1
|
Chong-Nguyen C, Yilmaz B, Coles B, Sokol H, MacPherson A, Siepe M, Reineke D, Mosbahi S, Tomii D, Nakase M, Atighetchi S, Ferro C, Wingert C, Gräni C, Pilgrim T, Windecker S, Blasco H, Dupuy C, Emond P, Banz Y, Losmanovà T, Döring Y, Siontis GCM. A scoping review evaluating the current state of gut microbiota and its metabolites in valvular heart disease physiopathology. Eur J Clin Invest 2025; 55:e14381. [PMID: 39797472 DOI: 10.1111/eci.14381] [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: 11/04/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND The human microbiome is crucial in regulating intestinal and systemic functions. While its role in cardiovascular disease is better understood, the link between intestinal microbiota and valvular heart diseases (VHD) remains largely unexplored. METHODS Peer-reviewed studies on human, animal or cell models analysing gut microbiota profiles published up to April 2024 were included. Eligible studies used 16S rRNA or shotgun sequencing, metabolite profiling by mass spectrometry, and examined osteogenesis or fibrosis signalling in valve cells. Methods and findings were qualitatively analysed, with data charted to summarize study design, materials and outcomes. RESULTS Thirteen studies were included in the review: five human, three animal and five in vitro. Of the nine studies on calcific aortic stenosis (CAS), elevated trimethylamine N-oxide (TMAO) levels were linked to an increased risk of cardiovascular events in cohort studies, with CAS patients showing higher levels of Bacteroides plebeius, Enterobacteriaceae, Veillonella dispar and Prevotella copri. In vivo, TMAO promoted aortic valve fibrosis, while tryptophan derivatives stimulated osteogenic differentiation and interleukin-6 secretion in valvular interstitial cells. Two studies on rheumatic mitral valve disease found altered microbiota profiles and lower short-chain fatty acid levels, suggesting potential impacts on immune regulation. Two studies on Barlow's mitral valve disease in animal models revealed elevated TMAO levels in dogs with congestive heart failure, reduced Paraprevotellaceae, increased Actinomycetaceae and dysbiosis involving Turicibacter and E. coli. CONCLUSIONS TMAO has been mainly identified as a prognostic marker in VHD. Gut microbiota dysbiosis has been observed in various forms of VHD and deserve further study.
Collapse
Affiliation(s)
| | - Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Bernadette Coles
- Velindre University NHS Trust Library and Knowledge Service, Cardiff, UK
| | - Harry Sokol
- Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique-Hopitaux de Paris (APHP), Paris, France
| | - Andrew MacPherson
- Department of Visceral Surgery and Medicine, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Matthias Siepe
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - David Reineke
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Selim Mosbahi
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Daijiro Tomii
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Masaaki Nakase
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Sarah Atighetchi
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Cyril Ferro
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Christoph Wingert
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Christoph Gräni
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Thomas Pilgrim
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Stephan Windecker
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Hélène Blasco
- Faculté de médecine, Equipe neurogénétique et neurométabolomique, INSERM U930, Université François Rabelais, Tours, France
| | - Camille Dupuy
- Faculté de médecine, Equipe neurogénétique et neurométabolomique, INSERM U930, Université François Rabelais, Tours, France
| | - Patrick Emond
- Faculté de médecine, Equipe neurogénétique et neurométabolomique, INSERM U930, Université François Rabelais, Tours, France
| | - Yara Banz
- Institute of Tissue Medicine and Pathology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Tereza Losmanovà
- Institute of Tissue Medicine and Pathology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Yvonne Döring
- Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
| | - George C M Siontis
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| |
Collapse
|
2
|
Shah T, Zhang Z, Shah H, Fanaroff AC, Nathan AS, Parise H, Lutz J, Sugeng L, Bellumkonda L, Redfors B, Omerovic E, Petrie MC, Vora AN, Fiorilli PN, Kobayashi T, Ahmad Y, Forrest JK, Giri JS, Herrmann HC, Lansky AJ. Effect of Sodium-Glucose Cotransporter-2 Inhibitors on the Progression of Aortic Stenosis. JACC Cardiovasc Interv 2025; 18:738-748. [PMID: 39985508 DOI: 10.1016/j.jcin.2024.11.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 02/24/2025]
Abstract
BACKGROUND Aortic stenosis (AS) is the leading cause of valvular heart disease-related morbidity and mortality, but there are no medical treatments to slow its progression. Sodium-glucose cotransporter-2 inhibitors (SGLT2i) have pleiotropic effects which could be disease modifying in AS. OBJECTIVES The purpose of this study was to determine if SGLT2i usage is associated with slower progression of AS. METHODS A target trial emulation comparing the effect of the initiation of SGLT2i compared with no SGLT2i in patients with nonsevere AS was performed using retrospective electronic medical record data from the Yale New Haven Health System from January 2016 to September 2022. Patients with native aortic valve sclerosis or nonsevere AS with at least 12 months of echocardiographic follow-up were included. Patients were excluded if they had an estimated glomerular filtration rate <30 mL/min/1.73 m2 or had initiated SGLT2i >1 year before the index echocardiogram. The prespecified primary outcome was progression to severe AS. RESULTS A total of 458 patients prescribed SGLT2i and 11,240 patients never prescribed SGLT2i were included. Patients were on SGLT2i for a median of 0.9 years. Patients on SGLT2i were younger and had higher rates of diabetes and chronic kidney disease. Patients on SGLT2i were more likely to have ejection fraction ≤40%. There were no differences between groups in baseline AS severity (66% sclerosis, 23% mild stenosis, and 11% moderate in overall cohort). Patients ever prescribed SGLT2i were less likely to progress to severe AS (HR: 0.61; 95% CI: 0.39-0.94; P = 0.03) with a progressively lower risk among patients on SGLT2i for >3, 6, and 12 months (HR: 0.54, 0.48, and 0.27, respectively). CONCLUSIONS This retrospective, multicenter, observational study suggests that SGLT2i may slow the progression of nonsevere AS.
