1
|
Satomi N, Miyasaka M, Enta Y, Munehisa Y, Toki Y, Nakashima M, Saigan M, Kobayashi Y, Teng Y, Maeda M, Kosuga M, Abe K, Taguri M, Hayatsu Y, Hata M, Tada N. Transcatheter Aortic Valve Replacement for Structural Valve Deterioration After Aortic Valve Neocuspidization. Catheter Cardiovasc Interv 2025. [PMID: 40370003 DOI: 10.1002/ccd.31583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/07/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Evidence on structural valve deterioration (SVD) after aortic valve neocuspidization (AVNeo) remains limited. While transcatheter aortic valve replacement (TAVR) offers a less invasive option, its feasibility is unclear. This study aimed to characterize SVD patients post-AVNeo and to assess TAVR feasibility. METHODS This retrospective study included 11 patients who underwent TAVR for SVD after AVNeo in Sendai Kousei Hospital between June 2017 and August 2024. Four patients with aortic stenosis (AS) and seven with aortic regurgitation (AR) due to SVD were included. RESULTS AS-SVD group showed shorter time interval from AVNeo to TAVR (50.5 [28.3-98.3] vs. 107.0 [97.0-131.0] months, p = 0.04) and smaller aortic annulus area (316.5 [259.8-375.0] vs. 538.0 [440.0-582.0] mm2, p = 0.005) than AR-SVD group. In TAVR, technical success was achieved in 91% of cases. Although coronary obstruction is a concern due to the long leaflet, no case occurred. Misidentification of the basal ring plane caused by the slack cusp resulted in second valve implantation (one case) and new conduction disturbance (two cases) due to deep implantation, and aortic contained rupture (one case) for oversized valve selection. CONCLUSIONS Rapid AS-SVD progression and AR-SVD occurring later than previously reported were observed, highlighting the need for further studies on AVNeo durability. TAVR after AVNeo was feasible. Special precautions for coronary obstruction may be unnecessary. Careful hinge point placement at the boundary between pericardial cusps and the annulus was a key to CT-based basal ring determination in patients after AVNeo.
Collapse
Affiliation(s)
- Natsuko Satomi
- Department of Cardiology, Sendai Kousei Hospital, Miyagi, Japan
| | - Masaki Miyasaka
- Department of Cardiology, Sendai Kousei Hospital, Miyagi, Japan
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yusuke Enta
- Department of Cardiology, Sendai Kousei Hospital, Miyagi, Japan
| | | | - Yusuke Toki
- Department of Cardiology, Sendai Kousei Hospital, Miyagi, Japan
| | | | - Makoto Saigan
- Department of Cardiology, Sendai Kousei Hospital, Miyagi, Japan
| | - Yuta Kobayashi
- Department of Cardiology, Sendai Kousei Hospital, Miyagi, Japan
| | - Yun Teng
- Department of Cardiology, Sendai Kousei Hospital, Miyagi, Japan
| | - Manabu Maeda
- Department of Cardiology, Sendai Kousei Hospital, Miyagi, Japan
| | - Momo Kosuga
- Department of Cardiology, Sendai Kousei Hospital, Miyagi, Japan
| | - Kazuo Abe
- Department of Cardiovascular Surgery, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Masataka Taguri
- Department of Health Data Science, Tokyo Medical University, Tokyo, Japan
| | - Yukihiro Hayatsu
- Department of Cardiovascular Surgery, Sendai Kousei Hospital, Miyagi, Japan
| | - Masaki Hata
- Department of Cardiovascular Surgery, Sendai Kousei Hospital, Miyagi, Japan
| | - Norio Tada
- Department of Cardiology, Sendai Kousei Hospital, Miyagi, Japan
| |
Collapse
|
2
|
Maller T, Bruoha S, Loutati R, Carasso S, Taha L, Sabouret P, Galli M, Zoccai GB, Spadafora L, Dvir D, Shuvy M, Jubeh R, Marmor D, Perel N, Levi N, Amsalem I, Hitter R, Shrem M, Glikson M, Asher E. Correlation between staging classification of aortic stenosis based on the extent of cardiac damage and platelet indices. BMC Cardiovasc Disord 2024; 24:555. [PMID: 39402456 PMCID: PMC11475608 DOI: 10.1186/s12872-024-04246-7] [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: 06/30/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Platelets play a key role in the natural history of aortic stenosis (AS) and after transcatheter aortic valve implantation (TAVI). An echo-based staging system stratifies patients with severe AS into 5 groups according to the associated cardiac damage phenotype. We aimed to correlate these AS stages with platelet indices in post-TAVI patients. METHODS Patients with severe AS who underwent TAVI and were admitted to intensive cardiac care unit (ICCU) were prospectively identified and divided into 5 groups according to extra-valvular cardiac damage [no extravalvular cardiac damage (Stage 0), left ventricular damage (Stage 1), left atrial or mitral valve damage (Stage 2), pulmonary vasculature or tricuspid valve damage (Stage 3), or right ventricular damage (Stage 4)]. Baseline characteristics and complete blood count including mean platelet volume (MPV) and immature platelet fraction (IPF) were collected within 2 h after the procedure and analyzed in relation to aortic stenosis staging. RESULTS A total of 220 patients were included. The mean age was 81 years old and 112 (50.9%) were female. Two (1%) patients were classified in stage 0; 34 (15%) in stage 1; 48 (22%) in stage 2; 49 (22%) in stage 3 and 87 (40%) in stage 4. Higher mean MPV values were correlated with higher AS staging (10.8 fL, 11 fL, 11.3 fL and 10.8 fL in stages 1, 2, 3 and 4, respectively, P = 0.02) as well as lower hemoglobin values (12 mg/dl, 11.6 mg/dl, 11 mg/dl and 11.3 mg/dl in stages 1, 2, 3 and 4, respectively P = 0.04). Mean IPF values were 5.3%, 5.58%, 5.57% and 4.83% in stage 1, 2, 3 and 4, respectively (P = 0.4). In a multivariate logistic regression model only MPV (OR = 2.6, P = 0.03) and body mass index (BMI) (OR = 1.17, P = 0.004) were correlated with higher staging (0-3) of AS. CONCLUSIONS Although IPF and MPV levels increased in stages 0-3, there was a decrease in indices in stage 4, (probably due to bone marrow dysfunction) in this end-stage population. Higher levels of MPV and lower levels of hemoglobin were independently correlated with higher stages (0-3) of AS.
Collapse
Affiliation(s)
- Tomer Maller
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sharon Bruoha
- Department of Cardiology, Barzilai Medical Center, Ashkelon and Ben-Gurion University of the Negev, Ashkelon, Israel
| | - Ranel Loutati
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shemy Carasso
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Louay Taha
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Pierre Sabouret
- ACTION Study Group, Institut de Cardiologie, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
- National College of French Cardiologists, 13 Rue Niepce, Paris, 75014, France
| | - Mattia Galli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033, Italy
| | - Giuseppe Biondi Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, 00185, Italy
- Mediterranea Cardiocentro, Naples, 80122, Italy
| | - Luigi Spadafora
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Rome and Sapienza University of Rome, Rome, 00185, Italy
| | - Danny Dvir
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mony Shuvy
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rami Jubeh
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - David Marmor
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nimrod Perel
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nir Levi
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Itshak Amsalem
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rafael Hitter
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maayan Shrem
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael Glikson
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Elad Asher
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center Jerusalem and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
3
|
Xu L, Zhao Y, Zhang X, Gang X, Han J, Zhou T, Qi B, Song S, Ren R, Liang Y. Low Intraocular Pressure Induces Fibrotic Changes in the Trabecular Meshwork and Schlemm's Canal of Sprague Dawley Rats. Transl Vis Sci Technol 2024; 13:10. [PMID: 39374003 PMCID: PMC11463712 DOI: 10.1167/tvst.13.10.10] [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: 12/23/2023] [Accepted: 08/14/2024] [Indexed: 10/08/2024] Open
Abstract
Purpose Continuous artificial aqueous humor drainage in the eyes of patients with glaucoma undergoing trabeculectomy likely exerts abnormal shear stress. However, it remains unknown how changes in intraocular pressure (IOP) can affect aqueous humor outflow (AHO). Methods Here, we induced and maintained low intraocular pressure (L-IOP) in healthy Sprague Dawley (SD) rats by puncturing their eyes using a tube (200-µm diameter) for 2 weeks. After the rats were euthanized, their eyes were removed, fixed, embedded, stained, and scanned to analyze the physiological and pathological changes in the trabecular meshwork (TM) and Schlemm's canal (SC). We measured SC parameters using ImageJ software and assessed the expression of various markers related to flow shear stress (KLF4), fibrosis (TGF-β1, TGF-β2, α-SMA, pSmad1/5, pSmad2/3, and fibronectin), cytoskeleton (integrin β1 and F-actin), diastolic function (nitric oxide synthase and endothelial nitric oxide synthase [eNOS]), apoptosis (cleaved caspase-3), and proliferation (Ki-67) using immunofluorescence or immunohistochemistry. Results L-IOP eyes showed a larger SC area, higher eNOS expression, and lower KLF4 and F-actin expression in the TM and SC (both P < 0.05) than control eyes. The aqueous humor of L-IOP eyes had a higher abundance of fibrotic proteins and apoptotic cells than that of control eyes, with significantly higher TGF-β1, α-SMA, fibronectin, and cleaved caspase-3 expression (all P < 0.05). Conclusions In conclusion, a persistence of L-IOP for 2 weeks may contribute to fibrosis in the TM and SC and might be detrimental to conventional AHO in SD rat eyes. Translational Relevance Clinicians should consider that aberrant shear force induced by aqueous humor fluctuation may damage AHO outflow channel when treating patients.
Collapse
Affiliation(s)
- Lijuan Xu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, China
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Glaucoma Research Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yin Zhao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, China
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinyao Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, China
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaorui Gang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, China
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jialing Han
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, China
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tao Zhou
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, China
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Binyan Qi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, China
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuning Song
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, China
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ruiyi Ren
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, China
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Glaucoma Research Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuanbo Liang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, China
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Glaucoma Research Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
4
|
Takeji Y, Tada H, Taniguchi T, Sakata K, Kitai T, Shirai S, Takamura M. Current Management and Therapy of Severe Aortic Stenosis and Future Perspective. J Atheroscler Thromb 2024; 31:1353-1364. [PMID: 39111841 PMCID: PMC11456350 DOI: 10.5551/jat.rv22023] [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: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 10/04/2024] Open
Abstract
Intervention for severe aortic stenosis (AS) has dramatically progressed since the introduction of transcatheter aortic valve replacement (TAVR). Decades ago, controversies existed regarding comparing clinical outcomes between TAVR and surgical aortic valve replacement (SAVR) in various risk profiles. Recently, we discussed the durability of transcatheter heart valves and their lifetime management after aortic valve replacement (AVR). Regarding the management of AS, we discuss the appropriate timing of intervention for severe aortic stenosis, especially in asymptomatic patients. In spite of dramatic progression of intervention for AS, there are no established medications available to prevent or slow the progression of AS at present. Basic research and genome studies have suggested several targets associated with the progression of aortic valve calcification. Randomized controlled trials evaluating the efficacy of medications to prevent AS progression are ongoing, which might lead to new strategies for AS management. In this review, we summarize the current management of AS and the drugs expected to prevent the progression of AS.
Collapse
Affiliation(s)
- Yasuaki Takeji
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Hayato Tada
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tomohiko Taniguchi
- Department of Cardiovascular Medicine, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Kenji Sakata
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Takeshi Kitai
- Department of Heart Failure and Transplantation, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Shinichi Shirai
- Division of Cardiology, Kokura Memorial Hospital, Fukuoka, Japan
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
5
|
Fan L, Yao D, Fan Z, Zhang T, Shen Q, Tong F, Qian X, Xu L, Jiang C, Dong N. Beyond VICs: Shedding light on the overlooked VECs in calcific aortic valve disease. Biomed Pharmacother 2024; 178:117143. [PMID: 39024838 DOI: 10.1016/j.biopha.2024.117143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024] Open
Abstract
Calcific aortic valve disease (CAVD) is prevalent in developed nations and has emerged as a pressing global public health concern due to population aging. The precise etiology of this disease remains uncertain, and recent research has primarily focused on examining the role of valvular interstitial cells (VICs) in the development of CAVD. The predominant treatment options currently available involve open surgery and minimally invasive interventional surgery, with no efficacious pharmacological treatment. This article seeks to provide a comprehensive understanding of valvular endothelial cells (VECs) from the aspects of valvular endothelium-derived nitric oxide (NO), valvular endothelial mechanotransduction, valvular endothelial injury, valvular endothelial-mesenchymal transition (EndMT), and valvular neovascularization, which have received less attention, and aims to establish their role and interaction with VICs in CAVD. The ultimate goal is to provide new perspectives for the investigation of non-invasive treatment options for this disease.
