomatic severe aortic stenosis in patients non-eligible for transcatheter aortic valve implantation. J Geriatr Cardiol 2020; 17:704-709. [PMID: 33343649 PMCID: PMC7729185 DOI: 10.11909/j.issn.1671-5411.2020.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
33
|
Donato M, Ferri N, Lupo MG, Faggin E, Rattazzi M. Current Evidence and Future Perspectives on Pharmacological Treatment of Calcific Aortic Valve Stenosis. Int J Mol Sci 2020; 21:ijms21218263. [PMID: 33158204 PMCID: PMC7663524 DOI: 10.3390/ijms21218263] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Calcific aortic valve stenosis (CAVS), the most common heart valve disease, is characterized by the slow progressive fibro-calcific remodeling of the valve leaflets, leading to progressive obstruction to the blood flow. CAVS is an increasing health care burden and the development of an effective medical treatment is a major medical need. To date, no effective pharmacological therapies have proven to halt or delay its progression to the severe symptomatic stage and aortic valve replacement represents the only available option to improve clinical outcomes and to increase survival. In the present report, the current knowledge and latest advances in the medical management of patients with CAVS are summarized, placing emphasis on lipid-lowering agents, vasoactive drugs, and anti-calcific treatments. In addition, novel potential therapeutic targets recently identified and currently under investigation are reported.
Collapse
Affiliation(s)
- Maristella Donato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Maria Giovanna Lupo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Elisabetta Faggin
- Department of Medicine—DIMED, University of Padova, 35122 Padova, Italy;
| | - Marcello Rattazzi
- Department of Medicine—DIMED, University of Padova, 35122 Padova, Italy;
- Correspondence: ; Tel.: +39-0498-211-867 or +39-0422-322-207
| |
Collapse
|
34
|
Nazarzadeh M, Pinho-Gomes AC, Bidel Z, Dehghan A, Canoy D, Hassaine A, Ayala Solares JR, Salimi-Khorshidi G, Smith GD, Otto CM, Rahimi K. Plasma lipids and risk of aortic valve stenosis: a Mendelian randomization study. Eur Heart J 2020; 41:3913-3920. [PMID: 32076698 PMCID: PMC7654932 DOI: 10.1093/eurheartj/ehaa070] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/09/2019] [Accepted: 01/29/2020] [Indexed: 01/09/2023] Open
Abstract
AIMS Aortic valve stenosis is commonly considered a degenerative disorder with no recommended preventive intervention, with only valve replacement surgery or catheter intervention as treatment options. We sought to assess the causal association between exposure to lipid levels and risk of aortic stenosis. METHODS AND RESULTS Causality of association was assessed using two-sample Mendelian randomization framework through different statistical methods. We retrieved summary estimations of 157 genetic variants that have been shown to be associated with plasma lipid levels in the Global Lipids Genetics Consortium that included 188 577 participants, mostly European ancestry, and genetic association with aortic stenosis as the main outcome from a total of 432 173 participants in the UK Biobank. Secondary negative control outcomes included aortic regurgitation and mitral regurgitation. The odds ratio for developing aortic stenosis per unit increase in lipid parameter was 1.52 [95% confidence interval (CI) 1.22-1.90; per 0.98 mmol/L] for low density lipoprotein (LDL)-cholesterol, 1.03 (95% CI 0.80-1.31; per 0.41 mmol/L) for high density lipoprotein (HDL)-cholesterol, and 1.38 (95% CI 0.92-2.07; per 1 mmol/L) for triglycerides. There was no evidence of a causal association between any of the lipid parameters and aortic or mitral regurgitation. CONCLUSION Lifelong exposure to high LDL-cholesterol increases the risk of symptomatic aortic stenosis, suggesting that LDL-lowering treatment may be effective in its prevention.
