1
|
Myasoedova VA, Franchi M, De Giorgi D, Bonomi A, Valerio V, Pirola S, Andreani N, Rusconi V, Bertolini F, Massaiu I, Pontone G, Poggio P. High-Intensity Statins Promote PCSK9 Secretion and aortic valve calcification in patients with severe aortic stenosis: In vitro and clinical evidence. Pharmacol Res 2025; 215:107737. [PMID: 40239750 DOI: 10.1016/j.phrs.2025.107737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
Aortic stenosis (AS) is the most common valvular disease, characterized by progressive fibro-calcific remodeling of the aortic leaflets, leading to increased morbidity and mortality. It is now well known that statins influence the production of proprotein convertase subtilisin/kexin type 9 (PCSK9), which in turn is linked to calcification. Here, we found that statins significantly increased, in a dose dependent manner, both PCSK9 secretion and valve interstitial cell (VIC) calcification, in vitro. These effects were blunted by PCSK9 genetic knock-down or by PCSK9 antibody neutralization. In AS patients, contrast-enhanced computed tomography evaluation showed a higher aortic valve calcium (AVC) content in patients on high-intensity statins compared to low-intensity ones, with no significant difference between low-intensity statin and non-users. At follow-up, high-intensity statin users exhibited a higher annual AVC accumulation compared to low-intensity statins and non-users. In a real-world scenario, high-intensity statin therapy was associated with a 30 % increased rate of hospitalization for non-rheumatic aortic valve disease. Our findings highlight the need for further investigation into the intricate relationship between statin therapy and aortic valve health to identify the optimal lipid-lowering strategy in the management of patients at risk of developing or afflicted by AS.
Collapse
Affiliation(s)
| | - Matteo Franchi
- Centro Cardiologico Monzino IRCCS, Milan 20138, Italy; Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan 20126, Italy
| | | | - Alice Bonomi
- Centro Cardiologico Monzino IRCCS, Milan 20138, Italy
| | | | - Sergio Pirola
- Centro Cardiologico Monzino IRCCS, Milan 20138, Italy
| | - Niccolò Andreani
- Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan 20126, Italy
| | | | | | | | - Gianluca Pontone
- Centro Cardiologico Monzino IRCCS, Milan 20138, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan 20122, Italy
| | - Paolo Poggio
- Centro Cardiologico Monzino IRCCS, Milan 20138, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan 20122, Italy.
| |
Collapse
|
2
|
Gramling DP, van Veldhuisen AL, Damen FW, Thatcher K, Liu F, McComb D, Lincoln J, Breuer CK, Goergen CJ, Sacks MS. In Vivo Three-Dimensional Geometric Reconstruction of the Mouse Aortic Heart Valve. Ann Biomed Eng 2024; 52:2596-2609. [PMID: 38874705 DOI: 10.1007/s10439-024-03555-4] [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: 04/30/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024]
Abstract
Aortic valve (AV) disease is a common valvular lesion in the United States, present in about 5% of the population at age 65 with increasing prevalence with advancing age. While current replacement heart valves have extended life for many, their long-term use remains hampered by limited durability. Non-surgical treatments for AV disease do not yet exist, in large part because our understanding of AV disease etiology remains incomplete. The direct study of human AV disease remains hampered by the fact that clinical data is only available at the time of treatment, where the disease is at or near end stage and any time progression information has been lost. Large animal models, long used to assess replacement AV devices, cannot yet reproduce AV disease processes. As an important alternative mouse animal models are attractive for their ability to perform genetic studies of the AV disease processes and test potential pharmaceutical treatments. While mouse models have been used for cellular and genetic studies of AV disease, their small size and fast heart rates have hindered their use for tissue- and organ-level studies. We have recently developed a novel ex vivo micro-CT-based methodology to 3D reconstruct murine heart valves and estimate the leaflet mechanical behaviors (Feng et al. in Sci Rep 13(1):12852, 2023). In the present study, we extended our approach to 3D reconstruction of the in vivo functional murine AV (mAV) geometry using high-frequency four-dimensional ultrasound (4DUS). From the resulting 4DUS images we digitized the mAV mid-surface coordinates in the fully closed and fully opened states. We then utilized matched high-resolution µCT images of ex vivo mouse mAV to develop mAV NURBS-based geometric model. We then fitted the mAV geometric model to the in vivo data to reconstruct the 3D in vivo mAV geometry in the closed and open states in n = 3 mAV. Results demonstrated high fidelity geometric results. To our knowledge, this is the first time such reconstruction was ever achieved. This robust assessment of in vivo mAV leaflet kinematics in 3D opens up the possibility for longitudinal characterization of murine models that develop aortic valve disease.
Collapse
Affiliation(s)
- Daniel P Gramling
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | | | - Frederick W Damen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Kaitlyn Thatcher
- Department of Pediatrics, Medical College of Wisconsin, Herma Heart Institute, Children's Wisconsin Milwaukee, Milwaukee, WI, USA
| | - Felix Liu
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, USA
| | - David McComb
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, USA
| | - Joy Lincoln
- Department of Pediatrics, Medical College of Wisconsin, Herma Heart Institute, Children's Wisconsin Milwaukee, Milwaukee, WI, USA
| | - Christopher K Breuer
- Tissue Engineering and Surgical Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Michael S Sacks
- Department of Biomedical Engineering, James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
3
|
Rana A, Singh TU, Sharma M, Gari M, Kumar T, Parida S, Lingaraju MC, Kumar Mariappan A, Kumar A, Kumar D. Pravastatin attenuates isoprenaline induced cardiac fibrosis in a mouse model. Biotech Histochem 2023; 98:567-577. [PMID: 37814775 DOI: 10.1080/10520295.2023.2260303] [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] [Indexed: 10/11/2023] Open
Abstract
We investigated the effects of pravastatin (PRAVA) on isoprenaline (ISP) induced cardiac fibrosis using four groups of mice: untreated control, PRAVA, ISP, ISP + PRAVA groups. ISP, 20 mg/kg, was administered subcutaneously daily for 14 days. PRAVA, 20 mg/kg, was administered orally daily for 14 days. Mice were sacrificed on day15 and heart and blood samples were collected to investigate cardiac injury markers. The mean body weight for the ISP group on day 15 was decreased significantly compared to day 0; PRAVA increased the mean body weight slightly on day 15 of treatment compared to day 0. The heart:body weight ratio was increased in the ISP group compared to the control group, but the ratio was returned to near control ratio in the PRAVA + ISP group. The serum creatine kinase-myocardial band (CK-MB) level was reduced significantly in the PRAVA + ISP group compared to the ISP group. Serum triglyceride level was decreased significantly in ISP + PRAVA group compared to the ISP group. PRAVA administration significantly reduced tissue collagen I and III levels in the ISP + PRAVA group compared to the ISP group. Lipid oxidation was decreased and reduced glutathione activity was increased in the PRAVA + ISP group compared to the ISP group. IL-6, α-SMA, CTGF, TGF-β and SMAD-3 gene expressions were decreased in the PRAVA + ISP group compared to the ISP group. We found fewer inflammatory cells and less fibrosis in heart tissue in the PRAVA + ISP group compared to the ISP group. PRAVA decreased ISP induced cardiac fibrosis by reducing oxidative stress, collagen deposition and inflammation, as well as by decreasing expression of TGF-β, SMAD-3 and CTGF genes.
Collapse
Affiliation(s)
- Abhinav Rana
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Thakur Uttam Singh
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Meemansha Sharma
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Manju Gari
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Tarun Kumar
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Subhashree Parida
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | | | - Asok Kumar Mariappan
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Akhilesh Kumar
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Dinesh Kumar
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| |
Collapse
|
4
|
Lu W, Sun C, Hou J. Predicting key gene related to immune infiltration and myofibroblast-like valve interstitial cells in patients with calcified aortic valve disease based on bioinformatics analysis. J Thorac Dis 2023; 15:3726-3740. [PMID: 37559614 PMCID: PMC10407485 DOI: 10.21037/jtd-23-72] [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: 01/14/2023] [Accepted: 06/09/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND Calcified aortic valve disease (CAVD) is the most prevalent valvular disease that can be treated only through valve replacement. We aimed to explore potential biomarkers and the role of immune cell infiltration in CAVD progression through bioinformatics analysis. METHODS Differentially ex-pressed genes (DEGs) were screened out based on three microarray datasets: GSE12644, GSE51472 and GSE83453. Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed to evaluate gene expression differences. Machine learning algorithms and DEGs were used to screen key gene. We used CIBERSORT to evaluate the immune cell infiltration of CAVD and evaluated the correlation between the biomarkers and infiltrating immune cells. We also compared bioinformatics analysis results with the valve interstitial cells (VICs) gene expression in single-cell RNA sequencing. RESULTS Collagen triple helix repeat containing 1 (CTHRC1) was identified as the key gene of CAVD. We identified a cell subtype valve interstitial cells-fibroblast, which was closely associated with fibro-calcific progress of aortic valve. CTHRC1 highly expressed in the VIC subpopulation. Immune infiltration analysis demonstrated that mast cells, B cells, dendritic cells and eosinophils were involved in pathogenesis of CAVD. Correlation analysis demonstrated that CTHRC1 was correlated with mast cells mostly. CONCLUSIONS In summary, the study suggested that CTHRC1 was a key gene of CAVD and CTHRC1 might participate in the potential molecular pathways involved in the connection between infiltrating immune cells and myofibroblast phenotype VICs.
Collapse
Affiliation(s)
- Wenyuan Lu
- Cardiac Surgery Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng Sun
- Cardiac Surgery Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianfeng Hou
- Cardiac Surgery Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Böttner J, Werner S, Feistner L, Fischer-Schaepmann T, Neussl K, Borger MA, Thiele H, Büttner P, Schlotter F. High resolution monitoring of valvular interstitial cell driven pathomechanisms in procalcific environment using label-free impedance spectroscopy. Front Cardiovasc Med 2023; 10:1155371. [PMID: 37408660 PMCID: PMC10319251 DOI: 10.3389/fcvm.2023.1155371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/30/2023] [Indexed: 07/07/2023] Open
Abstract
Introduction Fibro-calcific aortic valve disease has high prevalence and is associated with significant mortality. Fibrotic extracellular matrix (ECM) remodeling and calcific mineral deposition change the valvular microarchitecture and deteriorate valvular function. Valvular interstitial cells (VICs) in profibrotic or procalcifying environment are frequently used in vitro models. However, remodeling processes take several days to weeks to develop, even in vitro. Continuous monitoring by real-time impedance spectroscopy (EIS) may reveal new insights into this process. Methods VIC-driven ECM remodeling stimulated by procalcifying (PM) or profibrotic medium (FM) was monitored by label-free EIS. Collagen secretion, matrix mineralization, viability, mitochondrial damage, myofibroblastic gene expression and cytoskeletal alterations were analyzed. Results and Discussion EIS profiles of VICs in control medium (CM) and FM were comparable. PM reproducibly induced a specific, biphasic EIS profile. Phase 1 showed an initial impedance drop, which moderately correlated with decreasing collagen secretion (r = 0.67, p = 0.22), accompanied by mitochondrial membrane hyperpolarization and cell death. Phase 2 EIS signal increase was positively correlated with augmented ECM mineralization (r = 0.97, p = 0.008). VICs in PM decreased myofibroblastic gene expression (p < 0.001) and stress fiber assembly compared to CM. EIS revealed sex-specific differences. Male VICs showed higher proliferation and in PM EIS decrease in phase 1 was significantly pronounced compared to female VICs (male minimum: 7.4 ± 4.2%, female minimum: 26.5 ± 4.4%, p < 0.01). VICs in PM reproduced disease characteristics in vitro remarkably fast with significant impact of donor sex. PM suppressed myofibroblastogenesis and favored ECM mineralization. In summary, EIS represents an efficient, easy-to-use, high-content screening tool enabling patient-specific, subgroup- and temporal resolution.