Collapse
Affiliation(s)
- Lin Fan
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dingyi Yao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengfeng Fan
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tailong Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Shen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuqiang Tong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingyu Qian
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chen Jiang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Zhao K, Zeng Z, He Y, Zhao R, Niu J, Sun H, Li S, Dong J, Jing Z, Zhou J. Recent advances in targeted therapy for inflammatory vascular diseases. J Control Release 2024; 372:730-750. [PMID: 38945301 DOI: 10.1016/j.jconrel.2024.06.063] [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: 01/07/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
Vascular diseases constitute a significant contributor to worldwide mortality rates, placing a substantial strain on healthcare systems and socio-economic aspects. They are closely associated with inflammatory responses, as sustained inflammation could impact endothelial function, the release of inflammatory mediators, and platelet activation, thus accelerating the progression of vascular diseases. Consequently, directing therapeutic efforts towards mitigating inflammation represents a crucial approach in the management of vascular diseases. Traditional anti-inflammatory medications may have extensive effects on multiple tissues and organs when absorbed through the bloodstream. Conversely, treatments targeting inflammatory vascular diseases, such as monoclonal antibodies, drug-eluting stents, and nano-drugs, can achieve more precise effects, including precise intervention, minimal non-specific effects, and prolonged efficacy. In addition, personalized therapy is an important development trend in targeted therapy for inflammatory vascular diseases. Leveraging advanced simulation algorithms and clinical trial data, treatment strategies are gradually being personalized based on patients' genetic, biomarker, and clinical profiles. It is expected that the application of precision medicine in the field of vascular diseases will have a broader future. In conclusion, targeting therapies offer enhanced safety and efficacy compared to conventional medications; investigating novel targeting therapies and promoting clinical transformation may be a promising direction in improving the prognosis of patients with inflammatory vascular diseases. This article reviews the pathogenesis of inflammatory vascular diseases and presents a comprehensive overview of the potential for targeted therapies in managing this condition.
Collapse
Affiliation(s)
- Kaiwen Zhao
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Zan Zeng
- Department of Vascular Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Yuzhen He
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Rong Zhao
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Jinzhu Niu
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Huiying Sun
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Shuangshuang Li
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Jian Dong
- Department of Vascular Surgery, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zaiping Jing
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Jian Zhou
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China; Department of Vascular Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China; Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai, China.
| |
Collapse
|
7
|
Florido MHC, Ziats NP. Endothelial dysfunction and cardiovascular diseases: The role of human induced pluripotent stem cells and tissue engineering. J Biomed Mater Res A 2024; 112:1286-1304. [PMID: 38230548 DOI: 10.1002/jbm.a.37669] [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: 08/28/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024]
Abstract
Cardiovascular disease (CVD) remains to be the leading cause of death globally today and therefore the need for the development of novel therapies has become increasingly important in the cardiovascular field. The mechanism(s) behind the pathophysiology of CVD have been laboriously investigated in both stem cell and bioengineering laboratories. Scientific breakthroughs have paved the way to better mimic cell types of interest in recent years, with the ability to generate any cell type from reprogrammed human pluripotent stem cells. Mimicking the native extracellular matrix using both organic and inorganic biomaterials has allowed full organs to be recapitulated in vitro. In this paper, we will review techniques from both stem cell biology and bioengineering which have been fruitfully combined and have fueled advances in the cardiovascular disease field. We will provide a brief introduction to CVD, reviewing some of the recent studies as related to the role of endothelial cells and endothelial cell dysfunction. Recent advances and the techniques widely used in both bioengineering and stem cell biology will be discussed, providing a broad overview of the collaboration between these two fields and their overall impact on tissue engineering in the cardiovascular devices and implications for treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Mary H C Florido
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- Harvard Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Nicholas P Ziats
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Departments of Biomedical Engineering and Anatomy, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
8
|
Hu T, Jiang Y, Yang JS, Hu FJ, Yuan Y, Liu JC, Wang LJ. Investigation of autophagy‑related genes and immune infiltration in calcific aortic valve disease: A bioinformatics analysis and experimental validation. Exp Ther Med 2024; 27:233. [PMID: 38628660 PMCID: PMC11019644 DOI: 10.3892/etm.2024.12521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/11/2024] [Indexed: 04/19/2024] Open
Abstract
The present study aimed to elucidate the role of autophagy-related genes (ARGs) in calcific aortic valve disease (CAVD) and their potential interactions with immune infiltration via experimental verification and bioinformatics analysis. A total of three microarray datasets (GSE12644, GSE51472 and GSE77287) were obtained from the Gene Expression Omnibus database, and gene set enrichment analysis was performed to identify the relationship between autophagy and CAVD. After differentially expressed genes and differentially expressed ARGs (DEARGs) were identified using CAVD samples and normal aortic valve samples, a functional analysis was performed, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses, protein-protein interaction network construction, hub gene identification and validation, immune infiltration and drug prediction. The results of the present study indicated a significant relationship between autophagy and CAVD. A total of 46 DEARGs were identified. GO and pathway enrichment analyses revealed the complex roles of DEARGs in regulating CAVD, including multiple gene functions and pathways. A total of 10 hub genes were identified, with three (SPP1, CXCL12 and CXCR4) consistently upregulated in CAVD samples compared with normal aortic valve samples in multiple datasets and experimental validation. Immune infiltration analyses demonstrated significant differences in immune cell proportions between CAVD samples and normal aortic valve samples, thus showing the crucial role of immune infiltration in CAVD development. Furthermore, therapeutic drugs were predicted that could target the identified hub genes, including bisphenol A, resveratrol, progesterone and estradiol. In summary, the present study illuminated the crucial role of autophagy in CAVD development and identified key ARGs as potential therapeutic targets. In addition, the observed immune cell infiltration and predicted autophagy-related drugs suggest promising avenues for future research and novel CAVD treatments.
Collapse
Affiliation(s)
- Tie Hu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ying Jiang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jue-Sheng Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fa-Jia Hu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yong Yuan
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ji-Chun Liu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Li-Jun Wang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
9
|
Kazim M, Razian SA, Zamani E, Varandani D, Shahbad R, Desyatova A, Jadidi M. Variability in structure, morphology, and mechanical properties of the descending thoracic and infrarenal aorta around their circumference. J Mech Behav Biomed Mater 2024; 150:106332. [PMID: 38160644 DOI: 10.1016/j.jmbbm.2023.106332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024]
Abstract
Aortic diseases, such as aneurysms, atherosclerosis, and dissections, demonstrate a preferential development and progression around the aortic circumference, resulting in a highly heterogeneous disease state around the circumference. Differences in the aorta's structural composition and mechanical properties may be partly responsible for this phenomenon. Our goal in this study was to analyze the mechanical and structural properties of the human aorta at its lateral, anterior, posterior, and medial quadrants in two regions prone to circumferentially inhomogeneous diseases, descending Thoracic Aorta (TA) and Infrarenal Aorta (IFR). Human aortas were obtained from 10 donors (64 ± 11 years) and dissected from their loose surrounding tissue. Mechanical properties were determined in all four quadrants of TA and IFR using planar biaxial testing and fitted to three common constitutive models. The structure of tissues was assessed using Movat Pentachrome stained histology slides. We observed that the anterior quadrant exhibited the greatest thickness, followed by the lateral region, in both the TA and IFR. In TA, the posterior wall appeared as the stiffest location in most samples, while in IFR, the anterior wall was the stiffest. We observed a higher glycosaminoglycans content in the lateral and posterior regions of the IFR. We found elastin density to be similar in TA lateral, anterior, and posterior quadrants, while in IFR, the anterior region demonstrated the highest elastin density. Despite significant variations between subjects, this study highlights the distinct morphometrical, mechanical, and structural properties between the quadrants of both TA and IFR.
Collapse
Affiliation(s)
- Madihah Kazim
- Department of Biomechanics, University of Nebraska Omaha, Omaha, NE, USA
| | | | - Elham Zamani
- Department of Biomechanics, University of Nebraska Omaha, Omaha, NE, USA
| | - Dheeraj Varandani
- Department of Computer Science, University of Nebraska Omaha, Omaha, NE, USA
| | - Ramin Shahbad
- Department of Biomechanics, University of Nebraska Omaha, Omaha, NE, USA
| | | | - Majid Jadidi
- Department of Biomechanics, University of Nebraska Omaha, Omaha, NE, USA.
| |
Collapse
|
10
|
Savini C, Tenti E, Mikus E, Eligini S, Munno M, Gaspardo A, Gianazza E, Greco A, Ghilardi S, Aldini G, Tremoli E, Banfi C. Albumin Thiolation and Oxidative Stress Status in Patients with Aortic Valve Stenosis. Biomolecules 2023; 13:1713. [PMID: 38136584 PMCID: PMC10742097 DOI: 10.3390/biom13121713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/08/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Recent evidence indicates that reactive oxygen species play an important causative role in the onset and progression of valvular diseases. Here, we analyzed the oxidative modifications of albumin (HSA) occurring on Cysteine 34 and the antioxidant capacity of the serum in 44 patients with severe aortic stenosis (36 patients underwent aortic valve replacement and 8 underwent a second aortic valve substitution due to a degenerated bioprosthetic valve), and in 10 healthy donors (controls). Before surgical intervention, patients showed an increase in the oxidized form of albumin (HSA-Cys), a decrease in the native reduced form (HSA-SH), and a significant reduction in serum free sulfhydryl groups and in the total serum antioxidant activity. Patients undergoing a second valve replacement showed levels of HSA-Cys, free sulfhydryl groups, and total antioxidant activity similar to those of controls. In vitro incubation of whole blood with aspirin (ASA) significantly increased the free sulfhydryl groups, suggesting that the in vivo treatment with ASA may contribute to reducing oxidative stress. We also found that N-acetylcysteine and its amide derivative were able to regenerate HSA-SH. In conclusion, the systemic oxidative stress reflected by high levels of HSA-Cys is increased in patients with aortic valve stenosis. Thiol-disulfide breaking agents regenerate HSA-SH, thus paving the way to the use these compounds to mitigate the oxidative stress occurring in the disease.
Collapse
Affiliation(s)
- Carlo Savini
- GVM Care and Research, Maria Cecilia Hospital, 48033 Cotignola, Italy; (C.S.); (E.T.); (E.M.); (E.T.)
- Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum, Università di Bologna, 40126 Bologna, Italy
| | - Elena Tenti
- GVM Care and Research, Maria Cecilia Hospital, 48033 Cotignola, Italy; (C.S.); (E.T.); (E.M.); (E.T.)
| | - Elisa Mikus
- GVM Care and Research, Maria Cecilia Hospital, 48033 Cotignola, Italy; (C.S.); (E.T.); (E.M.); (E.T.)
| | - Sonia Eligini
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy; (S.E.); (M.M.); (A.G.); (E.G.); (A.G.); (S.G.)
| | - Marco Munno
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy; (S.E.); (M.M.); (A.G.); (E.G.); (A.G.); (S.G.)
| | - Anna Gaspardo
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy; (S.E.); (M.M.); (A.G.); (E.G.); (A.G.); (S.G.)
| | - Erica Gianazza
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy; (S.E.); (M.M.); (A.G.); (E.G.); (A.G.); (S.G.)
| | - Arianna Greco
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy; (S.E.); (M.M.); (A.G.); (E.G.); (A.G.); (S.G.)
| | - Stefania Ghilardi
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy; (S.E.); (M.M.); (A.G.); (E.G.); (A.G.); (S.G.)