Collapse
Affiliation(s)
- Milad Nazarzadeh
- The George Institute for Global Health, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford OX1 2BQ, UK
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- The Collaboration Center of Meta-Analysis Research, School of Health, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Ana-Catarina Pinho-Gomes
- The George Institute for Global Health, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford OX1 2BQ, UK
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
| | - Zeinab Bidel
- The George Institute for Global Health, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford OX1 2BQ, UK
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- The Collaboration Center of Meta-Analysis Research, School of Health, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Abbas Dehghan
- Department of Biostatistics and Epidemiology, School of Public Health, Imperial College London, London, UK
| | - Dexter Canoy
- The George Institute for Global Health, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford OX1 2BQ, UK
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Abdelaali Hassaine
- The George Institute for Global Health, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford OX1 2BQ, UK
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
| | - Jose Roberto Ayala Solares
- The George Institute for Global Health, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford OX1 2BQ, UK
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
| | - Gholamreza Salimi-Khorshidi
- The George Institute for Global Health, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford OX1 2BQ, UK
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
| | | | | | - Kazem Rahimi
- The George Institute for Global Health, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford OX1 2BQ, UK
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
35
|
Nienaber CA, Yuan X. Lipid levels linked to symptomatic aortic valve stenosis: evidence from Mendelian randomization? Eur Heart J 2020; 41:3921-3924. [PMID: 32350526 DOI: 10.1093/eurheartj/ehaa225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract
Collapse
Affiliation(s)
- Christoph A Nienaber
- Cardiology and Aortic Centre, Royal Brompton and Harefield Hospital NHS Foundation Trust, London, UK
| | - Xun Yuan
- Department of Cardiology, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
36
|
Pedriali G, Morciano G, Patergnani S, Cimaglia P, Morelli C, Mikus E, Ferrari R, Gasbarro V, Giorgi C, Wieckowski MR, Pinton P. Aortic Valve Stenosis and Mitochondrial Dysfunctions: Clinical and Molecular Perspectives. Int J Mol Sci 2020; 21:ijms21144899. [PMID: 32664529 PMCID: PMC7402290 DOI: 10.3390/ijms21144899] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 01/08/2023] Open
Abstract
Calcific aortic stenosis is a disorder that impacts the physiology of heart valves. Fibrocalcific events progress in conjunction with thickening of the valve leaflets. Over the years, these events promote stenosis and obstruction of blood flow. Known and common risk factors are congenital defects, aging and metabolic syndromes linked to high plasma levels of lipoproteins. Inflammation and oxidative stress are the main molecular mediators of the evolution of aortic stenosis in patients and these mediators regulate both the degradation and remodeling processes. Mitochondrial dysfunction and dysregulation of autophagy also contribute to the disease. A better understanding of these cellular impairments might help to develop new ways to treat patients since, at the moment, there is no effective medical treatment to diminish neither the advancement of valve stenosis nor the left ventricular function impairments, and the current approaches are surgical treatment or transcatheter aortic valve replacement with prosthesis.
Collapse
Affiliation(s)
- Gaia Pedriali
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; (G.P.); (G.M.); (S.P.); (R.F.)
| | - Giampaolo Morciano
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; (G.P.); (G.M.); (S.P.); (R.F.)
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (V.G.); (C.G.)
| | - Simone Patergnani
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; (G.P.); (G.M.); (S.P.); (R.F.)
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (V.G.); (C.G.)
| | - Paolo Cimaglia
- Cardiovascular Department, Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; (P.C.); (E.M.)
| | - Cristina Morelli
- Cardiology Unit, Azienda Ospedaliero Universitaria di Ferrara, 44121 Ferrara, Italy;
| | - Elisa Mikus
- Cardiovascular Department, Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; (P.C.); (E.M.)
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; (G.P.); (G.M.); (S.P.); (R.F.)
- Cardiology Unit, Azienda Ospedaliero Universitaria di Ferrara, 44121 Ferrara, Italy;
| | - Vincenzo Gasbarro
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (V.G.); (C.G.)
| | - Carlotta Giorgi
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (V.G.); (C.G.)
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Pasteur 3, 02-093 Warsaw, Poland;
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; (G.P.); (G.M.); (S.P.); (R.F.)
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (V.G.); (C.G.)