Collapse
Affiliation(s)
- Julia Böttner
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Sarah Werner
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Lukas Feistner
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | | | - Katherina Neussl
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Michael A. Borger
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Holger Thiele
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Petra Büttner
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Florian Schlotter
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| |
Collapse
|
6
|
Lei Y, Bortolin L, Benesch-Lee F, Oguntolu T, Dong Z, Bondah N, Billiar K. Hyaluronic acid regulates heart valve interstitial cell contraction in fibrin-based scaffolds. Acta Biomater 2021; 136:124-136. [PMID: 34592445 DOI: 10.1016/j.actbio.2021.09.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 11/30/2022]
Abstract
Heart valve disease is associated with high morbidity and mortality worldwide resulting in hundreds of thousands of heart valve replacements each year. Tissue engineered heart valves (TEHVs) have the potential to overcome the major limitations of traditional replacement valves; however, leaflet retraction has led to the failure of TEHVs in preclinical studies. As native unmodified hyaluronic acid (HA) is known to promote healthy tissue development in native heart valves, we hypothesize that adding unmodified HA to fibrin-based scaffolds common to tissue engineering will reduce retraction by increasing cell-scaffold interactions and density of the scaffolds. Using a custom high-throughput culture system, we found that incorporating HA into millimeter-scale fibrin-based cell-populated scaffolds increases initial fiber diameter and cell-scaffold interactions, causing a cascade of mechanical, morphological, and cellular responses. These changes lead to higher levels of scaffold compaction and stiffness, increased cell alignment, and less bundling of fibrin fibers by the cells during culture. These effects significantly reduce scaffold retraction and total contractile force each by around 25%. These findings increase our understanding of how HA alters tissue remodeling and could inform the design of the next generation of tissue engineered heart valves to help reduce retraction. STATEMENT OF SIGNIFICANCE: Tissue engineered heart valves (TEHVs) have the potential to overcome the major limitations of traditional replacement valves; however, leaflet retraction induced by excessive myofibroblast activation has led to failure in preclinical studies. Developing valves are rich in hyaluronic acid (HA), which helps maintain a physiological environment for tissue remodeling without retraction. We hypothesized that adding unmodified HA to TEHVs would reduce retraction by increasing cell-scaffold interactions and density of the scaffolds. Using a high-throughput tissue culture platform, we demonstrate that HA incorporation into a fibrin-based scaffold can significantly reduce tissue retraction and total contractile force by increasing fiber bundling and altering cell-mediated matrix remodeling, therefore increasing gel density and stiffness. These finding increase our knowledge of native HA's effects within the extracellular matrix, and provide a new tool for TEHV design.
Collapse
Affiliation(s)
- Ying Lei
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Luciano Bortolin
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Frank Benesch-Lee
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Teniola Oguntolu
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Zhijie Dong
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Narda Bondah
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Kristen Billiar
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA.
| |
Collapse
|
7
|
Chester AH, Sarathchandra P, McCormack A, Yacoub MH. Organ Culture Model of Aortic Valve Calcification. Front Cardiovasc Med 2021; 8:734692. [PMID: 34660737 PMCID: PMC8517236 DOI: 10.3389/fcvm.2021.734692] [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: 07/01/2021] [Accepted: 09/02/2021] [Indexed: 01/10/2023] Open
Abstract
A significant amount of knowledge has been gained with the use of cell-based assays to elucidate the mechanisms that mediate heart valve calcification. However, cells used in these studies lack their association with the extra-cellular matrix or the influence of other cellular components of valve leaflets. We have developed a model of calcification using intact porcine valve leaflets, that relies upon a biological stimulus to drive the formation of calcified nodules within the valve leaflets. Alizarin Red positive regions were formed in response to lipopolysaccharide and inorganic phosphate, which could be quantified when viewed under polarized light. Point analysis and elemental mapping analysis of electron microscope images confirmed the presence of nodules containing calcium and phosphorus. Immunohistochemical staining showed that the development of these calcified regions corresponded with the expression of RUNX2, osteocalcin, NF-kB and the apoptosis marker caspase 3. The formation of calcified nodules and the expression of bone markers were both inhibited by adenosine in a concentration-dependent manner, illustrating that the model is amenable to pharmacological manipulation. This organ culture model offers an increased level of tissue complexity in which to study the mechanisms that are involved in heart valve calcification.
Collapse
Affiliation(s)
- Adrian H Chester
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom.,National Heart & Lung Institute, Imperial College, Imperial College London, London, United Kingdom
| | - Padmini Sarathchandra
- National Heart & Lung Institute, Imperial College, Imperial College London, London, United Kingdom
| | - Ann McCormack
- National Heart & Lung Institute, Imperial College, Imperial College London, London, United Kingdom
| | - Magdi H Yacoub
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom.,National Heart & Lung Institute, Imperial College, Imperial College London, London, United Kingdom
| |
Collapse
|
8
|
Gonzalez Rodriguez A, Schroeder ME, Grim JC, Walker CJ, Speckl KF, Weiss RM, Anseth KS. Tumor necrosis factor-α promotes and exacerbates calcification in heart valve myofibroblast populations. FASEB J 2021; 35:e21382. [PMID: 33554387 DOI: 10.1096/fj.202002013rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/16/2022]
Abstract
Pro-inflammatory cytokines play critical roles in regulating valvular interstitial cell (VIC) phenotypic changes that can cause heart valve fibrosis and calcification. Tumor necrosis factor alpha (TNF-α) is a cytokine known to influence VIC behavior and has been reported at high levels in calcified valves ex vivo. We sought to understand the specific effects of TNF-α on VIC phenotypes (eg, fibroblast, profibrotic activated myofibroblasts) and its link with heart valve disorders. We characterize human aortic valve tissue from patients with valve disorders and identify a high variability of fibrotic and calcific markers between tissues. These results motivated in vitro studies to explore the effects of TNF-α on defined VIC fibroblasts and profibrotic activated myofibroblasts, induced via FGF-2 and TGF-β1 treatment. Using 3D hydrogels to culture VICs, we measure the effect of TNF-α (0.1-10 ng/mL) on key markers of fibrosis (eg, αSMA, COL1A1) and calcification (eg, RUNX2, BMP2, and calcium deposits). We observe calcification in TNF-α-treated VIC activated myofibroblasts and identify the MAPK/ERK signaling cascade as a potential pathway for TNF-α mediated calcification. Conversely, VIC fibroblasts respond to TNF-α with decreased calcification. Treatment of VIC profibrotic activated myofibroblast populations with TNF-α leads to increased calcification. Our in vitro findings correlate with findings in diseased human valves and highlight the importance of understanding the effect of cytokines and signaling pathways on specific VIC phenotypes. Finally, we reveal MAPK/ERK as a potential pathway involved in VIC-mediated matrix calcification with TNF-α treatment, suggesting this pathway as a potential pharmaceutical target for aortic valve disease.
Collapse
Affiliation(s)
- Andrea Gonzalez Rodriguez
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA.,BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Megan E Schroeder
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.,Materials Science and Engineering Program, University of Colorado Boulder, Boulder, CO, USA
| | - Joseph C Grim
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA.,BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Cierra J Walker
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.,Materials Science and Engineering Program, University of Colorado Boulder, Boulder, CO, USA
| | - Kelly F Speckl
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Robert M Weiss
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA.,BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.,Materials Science and Engineering Program, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
9
|
Büttner P, Feistner L, Lurz P, Thiele H, Hutcheson JD, Schlotter F. Dissecting Calcific Aortic Valve Disease-The Role, Etiology, and Drivers of Valvular Fibrosis. Front Cardiovasc Med 2021; 8:660797. [PMID: 34041283 PMCID: PMC8143377 DOI: 10.3389/fcvm.2021.660797] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is a highly prevalent and progressive disorder that ultimately causes gradual narrowing of the left ventricular outflow orifice with ensuing devastating hemodynamic effects on the heart. Calcific mineral accumulation is the hallmark pathology defining this process; however, fibrotic extracellular matrix (ECM) remodeling that leads to extensive deposition of fibrous connective tissue and distortion of the valvular microarchitecture similarly has major biomechanical and functional consequences for heart valve function. Significant advances have been made to unravel the complex mechanisms that govern these active, cell-mediated processes, yet the interplay between fibrosis and calcification and the individual contribution to progressive extracellular matrix stiffening require further clarification. Specifically, we discuss (1) the valvular biomechanics and layered ECM composition, (2) patterns in the cellular contribution, temporal onset, and risk factors for valvular fibrosis, (3) imaging valvular fibrosis, (4) biomechanical implications of valvular fibrosis, and (5) molecular mechanisms promoting fibrotic tissue remodeling and the possibility of reverse remodeling. This review explores our current understanding of the cellular and molecular drivers of fibrogenesis and the pathophysiological role of fibrosis in CAVD.
Collapse
Affiliation(s)
- Petra Büttner
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Lukas Feistner
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Philipp Lurz
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Holger Thiele
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Joshua D. Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL, United States
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Florian Schlotter
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| |
Collapse
|
10
|
Niazy N, Barth M, Selig JI, Feichtner S, Shakiba B, Candan A, Albert A, Preuß K, Lichtenberg A, Akhyari P. Degeneration of Aortic Valves in a Bioreactor System with Pulsatile Flow. Biomedicines 2021; 9:biomedicines9050462. [PMID: 33922670 PMCID: PMC8145810 DOI: 10.3390/biomedicines9050462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023] Open
Abstract
Calcific aortic valve disease is the most common valvular heart disease in industrialized countries. Pulsatile pressure, sheer and bending stress promote initiation and progression of aortic valve degeneration. The aim of this work is to establish an ex vivo model to study the therein involved processes. Ovine aortic roots bearing aortic valve leaflets were cultivated in an elaborated bioreactor system with pulsatile flow, physiological temperature, and controlled pressure and pH values. Standard and pro-degenerative treatment were studied regarding the impact on morphology, calcification, and gene expression. In particular, differentiation, matrix remodeling, and degeneration were also compared to a static cultivation model. Bioreactor cultivation led to shrinking and thickening of the valve leaflets compared to native leaflets while gross morphology and the presence of valvular interstitial cells were preserved. Degenerative conditions induced considerable leaflet calcification. In comparison to static cultivation, collagen gene expression was stable under bioreactor cultivation, whereas expression of hypoxia-related markers was increased. Osteopontin gene expression was differentially altered compared to protein expression, indicating an enhanced protein turnover. The present ex vivo model is an adequate and effective system to analyze aortic valve degeneration under controlled physiological conditions without the need of additional growth factors.