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milano, Italy;
| | - Elena Tremoli
- GVM Care and Research, Maria Cecilia Hospital, 48033 Cotignola, Italy; (C.S.); (E.T.); (E.M.); (E.T.)
| | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy; (S.E.); (M.M.); (A.G.); (E.G.); (A.G.); (S.G.)
| |
Collapse
|
11
|
Liu X, Li T, Sun J, Wang Z. The Role of Endoplasmic Reticulum Stress in Calcific Aortic Valve Disease. Can J Cardiol 2023; 39:1571-1580. [PMID: 37516250 DOI: 10.1016/j.cjca.2023.07.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/28/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023] Open
Abstract
Calcific aortic valve disease (CAVD), which is involved in osteogenic reprogramming of valvular interstitial cells, is the most common form of valve disease. It still lacks effective pharmacologic intervention, as its cellular biological mechanisms remain unclear. Congenital abnormality (bicuspid valve) and older age are considered to be the most powerful risk factors for CAVD. Aortic valve sclerosis (AVS) and calcific aortic stenosis (CAS), 2 subclinical forms of CAVD, represent 2 distinct stages of aortic valve calcification. During the AVS stage, the disease is characterised by endothelial activation/damage, inflammatory response, and lipid infiltration accompanied by microcalcification. The CAS stage is dominated by calcification, resulting in valvular dysfunction and severe obstruction to cardiac outflow, which is life threatening if surgery is not performed in time. Endoplasmic reticulum (ER) stress, a state in which conditions disrupting ER homeostasis cause an accumulation of unfolded and misfolded proteins in the ER lumen, has been shown to promote osteogenic differentiation and aortic valve calcification. Therefore, identifying targets or drugs for suppressing ER stress may be a novel approach for CAVD treatment.
Collapse
Affiliation(s)
- Xiaolin Liu
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Medicial Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong, China
| | - Ting Li
- School of Life Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, Shandong, China
| | - Jun Sun
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhengjun Wang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
12
|
Di Costanzo A, Indolfi C, Franzone A, Esposito G, Spaccarotella CAM. Lp(a) in the Pathogenesis of Aortic Stenosis and Approach to Therapy with Antisense Oligonucleotides or Short Interfering RNA. Int J Mol Sci 2023; 24:14939. [PMID: 37834387 PMCID: PMC10573862 DOI: 10.3390/ijms241914939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
To date, no medical therapy can slow the progression of aortic stenosis. Fibrocalcific stenosis is the most frequent form in the general population and affects about 6% of the elderly population. Over the years, diagnosis has evolved thanks to echocardiography and computed tomography assessments. The application of artificial intelligence to electrocardiography could further implement early diagnosis. Patients with severe aortic stenosis, especially symptomatic patients, have valve repair as their only therapeutic option by surgical or percutaneous technique (TAVI). The discovery that the pathogenetic mechanism of aortic stenosis is similar to the atherosclerosis process has made it possible to evaluate the hypothesis of medical therapy for aortic stenosis. Several drugs have been tested to reduce low-density lipoprotein (LDL) and lipoprotein(a) (Lp(a)) levels, inflammation, and calcification. The Proprotein Convertase Subtilisin/Kexin type 9 inhibitors (PCSK9-i) could decrease the progression of aortic stenosis and the requirement for valve implantation. Great interest is related to circulating Lp(a) levels as causally linked to degenerative aortic stenosis. New therapies with ASO (antisense oligonucleotides) and siRNA (small interfering RNA) are currently being tested. Olpasiran and pelacarsen reduce circulating Lp(a) levels by 85-90%. Phase 3 studies are underway to evaluate the effect of these drugs on cardiovascular events (cardiovascular death, non-fatal myocardial injury, and non-fatal stroke) in patients with elevated Lp(a) and CVD (cardiovascular diseases). For instance, if a reduction in Lp(a) levels is associated with aortic stenosis prevention or progression, further prospective clinical trials are warranted to confirm this observation in this high-risk population.
Collapse
Affiliation(s)
- Assunta Di Costanzo
- Division of Cardiology, Cardiovascular Research Center, University Magna Graecia Catanzaro, 88100 Catanzaro, Italy;
| | - Ciro Indolfi
- Division of Cardiology, Cardiovascular Research Center, University Magna Graecia Catanzaro, 88100 Catanzaro, Italy;
| | - Anna Franzone
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.F.); (G.E.); (C.A.M.S.)
| | - Giovanni Esposito
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.F.); (G.E.); (C.A.M.S.)
| | - Carmen Anna Maria Spaccarotella
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.F.); (G.E.); (C.A.M.S.)
| |
Collapse
|
13
|
Bańka P, Wybraniec M, Bochenek T, Gruchlik B, Burchacka A, Swinarew A, Mizia-Stec K. Influence of Aortic Valve Stenosis and Wall Shear Stress on Platelets Function. J Clin Med 2023; 12:6301. [PMID: 37834945 PMCID: PMC10573628 DOI: 10.3390/jcm12196301] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Aortic valve stenosis (AS) is a common heart valve disease in the elderly population, and its pathogenesis remains an interesting area of research. The degeneration of the aortic valve leaflets gradually progresses to valve sclerosis. The advanced phase is marked by the presence of extracellular fibrosis and calcification. Turbulent, accelerated blood flow generated by the stenotic valve causes excessive damage to the aortic wall. Elevated shear stress due to AS leads to the degradation of high-molecular weight multimers of von Willebrand factor, which may involve bleeding in the mucosal tissues. Conversely, elevated shear stress has been associated with the release of thrombin and the activation of platelets, even in individuals with acquired von Willebrand syndrome. Moreover, turbulent blood flow in the aorta may activate the endothelium and promote platelet adhesion and activation on the aortic valve surface. Platelets release a wide range of mediators, including lysophosphatidic acid, which have pro-osteogenic effects in AS. All of these interactions result in blood coagulation, fibrinolysis, and the hemostatic process. This review summarizes the current knowledge on high shear stress-induced hemostatic disorders, the influence of AS on platelets and antiplatelet therapy.
Collapse
Affiliation(s)
- Paweł Bańka
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Maciej Wybraniec
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Tomasz Bochenek
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Bartosz Gruchlik
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Aleksandra Burchacka
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Andrzej Swinarew
- Faculty of Science and Technology, University of Silesia in Katowice, 40-007 Katowice, Poland
- Department of Swimming and Water Rescue, Institute of Sport Science, The Jerzy Kukuczka Academy of Physical Education, 40-065 Katowice, Poland
| | - Katarzyna Mizia-Stec
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| |
Collapse
|
14
|
Shu L, Yuan Z, Li F, Cai Z. Oxidative stress and valvular endothelial cells in aortic valve calcification. Biomed Pharmacother 2023; 163:114775. [PMID: 37116353 DOI: 10.1016/j.biopha.2023.114775] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/15/2023] [Accepted: 04/23/2023] [Indexed: 04/30/2023] Open
Abstract
Calcified aortic valve disease (CAVD) is a common cardiovascular disease in elderly individuals. Although it was previously considered a degenerative disease, it is, in fact, a progressive disease involving multiple mechanisms. Aortic valve endothelial cells, which cover the outermost layer of the aortic valve and are directly exposed to various pathogenic factors, play a significant role in the onset and progression of CAVD. Hemodynamic changes can directly damage the structure and function of valvular endothelial cells (VECs). This leads to inflammatory infiltration and oxidative stress, which promote the progression of CAVD. VECs can regulate the pathological differentiation of valvular interstitial cells (VICs) through NO and thus affect the process of CAVD. Under the influence of pathological factors, VECs can also be transformed into VICs through EndMT, and then the pathological differentiation of VICs eventually leads to the formation of calcification. This review discusses the role of VECs, especially the role of oxidative stress in VECs, in the process of aortic valve calcification.
Collapse
Affiliation(s)
- Li Shu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Zhen Yuan
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China.
| | - Zhejun Cai
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China.
| |
Collapse
|
15
|
Liu H, Yin H, Wang Z, Yuan Q, Xu F, Chen Y, Li C. Rho A/ROCK1 signaling-mediated metabolic reprogramming of valvular interstitial cells toward Warburg effect accelerates aortic valve calcification via AMPK/RUNX2 axis. Cell Death Dis 2023; 14:108. [PMID: 36774349 PMCID: PMC9922265 DOI: 10.1038/s41419-023-05642-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/13/2023]
Abstract
The aberrant differentiation of valvular interstitial cells (VICs) to osteogenic lineages promotes calcified aortic valves disease (CAVD), partly activated by potentially destructive hemodynamic forces. These involve Rho A/ROCK1 signaling, a mechano-sensing pathway. However, how Rho A/ROCK1 signaling transduces mechanical signals into cellular responses and disrupts normal VIC homeostasis remain unclear. We examined Rho A/ROCK1 signaling in human aortic valves, and further detected how Rho A/ROCK1 signaling regulates mineralization in human VICs. Aortic valves (CAVD n = 22, normal control (NC) n = 12) from patients undergoing valve replacement were investigated. Immunostaining and western blotting analysis indicated that Rho A/ROCK1 signaling, as well as key transporters and enzymes involved in the Warburg effect, were markedly upregulated in human calcified aortic valves compared with those in the controls. In vitro, Rho A/ROCK1-induced calcification was confirmed as AMPK-dependent, via a mechanism involving metabolic reprogramming of human VICs to Warburg effect. Y-27632, a selective ROCK1 inhibitor, suppressed the Warburg effect, rescued AMPK activity and subsequently increased RUNX2 ubiquitin-proteasome degradation, leading to decreased RUNX2 protein accumulation in human VICs under pathological osteogenic stimulus. Rho A/ROCK1 signaling, which is elevated in human calcified aortic valves, plays a positive role in valvular calcification, partially through its ability to drive metabolic switching of VICs to the Warburg effect, leading to altered AMPK activity and RUNX2 protein accumulation. Thus, Rho A/ROCK1 signaling could be an important and unrecognized hub of destructive hemodynamics and cellular aerobic glycolysis that is essential to promote the CAVD process.
Collapse
Affiliation(s)
- Huiruo Liu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hang Yin
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhen Wang
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qiuhuan Yuan
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Feng Xu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yuguo Chen
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Chuanbao Li
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
16
|
Gollmann-Tepeköylü C, Nägele F, Engler C, Stoessel L, Zellmer B, Graber M, Hirsch J, Pölzl L, Ruttmann E, Tancevski I, Tiller C, Barbieri F, Stastny L, Reinstadler SJ, Oezpeker UC, Semsroth S, Bonaros N, Grimm M, Feuchtner G, Holfeld J. Different calcification patterns of tricuspid and bicuspid aortic valves and their clinical impact. Interact Cardiovasc Thorac Surg 2022; 35:ivac274. [PMID: 36383200 PMCID: PMC10906007 DOI: 10.1093/icvts/ivac274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/15/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES Mechanical strain plays a major role in the development of aortic calcification. We hypothesized that (i) valvular calcifications are most pronounced at the localizations subjected to the highest mechanical strain and (ii) calcification patterns are different in patients with bicuspid and tricuspid aortic valves. METHODS Multislice computed tomography scans of 101 patients with severe aortic stenosis were analysed using a 3-dimensional post-processing software to quantify calcification of tricuspid aortic valves (n = 51) and bicuspid aortic valves (n = 50) after matching. RESULTS Bicuspid aortic valves exhibited higher calcification volumes and increased calcification of the non-coronary cusp with significantly higher calcification of the free leaflet edge. The non-coronary cusp showed the highest calcium load compared to the other leaflets. Patients with annular calcification above the median had an impaired survival compared to patients with low annular calcification, whereas patients with calcification of the free leaflet edge above the median did not (P = 0.53). CONCLUSIONS Calcification patterns are different in patients with aortic stenosis with bicuspid and tricuspid aortic valves. Patients with high annular calcification might have an impaired prognosis.