- Correspondence: ; Tel.: +0532-455802
| |
Collapse
|
37
|
Kaltoft M, Langsted A, Nordestgaard BG. Triglycerides and remnant cholesterol associated with risk of aortic valve stenosis: Mendelian randomization in the Copenhagen General Population Study. Eur Heart J 2020; 41:2288-2299. [DOI: 10.1093/eurheartj/ehaa172] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/17/2019] [Accepted: 03/09/2020] [Indexed: 01/12/2023] Open
Abstract
AbstractAimsWe tested the hypothesis that higher levels of plasma triglycerides and remnant cholesterol are observationally and genetically associated with increased risk of aortic valve stenosis.Methods and resultsWe included 108 559 individuals from the Copenhagen General Population Study. Plasma triglycerides, remnant cholesterol (total cholesterol minus low-density lipoprotein and high-density lipoprotein cholesterol), and 16 genetic variants causing such increased or decreased levels were determined. Incident aortic valve stenosis occurred in 1593 individuals. Observationally compared to individuals with triglycerides <1 mmol/L (<89 mg/dL), the multifactorially adjusted hazard ratio for aortic valve stenosis was 1.02 [95% confidence interval (CI) 0.87–1.19] for individuals with triglycerides of 1.0–1.9 mmol/L (89–176 mg/dL), 1.22 (1.02–1.46) for 2.0–2.9 mmol/L (177–265 mg/dL), 1.40 (1.11–1.77) for 3.0–3.9 mmol/L (266–353 mg/dL), 1.29 (0.88–1.90) for 4.0–4.9 mmol/L (354–442 mg/dL), and 1.52 (1.02–2.27) for individuals with triglycerides ≥5 mmol/L (≥443 mg/dL). By age 85, the cumulative incidence of aortic valve stenosis was 5.1% for individuals with plasma triglycerides <2.0 mmol/L (77 mg/dL), 6.5% at 2.0–4.9 mmol/L (177–442 mg/dL), and 8.2% for individuals with plasma triglycerides ≥5.0 mmol/L (443 mg/dL). The corresponding values for remnant cholesterol categories were 4.8% for <0.5 mmol/L (19 mg/dL), 5.6% for 0.5–1.4 mmol/L (19–57 mg/dL), and 7.4% for ≥1.5 mmol/L (58 mg/dL). Genetically, compared to individuals with allele score 13–16, odds ratios for aortic valve stenosis were 1.30 (95% CI 1.20–1.42; Δtriglycerides +12%; Δremnant cholesterol +11%) for allele score 17–18, 1.41 (1.31–1.52; +25%; +22%) for allele score 19–20, and 1.51 (1.22–1.86; +51%; +44%) for individuals with allele score 21–23.ConclusionHigher triglycerides and remnant cholesterol were observationally and genetically associated with increased risk of aortic valve stenosis.
Collapse
Affiliation(s)
- Morten Kaltoft
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Elevator 7, DK-2730 Herlev, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Anne Langsted
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Elevator 7, DK-2730 Herlev, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Elevator 7, DK-2730 Herlev, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
38
|
Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020; 41:111-188. [PMID: 31504418 DOI: 10.1093/eurheartj/ehz455] [Citation(s) in RCA: 5188] [Impact Index Per Article: 1037.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
39
|
Barbieri F, Senoner T, Adukauskaite A, Dobner S, Holfeld J, Semsroth S, Lambert T, Zweiker D, Theurl T, Rainer PP, Schmidt A, Feuchtner GM, Steinwender C, Hoppe UC, Hintringer F, Bauer A, Müller S, Grimm M, Pfeifer BE, Dichtl W. Long-Term Prognostic Value of High-Sensitivity Troponin T Added to N-Terminal Pro Brain Natriuretic Peptide Plasma Levels Before Valve Replacement for Severe Aortic Stenosis. Am J Cardiol 2019; 124:1932-1939. [PMID: 31699359 DOI: 10.1016/j.amjcard.2019.09.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/12/2019] [Accepted: 09/12/2019] [Indexed: 11/18/2022]
Abstract
Natriuretic peptide plasma levels help to manage patients with severe aortic stenosis (AS). The role of troponin plasma levels in this patient cohort remains speculative. A consortium of 4 university hospital centers in Austria analyzed retrospectively 3,595 patients admitted for valve replacement because of severe AS since 2007. The aim was to compare the additive preprocedural value of high-sensitivity troponin T (hsTnT) to N-terminal pro brain natriuretic peptide (NT-proBNP) plasma levels in predicting postoperative long-term survival in a large cohort undergoing either surgical (57.8%) or transcatheter (42.2%) aortic valve replacement. During a median follow-up of 2.93 (1.91 to 4.92) years, 919 patients (25.6%) died, in them 556 (15.5%) due to cardiovascular causes. Both normal hsTnT (<14 ng/l) and NT-proBNP (within age- and sex-corrected normal range) plasma levels were found in 481 patients (14.3%, group 1). Normal hsTnT but elevated NT-proBNP plasma levels were found in 748 patients (22.3%, group 2). Elevated hsTnT but normal NT-proBNP plasma levels were found in 258 patients (7.7%, group 3). Both elevated hsTnT and elevated NT-proBNP plasma levels were found in 1,869 patients (55.7%, group 4). Using Log Rank tests for comparison there was a highly significant difference in both cardiovascular mortality (p <0.0001) and all-cause mortality (p <0.0001). All-cause mortality rates after 1, 3, and 5 years were 2.1%, 5.4%, 7.7% in group 1; 4.0%, 7.5%, 11.5% in group 2; 5.8%, 8.9%, 14.0% in group 3; and 12.3%, 22.6%, 28.4% in group 4. In conclusion, hsTnT adds additional impact to NT-proBNP as a routinely available biomarker for risk stratification concerning postoperative survival in patients with severe AS admitted for valve replacement. The present study supports the concept to integrate hsTnT plasma levels in the management of severe AS.