Collapse
Affiliation(s)
- Naima Niazy
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Mareike Barth
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Jessica I. Selig
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Sabine Feichtner
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Babak Shakiba
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Asya Candan
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| | - Alexander Albert
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
- Department of Cardiovascular Surgery, Klinikum Dortmund gGmbH, Beurhausstraße 40, 44137 Dortmund, Germany
| | - Karlheinz Preuß
- Faculty of Biotechnology, Bioprocessing, Modulation and Simulation, University of Applied Sciences Mannheim, Paul-Wittsack-Straße 10, 68163 Mannheim, Germany;
| | - Artur Lichtenberg
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
- Correspondence:
| | - Payam Akhyari
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (N.N.); (M.B.); (J.I.S.); (S.F.); (B.S.); (A.C.); (A.A.); (P.A.)
| |
Collapse
|
11
|
Badria AF, Koutsoukos PG, Mavrilas D. Decellularized tissue-engineered heart valves calcification: what do animal and clinical studies tell us? JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:132. [PMID: 33278023 PMCID: PMC7719105 DOI: 10.1007/s10856-020-06462-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/31/2020] [Indexed: 06/12/2023]
Abstract
Cardiovascular diseases are the first cause of death worldwide. Among different heart malfunctions, heart valve failure due to calcification is still a challenging problem. While drug-dependent treatment for the early stage calcification could slow down its progression, heart valve replacement is inevitable in the late stages. Currently, heart valve replacements involve mainly two types of substitutes: mechanical and biological heart valves. Despite their significant advantages in restoring the cardiac function, both types of valves suffered from serious drawbacks in the long term. On the one hand, the mechanical one showed non-physiological hemodynamics and the need for the chronic anticoagulation therapy. On the other hand, the biological one showed stenosis and/or regurgitation due to calcification. Nowadays, new promising heart valve substitutes have emerged, known as decellularized tissue-engineered heart valves (dTEHV). Decellularized tissues of different types have been widely tested in bioprosthetic and tissue-engineered valves because of their superior biomechanics, biocompatibility, and biomimetic material composition. Such advantages allow successful cell attachment, growth and function leading finally to a living regenerative valvular tissue in vivo. Yet, there are no comprehensive studies that are covering the performance of dTEHV scaffolds in terms of their efficiency for the calcification problem. In this review article, we sought to answer the question of whether decellularized heart valves calcify or not. Also, which factors make them calcify and which ones lower and/or prevent their calcification. In addition, the review discussed the possible mechanisms for dTEHV calcification in comparison to the calcification in the native and bioprosthetic heart valves. For this purpose, we did a retrospective study for all the published work of decellularized heart valves. Only animal and clinical studies were included in this review. Those animal and clinical studies were further subcategorized into 4 categories for each depending on the effect of decellularization on calcification. Due to the complex nature of calcification in heart valves, other in vitro and in silico studies were not included. Finally, we compared the different results and summed up all the solid findings of whether decellularized heart valves calcify or not. Based on our review, the selection of the proper heart valve tissue sources (no immunological provoking residues), decellularization technique (no damaged exposed residues of the decellularized tissues, no remnants of dead cells, no remnants of decellularizing agents) and implantation techniques (avoiding suturing during the surgical implantation) could provide a perfect anticalcification potential even without in vitro cell seeding or additional scaffold treatment.
Collapse
Affiliation(s)
- Adel F Badria
- Department of Fiber and Polymer Technology, Division of Coating Technology, KTH Royal Institute of Technology, Stockholm, Sweden.
- Department of Mechanical Engineering and Aeronautics, Division of Applied Mechanics, Technology of Materials and Biomechanics, University of Patras, Patras, Greece.
| | - Petros G Koutsoukos
- Department of Chemical Engineering, University of Patras, Patras University Campus, 26504, Patras, Greece
| | - Dimosthenis Mavrilas
- Department of Mechanical Engineering and Aeronautics, Division of Applied Mechanics, Technology of Materials and Biomechanics, University of Patras, Patras, Greece
| |
Collapse
|
12
|
Vieceli Dalla Sega F, Fortini F, Cimaglia P, Marracino L, Tonet E, Antonucci A, Moscarelli M, Campo G, Rizzo P, Ferrari R. COX-2 Is Downregulated in Human Stenotic Aortic Valves and Its Inhibition Promotes Dystrophic Calcification. Int J Mol Sci 2020; 21:ijms21238917. [PMID: 33255450 PMCID: PMC7727817 DOI: 10.3390/ijms21238917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/12/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the result of maladaptive fibrocalcific processes leading to a progressive thickening and stiffening of aortic valve (AV) leaflets. CAVD is the most common cause of aortic stenosis (AS). At present, there is no effective pharmacotherapy in reducing CAVD progression; when CAVD becomes symptomatic it can only be treated with valve replacement. Inflammation has a key role in AV pathological remodeling; hence, anti-inflammatory therapy has been proposed as a strategy to prevent CAVD. Cyclooxygenase 2 (COX-2) is a key mediator of the inflammation and it is the target of widely used anti-inflammatory drugs. COX-2-inhibitor celecoxib was initially shown to reduce AV calcification in a murine model. However, in contrast to these findings, a recent retrospective clinical analysis found an association between AS and celecoxib use. In the present study, we investigated whether variations in COX-2 expression levels in human AVs may be linked to CAVD. We extracted total RNA from surgically explanted AVs from patients without CAVD or with CAVD. We found that COX-2 mRNA was higher in non-calcific AVs compared to calcific AVs (0.013 ± 0.002 vs. 0.006 ± 0.0004; p < 0.0001). Moreover, we isolated human aortic valve interstitial cells (AVICs) from AVs and found that COX-2 expression is decreased in AVICs from calcific valves compared to AVICs from non-calcific AVs. Furthermore, we observed that COX-2 inhibition with celecoxib induces AVICs trans-differentiation towards a myofibroblast phenotype, and increases the levels of TGF-β-induced apoptosis, both processes able to promote the formation of calcific nodules. We conclude that reduced COX-2 expression is a characteristic of human AVICs prone to calcification and that COX-2 inhibition may promote aortic valve calcification. Our findings support the notion that celecoxib may facilitate CAVD progression.
Collapse
Affiliation(s)
| | - Francesca Fortini
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.V.D.S.); (F.F.); (P.C.); (M.M.); (R.F.)
| | - Paolo Cimaglia
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.V.D.S.); (F.F.); (P.C.); (M.M.); (R.F.)
| | - Luisa Marracino
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy;
| | - Elisabetta Tonet
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Cona, Italy; (E.T.); (A.A.); (G.C.)
| | - Antonio Antonucci
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Cona, Italy; (E.T.); (A.A.); (G.C.)
| | - Marco Moscarelli
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.V.D.S.); (F.F.); (P.C.); (M.M.); (R.F.)
| | - Gianluca Campo
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Cona, Italy; (E.T.); (A.A.); (G.C.)
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.V.D.S.); (F.F.); (P.C.); (M.M.); (R.F.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy;
- Correspondence: ; Tel.: +39-0532-455-508
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.V.D.S.); (F.F.); (P.C.); (M.M.); (R.F.)
| |
Collapse
|
13
|
Abstract
Familial hypercholesterolemia (FH) is a rare autosomal gene deficiency disease with increased low-density lipoprotein cholesterol, xanthoma, and premature coronary heart disease. Calcified aortic valve disease (CAVD) is prevalent in FH patients, resulting in adverse events and heavy health care burden. Aortic valve calcification is currently considered an active biological process, which shares several common risk factors with atherosclerosis, including aging, hypertension, dyslipidemia, and so on. Unfortunately, the pathogenesis and therapy of CAVD in FH are still controversial. There is no pharmacological intervention recommended to delay the development of CAVD in FH, and the only effective treatment for severe CAVD is aortic valve replacement. In this review, we summarize the detailed description of the pathophysiology, molecular mechanism, risk factors, and treatment of CAVD in FH patients.
Collapse
|
14
|
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
|
15
|
Tremoulet AH, Jain S, Jone PN, Best BM, Duxbury EH, Franco A, Printz B, Dominguez SR, Heizer H, Anderson MS, Glodé MP, He F, Padilla RL, Shimizu C, Bainto E, Pancheri J, Cohen HJ, Whitin JC, Burns JC. Phase I/IIa Trial of Atorvastatin in Patients with Acute Kawasaki Disease with Coronary Artery Aneurysm. J Pediatr 2019; 215:107-117.e12. [PMID: 31561960 PMCID: PMC6878161 DOI: 10.1016/j.jpeds.2019.07.064] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/17/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To determine the safety, tolerability, pharmacokinetics, and immunomodulatory effects of a 6-week course of atorvastatin in patients with acute Kawasaki disease with coronary artery (CA) aneurysm (CAA). STUDY DESIGN This was a Phase I/IIa 2-center dose-escalation study of atorvastatin (0.125-0.75 mg/kg/day) in 34 patients with Kawasaki disease (aged 2-17 years) with echocardiographic evidence of CAA. We measured levels of the brain metabolite 24(S)-hydroxycholesterol (24-OHC), serum lipids, acute-phase reactants, liver enzymes, and creatine phosphokinase; peripheral blood mononuclear cell populations; and CA internal diameter normalized for body surface area before atorvastatin treatment and at 2 and 6 weeks after initiation of atorvastatin treatment. RESULTS A 6-week course of up to 0.75 mg/kg/day of atorvastatin was well tolerated by the 34 subjects (median age, 5.3 years; IQR, 2.6-6.4 years), with no serious adverse events attributable to the study drug. The areas under the curve for atorvastatin and its metabolite were larger in the study subjects compared with those reported in adults, suggesting a slower rate of metabolism in children. The 24-OHC levels were similar between the atorvastatin-treated subjects and matched controls. CONCLUSIONS Atorvastatin was safe and well tolerated in our cohort of children with acute Kawasaki disease and CAA. A Phase III efficacy trial is warranted in this patient population, which may benefit from the known anti-inflammatory and immunomodulatory effects of this drug.
Collapse
Affiliation(s)
- Adriana H. Tremoulet
- Kawasaki Disease Research Center, Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Rady Children’s Hospital San Diego, San Diego, California, USA
| | - Sonia Jain
- Biostatistics Research Center, Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California
| | - Pei-Ni Jone
- Pediatric Cardiology, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO
| | - Brookie M. Best
- Kawasaki Disease Research Center, Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Rady Children’s Hospital San Diego, San Diego, California, USA,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA
| | - Elizabeth H. Duxbury
- Kawasaki Disease Research Center, Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Rady Children’s Hospital San Diego, San Diego, California, USA,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA
| | - Alessandra Franco
- Kawasaki Disease Research Center, Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Rady Children’s Hospital San Diego, San Diego, California, USA
| | - Beth Printz
- Kawasaki Disease Research Center, Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Rady Children’s Hospital San Diego, San Diego, California, USA
| | - Samuel R. Dominguez
- Pediatric Infectious Disease, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO
| | - Heather Heizer
- Pediatric Infectious Disease, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO
| | - Marsha S. Anderson
- Pediatric Infectious Disease, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO
| | - Mary P. Glodé
- Pediatric Infectious Disease, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO
| | - Feng He
- Biostatistics Research Center, Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California
| | - Robert L. Padilla
- Kawasaki Disease Research Center, Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Rady Children’s Hospital San Diego, San Diego, California, USA
| | - Chisato Shimizu
- Kawasaki Disease Research Center, Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Rady Children’s Hospital San Diego, San Diego, California, USA
| | - Emelia Bainto
- Kawasaki Disease Research Center, Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Rady Children’s Hospital San Diego, San Diego, California, USA
| | - Joan Pancheri
- Kawasaki Disease Research Center, Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Rady Children’s Hospital San Diego, San Diego, California, USA
| | | | - John C. Whitin
- Department of Pediatrics, Stanford University, Stanford, CA
| | - Jane C. Burns
- Kawasaki Disease Research Center, Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Rady Children’s Hospital San Diego, San Diego, California, USA
| |
Collapse
|
16
|
Wang X, Ali MS, Lacerda CMR. Osteogenesis inducers promote distinct biological responses in aortic and mitral valve interstitial cells. J Cell Biochem 2019; 120:11158-11171. [PMID: 30746757 DOI: 10.1002/jcb.28392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/27/2018] [Accepted: 01/09/2019] [Indexed: 01/24/2023]
Abstract
Both aortic and mitral valves calcify in pathological conditions; however, the prevalence of aortic valve calcification is high whereas mitral valve leaflet calcification is somewhat rare. Patterns of valvular calcification may differ due to valvular architecture, but little is known to that effect. In this study, we investigated the intrinsic osteogenic differentiation potential of aortic versus mitral valve interstitial cells provided minimal differentiation conditions. For the assessment of calcification at the cellular level, we used classic inducers of osteogenesis in stem cells: β-glycerophosphate (β-Gly), dexamethasone (Dex), and ascorbate (Asc). In addition to proteomic analyses, osteogenic markers and calcium precipitates were evaluated across treatments of aortic and mitral valve cells. The combination of β-Gly, Asc, and Dex induced aortic valve interstitial cells to synthesize extracellular matrix, overexpress osteoblastic markers, and deposit calcium. However, no strong evidence showed the calcification of mitral valve interstitial cells. Mitral cells mainly responded to Asc and Dex by cell activation. These findings provide a deeper understanding of the physiological properties of aortic and mitral valves and tendencies for calcific changes within each valve type, contributing to the development of future therapeutics for heart valve diseases.