Collapse
Affiliation(s)
| | - Felix Nägele
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | - Clemens Engler
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | - Leon Stoessel
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | - Berit Zellmer
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | - Michael Graber
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | - Jakob Hirsch
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | - Leo Pölzl
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | - Elfriede Ruttmann
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University of Innsbruck, Austria
| | - Christina Tiller
- Deparment of Internal Medicine III, Medical University of Innsbruck, Austria
| | - Fabian Barbieri
- Deparment of Internal Medicine III, Medical University of Innsbruck, Austria
| | - Lukas Stastny
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | | | | | - Severin Semsroth
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | - Nikolaos Bonaros
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | - Michael Grimm
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| | - Gudrun Feuchtner
- Department of Radiology, Medical University of Innsbruck, Austria
| | - Johannes Holfeld
- Department of Cardiac Surgery, Medical University of Innsbruck, Austria
| |
Collapse
|
17
|
[Optimal time window for observation of calcific aortic valve disease in mice following catheter-induced valve injury]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1532-1538. [PMID: 36329588 PMCID: PMC9637488 DOI: 10.12122/j.issn.1673-4254.2022.10.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To investigate the optimal time window for observation of catheter-induced valve injury that mimics calcified aortic valve disease in mice. METHODS A catheter was inserted into the right common carotid artery of 8-week-old C57BL6 mice under ultrasound guidance, and aortic valve injury was induced using the guide wire.At 4, 8 and 16 weeks after modeling, the mice were subjected to ultrasound measurement of the heart short axial shortening rate, aortic valve peak velocity and aortic valve orifice area.Grain-Eosin staining was used to observe the changes in the thickness of the aortic valve, and calcium deposition in the aortic valve was assessed using Alizarin red staining.Immunofluorescence assay was performed to detect the expression of alkaline phosphatase (ALP) in the aortic valve. RESULTS At 4, 8 and 16 weeks after modeling, valve thickness (P=0.002), calcium deposition (P < 0.0001) and the expression of osteogenic protein ALP (P=0.0016) were significantly increased, but their increments were comparable at the 3 time points of observation. CONCLUSION In mouse models of calcific aortic valve disease induced by catheter valve injury, 4 weeks after the injury appears to be the optimal time window for observation of pathophysiological changes in the aortic valves to avoid further increase of the death rate of the mice over time.
Collapse
|
18
|
Li XZ, Xiong ZC, Zhang SL, Hao QY, Gao M, Wang JF, Gao JW, Liu PM. Potential ferroptosis key genes in calcific aortic valve disease. Front Cardiovasc Med 2022; 9:916841. [PMID: 36003913 PMCID: PMC9395208 DOI: 10.3389/fcvm.2022.916841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] 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 remodeling, culminating in aortic stenosis, heart failure, and ultimately premature death. Ferroptosis has been hypothesized to contribute to the pathogenesis of CAVD. We aimed to study the association between ferroptosis genes and CAVD and reveal the potential roles of ferroptosis in CAVD. CAVD-related differentially expressed genes (DEGs) were identified via bioinformatic analysis of Datasets GSE51472 and GSE12644 obtained from Gene Expression Omnibus. A ferroptosis dataset containing 259 genes was obtained from the Ferroptosis Database. We then intersected with CAVD-related DEGs to identify the ferroptosis DEGs. Subsequently, protein–protein interaction networks and functional enrichment analyses were performed for ferroptosis DEGs. Then, we used miRWalk3.0 to predict the target pivotal microRNAs. An in vitro model of CAVD was constructed using human aortic valve interstitial cells. The qRT-PCR and western blotting methods were used to validate the ferroptosis DEGs identified by the microarray data. A total of 21 ferroptosis DEGs in CAVD containing 12 upregulated and nine downregulated genes were identified. The results of the Gene Set Enrichment Analysis (GSEA) and analysis of the KEGG pathway by WebGestalt indicated that the ferroptosis DEGs were enriched in six signaling pathways among which NAFLD (including IL-6, BID, and PRKAA2 genes) and HIF-1 (including IL-6, HIF-1, and HMOX1 genes) signaling pathways were also verified by DAVID and/or Metascape. Finally, the in vitro results showed that the mRNA and protein expression levels of IL-6, HIF-1α, HMOX1, and BID were higher, while the levels of PRKAA2 were lower in the Pi-treated group than those in the control group. However, the addition of ferrostatin-1 (a selective ferroptosis inhibitor) significantly reversed the above changes. Therefore, IL-6, HIF-1α, HMOX1, BID, and PRKAA2 are potential key genes closely associated with ferroptosis in CAVD. Further work is required to explore the underlying ferroptosis-related molecular mechanisms and provide possible therapeutic targets for CAVD.
Collapse
Affiliation(s)
- Xiong-Zhi Li
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhuo-Chao Xiong
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shao-Ling Zhang
- Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qing-Yun Hao
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ming Gao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jing-Feng Wang
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jing-Wei Gao
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Jing-Wei Gao
| | - Pin-Ming Liu
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Pin-Ming Liu
| |
Collapse
|
19
|
Hu P, Chen H, Wang LH, Jiang JB, Li JM, Tang MY, Guo YC, Zhu QF, Pu ZX, Lin XP, Ng S, Liu XB, Wang JA. Elevated N-terminal pro C-type natriuretic peptide is associated with mortality in patients undergoing transcatheter aortic valve replacement. BMC Cardiovasc Disord 2022; 22:164. [PMID: 35413789 PMCID: PMC9004019 DOI: 10.1186/s12872-022-02615-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/01/2022] [Indexed: 12/02/2022] Open
Abstract
Background Unlike N-terminal pro-B-type natriuretic peptide (NT-proBNP), which have been extensively studied, little is known about the role of N-terminal pro-C-type natriuretic peptide (NT-proCNP) for predicting survival post transcatheter aortic valve replacement (TAVR). Methods A total of 309 patients were included in the analysis. Patients were grouped into quartiles (Q1–4) according to the baseline NT-proCNP value. Blood for NT-proCNP analysis was obtained prior to TAVR procedure. The primary endpoint was mortality after a median follow-up of 32 months. Multivariable Cox proportional hazards regression models analyzed prognostic factors. The predictive capability was compared between NT-proBNP and NT-proCNP using receiver operator curve (ROC) analysis. Results A total of 309 subjects with the mean age of 76.8 ± 6.3 years, among whom 58.6% were male, were included in the analysis. A total of 58 (18.8%) patients died during follow-up. Cox multivariable analyses indicated society of thoracic surgeons (STS)-score was a strong independent predictor for mortality (hazard ratio (HR) 1.08, 95% confidential interval (CI) 1.05–1.12, P < 0.001). Elevated NT-proCNP was associated with a higher risk of cardiovascular mortality (HR 1.02, 95% CI 1.00–1.03, P = 0.025) and All-cause mortality (HR 1.01, 95% CI 1.00–1.03, P = 0.027), whereas NT-proBNP showed a small effect size on mortality. ROC analysis indicated that NT-proCNP was superior to NT-proBNP for TAVR risk evaluation in patients with left ventricular ejection fraction (LVEF) < 50% [(Area under the curve (AUC)-values of 0.79 (0.69; 0.87) vs. 0.59 (0.48; 0.69), P = 0.0453]. Conclusions NT-proCNP and STS-Score were the independent prognostic factors of mortality among TAVR patients. Furthermore, NT-proCNP was superior to NT-proBNP for TAVR risk evaluation in patients with LVEF < 50%. Trial registration NCT02803294, 16/06/2016. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02615-8.
Collapse
Affiliation(s)
- Po Hu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Han Chen
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Li-Han Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Ju-Bo Jiang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Jia-Min Li
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China
| | - Meng-Yao Tang
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yu-Chao Guo
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Qi-Feng Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Zhao-Xia Pu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Department of Echocardiography, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Xin-Ping Lin
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Department of Echocardiography, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Stella Ng
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China
| | - Xian-Bao Liu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China. .,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China.
| | - Jian-An Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China. .,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
20
|
Xiong T, Han S, Pu L, Zhang TC, Zhan X, Fu T, Dai YH, Li YX. Bioinformatics and Machine Learning Methods to Identify FN1 as a Novel Biomarker of Aortic Valve Calcification. Front Cardiovasc Med 2022; 9:832591. [PMID: 35295271 PMCID: PMC8918776 DOI: 10.3389/fcvm.2022.832591] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/28/2022] [Indexed: 12/14/2022] Open
Abstract
AimThe purpose of this study was to identify potential diagnostic markers for aortic valve calcification (AVC) and to investigate the function of immune cell infiltration in this disease.MethodsThe AVC data sets were obtained from the Gene Expression Omnibus. The identification of differentially expressed genes (DEGs) and the performance of functional correlation analysis were carried out using the R software. To explore hub genes related to AVC, a protein–protein interaction network was created. Diagnostic markers for AVC were then screened and verified using the least absolute shrinkage and selection operator, logistic regression, support vector machine-recursive feature elimination algorithms, and hub genes. The infiltration of immune cells into AVC tissues was evaluated using CIBERSORT, and the correlation between diagnostic markers and infiltrating immune cells was analyzed. Finally, the Connectivity Map database was used to forecast the candidate small molecule drugs that might be used as prospective medications to treat AVC.ResultsA total of 337 DEGs were screened. The DEGs that were discovered were mostly related with atherosclerosis and arteriosclerotic cardiovascular disease, according to the analyses. Gene sets involved in the chemokine signaling pathway and cytokine–cytokine receptor interaction were differently active in AVC compared with control. As the diagnostic marker for AVC, fibronectin 1 (FN1) (area the curve = 0.958) was discovered. Immune cell infiltration analysis revealed that the AVC process may be mediated by naïve B cells, memory B cells, plasma cells, activated natural killer cells, monocytes, and macrophages M0. Additionally, FN1 expression was associated with memory B cells, M0 macrophages, activated mast cells, resting mast cells, monocytes, and activated natural killer cells. AVC may be reversed with the use of yohimbic acid, the most promising small molecule discovered so far.ConclusionFN1 can be used as a diagnostic marker for AVC. It has been shown that immune cell infiltration is important in the onset and progression of AVC, which may benefit in the improvement of AVC diagnosis and treatment.
Collapse
Affiliation(s)
- Tao Xiong
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Shen Han
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Lei Pu
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Tian-Chen Zhang
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Xu Zhan
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Fu
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Ying-Hai Dai
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Ya-Xiong Li
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
- *Correspondence: Ya-Xiong Li ;
| |
Collapse
|
21
|
Xiong JQ, Chen XM, Liang CT, Guo W, Wu BL, Du XG. Prognosis and risk factors for cardiac valve calcification in Chinese end-stage kidney disease patients on combination therapy with hemodialysis and hemodiafiltration. Ren Fail 2022; 44:224-232. [PMID: 35166181 PMCID: PMC8856035 DOI: 10.1080/0886022x.2022.2032742] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
BACKGROUND Cardiac valve calcification (CVC) is an important risk factor for cardiovascular complications. However, limited data are available concerning the prevalence, clinical features and risk factors for CVC in end-stage kidney disease (ESKD) patients. In this study, we aimed to assess these parameters in Chinese ESKD patients receiving combination therapy with hemodialysis and hemodiafiltration. METHODS We conducted a cross-sectional study on 293 ESKD patients undergoing combination therapy of hemodialysis and hemodiafiltration at the First Affiliated Hospital of Chongqing Medical University from October 2014 to December 2015. CVC was evaluated via echocardiography. RESULTS ESKD patients with CVC had a higher prevalence of diabetes mellitus, aortic and/or coronary artery calcification, arrhythmia, heart failure and coronary heart disease; increased systolic, diastolic and pulse pressure; longer duration of hemodialysis and hypertension; reduced hemoglobin, albumin and high-density lipoprotein cholesterol levels; and increased serum calcium and calcium-phosphorus product levels compared with those without CVC. Logistic regression analysis showed that increased dialysis duration (p = 0.006, OR = 2.25), serum calcium levels (p = 0.046, OR = 2.04) and pulse pressure (p < 0.001, OR = 3.22), the presence of diabetes (p = 0.037, OR = 1.81) and decreased serum albumin levels (p = 0.047, OR = 0.54) were risk factors for CVC. The correlation analysis indicated a significantly increased CVCs prevalence with an increase prevalence of heart failure, aortic and coronary artery calcification. CONCLUSIONS CVC represents a common complication and a danger signal for cardiovascular events in ESKD patients undergoing combination therapy of hemodialysis and hemodiafiltration. The presence of diabetes, increased pulse pressure, long dialysis duration, hypoalbuminemia and high serum calcium levels were independent risk factors for CVC.