Collapse
Affiliation(s)
- Fabian Barbieri
- University Clinic of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria
| | - Thomas Senoner
- University Clinic of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria
| | - Agne Adukauskaite
- University Clinic of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria
| | - Stephan Dobner
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Johannes Holfeld
- University Clinic of Heart Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Severin Semsroth
- University Clinic of Heart Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Thomas Lambert
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Austria
| | - David Zweiker
- Department of Internal Medicine, Division of Cardiology, Medical University Graz, Graz, Austria
| | - Thomas Theurl
- University Clinic of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - Peter Paul Rainer
- Department of Internal Medicine, Division of Cardiology, Medical University Graz, Graz, Austria
| | - Albrecht Schmidt
- Department of Internal Medicine, Division of Cardiology, Medical University Graz, Graz, Austria
| | | | - Clemens Steinwender
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Austria
| | - Uta Charlotte Hoppe
- University Clinic of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - Florian Hintringer
- University Clinic of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria
| | - Axel Bauer
- University Clinic of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria
| | - Silvana Müller
- University Clinic of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria
| | - Michael Grimm
- University Clinic of Heart Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Bernhard Erich Pfeifer
- Institute of Clinical Epidemiology, Tirol Kliniken, Innsbruck, Austria; Institute of Electrical and Biomedical Engineering, University for Health Sciences, Medical Informatics and Technology (UMIT), Hall in Tirol, Austria
| | - Wolfgang Dichtl
- University Clinic of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
40
|
Tsimikas S. Potential Causality and Emerging Medical Therapies for Lipoprotein(a) and Its Associated Oxidized Phospholipids in Calcific Aortic Valve Stenosis. Circ Res 2019; 124:405-415. [PMID: 30702993 DOI: 10.1161/circresaha.118.313864] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The prevalence of calcific aortic valve disease is increasing with aging of the population. Current treatment options for advanced or symptomatic aortic stenosis are limited to traditional surgical or percutaneous aortic valve replacement. Medical therapies that impact the progression of calcific aortic valve disease do not currently exist. New pathophysiological insights suggest that the processes leading to calcific aortic valve disease are metabolically active for many years before and during the clinical expression of disease. The identification of genetic and potentially causal mediators of calcific aortic valve disease allows opportunities for therapies that may slow progression to the point where aortic valve replacement can be avoided. Recent studies suggest that approximately one-third of aortic stenosis cases are associated with highly elevated lipoprotein(a) [Lp(a)] and pathways related to the metabolism of procalcifying oxidized phospholipids. Oxidized phospholipids can be carried by Lp(a) into valve leaflets but can also be formed in situ from cell membranes, lipoproteins, and apoptotic cells. This review will summarize the clinical data implicating the potential causality of Lp(a)/oxidized phospholipids, describe emerging therapeutic agents, and propose clinical trial designs to test the hypothesis that lowering Lp(a) will reduce progression aortic stenosis and the need for aortic valve replacement.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- From the Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla
| |
Collapse
|
41
|
Kontogeorgos S, Thunström E, Basic C, Hansson PO, Zhong Y, Ergatoudes C, Morales D, Mandalenakis Z, Rosengren A, Caidahl K, Fu M. Prevalence and risk factors of aortic stenosis and aortic sclerosis: a 21-year follow-up of middle-aged men. SCAND CARDIOVASC J 2019; 54:115-123. [PMID: 31674218 DOI: 10.1080/14017431.2019.1685126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction. There is limited knowledge about factors associated with the development of aortic stenosis. This study aimed to examine the prevalence of aortic sclerosis or stenosis in 71-years-old men and determine which risk factors at 50 years of age predict the development of aortic sclerosis or aortic stenosis. Methods. A random sample of Swedish men from the general population, born in 1943 (n = 798) were followed for 21 years. Data on clinical characteristics and laboratory values were collected in 1993. An echocardiography was performed in 2014. We used logistic regression to examine the association between baseline data and the outcome. Results. Echocardiography was performed in 535 men, and aortic sclerosis or aortic stenosis was diagnosed in 27 (5.0%). 14 persons developed aortic stenosis (2.6%). Among men with aortic sclerosis or aortic stenosis, 29.6% were obese. In multivariable stepwise regression model, body mass index (odds ratio per unit increase 1.23 (95% CI 1.10-1.38; p = .0003)) and hypercholesterolemia, combined with high sensitive C-reactive protein (odds ratio versus all other 2.66 (1.18-6.00; p = .019)) were significantly associated with increased risk of developing aortic sclerosis or aortic stenosis. Body mass index was the only factor significantly associated with a higher risk of developing aortic stenosis. Conclusion. The prevalence of either aortic sclerosis or aortic stenosis was 5% and of aortic stenosis 2.6%. Obesity and hypercholesterolemia combined with elevated high sensitive C-reactive protein at the age of 50 predicted the development of degenerative aortic sclerosis or stenosis, whilst only obesity was correlated with the occurrence of aortic stenosis.