Collapse
Affiliation(s)
- Xinmei Wang
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas
| | - Mir S Ali
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas
| | - Carla M R Lacerda
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas
| |
Collapse
|
17
|
Go JL, Prem K, Al-Hijji MA, Qin Q, Noble C, Young MD, Lerman LO, Lerman A. Experimental Metabolic Syndrome Model Associated with Mechanical and Structural Degenerative Changes of the Aortic Valve. Sci Rep 2018; 8:17835. [PMID: 30546028 PMCID: PMC6292876 DOI: 10.1038/s41598-018-36388-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/15/2018] [Indexed: 12/26/2022] Open
Abstract
The purpose of this study was to test the hypothesis that an experimental high fat (HF) animal with metabolic syndrome results in structural degeneration of the aortic valve. Domestic pigs were divided (n = 12) and administered either a normal or HF diet. After 16-weeks, the HF diet group had increased weight (p ≤ 0.05), total cholesterol (p ≤ 0.05), and systolic and diastolic pressure (p ≤ 0.05). The aortic valve extracellular matrix showed loss of elastin fibers and increased collagen deposition in the HF diet group. Collagen was quantified with ELISA, which showed an increased concentration of collagen types 1 and 3 (p ≤ 0.05). In the HF diet group, the initial stages of microcalcification were observed. Uniaxial mechanical testing of aortic cusps revealed that the HF diet group expressed a decrease in ultimate tensile strength and elastic modulus compared to the control diet group (p ≤ 0.05). Western blot and immunohistochemistry indicated the presence of proteins: lipoprotein-associated phospholipase A2, osteopontin, and osteocalcin with an increased expression in the HF diet group. The current study demonstrates that experimental metabolic syndrome induced by a 16-week HF diet was associated with a statistically significant alteration to the physical architecture of the aortic valve.
Collapse
Affiliation(s)
- Jason L Go
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, 55905, MN, USA
| | - Komal Prem
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, 55905, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, 55905, MN, USA
| | - Mohammed A Al-Hijji
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, 55905, MN, USA
| | - Qing Qin
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, 55905, MN, USA
| | - Christopher Noble
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, 55905, MN, USA
| | - Melissa D Young
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, 55905, MN, USA
| | - Lilach O Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, 55905, MN, USA
- Department of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, 55905, MN, USA
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, 55905, MN, USA.
| |
Collapse
|
18
|
Menon V, Lincoln J. The Genetic Regulation of Aortic Valve Development and Calcific Disease. Front Cardiovasc Med 2018; 5:162. [PMID: 30460247 PMCID: PMC6232166 DOI: 10.3389/fcvm.2018.00162] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/19/2018] [Indexed: 12/19/2022] Open
Abstract
Heart valves are dynamic, highly organized structures required for unidirectional blood flow through the heart. Over an average lifetime, the valve leaflets or cusps open and close over a billion times, however in over 5 million Americans, leaflet function fails due to biomechanical insufficiency in response to wear-and-tear or pathological stimulus. Calcific aortic valve disease (CAVD) is the most common valve pathology and leads to stiffening of the cusp and narrowing of the aortic orifice leading to stenosis and insufficiency. At the cellular level, CAVD is characterized by valve endothelial cell dysfunction and osteoblast-like differentiation of valve interstitial cells. These processes are associated with dysregulation of several molecular pathways important for valve development including Notch, Sox9, Tgfβ, Bmp, Wnt, as well as additional epigenetic regulators. In this review, we discuss the multifactorial mechanisms that contribute to CAVD pathogenesis and the potential of targeting these for the development of novel, alternative therapeutics beyond surgical intervention.
Collapse
Affiliation(s)
- Vinal Menon
- Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,The Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
| | - Joy Lincoln
- Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,The Heart Center, Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, Ohio State University, Columbus, OH, United States
| |
Collapse
|
19
|
Venardos N, Deng XS, Yao Q, Weyant MJ, Reece TB, Meng X, Fullerton DA. Simvastatin reduces the TLR4-induced inflammatory response in human aortic valve interstitial cells. J Surg Res 2018; 230:101-109. [PMID: 30100024 DOI: 10.1016/j.jss.2018.04.054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 04/18/2018] [Accepted: 04/24/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Calcific aortic stenosis is a chronic inflammatory disease. Proinflammatory stimulation via toll-like receptor 4 (TLR4) causes the aortic valve interstitial cell (AVIC) to undergo phenotypic change. The AVIC first assumes an inflammatory phenotype characterized by the production of inflammatory mediators such as intercellular adhesion molecule-1 (ICAM-1), interleukin-8 (IL-8), and monocyte chemoattractant protein-1 (MCP-1). This change has been linked with an osteogenic phenotypic response. Statins have recently been shown to have anti-inflammatory properties. We therefore hypothesized that statins may have an anti-inflammatory effect on human AVICs by downregulation of TLR4-stimulated inflammatory responses. Our purposes were (1) to determine the effect of simvastatin on TLR4-induced expression of inflammatory mediators in human AVICs and (2) to determine the mechanism(s) through which simvastatin exert this effect. MATERIALS AND METHODS Human AVICs were isolated from the explanted hearts of four patients undergoing cardiac transplantation. Cells were treated with simvastatin (50 μM) for 1 h before stimulation with TLR4 agonist lipopolysaccharide (LPS, 0.2 μg/mL). Immunoblotting (IB) was used to analyze cell lysates for ICAM-1 expression, and enzyme-linked immunosorbent assay was used to detect IL-8 and MCP-1 in cell culture media. Likewise, lysates were analyzed for TLR4 and nuclear factor-kappa B activation (IB). After simvastatin treatment, lysates were analyzed for TLR4 levels (IB). Statistics were by analysis of variance (P < 0.05). RESULTS Simvastatin reduced TLR4-induced ICAM-1, IL-8, and MCP-1 expression in AVICs. Simvastatin down-regulated TLR4 levels and suppressed TLR4-induced phosphorylation of nuclear factor-kappa B. CONCLUSIONS These data demonstrate the potential of a medical therapy (simvastatin) to impact the pathogenesis of aortic stenosis.
Collapse
Affiliation(s)
- Neil Venardos
- The Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado.
| | - Xin-Sheng Deng
- The Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Quinzhou Yao
- The Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Michael J Weyant
- The Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - T Brett Reece
- The Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Xianzhong Meng
- The Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - David A Fullerton
- The Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
20
|
Rutkovskiy A, Malashicheva A, Sullivan G, Bogdanova M, Kostareva A, Stensløkken KO, Fiane A, Vaage J. Valve Interstitial Cells: The Key to Understanding the Pathophysiology of Heart Valve Calcification. J Am Heart Assoc 2017; 6:e006339. [PMID: 28912209 PMCID: PMC5634284 DOI: 10.1161/jaha.117.006339] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Arkady Rutkovskiy
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Norway
- Centre for Heart Failure Research, University of Oslo, Norway
- Department of Emergency Medicine and Intensive Care, Oslo University Hospital, Oslo, Norway
- Division of Medicine, Akershus University Hospital, Lørenskog, Norway
- ITMO University, St. Petersburg, Russia
| | - Anna Malashicheva
- Almazov National Medical Research Centre, St. Petersburg, Russia
- ITMO University, St. Petersburg, Russia
| | - Gareth Sullivan
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Norway
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Norway
- Institute of Immunology, Oslo University Hospital, Oslo, Norway
- Norwegian Center for Stem Cell Research, Oslo, Norway
| | - Maria Bogdanova
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Anna Kostareva
- Almazov National Medical Research Centre, St. Petersburg, Russia
- ITMO University, St. Petersburg, Russia
| | - Kåre-Olav Stensløkken
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Norway
- Centre for Heart Failure Research, University of Oslo, Norway
| | - Arnt Fiane
- Institute of Clinical Medicine, University of Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Jarle Vaage
- Institute of Clinical Medicine, University of Oslo, Norway
- Department of Emergency Medicine and Intensive Care, Oslo University Hospital, Oslo, Norway
- ITMO University, St. Petersburg, Russia
| |
Collapse
|
21
|
Li F, Song R, Ao L, Reece TB, Cleveland JC, Dong N, Fullerton DA, Meng X. ADAMTS5 Deficiency in Calcified Aortic Valves Is Associated With Elevated Pro-Osteogenic Activity in Valvular Interstitial Cells. Arterioscler Thromb Vasc Biol 2017; 37:1339-1351. [PMID: 28546218 DOI: 10.1161/atvbaha.117.309021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 05/09/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Extracellular matrix proteinases are implicated in the pathogenesis of calcific aortic valve disease. The ADAMTS5 (a disintegrin and metalloproteinase with thrombospondin motifs 5) enzyme is secreted, matrix-associated metalloendopeptidase, capable of degrading extracellular matrix proteins, particularly matrilin 2. We sought to determine the role of the ADAMTS5/matrilin 2 axis in mediating the phenotype transition of valvular interstitial cells (VICs) associated with calcific aortic valve disease. APPROACH AND RESULTS Levels of ADAMTS5, matrilin 2, and α-SMA (α-smooth muscle actin) were evaluated in calcified and normal human aortic valve tissues and VICs. Calcified aortic valves have reduced levels of ADAMTS5 and higher levels of matrilin 2 and α-SMA. Treatment of normal VICs with soluble matrilin 2 caused an increase in α-SMA level through Toll-like receptors 2 and 4, which was accompanied by upregulation of runt-related transcription factor 2 and alkaline phosphatase. In addition, ADAMTS5 knockdown in normal VICs enhanced the effect of matrilin 2. Matrilin 2 activated nuclear factor (NF) κB and NF of activated T cells complex 1 and induced the interaction of these 2 NFs. Inhibition of either NF-κB or NF of activated T cells complex 1 suppressed matrilin 2's effect on VIC phenotype change. Knockdown of α-SMA reduced and overexpression of α-SMA enhanced the expression of pro-osteogenic factors and calcium deposit formation in human VICs. CONCLUSIONS Matrilin 2 induces myofibroblastic transition and elevates pro-osteogenic activity in human VICs via activation of NF-κB and NF of activated T cells complex 1. Myofibroblastic transition in human VICs is an important mechanism of elevating the pro-osteogenic activity. Matrilin 2 accumulation associated with relative ADAMTS5 deficiency may contribute to the mechanism underlying calcific aortic valve disease progression.