Collapse
Affiliation(s)
- Jian-Qiong Xiong
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Hospital, Chongqing University, Chongqing, China
| | - Xue-Mei Chen
- Emergency Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chun-Ting Liang
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Nephrology, Longchang People's Hospital, Neijiang, Sichuan, China
| | - Wen Guo
- Emergency Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bai-Li Wu
- Emergency Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-Gang Du
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Hu J, Lei H, Liu L, Xu D. Lipoprotein(a), a Lethal Player in Calcific Aortic Valve Disease. Front Cell Dev Biol 2022; 10:812368. [PMID: 35155427 PMCID: PMC8830536 DOI: 10.3389/fcell.2022.812368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/07/2022] [Indexed: 12/11/2022] Open
Abstract
Calcified aortic valve disease (CAVD) is the most common valvular cardiovascular disease with increasing incidence and mortality. The primary treatment for CAVD is surgical or transcatheter aortic valve replacement and there remains a lack of effective drug treatment. Recently, lipoprotein (a) (Lp(a)) has been considered to play a crucial role in CAVD pathophysiology. Multiple studies have shown that Lp(a) represents an independent risk factor for CAVD. Moreover, Lp(a) mediates the occurrence and development of CAVD by affecting aortic valve endothelial dysfunction, indirectly promoting foam cell formation through oxidized phospholipids (OxPL), inflammation, oxidative stress, and directly promotes valve calcification. However, there is a lack of clinical trials with Lp(a) reduction as a primary endpoint. This review aims to explore the relationship and mechanism between Lp(a) and CAVD, and focuses on the current drugs that can be used as potential therapeutic targets for CAVD.
Collapse
Affiliation(s)
- Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Hao Lei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Leiling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
- *Correspondence: Danyan Xu,
| |
Collapse
|
23
|
Tandon I, Ozkizilcik A, Ravishankar P, Balachandran K. Aortic valve cell microenvironment: Considerations for developing a valve-on-chip. BIOPHYSICS REVIEWS 2021; 2:041303. [PMID: 38504720 PMCID: PMC10903420 DOI: 10.1063/5.0063608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/15/2021] [Indexed: 03/21/2024]
Abstract
Cardiac valves are sophisticated, dynamic structures residing in a complex mechanical and hemodynamic environment. Cardiac valve disease is an active and progressive disease resulting in severe socioeconomic burden, especially in the elderly. Valve disease also leads to a 50% increase in the possibility of associated cardiovascular events. Yet, valve replacement remains the standard of treatment with early detection, mitigation, and alternate therapeutic strategies still lacking. Effective study models are required to further elucidate disease mechanisms and diagnostic and therapeutic strategies. Organ-on-chip models offer a unique and powerful environment that incorporates the ease and reproducibility of in vitro systems along with the complexity and physiological recapitulation of the in vivo system. The key to developing effective valve-on-chip models is maintaining the cell and tissue-level microenvironment relevant to the study application. This review outlines the various components and factors that comprise and/or affect the cell microenvironment that ought to be considered while constructing a valve-on-chip model. This review also dives into the advancements made toward constructing valve-on-chip models with a specific focus on the aortic valve, that is, in vitro studies incorporating three-dimensional co-culture models that incorporate relevant extracellular matrices and mechanical and hemodynamic cues.
Collapse
Affiliation(s)
- Ishita Tandon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, USA
| | - Asya Ozkizilcik
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, USA
| | - Prashanth Ravishankar
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, USA
| | - Kartik Balachandran
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, USA
| |
Collapse
|
24
|
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
|
25
|
Dharmarajan S, Speer MY, Pierce K, Lally J, Leaf EM, Lin ME, Scatena M, Giachelli CM. Role of Runx2 in Calcific Aortic Valve Disease in Mouse Models. Front Cardiovasc Med 2021; 8:687210. [PMID: 34778386 PMCID: PMC8585763 DOI: 10.3389/fcvm.2021.687210] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/28/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Calcific aortic valve disease is common in the aging population and is characterized by the histological changes of the aortic valves including extracellular matrix remodeling, osteochondrogenic differentiation, and calcification. Combined, these changes lead to aortic sclerosis, aortic stenosis (AS), and eventually to heart failure. Runt-related transcription factor 2 (Runx2) is a transcription factor highly expressed in the calcified aortic valves. However, its definitive role in the progression of calcific aortic valve disease (CAVD) has not been determined. In this study, we utilized constitutive and transient conditional knockout mouse models to assess the molecular, histological, and functional changes in the aortic valve due to Runx2 depletion. Methods: Lineage tracing studies were performed to determine the provenance of the cells giving rise to Runx2+ osteochondrogenic cells in the aortic valves of LDLr-/- mice. Hyperlipidemic mice with a constitutive or temporal depletion of Runx2 in the activated valvular interstitial cells (aVICs) and sinus wall cells were further investigated. Following feeding with a diabetogenic diet, the mice were examined for changes in gene expression, blood flow dynamics, calcification, and histology. Results: The aVICs and sinus wall cells gave rise to Runx2+ osteochondrogenic cells in diseased mouse aortic valves. The conditional depletion of Runx2 in the SM22α+ aVICs and sinus wall cells led to the decreased osteochondrogenic gene expression in diabetic LDLr-/- mice. The transient conditional depletion of Runx2 in the aVICs and sinus wall cells of LDLr-/-ApoB100 CAVD mice early in disease led to a significant reduction in the aortic peak velocity, mean velocity, and mean gradient, suggesting the causal role of Runx2 on the progression of AS. Finally, the leaflet hinge and sinus wall calcification were significantly decreased in the aortic valve following the conditional and temporal Runx2 depletion, but no significant effect on the valve cusp calcification or thickness was observed. Conclusions: In the aortic valve disease, Runx2 was expressed early and was required for the osteochondrogenic differentiation of the aVICs and sinus wall cells. The transient depletion of Runx2 in the aVICs and sinus wall cells in a mouse model of CAVD with a high prevalence of hemodynamic valve dysfunction led to an improved aortic valve function. Our studies also suggest that leaflet hinge and sinus wall calcification, even in the absence of significant leaflet cusp calcification, may be sufficient to cause significant valve dysfunctions in mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Cecilia M. Giachelli
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| |
Collapse
|
26
|
Zebhi B, Lazkani M, Bark D. Calcific Aortic Stenosis-A Review on Acquired Mechanisms of the Disease and Treatments. Front Cardiovasc Med 2021; 8:734175. [PMID: 34604358 PMCID: PMC8486019 DOI: 10.3389/fcvm.2021.734175] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/18/2021] [Indexed: 11/13/2022] Open
Abstract
Calcific aortic stenosis is a progressive disease that has become more prevalent in recent decades. Despite advances in research to uncover underlying biomechanisms, and development of new generations of prosthetic valves and replacement techniques, management of calcific aortic stenosis still comes with unresolved complications. In this review, we highlight underlying molecular mechanisms of acquired aortic stenosis calcification in relation to hemodynamics, complications related to the disease, diagnostic methods, and evolving treatment practices for calcific aortic stenosis.
Collapse
Affiliation(s)
- Banafsheh Zebhi
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Mohamad Lazkani
- Medical Center of the Rockies, University of Colorado Health, Loveland, CO, United States
| | - David Bark
- Department of Pediatrics, Washington University in Saint Louis, Saint Louis, MO, United States.,Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, United States
| |
Collapse
|
27
|
Computational Analysis of Wall Shear Stress Patterns on Calcified and Bicuspid Aortic Valves: Focus on Radial and Coaptation Patterns. FLUIDS 2021. [DOI: 10.3390/fluids6080287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Calcification and bicuspid valve formation are important aortic valve disorders that disturb the hemodynamics and the valve function. The detailed analysis of aortic valve hemodynamics would lead to a better understanding of the disease’s etiology. We computationally modeled the aortic valve using simplified three-dimensional geometry and inlet velocity conditions obtained via echocardiography. We examined various calcification severities and bicuspid valve formation. Fluid-structure interaction (FSI) analyses were adapted using ANSYS Workbench to incorporate both flow dynamics and leaflet deformation accurately. Simulation results were validated by comparing leaflet movements in B-mode echo recordings. Results indicate that the biomechanical environment is significantly changed for calcified and bicuspid valves. High flow jet velocities are observed in the calcified valves which results in high transvalvular pressure difference (TPG). Wall shear stresses (WSS) increased with the calcification on both fibrosa (aorta side) and ventricularis (left ventricle side) surfaces of the leaflet. The WSS distribution is regular on the ventricularis, as the WSS values proportionally increase from the base to the tip of the leaflet. However, WSS patterns are spatially complex on the fibrosa side. Low WSS levels and spatially complex WSS patterns on the fibrosa side are considered as promoting factors for further calcification and valvular diseases.
Collapse
|
28
|
Jiang Y, Chen J, Wei F, Wang Y, Chen S, Li G, Dong N. Micromechanical force promotes aortic valvular calcification. J Thorac Cardiovasc Surg 2021; 164:e313-e329. [PMID: 34507817 DOI: 10.1016/j.jtcvs.2021.08.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Calcified aortic valvular disease is known as an inflammation-related process related to force. The purpose of this study was to determine whether micromechanical force could induce valve calcification of porcine valvular interstitial cells and to examine the role of integrin αvβ3 in valvular calcification by using a novel method: magnetic twisting cytometry. METHODS Porcine valvular interstitial cells were cultured in vitro, and micromechanical force was applied to porcine valvular interstitial cells using magnetic twisting cytometry. Changes in calcification-related factors osteopontin and RUNX2 were detected. By using the calcification medium, the optimal magnetic twisting cytometry parameters for inducing valvular interstitial cell calcification were determined, and a magnetic twisting cytometry calcification promotion model was established. The role of αvβ3 in calcification was studied by using αvβ3 antagonists to block the function of αvβ3. RESULTS Reverse transcription polymerase chain reaction assays showed that the expression of osteopontin was enhanced 30 minutes after 25G-1Hz 5 minutes of stimulation. Western blotting assays showed that the expression of osteopontin and RUNX2 was upregulated 24 hours after 25G-1Hz 5 minutes of stimulation. The optimal magnetic twisting cytometry parameter for inducing porcine valvular interstitial cell calcification was 25G-2Hz for 10 minutes. The expression of osteopontin and RUNX2 decreased significantly after the addition of αvβ3 antagonist. Clinically, patients with bicuspid aortic valves had high expression of RUNX2 and β3 in the aortic valve, and β3 significantly correlated with RUNX2. CONCLUSIONS By using magnetic twisting cytometry, we established a porcine valvular interstitial cell calcification model by micromechanical force stimulation and obtained the optimal parameters. Integrin αvβ3 plays a key role in the aortic valve calcification process.
Collapse
Affiliation(s)
- Yefan Jiang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinjie Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fuxiang Wei
- Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yixuan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Si Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Geng Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
29
|
Fan Y, Shao J, Wei S, Song C, Li Y, Jiang S. Self-eating and Heart: The Emerging Roles of Autophagy in Calcific Aortic Valve Disease. Aging Dis 2021; 12:1287-1303. [PMID: 34341709 PMCID: PMC8279526 DOI: 10.14336/ad.2021.0101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 01/01/2021] [Indexed: 12/16/2022] Open
Abstract
Autophagy is a self-degradative pathway by which subcellular elements are broken down intracellularly to maintain cellular homeostasis. Cardiac autophagy commonly decreases with aging and is accompanied by the accumulation of misfolded proteins and dysfunctional organelles, which are undesirable to the cell. Reduction of autophagy over time leads to aging-related cardiac dysfunction and is inversely related to longevity. However, despite the increasing interest in autophagy in cardiac diseases and aging, the process remains an undervalued and disregarded object in calcific valvular disease. Neither the nature through which autophagy is triggered nor the interplay between autophagic machinery and targeted molecules during aortic valve calcification are fully understood. Recently, the upregulation of autophagy has been shown to result in cardioprotective effects against cell death as well as its origin. Here, we review the evidence that shows how autophagy can be both beneficial and detrimental as it pertains to aortic valve calcification in the heart.