Collapse
Affiliation(s)
- Silvana Kontogeorgos
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
| | - Erik Thunström
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
| | - Carmen Basic
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
| | - Per-Olof Hansson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
| | - You Zhong
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Constantinos Ergatoudes
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
| | - David Morales
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
| | - Zacharias Mandalenakis
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
| | - Annika Rosengren
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
| | - Kenneth Caidahl
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Michael Fu
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
| |
Collapse
|
42
|
Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O, Windecker S, Aboyans V, Baigent C, Collet JP, Dean V, Delgado V, Fitzsimons D, Gale CP, Grobbee D, Halvorsen S, Hindricks G, Iung B, Jüni P, Katus HA, Landmesser U, Leclercq C, Lettino M, Lewis BS, Merkely B, Mueller C, Petersen S, Petronio AS, Richter DJ, Roffi M, Shlyakhto E, Simpson IA, Sousa-Uva M, Touyz RM, Nibouche D, Zelveian PH, Siostrzonek P, Najafov R, van de Borne P, Pojskic B, Postadzhiyan A, Kypris L, Špinar J, Larsen ML, Eldin HS, Viigimaa M, Strandberg TE, Ferrières J, Agladze R, Laufs U, Rallidis L, Bajnok L, Gudjónsson T, Maher V, Henkin Y, Gulizia MM, Mussagaliyeva A, Bajraktari G, Kerimkulova A, Latkovskis G, Hamoui O, Slapikas R, Visser L, Dingli P, Ivanov V, Boskovic A, Nazzi M, Visseren F, Mitevska I, Retterstøl K, Jankowski P, Fontes-Carvalho R, Gaita D, Ezhov M, Foscoli M, Giga V, Pella D, Fras Z, Perez de Isla L, Hagström E, Lehmann R, Abid L, Ozdogan O, Mitchenko O, Patel RS. 2019 ESC/EAS guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk. Atherosclerosis 2019; 290:140-205. [PMID: 31591002 DOI: 10.1016/j.atherosclerosis.2019.08.014] [Citation(s) in RCA: 658] [Impact Index Per Article: 109.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
43
|
Development of calcific aortic valve disease: Do we know enough for new clinical trials? J Mol Cell Cardiol 2019; 132:189-209. [PMID: 31136747 DOI: 10.1016/j.yjmcc.2019.05.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/11/2019] [Accepted: 05/19/2019] [Indexed: 12/19/2022]
Abstract
Calcific aortic valve disease (CAVD), previously thought to represent a passive degeneration of the valvular extracellular matrix (VECM), is now regarded as an intricate multistage disorder with sequential yet intertangled and interacting underlying processes. Endothelial dysfunction and injury, initiated by disturbed blood flow and metabolic disorders, lead to the deposition of low-density lipoprotein cholesterol in the VECM further provoking macrophage infiltration, oxidative stress, and release of pro-inflammatory cytokines. Such changes in the valvular homeostasis induce differentiation of normally quiescent valvular interstitial cells (VICs) into synthetically active myofibroblasts producing excessive quantities of the VECM and proteins responsible for its remodeling. As a result of constantly ongoing degradation and re-deposition, VECM becomes disorganised and rigid, additionally potentiating myofibroblastic differentiation of VICs and worsening adaptation of the valve to the blood flow. Moreover, disrupted and excessively vascularised VECM is susceptible to the dystrophic calcification caused by calcium and phosphate precipitating on damaged collagen fibers and concurrently accompanied by osteogenic differentiation of VICs. Being combined, passive calcification and biomineralisation synergistically induce ossification of the aortic valve ultimately resulting in its mechanical incompetence requiring surgical replacement. Unfortunately, multiple attempts have failed to find an efficient conservative treatment of CAVD; however, therapeutic regimens and clinical settings have also been far from the optimal. In this review, we focused on interactions and transitions between aforementioned mechanisms demarcating ascending stages of CAVD, suggesting a predisposing condition (bicuspid aortic valve) and drug combination (lipid-lowering drugs combined with angiotensin II antagonists and cytokine inhibitors) for the further testing in both preclinical and clinical trials.