Collapse
Affiliation(s)
- Tayyab Shah
- The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Yale Cardiovascular Research Group, New Haven, Connecticut, USA
| | - Zhiyuan Zhang
- Yale Cardiovascular Research Group, New Haven, Connecticut, USA
| | | | | | - Ashwin S Nathan
- The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Helen Parise
- Yale Cardiovascular Research Group, New Haven, Connecticut, USA
| | - John Lutz
- Yale New Haven Hospital, New Haven, Connecticut, USA
| | | | | | - Björn Redfors
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden; Cardiovascular Research Foundation, New York, New York, USA
| | - Elmir Omerovic
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Mark C Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Amit N Vora
- Yale Cardiovascular Research Group, New Haven, Connecticut, USA
| | - Paul N Fiorilli
- The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Taisei Kobayashi
- The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yousif Ahmad
- Yale Cardiovascular Research Group, New Haven, Connecticut, USA
| | - John K Forrest
- Yale Cardiovascular Research Group, New Haven, Connecticut, USA
| | - Jay S Giri
- The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Howard C Herrmann
- The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
3
|
Skenteris NT, Hemme E, Delfos L, Karadimou G, Karlöf E, Lengquist M, Kronqvist M, Zhang X, Maegdefessel L, Schurgers LJ, Arnardottir H, Biessen EAL, Bot I, Matic L. Mast cells participate in smooth muscle cell reprogramming and atherosclerotic plaque calcification. Vascul Pharmacol 2023; 150:107167. [PMID: 36958707 DOI: 10.1016/j.vph.2023.107167] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/14/2023] [Accepted: 03/19/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND Calcification, a key feature of advanced human atherosclerosis, is positively associated with vascular disease burden and adverse events. We showed that macrocalcification can be a stabilizing factor for carotid plaque molecular biology, due to inverse association with immune processes. Mast cells (MCs) are important contributors to plaque instability, but their relationship with macrocalcification is unexplored. With a hypothesis that MC activation negatively associates with carotid plaque macrocalcification, we aimed to investigate the link between MCs and carotid plaque vulnerability, and study MC role in plaque calcification via smooth muscle cells (SMCs). METHODS Pre-operative computed tomography angiographies of patients (n = 40) undergoing surgery for carotid stenosis were used to characterize plaque morphology. Plaque microarrays (n = 40 and n = 126) were used for bioinformatic deconvolution of immune cell populations. Tissue microarrays (n = 103) were used to histologically validate the contribution of activated and resting MCs in plaques. RESULTS Activated MCs and their typical markers were negatively correlated with macrocalcification. The ratio of activated vs. resting MCs was increased in low-calcified plaques from symptomatic patients. There was no modulating effect of medication on MC ratios. In vitro experiments showed that SMC calcification attenuated MC activation, while both active and resting MCs stimulated SMC calcification and induced dedifferentiation towards a pro-inflammatory-, osteochondrocyte-like phenotype, without modulating their migro-proliferative function. CONCLUSIONS Integrative analyses from human plaques showed that MC activation is inversely associated with macrocalcification and positively with parameters of plaque vulnerability. Mechanistically, MCs induce SMC osteogenic reprograming, while matrix calcification in turn attenuates MC activation, offering new therapeutic avenues for exploration.