Collapse
Affiliation(s)
- Yunlong Fan
- Medical School of Chinese PLA, Beijing 100853, China.
- Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Jiakang Shao
- Medical School of Chinese PLA, Beijing 100853, China.
| | - Shixiong Wei
- Medical School of Chinese PLA, Beijing 100853, China.
- Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Chao Song
- Medical School of Chinese PLA, Beijing 100853, China.
- Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Yanan Li
- Medical School of Chinese PLA, Beijing 100853, China.
| | - Shengli Jiang
- Medical School of Chinese PLA, Beijing 100853, China.
- Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
30
|
Superimposed Tissue Formation in Human Aortic Valve Disease: Differences between Regurgitant and Stenotic Valves. J Cardiovasc Dev Dis 2021; 8:jcdd8070079. [PMID: 34357322 PMCID: PMC8306480 DOI: 10.3390/jcdd8070079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/29/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
The formation of superimposed tissue (SIT), a layer on top of the original valve leaflet, has been described in patients with mitral regurgitation as a major contributor to valve thickening and possibly as a result of increased mechanical stresses. However, little is known whether SIT formation also occurs in aortic valve disease. We therefore performed histological analyses to assess SIT formation in aortic valve leaflets (n = 31) from patients with aortic stenosis (n = 17) or aortic regurgitation due to aortic dilatation (n = 14). SIT was observed in both stenotic and regurgitant aortic valves, both on the ventricular and aortic sides, but with significant differences in distribution and composition. Regurgitant aortic valves showed more SIT formation in the free edge, leading to a thicker leaflet at that level, while stenotic aortic valves showed relatively more SIT formation on the aortic side of the body part of the leaflet. SIT appeared to be a highly active area, as determined by large populations of myofibroblasts, with varied extracellular matrix composition (higher collagen content in stenotic valves). Further, the identification of the SIT revealed the presence of foldings of the free edge in the diseased aortic valves. Insights into SIT regulation may further help in understanding the pathophysiology of aortic valve disease and potentially lead to the development of new therapeutic treatments.
Collapse
|
31
|
Wang Y, Fang Y, Lu P, Wu B, Zhou B. NOTCH Signaling in Aortic Valve Development and Calcific Aortic Valve Disease. Front Cardiovasc Med 2021; 8:682298. [PMID: 34239905 PMCID: PMC8259786 DOI: 10.3389/fcvm.2021.682298] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/14/2021] [Indexed: 01/05/2023] Open
Abstract
NOTCH intercellular signaling mediates the communications between adjacent cells involved in multiple biological processes essential for tissue morphogenesis and homeostasis. The NOTCH1 mutations are the first identified human genetic variants that cause congenital bicuspid aortic valve (BAV) and calcific aortic valve disease (CAVD). Genetic variants affecting other genes in the NOTCH signaling pathway may also contribute to the development of BAV and the pathogenesis of CAVD. While CAVD occurs commonly in the elderly population with tri-leaflet aortic valve, patients with BAV have a high risk of developing CAVD at a young age. This observation indicates an important role of NOTCH signaling in the postnatal homeostasis of the aortic valve, in addition to its prenatal functions during aortic valve development. Over the last decade, animal studies, especially with the mouse models, have revealed detailed information in the developmental etiology of congenital aortic valve defects. In this review, we will discuss the molecular and cellular aspects of aortic valve development and examine the embryonic pathogenesis of BAV. We will focus our discussions on the NOTCH signaling during the endocardial-to-mesenchymal transformation (EMT) and the post-EMT remodeling of the aortic valve. We will further examine the involvement of the NOTCH mutations in the postnatal development of CAVD. We will emphasize the deleterious impact of the embryonic valve defects on the homeostatic mechanisms of the adult aortic valve for the purpose of identifying the potential therapeutic targets for disease intervention.
Collapse
Affiliation(s)
- Yidong Wang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yuan Fang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Bin Zhou
- Departments of Genetics, Pediatrics (Pediatric Genetic Medicine), and Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States
- The Einstein Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
32
|
Fluid Flow Characteristics of Healthy and Calcified Aortic Valves Using Three-Dimensional Lagrangian Coherent Structures Analysis. FLUIDS 2021. [DOI: 10.3390/fluids6060203] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Aortic valve calcification is an important cardiovascular disorder that deteriorates the accurate functioning of the valve leaflets. The increasing stiffness due to the calcification prevents the complete closure of the valve and therefore leads to significant hemodynamic alterations. Computational fluid dynamics (CFD) modeling enables the investigation of the entire flow domain by processing medical images from aortic valve patients. In this study, we computationally modeled and simulated a 3D aortic valve using patient-specific dimensions of the aortic root and aortic sinus. Leaflet stiffness is deteriorated in aortic valve disease due to calcification. In order to investigate the influence of leaflet calcification on flow dynamics, three different leaflet-stiffness values were considered for healthy, mildly calcified, and severely calcified leaflets. Time-dependent CFD results were used for applying the Lagrangian coherent structures (LCS) technique by performing finite-time Lyapunov exponent (FTLE) computations along with Lagrangian particle residence time (PRT) analysis to identify unique vortex structures at the front and backside of the leaflets. Obtained results indicated that the peak flow velocity at the valve orifice increased with the calcification rate. For the healthy aortic valve, a low-pressure field was observed at the leaflet tips. This low-pressure field gradually expanded through the entire aortic sinus as the calcification level increased. FTLE field plots of the healthy and calcified valves showed a variety of differences in terms of flow structures. When the number of fluid particles in the healthy valve model was taken as reference, 1.59 and 1.74 times more particles accumulated in the mildly and severely calcified valves, respectively, indicating that the calcified valves were not sufficiently opened to allow normal mass flow rates.
Collapse
|
33
|
Protective Effects of Fucoxanthin on Hydrogen Peroxide-Induced Calcification of Heart Valve Interstitial Cells. Mar Drugs 2021; 19:md19060307. [PMID: 34073219 PMCID: PMC8227531 DOI: 10.3390/md19060307] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases such as atherosclerosis and aortic valve sclerosis involve inflammatory reactions triggered by various stimuli, causing increased oxidative stress. This increased oxidative stress causes damage to the heart cells, with subsequent cell apoptosis or calcification. Currently, heart valve damage or heart valve diseases are treated by drugs or surgery. Natural antioxidant products are being investigated in related research, such as fucoxanthin (Fx), which is a marine carotenoid extracted from seaweed, with strong antioxidant, anti-inflammatory, and anti-tumor properties. This study aimed to explore the protective effect of Fx on heart valves under high oxidative stress, as well as the underlying mechanism of action. Rat heart valve interstitial cells under H2O2-induced oxidative stress were treated with Fx. Fx improved cell survival and reduced oxidative stress-induced DNA damage, which was assessed by cell viability analysis and staining with propidium iodide. Alizarin Red-S analysis indicated that Fx has a protective effect against calcification. Furthermore, Western blotting revealed that Fx abrogates oxidative stress-induced apoptosis via reducing the expression of apoptosis-related proteins as well as modulate Akt/ERK-related protein expression. Notably, in vivo experiments using 26 dogs treated with 60 mg/kg of Fx in combination with medical treatment for 0.5 to 2 years showed significant recovery in their echocardiographic parameters. Collectively, these in vitro and in vivo results highlight the potential of Fx to protect heart valve cells from high oxidative stress-induced damage.
Collapse
|
34
|
Wang D, Xiong T, Yu W, Liu B, Wang J, Xiao K, She Q. Predicting the Key Genes Involved in Aortic Valve Calcification Through Integrated Bioinformatics Analysis. Front Genet 2021; 12:650213. [PMID: 34046056 PMCID: PMC8144713 DOI: 10.3389/fgene.2021.650213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/05/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Valvular heart disease is obtaining growing attention in the cardiovascular field and it is believed that calcific aortic valve disease (CAVD) is the most common valvular heart disease (VHD) in the world. CAVD does not have a fully effective treatment to delay its progression and the specific molecular mechanism of aortic valve calcification remains unclear. Materials and Methods: We obtained the gene expression datasets GSE12644 and GSE51472 from the public comprehensive free database GEO. Then, a series of bioinformatics methods, such as GO and KEGG analysis, STING online tool, Cytoscape software, were used to identify differentially expressed genes in CAVD and healthy controls, construct a PPI network, and then identify key genes. In addition, immune infiltration analysis was used via CIBERSORT to observe the expression of various immune cells in CAVD. Results: A total of 144 differential expression genes were identified in the CAVD samples in comparison with the control samples, including 49 up-regulated genes and 95 down-regulated genes. GO analysis of DEGs were most observably enriched in the immune response, signal transduction, inflammatory response, proteolysis, innate immune response, and apoptotic process. The KEGG analysis revealed that the enrichment of DEGs in CAVD were remarkably observed in the chemokine signaling pathway, cytokine-cytokine receptor interaction, and PI3K-Akt signaling pathway. Chemokines CXCL13, CCL19, CCL8, CXCL8, CXCL16, MMP9, CCL18, CXCL5, VCAM1, and PPBP were identified as the hub genes of CAVD. It was macrophages that accounted for the maximal proportion among these immune cells. The expression of macrophages M0, B cells memory, and Plasma cells were higher in the CAVD valves than in healthy valves, however, the expression of B cells naïve, NK cells activated, and macrophages M2 were lower. Conclusion: We detected that chemokines CXCL13, CXCL8, CXCL16, and CXCL5, and CCL19, CCL8, and CCL18 are the most important markers of aortic valve disease. The regulatory macrophages M0, plasma cells, B cells memory, B cells naïve, NK cells activated, and macrophages M2 are probably related to the occurrence and the advancement of aortic valve stenosis. These identified chemokines and these immune cells may interact with a subtle adjustment relationship in the development of calcification in CAVD.
Collapse
Affiliation(s)
- Dinghui Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tianhua Xiong
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenlong Yu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kaihu Xiao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
35
|
Mirani B, Parvin Nejad S, Simmons CA. Recent Progress Toward Clinical Translation of Tissue-Engineered Heart Valves. Can J Cardiol 2021; 37:1064-1077. [PMID: 33839245 DOI: 10.1016/j.cjca.2021.03.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/04/2021] [Accepted: 03/14/2021] [Indexed: 01/02/2023] Open
Abstract
Surgical replacement remains the primary option to treat the rapidly growing number of patients with severe valvular heart disease. Although current valve replacements-mechanical, bioprosthetic, and cryopreserved homograft valves-enhance survival and quality of life for many patients, the ideal prosthetic heart valve that is abundantly available, immunocompatible, and capable of growth, self-repair, and life-long performance has yet to be developed. These features are essential for pediatric patients with congenital defects, children and young adult patients with rheumatic fever, and active adult patients with valve disease. Heart valve tissue engineering promises to address these needs by providing living valve replacements that function similarly to their native counterparts. This is best evidenced by the long-term clinical success of decellularised pulmonary and aortic homografts, but the supply of homografts cannot meet the demand for replacement valves. A more abundant and consistent source of replacement valves may come from cellularised valves grown in vitro or acellular off-the-shelf biomaterial/tissue constructs that recellularise in situ, but neither tissue engineering approach has yet achieved long-term success in preclinical testing. Beyond the technical challenges, heart valve tissue engineering faces logistical, economic, and regulatory challenges. In this review, we summarise recent progress in heart valve tissue engineering, highlight important outcomes from preclinical and clinical testing, and discuss challenges and future directions toward clinical translation.