Collapse
|
44
|
Clinical, sonographic characteristics and long-term prognosis of valvular heart disease in elderly patients. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2019; 16:33-41. [PMID: 30800149 PMCID: PMC6379235 DOI: 10.11909/j.issn.1671-5411.2019.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Valvular heart disease (VHD) is expected to become more prevail as the population ages and disproportionately affects older adults. However, direct comparison of clinical characteristics, sonographic diagnosis, and outcomes in VHD patients aged over 65 years is scarce. The objective of this study was to evaluate the differences in clinical characteristics and prognosis in two age-groups of geriatric patients with VHD. Methods We retrospectively enrolled consecutive individuals aged ≥ 65 years from Guangdong Provincial People's Hospital and screened for VHD using transthoracic echocardiography (TTE) or transesophageal echocardiography (TEE). Finally, 260 (48.9%) patients were in the 65-74 years group, and 272 (51.1%) were in the ≥ 75-year group. Factors that affected long-term survival was explored. A multivariable Cox hazards regression was performed to identify the predictors of major adverse cardiac events (MACEs) in each group. Results In our population, the older group were more likely to have chronic obstructive pulmonary disease (COPD), degenerative VHD, but with less rheumatic VHD, aortic stenosis (AS) and mitral stenosis (MS). Compared with those aged 65-74 years, the older group had a higher incidence of all-cause death (10.0% vs. 16.5%, P = 0.027), ischemic stroke (13.5% vs. 20.2%, P = 0.038) and MACEs (37.3% vs. 48.2%, P = 0.011) at long-term follow-up. In multivariable Cox regression analysis, mitral regurgitation, a history of COPD, chronic kidney disease, diabetes, hypertension, atrial fibrillation and New York Heart Association (NYHA) functional class were identified as independent predictors of MACEs in the older group. Conclusion Advanced age profoundly affect prognosis and different predictors were associated with MACEs in geriatric patients with VHD.
Collapse
|
45
|
Association between Comorbidities and Progression of Transvalvular Pressure Gradients in Patients with Moderate and Severe Aortic Valve Stenosis. Cardiol Res Pract 2018; 2018:3713897. [PMID: 30534431 PMCID: PMC6252229 DOI: 10.1155/2018/3713897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/05/2018] [Accepted: 10/15/2018] [Indexed: 01/04/2023] Open
Abstract
Background Fast progression of the transaortic mean gradient (P mean) is relevant for clinical decision making of valve replacement in patients with moderate and severe aortic stenosis (AS) patients. However, there is currently little knowledge regarding the determinants affecting progression of transvalvular gradient in AS patients. Methods This monocentric retrospective study included consecutive patients presenting with at least two transthoracic echocardiography examinations covering a time interval of one year or more between April 2006 and February 2016 and diagnosed as moderate or severe aortic stenosis at the final echocardiographic examination. Laboratory parameters, medication, and prevalence of eight known cardiac comorbidities and risk factors (hypertension, diabetes, coronary heart disease, peripheral artery occlusive disease, cerebrovascular disease, renal dysfunction, body mass index ≥30 Kg/m2, and history of smoking) were analyzed. Patients were divided into slow (P mean < 5 mmHg/year) or fast (P mean ≥ 5 mmHg/year) progression groups. Results A total of 402 patients (mean age 78 ± 9.4 years, 58% males) were included in the study. Mean follow-up duration was 3.4 ± 1.9 years. The average number of cardiac comorbidities and risk factors was 3.1 ± 1.6. Average number of cardiac comorbidities and risk factors was higher in patients in slow progression group than in fast progression group (3.3 ± 1.5 vs 2.9 ± 1.7; P=0.036). Patients in slow progression group had more often coronary heart disease (49.2% vs 33.6%; P=0.003) compared to patients in fast progression group. LDL-cholesterol values were lower in the slow progression group (100 ± 32.6 mg/dl vs 110.8 ± 36.6 mg/dl; P=0.005). Conclusion These findings suggest that disease progression of aortic valve stenosis is faster in patients with fewer cardiac comorbidities and risk factors, especially if they do not have coronary heart disease. Further prospective studies are warranted to investigate the outcome of patients with slow versus fast progression of transvalvular gradient with regards to comorbidities and risk factors.