Collapse
Affiliation(s)
- Fei Li
- From the Department of Surgery, University of Colorado Denver, Aurora (F.L., R.S., L.A., T.B.R., J.C.C., D.A.F., X.M.); and Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (F.L., N.D.)
| | - Rui Song
- From the Department of Surgery, University of Colorado Denver, Aurora (F.L., R.S., L.A., T.B.R., J.C.C., D.A.F., X.M.); and Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (F.L., N.D.)
| | - Lihua Ao
- From the Department of Surgery, University of Colorado Denver, Aurora (F.L., R.S., L.A., T.B.R., J.C.C., D.A.F., X.M.); and Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (F.L., N.D.)
| | - T Brett Reece
- From the Department of Surgery, University of Colorado Denver, Aurora (F.L., R.S., L.A., T.B.R., J.C.C., D.A.F., X.M.); and Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (F.L., N.D.)
| | - Joseph C Cleveland
- From the Department of Surgery, University of Colorado Denver, Aurora (F.L., R.S., L.A., T.B.R., J.C.C., D.A.F., X.M.); and Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (F.L., N.D.)
| | - Nianguo Dong
- From the Department of Surgery, University of Colorado Denver, Aurora (F.L., R.S., L.A., T.B.R., J.C.C., D.A.F., X.M.); and Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (F.L., N.D.)
| | - David A Fullerton
- From the Department of Surgery, University of Colorado Denver, Aurora (F.L., R.S., L.A., T.B.R., J.C.C., D.A.F., X.M.); and Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (F.L., N.D.)
| | - Xianzhong Meng
- From the Department of Surgery, University of Colorado Denver, Aurora (F.L., R.S., L.A., T.B.R., J.C.C., D.A.F., X.M.); and Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (F.L., N.D.).
| |
Collapse
|
22
|
Aortic calcified particles modulate valvular endothelial and interstitial cells. Cardiovasc Pathol 2017; 28:36-45. [DOI: 10.1016/j.carpath.2017.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 02/15/2017] [Accepted: 02/22/2017] [Indexed: 12/11/2022] Open
|
23
|
Cirka HA, Uribe J, Liang V, Schoen FJ, Billiar KL. Reproducible in vitro model for dystrophic calcification of cardiac valvular interstitial cells: insights into the mechanisms of calcific aortic valvular disease. LAB ON A CHIP 2017; 17:814-829. [PMID: 28128382 DOI: 10.1039/c6lc01226d] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Calcific aortic valvular disease (CAVD) is the most prevalent valvular pathology in the United States. Development of a pharmacologic agent to slow, halt, or reverse calcification has proven to be unsuccessful as still much remains unknown about the mechanisms of disease initiation. Although in vitro models of some features of CAVD exist, their utility is limited by the inconsistency of the size and time course of the calcified cell aggregates. In this study, we introduce and verify a highly reproducible in vitro method for studying dystrophic calcification of cardiac valvular interstitial cells, considered to be a key mechanism of clinical CAVD. By utilizing micro-contact printing, we were able to consistently reproduce cell aggregation, myofibroblastic markers, programmed cell death, and calcium accumulation within aggregates of 50-400 μm in diameter on substrates with moduli from 9.6 to 76.8 kPa. This method is highly repeatable, with 70% of aggregates staining positive for Alizarin Red S after one week in culture. Dense mineralized calcium-positive nanoparticles were found within the valvular interstitial cell aggregates as shown by scanning electron microscopy (SEM) and energy dispersive spectrometry (EDS). The area of micro-contact printed aggregates staining positive for caspase 3/7 activity increased from 5.9 ± 0.9% to 12.6 ± 4.5% over one week in culture. Z-VAD-FMK reduced aggregates staining positive for Alizarin Red S by 60%. The state of cell stress is hypothesized to play a role in the disease progression; traction force microscopy indicates high substrate stresses along the aggregate periphery which can be modulated by altering the size of the aggregates and the modulus of the substrate. Micro-contact printing is advantageous over the currently used in vitro model as it allows the independent study of how cytokines, substrate modulus, and pharmacologic agents affect calcification. This controlled method for aggregate creation has the potential to be used as an in vitro assay for the screening of promising therapeutics to mitigate CAVD.
Collapse
Affiliation(s)
- Heather A Cirka
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA.
| | - Johana Uribe
- Department of Bioengineering, University of Massachusetts at Dartmouth, Dartmouth, MA 02714, USA
| | - Vivian Liang
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA.
| | - Frederick J Schoen
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kristen L Billiar
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA.
| |
Collapse
|
24
|
Ayoub S, Ferrari G, Gorman RC, Gorman JH, Schoen FJ, Sacks MS. Heart Valve Biomechanics and Underlying Mechanobiology. Compr Physiol 2016; 6:1743-1780. [PMID: 27783858 PMCID: PMC5537387 DOI: 10.1002/cphy.c150048] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Heart valves control unidirectional blood flow within the heart during the cardiac cycle. They have a remarkable ability to withstand the demanding mechanical environment of the heart, achieving lifetime durability by processes involving the ongoing remodeling of the extracellular matrix. The focus of this review is on heart valve functional physiology, with insights into the link between disease-induced alterations in valve geometry, tissue stress, and the subsequent cell mechanobiological responses and tissue remodeling. We begin with an overview of the fundamentals of heart valve physiology and the characteristics and functions of valve interstitial cells (VICs). We then provide an overview of current experimental and computational approaches that connect VIC mechanobiological response to organ- and tissue-level deformations and improve our understanding of the underlying functional physiology of heart valves. We conclude with a summary of future trends and offer an outlook for the future of heart valve mechanobiology, specifically, multiscale modeling approaches, and the potential directions and possible challenges of research development. © 2016 American Physiological Society. Compr Physiol 6:1743-1780, 2016.
Collapse
Affiliation(s)
- Salma Ayoub
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences, Department of Biomedical Engineering, The University of Texas at Austin, Austin, USA
| | - Giovanni Ferrari
- Gorman Cardiovascular Research Group, University of Pennsylvania, Philadelphia, USA
| | - Robert C. Gorman
- Gorman Cardiovascular Research Group, University of Pennsylvania, Philadelphia, USA
| | - Joseph H. Gorman
- Gorman Cardiovascular Research Group, University of Pennsylvania, Philadelphia, USA
| | - Frederick J. Schoen
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Michael S. Sacks
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences, Department of Biomedical Engineering, The University of Texas at Austin, Austin, USA
| |
Collapse
|
25
|
Wang B, Wei G, Liu B, Zhou X, Xiao H, Dong N, Li F. The Role of High Mobility Group Box 1 Protein in Interleukin-18-Induced Myofibroblastic Transition of Valvular Interstitial Cells. Cardiology 2016; 135:168-178. [PMID: 27395056 DOI: 10.1159/000447483] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/07/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Increased levels of interleukin-18 (IL-18) and high mobility group box 1 protein (HMGB1) have been reported in patients with calcific aortic valve disease (CAVD). However, the role of IL-18 and HMGB1 in the modulation of the valvular interstitial cell (VIC) phenotype remains unclear. We hypothesized that HMGB1 mediates IL-18-induced myofibroblastic transition of VICs. METHODS The expression of IL-18, HMGB1 and α-smooth muscle actin (α-SMA) in human aortic valves was evaluated by immunohistochemical staining, real-time polymerase chain reaction and immunoblotting. Plasma concentrations of IL-18 and HMGB1 were measured using the ELISA kit. Cultured human aortic VICs were used as an in vitro model. RESULTS Immunohistochemistry and immunoblotting revealed increased levels of IL-18, HMGB1 and α-SMA in calcific valves. Circulating IL-18 and HMGB1 levels were also higher in CAVD patients. In vitro, IL-18 induced upregulation of HMGB1 and α-SMA in VICs. Moreover, IL-18 induced secretion of HMGB1 to the extracellular space and activation of nuclear factor kappa-B (NF-κB). Blockade of NF-κB abrogated the upregulation and release of HMGB1 induced by IL-18. Whereas HMGB1 inhibition attenuated the IL-18-induced expression of α-SMA, HMGB1 enhanced the effect of IL-18. CONCLUSIONS We demonstrated for the first time that both tissue and plasma levels of IL-18 and HMGB1 were increased in patients with CAVD. Mechanically, HMGB1 mediated IL-18-induced VIC myofibroblastic transition.
Collapse
Affiliation(s)
- Bo Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Zhou J, Zhu J, Jiang L, Zhang B, Zhu D, Wu Y. Interleukin 18 promotes myofibroblast activation of valvular interstitial cells. Int J Cardiol 2016; 221:998-1003. [PMID: 27441481 DOI: 10.1016/j.ijcard.2016.07.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/04/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND Calcific aortic valve disease is the main heart valve disease in the elderly. Valvular interstitial cells (VICs) play an important role in the process of valve calcification. Interleukin 18 (IL-18) is expressed in stenosis aortic valves and is positively related with the clinical severity of aortic stenosis. However, the role of IL-18 in aortic valve calcification remains unclear. This study examined whether IL-18 promotes myofibroblast and/or osteoblast transdifferention of VICs. Porcine VICs were isolated and treated with recombinant porcine IL-18. METHODS Porcine VICs were cultured and treated with IL-18. Gene and protein expression of myofibroblastic and osteoblastic markers were tested and nuclear factor κB (NF-κB) phosphorylation was also analyzed. Alkaline phosphatase (ALP) staining and activity assay were also performed. RESULTS Our experiments demonstrated that IL-18 significantly enhanced alpha-smooth muscle actin (α-SMA) gene and protein expression. IL-18 treatment also promoted the expression of osteopontin (OPN) and runt-related transcription factor 2 (Runx2) mRNA, although OPN and Runx2 protein expressions were not changed. IL-18 could induce ALP activity in the presence of conditioning medium. We also demonstrated that IL-18 markedly enhanced NF-κB p65 phosphorylation in VICs. CONCLUSIONS Together these results suggest that IL-18 promotes the myofibroblast differentiation of VICs and accelerates calcification in the presence of conditioning medium via the NF-κB pathway.
Collapse
Affiliation(s)
- Jingxin Zhou
- Department of Cardiovascular Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jinfu Zhu
- Department of Cardiovascular Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li Jiang
- Department of Cardiology, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Busheng Zhang
- Department of Cardiovascular Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Zhu
- Department of Cardiovascular Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Yanhu Wu
- Department of Cardiovascular Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
27
|
Madhu MN, Aguiar C, Hassan A, Brunt KR. Translating calcified aortic valve disease to the bench - Use of 3D matrices in the development of future treatment strategies. J Mol Cell Cardiol 2016; 98:58-61. [PMID: 27338001 DOI: 10.1016/j.yjmcc.2016.06.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 06/18/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Malav N Madhu
- Department of Pharmacology, Dalhousie Medicine New Brunswick, Canada; Faculty of Medicine, Dalhousie University, Canada
| | - Christie Aguiar
- Department of Cardiac Surgery, Saint John Regional Hospital, Canada
| | - Ansar Hassan
- Department of Cardiac Surgery, Saint John Regional Hospital, Canada; Faculty of Medicine, Dalhousie University, Canada
| | - Keith R Brunt
- Department of Pharmacology, Dalhousie Medicine New Brunswick, Canada; Faculty of Medicine, Dalhousie University, Canada.
| |
Collapse
|
28
|
Rush MN, Coombs KE, Hedberg-Dirk EL. Surface chemistry regulates valvular interstitial cell differentiation in vitro. Acta Biomater 2015; 28:76-85. [PMID: 26428193 DOI: 10.1016/j.actbio.2015.09.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/28/2015] [Accepted: 09/26/2015] [Indexed: 11/27/2022]
Abstract
The primary driver for valvular calcification is the differentiation of valvular interstitial cells (VICs) into a diseased phenotype. However, the factors leading to the onset of osteoblastic-like VICs (obVICs) and resulting calcification are not fully understood. This study isolates the effect of substrate surface chemistry on in vitro VIC differentiation and calcified tissue formation. Using ω-functionalized alkanethiol self-assembled monolayers (SAMs) on gold [CH3 (hydrophobic), OH (hydrophilic), COOH (COO(-), negative at physiological pH), and NH2 (NH3(+), positive at physiological pH)], we have demonstrated that surface chemistry modulates VIC phenotype and calcified tissue deposition independent of osteoblastic-inducing media additives. Over seven days VICs exhibited surface-dependent differences in cell proliferation (COO(-)=NH3(+)>OH>CH3), morphology, and osteoblastic potential. Both NH3(+)and CH3-terminated SAMs promoted calcified tissue formation while COO(-)-terminated SAMs showed no calcification. VICs on NH3(+)-SAMs exhibited the most osteoblastic phenotypic markers through robust nodule formation, up-regulated osteocalcin and α-smooth muscle actin expression, and adoption of a round/rhomboid morphology indicative of osteoblastic differentiation. With the slowest proliferation, VICs on CH3-SAMs promoted calcified aggregate formation through cell detachment and increased cell death indicative of dystrophic calcification. Furthermore, induction of calcified tissue deposition on NH3(+) and CH3-SAMs was distinctly different than that of media induced osteoblastic VICs. These results demonstrate that substrate surface chemistry alters VIC behavior and plays an important role in calcified tissue formation. In addition, we have identified two novel methods of calcified VIC induction in vitro. Further study of these environments may yield new models for in vitro testing of therapeutics for calcified valve stenosis, although additional studies need to be conducted to correlate results to in vivo models. STATEMENT OF SIGNIFICANCE Valvular interstitial cell (VIC) differentiation and aortic valve calcification is associated with increased risk of mortality and onset of other cardiovascular disorders. This research examines effects of in vitro substrate surface chemistry on VIC differentiation and has led to the identification of two materials-based initiation mechanisms of osteoblastic-like calcified tissue formation independent of soluble signaling methods. Such findings are important for their potential to study signaling cascades responsible for valvular heart disease initiation and progression as well providing in vitro disease models for drug development. We have also identified a VIC activating in vitro environment that does not exhibit confluence induced nodule formation with promise for the development of tissue regenerating scaffolds.