Collapse
Affiliation(s)
- Bahram Mirani
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Shouka Parvin Nejad
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Craig A Simmons
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
36
|
Kruithof BPT, van de Pol V, Los T, Lodder K, Gourabi BM, DeRuiter MC, Goumans MJ, Ajmone Marsan N. New calcification model for intact murine aortic valves. J Mol Cell Cardiol 2021; 156:95-104. [PMID: 33744308 DOI: 10.1016/j.yjmcc.2021.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023]
Abstract
Calcific aortic valve disease (CAVD) is a common progressive disease of the aortic valves, for which no medical treatment exists and surgery represents currently the only therapeutic solution. The development of novel pharmacological treatments for CAVD has been hampered by the lack of suitable test-systems, which require the preservation of the complex valve structure in a mechanically and biochemical controllable system. Therefore, we aimed at establishing a model which allows the study of calcification in intact mouse aortic valves by using the Miniature Tissue Culture System (MTCS), an ex vivo flow model for whole mouse hearts. Aortic valves of wild-type mice were cultured in the MTCS and exposed to osteogenic medium (OSM, containing ascorbic acid, β-glycerophosphate and dexamethasone) or inorganic phosphates (PI). Osteogenic calcification occurred in the aortic valve leaflets that were cultured ex vivo in the presence of PI, but not of OSM. In vitro cultured mouse and human valvular interstitial cells calcified in both OSM and PI conditions, revealing in vitro-ex vivo differences. Furthermore, endochondral differentiation occurred in the aortic root of ex vivo cultured mouse hearts near the hinge of the aortic valve in both PI and OSM conditions. Dexamethasone was found to induce endochondral differentiation in the aortic root, but to inhibit calcification and the expression of osteogenic markers in the aortic leaflet, partly explaining the absence of calcification in the aortic valve cultured with OSM. The osteogenic calcifications in the aortic leaflet and the endochondral differentiation in the aortic root resemble calcifications found in human CAVD. In conclusion, we have established an ex vivo calcification model for intact wild-type murine aortic valves in which the initiation and progression of aortic valve calcification can be studied. The in vitro-ex vivo differences found in our studies underline the importance of ex vivo models to facilitate pre-clinical translational studies.
Collapse
Affiliation(s)
- Boudewijn P T Kruithof
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands; Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands; Netherlands Heart Institute, Utrecht, The Netherlands.
| | - Vera van de Pol
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tamara Los
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kirsten Lodder
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Babak Mousavi Gourabi
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marco C DeRuiter
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nina Ajmone Marsan
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
37
|
Kanda H, Yamakuchi M, Matsumoto K, Mukaihara K, Shigehisa Y, Tachioka S, Okawa M, Takenouchi K, Oyama Y, Hashiguchi T, Imoto Y. Dynamic changes in platelets caused by shear stress in aortic valve stenosis. Clin Hemorheol Microcirc 2021; 77:71-81. [PMID: 32924997 PMCID: PMC7990459 DOI: 10.3233/ch-200928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND AND OBJECTIVE: Turbulent blood flow in patients with aortic valve stenosis (AS) results in morphological and functional changes in platelets and coagulation factors. The aim of this study is to determine how shear stress affects platelets and coagulation factors. METHODS: We retrospectively evaluated data from 78 patients who underwent AVR to treat AS between March 2008 and July 2017 at Kagoshima University Hospital. RESULTS: Platelet (PLT) count obviously decreased at three days after AVR, and increased above preoperative levels at the time of discharge. In contrast, platelet distribution width (PDW), mean platelet volume (MPV), and platelet large cell ratio (P-LCR) increased three days after AVR, then decreased to below preoperative levels. No differences were evident between groups with higher (HPPG > 100 mmHg) and lower (LPPG < 100 mmHg) peak pressure gradients (PPG) before AVR, whereas PLT count, PDW, MPV and P-LCR improved more in the HPPG group. Plateletcrit (PCT), which represents the total volume of platelets, increased after AVR due to decreased shear stress. High increasing rate of PCT was associated with lower PLT count, higher PDW and lower fibrinogen. CONCLUSION: Shear stress affects PLT count, PDW, and fibrinogen in patients with AS.
Collapse
Affiliation(s)
- Hideaki Kanda
- Cardiovascular and Gastroenterological Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Munekazu Yamakuchi
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kazuhisa Matsumoto
- Cardiovascular and Gastroenterological Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kosuke Mukaihara
- Cardiovascular and Gastroenterological Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yoshiya Shigehisa
- Cardiovascular and Gastroenterological Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shuji Tachioka
- Cardiovascular and Gastroenterological Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Masashi Okawa
- Cardiovascular and Gastroenterological Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kazunori Takenouchi
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yoko Oyama
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Teruto Hashiguchi
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yutaka Imoto
- Cardiovascular and Gastroenterological Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
38
|
Shahriyari HA, Nikmanesh Y, Jalali S, Tahery N, Zhiani Fard A, Hatamzadeh N, Zarea K, Cheraghi M, Mohammadi MJ. Air pollution and human health risks: mechanisms and clinical manifestations of cardiovascular and respiratory diseases. TOXIN REV 2021. [DOI: 10.1080/15569543.2021.1887261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
| | - Yousef Nikmanesh
- Gastroenterohepatology Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Saeid Jalali
- Department of Environmental Health Engineering, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Noorollah Tahery
- Department of Nursing, School of Nursing, Abadan Faculty of Medical Sciences, Abadan, Iran
| | - Akram Zhiani Fard
- Instructor Medical Education, Department of Public Health, Esfarayen Faculty of Medical Sciences, Esfarayen, Iran
| | - Nasser Hatamzadeh
- Department of Health Promotion and Education, School of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kourosh Zarea
- Department of Nursing, Nursing Care Research Center in Chronic Diseases, School of Nursing and Midwifery, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maria Cheraghi
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Mohammadi
- Department of Environmental Health Engineering, School of Public Health and Environmental Technologies Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
39
|
Wu S, Kumar V, Xiao P, Kuss M, Lim JY, Guda C, Butcher J, Duan B. Age related extracellular matrix and interstitial cell phenotype in pulmonary valves. Sci Rep 2020; 10:21338. [PMID: 33288823 PMCID: PMC7721746 DOI: 10.1038/s41598-020-78507-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/24/2020] [Indexed: 12/21/2022] Open
Abstract
Heart valve disease is a common manifestation of cardiovascular disease and is a significant cause of cardiovascular morbidity and mortality worldwide. The pulmonary valve (PV) is of primary concern because of its involvement in common congenital heart defects, and the PV is usually the site for prosthetic replacement following a Ross operation. Although effects of age on valve matrix components and mechanical properties for aortic and mitral valves have been studied, very little is known about the age-related alterations that occur in the PV. In this study, we isolated PV leaflets from porcine hearts in different age groups (~ 4-6 months, denoted as young versus ~ 2 years, denoted as adult) and studied the effects of age on PV leaflet thickness, extracellular matrix components, and mechanical properties. We also conducted proteomics and RNA sequencing to investigate the global changes of PV leaflets and passage zero PV interstitial cells in their protein and gene levels. We found that the size, thickness, elastic modulus, and ultimate stress in both the radial and circumferential directions and the collagen of PV leaflets increased from young to adult age, while the ultimate strain and amount of glycosaminoglycans decreased when age increased. Young and adult PV had both similar and distinct protein and gene expression patterns that are related to their inherent physiological properties. These findings are important for us to better understand the physiological microenvironments of PV leaflet and valve cells for correctively engineering age-specific heart valve tissues.
Collapse
Affiliation(s)
- Shaohua Wu
- College of Textiles & Clothing, Qingdao University, Qingdao, People's Republic of China
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Vikas Kumar
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peng Xiao
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jonathan Butcher
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA.
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
40
|
Flemister DC, Hatoum H, Guhan V, Zebhi B, Lincoln J, Crestanello J, Dasi LP. Effect of Left and Right Coronary Flow Waveforms on Aortic Sinus Hemodynamics and Leaflet Shear Stress: Correlation with Calcification Locations. Ann Biomed Eng 2020; 48:2796-2808. [PMID: 33145675 PMCID: PMC11022940 DOI: 10.1007/s10439-020-02677-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022]
Abstract
Coronary flow induces hemodynamic alterations in the aortic sinus region. The objectives of this study are to: (1) investigate the differences among sinus hemodynamics and leaflet wall shear stresses engendered by the left versus right versus non-coronary flow and (2) correlate respective wall shear stresses with leaflet calcification in patients. A left heart simulator flow loop with a tunable coronary circuit provided physiological coronary flow waveforms corresponding to the left coronary cusp case (LCC), right coronary cusp case (RCC), and non-coronary cusp case (NCC). High spatio-temporal resolution particle image velocimetry was conducted to quantify leaflet wall shear stress and sinus vorticity fields and to measure aortic leaflet tip kinematics. Thirty-one patients with severe calcific aortic valve disease were segmented from CT data for the calcific volumes in their respective left, right, and non-coronary cusps. Leaflet tip position during systole shows the RCC has a wider leaflet opening compared to LCC and NCC. Velocity and vorticity fields combined with leaflet position data show that sinus vorticity is diminished (peak ~ 43 s-1) in the LCC while RCC and NCC maintain high vorticity (~ 1200 and ~ 950 s-1 respectively). WSS magnitudes greater than 0.3 Pa show 20 and 81% greater occurrences in the LCC and RCC respectively compared to NCC. Significant differences [X2 (2, n = 31) = 7.31, p = 0.0258] between the calcification levels in each cusp of the patient population. Coronary flow differences between LCC, RCC, and NCC show significant impact on leaflet kinematics and sinus flow hemodynamics. Clinical data correlations of the coronary flow cases indicate the left coronary cusp has a higher likelihood of calcification compared to the right.
Collapse
Affiliation(s)
- Dorma C Flemister
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Hoda Hatoum
- Department of Biomedical Engineering, Georgia Institute of Technology, 387 Technology Circle, Atlanta, GA, 30313, USA
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, 49931, USA
| | - Varshini Guhan
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Banafsheh Zebhi
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Joy Lincoln
- Department of Pediatric Cardiology, Children's Hospital of Wisconsin, Milwaukee, WI, USA
| | | | - Lakshmi P Dasi
- Department of Biomedical Engineering, Georgia Institute of Technology, 387 Technology Circle, Atlanta, GA, 30313, USA.
| |
Collapse
|
41
|
miR-214 Attenuates Aortic Valve Calcification by Regulating Osteogenic Differentiation of Valvular Interstitial Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:971-980. [PMID: 33251046 PMCID: PMC7679242 DOI: 10.1016/j.omtn.2020.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/10/2020] [Indexed: 01/04/2023]
Abstract
Calcific aortic valve disease (CAVD) is a common heart valve disease in aging populations, and aberrant osteogenic differentiation of valvular interstitial cells (VICs) plays a critical role in the pathogenesis of ectopic ossification of the aortic valve. miR-214 has been validated to be involved in the osteogenesis process. Here, we aim to investigate the role and mechanism of miR-214 in CAVD progression. miR-214 expression was significantly downregulated in CAVD aortic valve leaflets, accompanied by upregulation of osteogenic markers. Overexpression of miR-214 suppressed osteogenic differentiation of VICs, while silencing the expression of miR-214 promoted this function. miR-214 directly targeted ATF4 and Sp7 to modulate osteoblastic differentiation of VICs, which was proved by dual luciferase reporter assay and rescue experiment. miR-214 knockout rats exhibited higher mean transvalvular velocity and gradient. The expression of osteogenic markers in aortic valve leaflets of miR-214 knockout rats was upregulated compared to that of the wild-type group. Taken together, our study showed that miR-214 inhibited aortic valve calcification via regulating osteogenic differentiation of VICs by directly targeting ATF4 and Sp7, indicating that miR-214 may act as a profound candidate of targeting therapy for CAVD.