Collapse
|
46
|
Novel pharmacological targets for calcific aortic valve disease: Prevention and treatments. Pharmacol Res 2018; 136:74-82. [DOI: 10.1016/j.phrs.2018.08.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/24/2022]
|
47
|
Cho KI, Sakuma I, Sohn IS, Jo SH, Koh KK. Inflammatory and metabolic mechanisms underlying the calcific aortic valve disease. Atherosclerosis 2018; 277:60-65. [PMID: 30173080 DOI: 10.1016/j.atherosclerosis.2018.08.029] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/04/2018] [Accepted: 08/24/2018] [Indexed: 12/19/2022]
Abstract
Although calcific aortic stenosis is a very common disease with major adverse cardiovascular events and healthcare costs, there are no effective medical interventions to delay or halt its progression. Cardiometabolic risk factors, including smoking and male sex, are linked to aortic stenosis. Emerging studies have identified important regulatory roles for immunological and inflammatory responses, including oxidized lipids, various cytokines, and biomineralization. Recent clinical and experimental studies in atherosclerosis and osteoporosis have demonstrated that oxidative stress and oxidized lipids decrease bone formation in the skeletal system while they increase bone formation in the cardiovascular system. Multidisciplinary factors contribute to vascular calcification, including inflammation and metabolic regulation of osteogenesis in the cardiovascular system via similar signaling pathways as bone formation. Calcific aortic valve disease (CAVD) is no longer considered a simple passive process of calcium deposition that occurs with advanced age. Biomineralization in CAVD is a complex, regulated process that involves valvular, circulating, bone marrow-derived cells, macrophage heterogeneity and genetic factors along with biochemical and mechanical factors. The current review will discuss the recently discovered important role of inflammation, metabolic risk factors, and molecular and cellular mechanisms that promote CAVD, as well as the link between osteogenic signals in the skeletal and cardiovascular systems. This may inform future therapeutic strategies for CAVD progression.
Collapse
Affiliation(s)
- Kyoung Im Cho
- Department of Cardiology, Kosin University Gospel Hospital, Busan, Republic of Korea
| | - Ichiro Sakuma
- Cardiovascular Medicine, Hokko Memorial Clinic, Sapporo, Japan; Health Science University of Hokkaido, Tobetsu, Japan
| | - Il Suk Sohn
- Department of Cardiology, Cardiovascular Center, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Sang-Ho Jo
- Department of Cardiology, Hanlym University Hospital at Pyungchon, Pyungchon, Republic of Korea
| | - Kwang Kon Koh
- Department of Cardiovascular Medicine, Heart Center, Gachon University Gil Medical Center, Incheon, Republic of Korea; Gachon Cardiovascular Research Institute, Incheon, Republic of Korea.
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW As the incidence of calcific aortic valve stenosis increases with the aging of the population, improved understanding and novel therapies to reduce its progression and need for aortic valve replacement are urgently needed. RECENT FINDINGS Lipoprotein(a) is the only monogenetic risk factor for calcific aortic stenosis. Elevated levels are a strong, causal, independent risk factor, as demonstrated in epidemiological, genome-wide association studies and Mendelian randomization studies. Lipoprotein(a) is the major lipoprotein carrier of oxidized phospholipids, which are proinflammatory and promote calcification of vascular cells, two key pathophysiological drivers of aortic stenosis. Elevated plasma lipoprotein(a) and oxidized phospholipids predict progression of pre-existing aortic stenosis and need for aortic valve replacement. The failure of statin trials in pre-existing aortic stenosis may be partially due to an increase in lipoprotein(a) and oxidized phospholipid levels caused by statins. Antisense oligonucleotides targeted to apo(a) are in Phase 2 clinical development and shown to lower both lipoprotein(a) and oxidized phospholipids. SUMMARY Lipoprotein(a) and oxidized phospholipids are key therapeutic targets in calcific aortic stenosis. Strategies aimed at potent lipoprotein(a) lowering to normalize levels and/or to suppress the proinflammatory effects of oxidized phospholipids may prevent progression of this disease.