Collapse
|
29
|
Hutcheson JD, Goettsch C, Rogers MA, Aikawa E. Revisiting cardiovascular calcification: A multifaceted disease requiring a multidisciplinary approach. Semin Cell Dev Biol 2015; 46:68-77. [PMID: 26358815 DOI: 10.1016/j.semcdb.2015.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 12/24/2022]
Abstract
The presence of cardiovascular calcification significantly predicts patients' morbidity and mortality. Calcific mineral deposition within the soft cardiovascular tissues disrupts the normal biomechanical function of these tissues, leading to complications such as heart failure, myocardial infarction, and stroke. The realization that calcification results from active cellular processes offers hope that therapeutic intervention may prevent or reverse the disease. To this point, however, no clinically viable therapies have emerged. This may be due to the lack of certainty that remains in the mechanisms by which mineral is deposited in cardiovascular tissues. Gaining new insight into this process requires a multidisciplinary approach. The pathological changes in cell phenotype that lead to the physicochemical deposition of mineral and the resultant effects on tissue biomechanics must all be considered when designing strategies to treat cardiovascular calcification. In this review, we overview the current cardiovascular calcification paradigm and discuss emerging techniques that are providing new insight into the mechanisms of ectopic calcification.
Collapse
Affiliation(s)
- Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences and Center for Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| | - Claudia Goettsch
- Center for Interdisciplinary Cardiovascular Sciences and Center for Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Maximillian A Rogers
- Center for Interdisciplinary Cardiovascular Sciences and Center for Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences and Center for Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
30
|
Wong FF, Ho ML, Yamagami M, Lam MT, Grande-Allen KJ, Suh J. Effective Gene Delivery to Valvular Interstitial Cells Using Adeno-Associated Virus Serotypes 2 and 3. Tissue Eng Part C Methods 2015; 21:808-15. [DOI: 10.1089/ten.tec.2014.0493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Fergus F. Wong
- Department of Bioengineering, Rice University, Houston, Texas
| | - Michelle L. Ho
- Department of Bioengineering, Rice University, Houston, Texas
| | - Momona Yamagami
- Department of Bioengineering, Rice University, Houston, Texas
| | - Michael T. Lam
- Department of Bioengineering, Rice University, Houston, Texas
| | | | - Junghae Suh
- Department of Bioengineering, Rice University, Houston, Texas
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, Texas
| |
Collapse
|
31
|
Schuster L, Seifert O, Vollmer S, Kontermann RE, Schlosshauer B, Hartmann H. Immunoliposomes for Targeted Delivery of an Antifibrotic Drug. Mol Pharm 2015; 12:3146-57. [DOI: 10.1021/acs.molpharmaceut.5b00012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Liane Schuster
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770 Reutlingen, Germany
| | - Oliver Seifert
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Stefanie Vollmer
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770 Reutlingen, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Burkhard Schlosshauer
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770 Reutlingen, Germany
| | - Hanna Hartmann
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770 Reutlingen, Germany
| |
Collapse
|
32
|
Chen J, Ryzhova LM, Sewell-Loftin MK, Brown CB, Huppert SS, Baldwin HS, Merryman WD. Notch1 Mutation Leads to Valvular Calcification Through Enhanced Myofibroblast Mechanotransduction. Arterioscler Thromb Vasc Biol 2015; 35:1597-605. [PMID: 26023079 DOI: 10.1161/atvbaha.114.305095] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/19/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Calcific aortic valve disease (CAVD) is a significant cardiovascular disorder, and controversy exists as to whether it is primarily a dystrophic or osteogenic process in vivo. In this study, we sought to clarify the mechanism of CAVD by assessing a genetic mutation, Notch1 heterozygosity, which leads to CAVD with 100% penetrance in humans. APPROACH AND RESULTS Murine immortalized Notch1(+/-) aortic valve interstitial cells (AVICs) were isolated and expanded in vitro. Molecular signaling of wild-type and Notch1(+/-) AVICs were compared to identify changes in pathways that have been linked to CAVD-transforming growth factor-β1/bone morphogenetic protein, mitogen-activated protein kinase, and phosphoinositide 3-kinase/protein kinase B-and assessed for calcification potential. Additionally, AVIC mechanobiology was studied in a physiologically relevant, dynamic mechanical environment (10% cyclic strain) to investigate differences in responses between the cell types. We found that Notch1(+/-) AVICs resembled a myofibroblast-like phenotype expressing higher amounts of cadherin-11, a known mediator of dystrophic calcification, and decreased Runx2, a known osteogenic marker. We determined that cadherin-11 expression is regulated by Akt activity, and inhibition of Akt phosphorylation significantly reduced cadherin-11 expression. Moreover, in the presence of cyclic strain, Notch1(+/-) AVICs exhibited significantly upregulated phosphorylation of Akt at Ser473 and smooth muscle α-actin expression, indicative of a fully activated myofibroblast. Finally, these Notch1-mediated alterations led to enhanced dystrophic calcific nodule formation. CONCLUSIONS This study presents novel insights in our understanding of Notch1-mediated CAVD by demonstrating that the mutation leads to AVICs that are fully activated myofibroblasts, resulting in dystrophic, but not osteogenic, calcification.
Collapse
Affiliation(s)
- Joseph Chen
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - Larisa M Ryzhova
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - M K Sewell-Loftin
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - Christopher B Brown
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - Stacey S Huppert
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - H Scott Baldwin
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - W David Merryman
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.).
| |
Collapse
|
33
|
|
34
|
Sapp MC, Fares HJ, Estrada AC, Grande-Allen KJ. Multilayer three-dimensional filter paper constructs for the culture and analysis of aortic valvular interstitial cells. Acta Biomater 2015; 13:199-206. [PMID: 25463506 PMCID: PMC10576252 DOI: 10.1016/j.actbio.2014.11.039] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 11/13/2014] [Accepted: 11/18/2014] [Indexed: 11/28/2022]
Abstract
Culturing aortic valvular interstitial cells in an environment that models the aortic valve is an essential step towards understanding the progression of calcific aortic valve disease. Here the adaption of a three-dimensional (3-D) stacked paper-based culture system is presented for analyzing valve cells in a thick collagen gel matrix. Filter paper layers, modeled after a 96-well plate design, were printed with a wax well-plate template and then seeded with valve cell and collagen mixtures that quickly gelled into 3-D cultures. Stacking these layers permitted extensive customization of culture thickness and cell density profiles to model the full thickness of native valve tissue. Aortic valvular interstitial cells seeded into the paper-based constructs consistently demonstrated high survival up to 14 days of culture with significant increases in cell number through the first 3 days of culture. After 4 days following seeding, valve cells in single layer cultures showed reduced smooth muscle α-actin expression with a stabilized cell density, suggesting a transition from an activated phenotype to a more quiescent state. Valve cells in multilayer cultures demonstrated the ability to migrate from layer to layer and had the highest smooth muscle α-actin expression in areas with predicted low oxygen tensions. These results establish the filter-paper-based method as a viable culture system for analyzing valve cells in an in vitro 3-D model of the aortic valve.
Collapse
Affiliation(s)
- Matthew C Sapp
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Hannelle J Fares
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Ana C Estrada
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | | |
Collapse
|
35
|
Chen J, Peacock JR, Branch J, David Merryman W. Biophysical analysis of dystrophic and osteogenic models of valvular calcification. J Biomech Eng 2015; 137:020903. [PMID: 25405546 DOI: 10.1115/1.4029115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Indexed: 12/27/2022]
Abstract
Calcific aortic valve disease (CAVD) is a significant cardiovascular disorder characterized by the formation of calcific nodules (CN) on the valve. In vitro assays studying the formation of these nodules were developed and have led to many significant mechanistic findings; however, the biophysical properties of CNs have not been clearly defined. A thorough analysis of dystrophic and osteogenic nodules utilizing scanning electron microscopy (SEM), energy dispersive spectrometry (EDS), and atomic force microscopy (AFM) was conducted to describe calcific nodule properties and provide a link between calcific nodule morphogenesis in vitro and in vivo. Unique nodule properties were observed for dystrophic and osteogenic nodules, highlighting the distinct mechanisms occurring in valvular calcification.
Collapse
|
36
|
Bowler MA, Merryman WD. In vitro models of aortic valve calcification: solidifying a system. Cardiovasc Pathol 2015; 24:1-10. [PMID: 25249188 PMCID: PMC4268061 DOI: 10.1016/j.carpath.2014.08.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 07/21/2014] [Accepted: 08/07/2014] [Indexed: 12/21/2022] Open
Abstract
Calcific aortic valve disease (CAVD) affects 25% of people over 65, and the late-stage stenotic state can only be treated with total valve replacement, requiring 85,000 surgeries annually in the US alone (University of Maryland Medical Center, 2013, http://umm.edu/programs/services/heart-center-programs/cardiothoracic-surgery/valve-surgery/facts). As CAVD is an age-related disease, many of the affected patients are unable to undergo the open-chest surgery that is its only current cure. This challenge motivates the elucidation of the mechanisms involved in calcification, with the eventual goal of alternative preventative and therapeutic strategies. There is no sufficient animal model of CAVD, so we turn to potential in vitro models. In general, in vitro models have the advantages of shortened experiment time and better control over multiple variables compared to in vivo models. As with all models, the hypothesis being tested dictates the most important characteristics of the in vivo physiology to recapitulate. Here, we collate the relevant pieces of designing and evaluating aortic valve calcification so that investigators can more effectively draw significant conclusions from their results.
Collapse
Affiliation(s)
- Meghan A Bowler
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212.
| |
Collapse
|
37
|
Drug delivery in aortic valve tissue engineering. J Control Release 2014; 196:307-23. [DOI: 10.1016/j.jconrel.2014.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/07/2014] [Accepted: 10/09/2014] [Indexed: 01/08/2023]
|
38
|
Abstract
During every heartbeat, cardiac valves open and close coordinately to control the unidirectional flow of blood. In this dynamically challenging environment, resident valve cells actively maintain homeostasis, but the signalling between cells and their microenvironment is complex. When homeostasis is disrupted and the valve opening obstructed, haemodynamic profiles can be altered and lead to impaired cardiac function. Currently, late stages of cardiac valve diseases are treated surgically, because no drug therapies exist to reverse or halt disease progression. Consequently, investigators have sought to understand the molecular and cellular mechanisms of valvular diseases using in vitro cell culture systems and biomaterial scaffolds that can mimic the extracellular microenvironment. In this Review, we describe how signals in the extracellular matrix regulate valve cell function. We propose that the cellular context is a critical factor when studying the molecular basis of valvular diseases in vitro, and one should consider how the surrounding matrix might influence cell signalling and functional outcomes in the valve. Investigators need to build a systems-level understanding of the complex signalling network involved in valve regulation, to facilitate drug target identification and promote in situ or ex vivo heart valve regeneration.