Collapse
|
42
|
Poulis N, Zaytseva P, Gähwiler EKN, Motta SE, Fioretta ES, Cesarovic N, Falk V, Hoerstrup SP, Emmert MY. Tissue engineered heart valves for transcatheter aortic valve implantation: current state, challenges, and future developments. Expert Rev Cardiovasc Ther 2020; 18:681-696. [DOI: 10.1080/14779072.2020.1792777] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Nikolaos Poulis
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Polina Zaytseva
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Eric K. N. Gähwiler
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Sarah E. Motta
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Wyss Translational Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | | | - Nikola Cesarovic
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology in Zurich, Zurich, Switzerland
| | - Volkmar Falk
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology in Zurich, Zurich, Switzerland
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- German Center of Cardiovascular Research, Partner Site Berlin, Berlin, Germany
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Wyss Translational Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Wyss Translational Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
| |
Collapse
|
43
|
Liu FY, Bai P, Jiang YF, Dong NG, Li G, Chu C. Role of Interleukin 17A in Aortic Valve Inflammation in Apolipoprotein E-deficient Mice. Curr Med Sci 2020; 40:729-738. [PMID: 32862384 DOI: 10.1007/s11596-020-2230-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 06/10/2020] [Indexed: 12/29/2022]
Abstract
Interleukin 17A (IL17A) is reported to be involved in many inflammatory processes, but its role in aortic valve diseases remains unknown. We examined the role of IL17A based on an ApoE-/- mouse model with strategies as fed with high-fat diet or treated with IL17A monoclonal antibody (mAb). 12 weeks of high-fat diet feeding can elevate cytokines secretion, inflammatory cells infiltration and myofibroblastic transition of valvular interstitial cells (VICs) in aortic valve. Moreover, diet-induction accelerated interleukin 17 receptor A (IL17RA) activation in VICs. In an IL17A inhibition model, the treatment group was intra-peritoneally injected with anti-IL17A mAb while controls received irrelevant antibody. Functional blockade of IL17A markedly reduced cellular infiltration and transition in aortic valve. To investigate potential mechanisms, NF-κB was co-stained in IL17RA+ VICs and IL17RA+ macrophages, and further confirmed by Western blotting in VICs. High-fat diet could activate NF-κB nuclear translocation in IL17RA+ VICs and IL17RA+ macrophages and this process was depressed after IL17A mAb-treatment. In conclusion, high-fat diet can lead to IL17A upregulation, VICs myofibroblastic transition and inflammatory cells infiltration in the aortic value of ApoE-/- mice. Blocking IL17A with IL17A mAb can alleviate aortic valve inflammatory states.
Collapse
Affiliation(s)
- Fa-Yuan Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Peng Bai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ye-Fan Jiang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nian-Guo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Geng Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chong Chu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
44
|
Greene CL, Jaatinen KJ, Wang H, Koyano TK, Bilbao MS, Woo YJ. Transcriptional Profiling of Normal, Stenotic, and Regurgitant Human Aortic Valves. Genes (Basel) 2020; 11:genes11070789. [PMID: 32674273 PMCID: PMC7397246 DOI: 10.3390/genes11070789] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/01/2020] [Accepted: 07/08/2020] [Indexed: 12/26/2022] Open
Abstract
The genetic mechanisms underlying aortic stenosis (AS) and aortic insufficiency (AI) disease progression remain unclear. We hypothesized that normal aortic valves and those with AS or AI all exhibit unique transcriptional profiles. Normal control (NC) aortic valves were collected from non-matched donor hearts that were otherwise acceptable for transplantation (n = 5). Valves with AS or AI (n = 5, each) were collected from patients undergoing surgical aortic valve replacement. High-throughput sequencing of total RNA revealed 6438 differentially expressed genes (DEGs) for AS vs. NC, 4994 DEGs for AI vs. NC, and 2771 DEGs for AS vs. AI. Among 21 DEGs of interest, APCDD1L, CDH6, COL10A1, HBB, IBSP, KRT14, PLEKHS1, PRSS35, and TDO2 were upregulated in both AS and AI compared to NC, whereas ALDH1L1, EPHB1, GPX3, HIF3A, and KCNT1 were downregulated in both AS and AI (p < 0.05). COL11A1, H19, HIF1A, KCNJ6, PRND, and SPP1 were upregulated only in AS, and NPY was downregulated only in AS (p < 0.05). The functional network for AS clustered around ion regulation, immune regulation, and lipid homeostasis, and that for AI clustered around ERK1/2 regulation. Overall, we report transcriptional profiling data for normal human aortic valves from non-matched donor hearts that were acceptable for transplantation and demonstrated that valves with AS and AI possess unique genetic signatures. These data create a roadmap for the development of novel therapeutics to treat AS and AI.
Collapse
Affiliation(s)
- Christina L. Greene
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA; (C.L.G.); (K.J.J.); (H.W.); (T.K.K.); (M.S.B.)
| | - Kevin J. Jaatinen
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA; (C.L.G.); (K.J.J.); (H.W.); (T.K.K.); (M.S.B.)
| | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA; (C.L.G.); (K.J.J.); (H.W.); (T.K.K.); (M.S.B.)
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| | - Tiffany K. Koyano
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA; (C.L.G.); (K.J.J.); (H.W.); (T.K.K.); (M.S.B.)
| | - Mary S. Bilbao
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA; (C.L.G.); (K.J.J.); (H.W.); (T.K.K.); (M.S.B.)
| | - Y. Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA; (C.L.G.); (K.J.J.); (H.W.); (T.K.K.); (M.S.B.)
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Correspondence: ; Tel.: +1-650-725-3828
| |
Collapse
|
45
|
Yang H, Song Y, Chen J, Pang Z, Zhang N, Cao J, Wang Q, Li Q, Zhang F, Dai Y, Li C, Huang Z, Qian J, Ge J. Platelet Membrane-Coated Nanoparticles Target Sclerotic Aortic Valves in ApoE -/- Mice by Multiple Binding Mechanisms Under Pathological Shear Stress. Int J Nanomedicine 2020; 15:901-912. [PMID: 32103945 PMCID: PMC7020933 DOI: 10.2147/ijn.s224024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 01/09/2020] [Indexed: 11/23/2022] Open
Abstract
Background Aortic valve disease is the most common valvular heart disease leading to valve replacement. The efficacy of pharmacological therapy for aortic valve disease is limited by the high mechanical stress at the aortic valves impairing the binding rate. We aimed to identify nanoparticle coating with entire platelet membranes to fully mimic their inherent multiple adhesive mechanisms and target the sclerotic aortic valve of apolipoprotein E-deficient (ApoE−/−) mice based on their multiple sites binding capacity under high shear stress. Methods Considering the potent interaction of platelet membrane glycoproteins with components present in sclerotic aortic valves, platelet membrane-coated nanoparticles (PNPs) were synthetized and the binding capacity under high shear stress was evaluated in vitro and in vivo. Results PNPs demonstrated effectively adhering to von Willebrand factor, collagen and fibrin under shear stresses in vitro. In an aortic valve disease model established in ApoE−/− mice, PNPs exhibited good targeting to sclerotic aortic valves by mimicking platelet multiple adhesive mechanisms. Conclusion PNPs could provide a promising platform for the molecular diagnosis and targeting treatment of aortic valve disease.
Collapse
Affiliation(s)
- Hongbo Yang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Yanan Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Jing Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Zhiqing Pang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, People's Republic of China
| | - Ning Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Jiatian Cao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Qiaozi Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Qiyu Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Feng Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Chenguang Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Zheyong Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, People's Republic of China
| |
Collapse
|
46
|
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: 11] [Impact Index Per Article: 1.8] [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
|
47
|
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
|
48
|
Simmons CA, Jo H. Editorial: Special Issue on Heart Valve Mechanobiology : New Insights into Mechanical Regulation of Valve Disease and Regeneration. Cardiovasc Eng Technol 2019; 9:121-125. [PMID: 29761407 DOI: 10.1007/s13239-018-0360-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Craig A Simmons
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada. .,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada. .,Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada.
| | - Hanjoong Jo
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA. .,Department of Cardiology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
49
|
Varshney R, Murphy B, Woolington S, Ghafoory S, Chen S, Robison T, Ahamed J. Inactivation of platelet-derived TGF-β1 attenuates aortic stenosis progression in a robust murine model. Blood Adv 2019; 3:777-788. [PMID: 30846427 PMCID: PMC6418501 DOI: 10.1182/bloodadvances.2018025817] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/24/2019] [Indexed: 11/20/2022] Open
Abstract
Aortic stenosis (AS) is a degenerative heart condition characterized by fibrosis and narrowing of aortic valves (AV), resulting in high wall shear stress (WSS) across valves. AS is associated with high plasma levels of transforming growth factor-β1 (TGF-β1), which can be activated by WSS to induce organ fibrosis, but the cellular source of TGF-β1 is not clear. Here, we show that platelet-derived TGF-β1 plays an important role in AS progression. We first established an aggressive and robust murine model of AS, using the existing Ldlr -/- Apob100/100 (LDLR) breed of mice, and accelerated AS progression by feeding them a high-fat diet (HFD). We then captured very high resolution images of AV movement and thickness and of blood flow velocity across the AV, using a modified ultrasound imaging technique, which revealed early evidence of AS and distinguished different stages of AS progression. More than 90% of LDLR animals developed AS within 6 months of HFD. Scanning electron microscopy and whole-mount immunostaining imaging of AV identified activated platelets physically attached to valvular endothelial cells (VEC) expressing high phosphorylated Smad2 (p-Smad2). To test the contribution of platelet-derived TGF-β1 in AS, we derived LDLR mice lacking platelet TGF-β1 (TGF-β1platelet-KO-LDLR) and showed reduced AS progression and lower p-Smad2 and myofibroblasts in their AV compared with littermate controls fed the HFD for 6 months. Our data suggest that platelet-derived TGF-β1 triggers AS progression by inducing signaling in VEC, and their subsequent transformation into collagen-producing-myofibroblasts. Thus, inhibiting platelet-derived TGF-β1 might attenuate or prevent fibrotic diseases characterized by platelet activation and high WSS, such as AS.
Collapse
Affiliation(s)
- Rohan Varshney
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK; and
| | - Brennah Murphy
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK; and
| | - Sean Woolington
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK; and
| | - Shahrouz Ghafoory
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK; and
| | - Sixia Chen
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Tyler Robison
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK; and
| | - Jasimuddin Ahamed
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK; and
| |
Collapse
|
50
|
Duan B, Xu C, Das S, Chen JM, Butcher JT. Spatial Regulation of Valve Interstitial Cell Phenotypes within Three-Dimensional Micropatterned Hydrogels. ACS Biomater Sci Eng 2019; 5:1416-1425. [PMID: 33405617 PMCID: PMC10951959 DOI: 10.1021/acsbiomaterials.8b01280] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcific aortic valve disease (CAVD) is the third leading cause of cardiovascular disease. CAVD exhibits progressive disruption of the normally highly organized and aligned extracellular matrix (ECM) structure within the valve leaflets simultaneously with myofibroblastic and/or osteogenic differentiation of indigenous endogenous valve interstitial cells (VIC). It is unclear how the alignment of VIC within their 3D microenvironment drives VIC phenotype or how alignment affects cellular responses to biochemical cues in physiological or pathological conditions. In this study, we implement a photolithographic technique to control the alignment and elongation of both normal and diseased human aortic VIC (HAVIC) within microengineered 3D hydrogels consisting of methacrylated hyaluronic acid and methacrylated gelatin. Stripe micropatterning created distinct alignment of HAVIC within a 3D culture system, which promoted spreading and enhanced their activation and osteogenic differentiation in pro-osteogenic conditions. HAVIC from a patient with CAVD exhibited greater susceptibility to myofibroblastic and osteogenic differentiation in culture. The roles of conjugated basic fibroblastic growth factor (bFGF) and RhoA/ROCK pathway in regulating HAVIC phenotypes were also investigated in the presence of aligned microtopography. The addition of bFGF was preventative to osteogenic differentiation for healthy HAVIC; however, it promoted osteogenic differentiation in diseased HAVIC. Inhibition of the ROCK pathway only decreased osteogenic differentiation for diseased HAVIC in the aligned formation. Collectively, these results improve our knowledge of the effects that VIC alignment has on VIC phenotypes and valve disease progression. The cell culture platform also enables a better understanding of the interplay between topography, biochemical cues, and VIC differentiation and provides information useful for directing differentiation as well as valve tissue regeneration.
Collapse
Affiliation(s)
- Bin Duan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
| | - Charlie Xu
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Shoshana Das
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jonathan M. Chen
- Department of Cardiac Surgery, Seattle Children’s Hospital, Seattle WA, USA
| | - Jonathan T. Butcher
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|