Collapse
|
49
|
Peeters FECM, van Mourik MJW, Meex SJR, Bucerius J, Schalla SM, Gerretsen SC, Mihl C, Dweck MR, Schurgers LJ, Wildberger JE, Crijns HJGM, Kietselaer BLJH. Bicuspid Aortic Valve Stenosis and the Effect of Vitamin K2 on Calcification Using 18F-Sodium Fluoride Positron Emission Tomography/Magnetic Resonance: The BASIK2 Rationale and Trial Design. Nutrients 2018; 10:E386. [PMID: 29561783 PMCID: PMC5946171 DOI: 10.3390/nu10040386] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/08/2018] [Accepted: 03/19/2018] [Indexed: 01/07/2023] Open
Abstract
BASIK2 is a prospective, double-blind, randomized placebo-controlled trial investigating the effect of vitamin K2 (menaquinone-7;MK7) on imaging measurements of calcification in the bicuspid aortic valve (BAV) and calcific aortic valve stenosis (CAVS). BAV is associated with early development of CAVS. Pathophysiologic mechanisms are incompletely defined, and the only treatment available is valve replacement upon progression to severe symptomatic stenosis. Matrix Gla protein (MGP) inactivity is suggested to be involved in progression. Being a vitamin K dependent protein, supplementation with MK7 is a pharmacological option for activating MGP and intervening in the progression of CAVS. Forty-four subjects with BAV and mild-moderate CAVS will be included in the study, and baseline 18F-sodiumfluoride (18F-NaF) positron emission tomography (PET)/ magnetic resonance (MR) and computed tomography (CT) assessments will be performed. Thereafter, subjects will be randomized (1:1) to MK7 (360 mcg/day) or placebo. During an 18-month follow-up period, subjects will visit the hospital every 6 months, undergoing a second 18F-NaF PET/MR after 6 months and CT after 6 and 18 months. The primary endpoint is the change in PET/MR 18F-NaF uptake (6 months minus baseline) compared to this delta change in the placebo arm. The main secondary endpoints are changes in calcium score (CT), progression of the left ventricularremodeling response and CAVS severity (echocardiography). We will also examine the association between early calcification activity (PET) and later changes in calcium score (CT).
Collapse
Affiliation(s)
- Frederique E C M Peeters
- Department of Cardiology, Maastricht University Medical Center+ and CARIM, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands.
| | - Manouk J W van Mourik
- Department of Cardiology, Maastricht University Medical Center+ and CARIM, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands.
| | - Steven J R Meex
- Department of Clinical Chemistry, Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands.
| | - Jan Bucerius
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center+ and CARIM, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands.
- Department of Nuclear Medicine University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany.
| | - Simon M Schalla
- Departments of Cardiology and Radiology, Maastricht University Medical Center+ and CARIM, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands.
| | - Suzanne C Gerretsen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center+ and CARIM, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands.
| | - Casper Mihl
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center+ and CARIM, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands.
| | - Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | - Leon J Schurgers
- Department of Biochemistry, Maastricht University and CARIM, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Joachim E Wildberger
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center+ and CARIM, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands.
| | - Harry J G M Crijns
- Department of Cardiology, Maastricht University Medical Center+ and CARIM, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands.
| | - Bas L J H Kietselaer
- Department of Cardiology, Maastricht University Medical Center+ and CARIM, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands.
- Department of Cardiology, Zuyderland Medisch Centrum Heerlen/Sittard, Henri Dunantstraat 5, 6419 PC Heerlen, The Netherlands.
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW This review aims to highlight the past and more current literature related to the multifaceted pathogenic programs that contribute to calcific aortic valve disease (CAVD) with a focus on the contribution of developmental programs. RECENT FINDINGS Calcification of the aortic valve is an active process characterized by calcific nodule formation on the aortic surface leading to a less supple and more stiffened cusp, thereby limiting movement and causing clinical stenosis. The mechanisms underlying these pathogenic changes are largely unknown, but emerging studies have suggested that signaling pathways common to valvulogenesis and bone development play significant roles and include Transforming Growth Factor-β (TGF-β), bone morphogenetic protein (BMP), Wnt, Notch, and Sox9. This comprehensive review of the literature highlights the complex nature of CAVD but concurrently identifies key regulators that can be targeted in the development of mechanistic-based therapies beyond surgical intervention to improve patient outcome.
Collapse
|