Collapse
|
39
|
Poggio P, Branchetti E, Grau JB, Lai EK, Gorman RC, Gorman JH, Sacks MS, Bavaria JE, Ferrari G. Osteopontin-CD44v6 interaction mediates calcium deposition via phospho-Akt in valve interstitial cells from patients with noncalcified aortic valve sclerosis. Arterioscler Thromb Vasc Biol 2014; 34:2086-94. [PMID: 25060796 DOI: 10.1161/atvbaha.113.303017] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The activation of valve interstitial cells (VICs) toward an osteogenic phenotype characterizes aortic valve sclerosis, the early asymptomatic phase of calcific aortic valve disease. Osteopontin is a phosphorylated acidic glycoprotein that accumulates within the aortic leaflets and labels VIC activation even in noncalcified asymptomatic patients. Despite this, osteopontin protects VICs against in vitro calcification. Here, we hypothesize that the specific interaction of osteopontin with CD44v6, and the related intracellular pathway, prevents calcium deposition in human-derived VICs from patients with aortic valve sclerosis. APPROACH AND RESULTS On informed consent, 23 patients and 4 controls were enrolled through the cardiac surgery and heart transplant programs. Human aortic valves and VICs were tested for osteogenic transdifferentiation, ex vivo and in vitro. Osteopontin-CD44 interaction was analyzed using proximity ligation assay and the signaling pathways investigated. A murine model based on angiotensin II infusion was used to mimic early pathological remodeling of the aortic valves. We report osteopontin-CD44 functional interaction as a hallmark of early stages of calcific aortic valve disease. We demonstrated that osteopontin-CD44 interaction mediates calcium deposition via phospho-Akt in VICs from patients with noncalcified aortic valve sclerosis. Finally, microdissection analysis of murine valves shows increased cusp thickness in angiotensin II-treated mice versus saline infused along with colocalization of osteopontin and CD44 as seen in human lesions. CONCLUSIONS Here, we unveil a specific protein-protein association and intracellular signaling mechanisms of osteopontin. Understanding the molecular mechanisms of early VIC activation and calcium deposition in asymptomatic stage of calcific aortic valve disease could open new prospective for diagnosis and therapeutic intervention.
Collapse
Affiliation(s)
- Paolo Poggio
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Emanuela Branchetti
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Juan B Grau
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Eric K Lai
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Robert C Gorman
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Joseph H Gorman
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Michael S Sacks
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Joseph E Bavaria
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Giovanni Ferrari
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.).
| |
Collapse
|
40
|
Witt W, Büttner P, Jannasch A, Matschke K, Waldow T. Reversal of myofibroblastic activation by polyunsaturated fatty acids in valvular interstitial cells from aortic valves. Role of RhoA/G-actin/MRTF signalling. J Mol Cell Cardiol 2014; 74:127-38. [PMID: 24839911 DOI: 10.1016/j.yjmcc.2014.05.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 12/19/2022]
Abstract
Valvular interstitial cells (VICs), the fibroblast-like cellular constituents of aortic heart valves, maintain structural integrity of valve tissue. Activation into contractile myofibroblasts occurs under pathological situations and under standard cell culture conditions of isolated VICs. Reversal of this phenotype switch would be of major importance in respect to fibrotic valve diseases. In this investigation, we found that exogenous polyunsaturated fatty acids (PUFAs) decreased contractility and expression of myofibroblastic markers like α-smooth muscle actin (αSMA) in cultured VICs from porcine aortic valves. The most active PUFAs, docosahexaenoic acid (DHA) and arachidonic acid (AA) reduced the level of active RhoA and increased the G/F-actin ratio. The G-actin-regulated nuclear translocation of myocardin-related transcription factors (MRTFs), co-activators of serum response factor, was also reduced by DHA and AA. The same effects were observed after blocking RhoA directly with C3 transferase. In addition, increased contractility after induction of actin polymerisation with jasplakinolide and concomitant expression of αSMA were ameliorated by active PUFAs. Furthermore, reduced αSMA expression under PUFA exposure was observed in valve tissue explants demonstrating physiological relevance. In conclusion, RhoA/G-actin/MRTF signalling is operative in VICs, and this pathway can be partially blocked by certain PUFAs whereby the activation into the myofibroblastic phenotype is reversed.
Collapse
Affiliation(s)
- Wolfgang Witt
- Department of Cardiac Surgery, Heart Center Dresden, Technical University Dresden, Dresden, Germany.
| | - Petra Büttner
- Department of Cardiac Surgery, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Anett Jannasch
- Department of Cardiac Surgery, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Klaus Matschke
- Department of Cardiac Surgery, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Thomas Waldow
- Department of Cardiac Surgery, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| |
Collapse
|
41
|
Abstract
Calcific aortic valve disease (CAVD) is a major contributor to cardiovascular morbidity and mortality and, given its association with age, the prevalence of CAVD is expected to continue to rise as global life expectancy increases. No drug strategies currently exist to prevent or treat CAVD. Given that valve replacement is the only available clinical option, patients often cope with a deteriorating quality of life until diminished valve function demands intervention. The recognition that CAVD results from active cellular mechanisms suggests that the underlying pathways might be targeted to treat the condition. However, no such therapeutic strategy has been successfully developed to date. One hope was that drugs already used to treat vascular complications might also improve CAVD outcomes, but the mechanisms of CAVD progression and the desired therapeutic outcomes are often different from those of vascular diseases. Therefore, we discuss the benchmarks that must be met by a CAVD treatment approach, and highlight advances in the understanding of CAVD mechanisms to identify potential novel therapeutic targets.
Collapse
Affiliation(s)
- Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences, 3 Blackfan Circle, 17th Floor, Center for Life Sciences Boston, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, 3 Blackfan Circle, 17th Floor, Center for Life Sciences Boston, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - W David Merryman
- Department of Biomedical Engineering, 2213 Garland Avenue, Vanderbilt University, Nashville, TN 37212, USA
| |
Collapse
|
42
|
El Accaoui RN, Gould ST, Hajj GP, Chu Y, Davis MK, Kraft DC, Lund DD, Brooks RM, Doshi H, Zimmerman KA, Kutschke W, Anseth KS, Heistad DD, Weiss RM. Aortic valve sclerosis in mice deficient in endothelial nitric oxide synthase. Am J Physiol Heart Circ Physiol 2014; 306:H1302-13. [PMID: 24610917 DOI: 10.1152/ajpheart.00392.2013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Risk factors for fibrocalcific aortic valve disease (FCAVD) are associated with systemic decreases in bioavailability of endothelium-derived nitric oxide (EDNO). In patients with bicuspid aortic valve (BAV), vascular expression of endothelial nitric oxide synthase (eNOS) is decreased, and eNOS(-/-) mice have increased prevalence of BAV. The goal of this study was to test the hypotheses that EDNO attenuates profibrotic actions of valve interstitial cells (VICs) in vitro and that EDNO deficiency accelerates development of FCAVD in vivo. As a result of the study, coculture of VICs with aortic valve endothelial cells (vlvECs) significantly decreased VIC activation, a critical early phase of FCAVD. Inhibition of VIC activation by vlvECs was attenuated by N(G)-nitro-l-arginine methyl ester or indomethacin. Coculture with vlvECs attenuated VIC expression of matrix metalloproteinase-9, which depended on stiffness of the culture matrix. Coculture with vlvECs preferentially inhibited collagen-3, compared with collagen-1, gene expression. BAV occurred in 30% of eNOS(-/-) mice. At age 6 mo, collagen was increased in both bicuspid and trileaflet eNOS(-/-) aortic valves, compared with wild-type valves. At 18 mo, total collagen was similar in eNOS(-/-) and wild-type mice, but collagen-3 was preferentially increased in eNOS(-/-) mice. Calcification and apoptosis were significantly increased in BAV of eNOS(-/-) mice at ages 6 and 18 mo. Remarkably, these histological changes were not accompanied by physiologically significant valve stenosis or regurgitation. In conclusion, coculture with vlvECs inhibits specific profibrotic VIC processes. In vivo, eNOS deficiency produces fibrosis in both trileaflet and BAVs but produces calcification only in BAVs.
Collapse
Affiliation(s)
- Ramzi N El Accaoui
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tseng H, Balaoing LR, Grigoryan B, Raphael RM, Killian TC, Souza GR, Grande-Allen KJ. A three-dimensional co-culture model of the aortic valve using magnetic levitation. Acta Biomater 2014; 10:173-82. [PMID: 24036238 PMCID: PMC10593146 DOI: 10.1016/j.actbio.2013.09.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 08/31/2013] [Accepted: 09/04/2013] [Indexed: 01/09/2023]
Abstract
The aortic valve consists of valvular interstitial cells (VICs) and endothelial cells (VECs). While these cells are understood to work synergistically to maintain leaflet structure and valvular function, few co-culture models of these cell types exist. In this study, aortic valve co-cultures (AVCCs) were assembled using magnetic levitation and cultured for 3 days. Immunohistochemistry and quantitative reverse-transcriptase polymerase chain reaction were used to assess the maintenance of cellular phenotype and function, and the formation of extracellular matrix. AVCCs stained positive for CD31 and α-smooth muscle actin (αSMA), demonstrating that the phenotype was maintained. Functional markers endothelial nitric oxide synthase (eNOS), von Willebrand factor (VWF) and prolyl-4-hydroxylase were present. Extracellular matrix components collagen type I, laminin and fibronectin also stained positive, with reduced gene expression of these proteins in three dimensions compared to two dimensions. Genes for collagen type I, lysyl oxidase and αSMA were expressed less in AVCCs than in 2-D cultures, indicating that VICs are quiescent. Co-localization of CD31 and αSMA in the AVCCs suggests that endothelial-mesenchymal transdifferentiation might be occurring. Differences in VWF and eNOS in VECs cultured in two and three dimensions also suggests that the AVCCs possibly have anti-thrombotic potential. Overall, a co-culture model of the aortic valve was designed, and serves as a basis for future experiments to understand heart valve biology.
Collapse
Affiliation(s)
- Hubert Tseng
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
- Nano3D Biosciences, Houston, TX 77030, USA
| | - Liezl R. Balaoing
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Bagrat Grigoryan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Robert M. Raphael
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
- Nano3D Biosciences, Houston, TX 77030, USA
| | - T. C. Killian
- Nano3D Biosciences, Houston, TX 77030, USA
- Department of Physics, Rice University, Houston, TX 77005, USA
| | | | | |
Collapse
|
44
|
Hydrogels preserve native phenotypes of valvular fibroblasts through an elasticity-regulated PI3K/AKT pathway. Proc Natl Acad Sci U S A 2013; 110:19336-41. [PMID: 24218588 DOI: 10.1073/pnas.1306369110] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Matrix elasticity regulates proliferation, apoptosis, and differentiation of many cell types across various tissues. In particular, stiffened matrix in fibrotic lesions has been shown to promote pathogenic myofibroblast activation. To better understand the underlying pathways by which fibroblasts mechano-sense matrix elasticity, we cultured primary valvular interstitial cells (VICs) isolated from porcine aortic valves on poly(ethylene glycol)-based hydrogels with physiologically relevant and tunable elasticities. We show that soft hydrogels preserve the quiescent fibroblast phenotype of VICs much better than stiff plastic plates. We demonstrate that the PI3K/AKT pathway is significantly up-regulated when VICs are cultured on stiff gels or tissue culture polystyrene compared with freshly isolated VICs. In contrast, myofibroblasts de-activate and pAKT/AKT decreases as early as 2 h after reducing the substrate modulus. When PI3K or AKT is inhibited on stiff substrates, myofibroblast activation is blocked. When constitutively active PI3K is overexpressed, the myofibroblast phenotype is promoted even on soft substrates. These data suggest that valvular fibroblasts are sensing the changes in matrix elasticity through the PI3K/AKT pathway. This mechanism may be used by other mechano-sensitive cells in response to substrate modulus, and this pathway may be a worthwhile target for treating matrix stiffness-associated diseases. Furthermore, hydrogels can be designed to recapitulate important mechanical cues in native tissues to preserve aspects of the native phenotype of primary cells for understanding basic cellular responses to biophysical and biochemical signals, and for tissue-engineering applications.
Collapse
|
45
|
Weiss RM, Miller JD, Heistad DD. Fibrocalcific aortic valve disease: opportunity to understand disease mechanisms using mouse models. Circ Res 2013; 113:209-22. [PMID: 23833295 DOI: 10.1161/circresaha.113.300153] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Studies in vitro and in vivo continue to identify complex-regulated mechanisms leading to overt fibrocalcific aortic valve disease (FCAVD). Assessment of the functional impact of those processes requires careful studies of models of FCAVD in vivo. Although the genetic basis for FCAVD is unknown for most patients with FCAVD, several disease-associated genes have been identified in humans and mice. Some gene products which regulate valve development in utero also protect against fibrocalcific disease during postnatal aging. Valve calcification can occur via processes that resemble bone formation. But valve calcification can also occur by nonosteogenic mechanisms, such as formation of calcific apoptotic nodules. Anticalcific interventions might preferentially target either osteogenic or nonosteogenic calcification. Although FCAVD and atherosclerosis share several risk factors and mechanisms, there are fundamental differences between arteries and the aortic valve, with respect to disease mechanisms and responses to therapeutic interventions. Both innate and acquired immunity are likely to contribute to FCAVD. Angiogenesis is a feature of inflammation, but may also contribute independently to progression of FCAVD, possibly by actions of pericytes that are associated with new blood vessels. Several therapeutic interventions seem to be effective in attenuating the development of FCAVD in mice. Therapies which are effective early in the course of FCAVD, however, are not necessarily effective in established disease.
Collapse
Affiliation(s)
- Robert M Weiss
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| | | | | |
Collapse
|
46
|
Yang HL, Lin KY, Juan YC, Kumar KJS, Way TD, Shen PC, Chen SC, Hseu YC. The anti-cancer activity of Antrodia camphorata against human ovarian carcinoma (SKOV-3) cells via modulation of HER-2/neu signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2013; 148:254-265. [PMID: 23619020 DOI: 10.1016/j.jep.2013.04.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/09/2013] [Accepted: 04/08/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Antrodia camphorata (AC) is well known in Taiwan as a traditional Chinese medicinal fungus. However, the anticancer activity of AC against human HER-2/neu-overexpressing ovarian cancers is poorly understood. MATERIALS AND METHODS The aim of this study is to investigate whether a submerged fermentation culture of AC can inhibit human ovarian carcinoma cell (SKOV-3) proliferation by suppressing the HER-2/neu signaling pathway. Cell viability, colony formation, DCFH-DA fluorescence microscopy, western blotting, HER-2/neu immunofluorescence imaging, flow cytometry, and TUNEL assays were carried out to determine the anti-cancer effects of AC. RESULTS MTT and colony formation assays showed that AC induced a dose-dependent reduction in SKOV-3 cell growth. Immunoblot analysis demonstrated that HER-2/neu activity and tyrosine phosphorylation were significantly inhibited by AC. Furthermore, AC treatment significantly inhibited the activation of PI3K/Akt and their downstream effector β-catenin. We also observed that AC caused G2/M arrest mediated by down-regulation of cyclin D1, cyclin A, cyclin B1, and Cdk1 and increased p27 expression. Notably, AC induced apoptosis, which was associated with DNA fragmentation, cytochrome c release, caspase-9/-3 activation, PARP degradation, and Bcl-2/Bax dysregulation. An increase in intracellular reactive oxygen species (ROS) was observed in AC-treated cells, whereas the antioxidant N-acetylcysteine (NAC) prevented AC-induced cell death, HER-2/neu depletion, PI3K/Akt inactivation, and Bcl-2/Bax dysregulation, indicating that AC-induced cell death was mediated by ROS generation. CONCLUSIONS These results suggest that AC may exert anti-tumor activity against human ovarian carcinoma by suppressing HER-2/neu signaling pathways.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Institute of Nutrition, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Poggio P, Sainger R, Branchetti E, Grau JB, Lai EK, Gorman RC, Sacks MS, Parolari A, Bavaria JE, Ferrari G. Noggin attenuates the osteogenic activation of human valve interstitial cells in aortic valve sclerosis. Cardiovasc Res 2013; 98:402-10. [PMID: 23483047 DOI: 10.1093/cvr/cvt055] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AIMS Aortic valve sclerosis (AVSc) is a hallmark of several cardiovascular conditions ranging from chronic heart failure and myocardial infarction to calcific aortic valve stenosis (AVS). AVSc, present in 25-30% of patients over 65 years of age, is characterized by thickening of the leaflets with marginal effects on the mechanical proprieties of the valve making its presentation asymptomatic. Despite its clinical prevalence, few studies have investigated the pathogenesis of this disease using human AVSc specimens. Here, we investigate in vitro and ex vivo BMP4-mediated transdifferentiation of human valve interstitial cells (VICs) towards an osteogenic-like phenotype in AVSc. METHODS AND RESULTS Human specimens from 60 patients were collected at the time of aortic valve replacement (AVS) or through the heart transplant programme (Controls and AVSc). We show that non-calcified leaflets from AVSc patients can be induced to express markers of osteogenic transdifferentiation and biomineralization through the combinatory effect of BMP4 and mechanical stimulation. We show that BMP4 antagonist Noggin attenuates VIC activation and biomineralization. Additionally, patient-derived VICs were induced to transdifferentiate using either cell culture or a Tissue Engineering (TE) Aortic Valve model. We determine that while BMP4 alone is not sufficient to induce osteogenic transdifferentiation of AVSc-derived cells, the combinatory effect of BMP4 and mechanical stretch induces VIC activation towards a phenotype typical of late calcified stage of the disease. CONCLUSION This work demonstrates, for the first time using AVSc specimens, that human sclerotic aortic valves can be induced to express marker of osteogenic-like phenotype typical of advanced severe aortic stenosis.
Collapse
Affiliation(s)
- Paolo Poggio
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Branchetti E, Sainger R, Poggio P, Grau JB, Patterson-Fortin J, Bavaria JE, Chorny M, Lai E, Gorman RC, Levy RJ, Ferrari G. Antioxidant enzymes reduce DNA damage and early activation of valvular interstitial cells in aortic valve sclerosis. Arterioscler Thromb Vasc Biol 2012; 33:e66-74. [PMID: 23241403 DOI: 10.1161/atvbaha.112.300177] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Accumulation of reactive oxygen species (ROS) and remodeling of the microstructure of the cusp characterize aortic valve sclerosis, the early phase of calcific aortic valve disease. These events are associated with activation of valvular interstitial cells (VICs) toward an osteogenic-like phenotype. Because ROS cause DNA damage and transcriptional activation we investigated the relationship between ROS, DNA damage response, and transdifferentiation of VICs. METHODS AND RESULTS Human aortic valve cusps and patient-matched VICs were collected from 39 patients both with and without calcific aortic valve disease. VICs were exposed to hydrogen peroxide (0.1-1 mmol/L) after cell transduction with extracellular superoxide dismutase/catalase adenoviruses and characterized for DNA-damage response, osteogenic transdifferentiation, and calcification. ROS induce relocalization of phosphorylated γH2AX, MRE11, and XRCC1 proteins with expression of osteogenic signaling molecule RUNX2 via AKT. We report a sustained activation of γH2AX in aortic valve sclerosis-derived VICs suggesting their impaired ability to repair DNA damage. Adenovirus superoxide dismutase/catalase transduction decreases ROS-induced DNA damage and VIC transdifferentiation in aortic valve sclerosis-derived cells. Finally, adenoviral transduction with catalase reverts ROS-mediated calcification and cellular transdifferentiation. CONCLUSIONS We conclude that the ROS-induced DNA damage response is dysfunctional in early asymptomatic stages of calcific aortic valve disease. We unveiled an association among ROS, DNA-damage response, and cellular transdifferentiation, reversible by antioxidant enzymes delivery.
Collapse
Affiliation(s)
- Emanuela Branchetti
- Department of Surgery, Division of Cardiovascular Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19036, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hutcheson JD, Chen J, Sewell-Loftin MK, Ryzhova LM, Fisher CI, Su YR, Merryman WD. Cadherin-11 regulates cell-cell tension necessary for calcific nodule formation by valvular myofibroblasts. Arterioscler Thromb Vasc Biol 2012; 33:114-20. [PMID: 23162011 DOI: 10.1161/atvbaha.112.300278] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Dystrophic calcific nodule formation in vitro involves differentiation of aortic valve interstitial cells (AVICs) into a myofibroblast phenotype. Interestingly, inhibition of the kinase MAPK Erk kinase (MEK)1/2 prevents calcific nodule formation despite leading to myofibroblast activation of AVICs, indicating the presence of an additional mechanotransductive component required for calcific nodule morphogenesis. In this study, we assess the role of transforming growth factor β1-induced cadherin-11 expression in calcific nodule formation. METHODS AND RESULTS As shown previously, porcine AVICs treated with transforming growth factor β1 before cyclic strain exhibit increased myofibroblast activation and significant calcific nodule formation. In addition to an increase in contractile myofibroblast markers, transforming growth factor β1-treated AVICs exhibit significantly increased expression of cadherin-11. This expression is inhibited by the addition of U0126, a specific MEK1/2 inhibitor. The role of increased cadherin-11 is revealed through a wound assay, which demonstrates increased intercellular tension in transforming growth factor β1-treated AVICs possessing cadherin-11. Furthermore, when small interfering RNA is used to knockdown cadherin-11, calcific nodule formation is abrogated, indicating that robust cell-cell connections are necessary in generating tension for calcific nodule morphogenesis. Finally, we demonstrate enrichment of cadherin-11 in human calcified leaflets. CONCLUSIONS These results indicate the necessity of cadherin-11 for dystrophic calcific nodule formation, which proceeds through an Erk1/2-dependent pathway.
Collapse
Affiliation(s)
- Joshua D Hutcheson
- Department of Biomedical Engineering, Vanderbilt University, 2213 Garland Ave, 9445 MRB IV, Nashville, TN 37232-0493, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Hutcheson JD, Ryzhova LM, Setola V, Merryman WD. 5-HT(2B) antagonism arrests non-canonical TGF-β1-induced valvular myofibroblast differentiation. J Mol Cell Cardiol 2012; 53:707-14. [PMID: 22940605 PMCID: PMC3472096 DOI: 10.1016/j.yjmcc.2012.08.012] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 07/20/2012] [Accepted: 08/10/2012] [Indexed: 01/12/2023]
Abstract
Transforming growth factor-β1 (TGF-β1) induces myofibroblast activation of quiescent aortic valve interstitial cells (AVICs), a differentiation process implicated in calcific aortic valve disease (CAVD). The ubiquity of TGF-β1 signaling makes it difficult to target in a tissue specific manner; however, the serotonin 2B receptor (5-HT(2B)) is highly localized to cardiopulmonary tissues and agonism of this receptor displays pro-fibrotic effects in a TGF-β1-dependent manner. Therefore, we hypothesized that antagonism of 5-HT(2B) opposes TGF-β1-induced pathologic differentiation of AVICs and may offer a druggable target to prevent CAVD. To test this hypothesis, we assessed the interaction of 5-HT(2B) antagonism with canonical and non-canonical TGF-β1 pathways to inhibit TGF-β1-induced activation of isolated porcine AVICs in vitro. Here we show that AVIC activation and subsequent calcific nodule formation is completely mitigated by 5-HT(2B) antagonism. Interestingly, 5-HT(2B) antagonism does not inhibit canonical TGF-β1 signaling as identified by Smad3 phosphorylation and activation of a partial plasminogen activator inhibitor-1 promoter (PAI-1, a transcriptional target of Smad3), but prevents non-canonical p38 MAPK phosphorylation. It was initially suspected that 5-HT(2B) antagonism prevents Src tyrosine kinase phosphorylation; however, we found that this is not the case and time-lapse microscopy indicates that 5-HT(2B) antagonism prevents non-canonical TGF-β1 signaling by physically arresting Src tyrosine kinase. This study demonstrates the necessity of non-canonical TGF-β1 signaling in leading to pathologic AVIC differentiation. Moreover, we believe that the results of this study suggest 5-HT(2B) antagonism as a novel therapeutic approach for CAVD that merits further investigation.
Collapse
Affiliation(s)
| | - Larisa M. Ryzhova
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Vincent Setola
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC
| | - W. David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| |
Collapse
|