Collapse
Affiliation(s)
- Nikolaos T Skenteris
- Cardiovascular Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden; Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, the Netherlands
| | - Esmeralda Hemme
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | - Lucie Delfos
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | - Glykeria Karadimou
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Eva Karlöf
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Mariette Lengquist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Malin Kronqvist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Xiang Zhang
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Lars Maegdefessel
- Cardiovascular Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden; Technical University Munich, Klinikum rechts der Isar, Department for Vascular and Endovascular Surgery, Germany
| | - Leon J Schurgers
- Department of Biochemistry and CARIM, School for Cardiovascular Diseases, Maastricht University, Netherlands; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Hildur Arnardottir
- Cardiovascular Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, the Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | - Ljubica Matic
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
4
|
Abourehab MAS, Baisakhiya S, Aggarwal A, Singh A, Abdelgawad MA, Deepak A, Ansari MJ, Pramanik S. Chondroitin sulfate-based composites: a tour d'horizon of their biomedical applications. J Mater Chem B 2022; 10:9125-9178. [PMID: 36342328 DOI: 10.1039/d2tb01514e] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chondroitin sulfate (CS), a natural anionic mucopolysaccharide, belonging to the glycosaminoglycan family, acts as the primary element of the extracellular matrix (ECM) of diverse organisms. It comprises repeating units of disaccharides possessing β-1,3-linked N-acetyl galactosamine (GalNAc), and β-1,4-linked D-glucuronic acid (GlcA), and exhibits antitumor, anti-inflammatory, anti-coagulant, anti-oxidant, and anti-thrombogenic activities. It is a naturally acquired bio-macromolecule with beneficial properties, such as biocompatibility, biodegradability, and immensely low toxicity, making it the center of attention in developing biomaterials for various biomedical applications. The authors have discussed the structure, unique properties, and extraction source of CS in the initial section of this review. Further, the current investigations on applications of CS-based composites in various biomedical fields, focusing on delivering active pharmaceutical compounds, tissue engineering, and wound healing, are discussed critically. In addition, the manuscript throws light on preclinical and clinical studies associated with CS composites. A short section on Chondroitinase ABC has also been canvassed. Finally, this review emphasizes the current challenges and prospects of CS in various biomedical fields.
Collapse
Affiliation(s)
- Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al Qura University, Makkah 21955, Saudi Arabia. .,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, Minia 11566, Egypt
| | - Shreya Baisakhiya
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Sector 1, Rourkela, Odisha 769008, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu 613401, India
| | - Akanksha Aggarwal
- Delhi Institute of Pharmaceutical Sciences and Research, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Anshul Singh
- Department of Chemistry, Baba Mastnath University, Rohtak-124021, India
| | - Mohamed A Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Al Jouf 72341, Saudi Arabia
| | - A Deepak
- Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 600128, Tamil Nadu, India.
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Sheersha Pramanik
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India.
| |
Collapse
|
5
|
The Haemodynamic and Pathophysiological Mechanisms of Calcific Aortic Valve Disease. Biomedicines 2022; 10:biomedicines10061317. [PMID: 35740339 PMCID: PMC9220142 DOI: 10.3390/biomedicines10061317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 11/17/2022] Open
|
6
|
Deb N, Lacerda CMR. Valvular Endothelial Cell Response to the Mechanical Environment-A Review. Cell Biochem Biophys 2021; 79:695-709. [PMID: 34661855 DOI: 10.1007/s12013-021-01039-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/02/2021] [Indexed: 01/08/2023]
Abstract
Heart valve leaflets are complex structures containing valve endothelial cells, interstitial cells, and extracellular matrix. Heart valve endothelial cells sense mechanical stimuli, and communicate amongst themselves and the surrounding cells and extracellular matrix to maintain tissue homeostasis. In the presence of abnormal mechanical stimuli, endothelial cell communication is triggered in defense and such processes may eventually lead to cardiac disease progression. This review focuses on the role of mechanical stimuli on heart valve endothelial surfaces-from heart valve development and maintenance of tissue integrity to disease progression with related signal pathways involved in this process.
Collapse
Affiliation(s)
- Nandini Deb
- Jasper Department of Chemical Engineering, The University of Texas at Tyler, 3900 University Blvd, Tyler, 75799, TX, US
| | - Carla M R Lacerda
- Jasper Department of Chemical Engineering, The University of Texas at Tyler, 3900 University Blvd, Tyler, 75799, TX, US.
| |
Collapse
|
7
|
Chen H, Wang Y, Sun B, Bao X, Tang Y, Huang F, Zhu S, Xu J. Negative correlation between endoglin levels and coronary atherosclerosis. Lipids Health Dis 2021; 20:127. [PMID: 34602076 PMCID: PMC8487532 DOI: 10.1186/s12944-021-01545-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/06/2021] [Indexed: 11/22/2022] Open
Abstract
Background Coronary artery disease (CAD) is a common cardiovascular disease, and abnormal blood lipid metabolism is an important risk factor. Transforming growth factor-ß (TGF-ß) and its receptor (TGF-ßR) can inhibit the release of inflammatory factors through the SMAD pathway-mediated immune response, thereby suppressing the progression of CAD. Endoglin (TGF-ßRIII), a TGF-ßR family homologous receptor protein, is directly involved in the immunoregulatory process, but the exact mechanism is unclear. This study aimed to clarify the pathophysiological effects of endoglin on the development of atherosclerosis and to explore the mechanism of the signalling pathway. Methods We downloaded the GEO dataset to perform a functional analysis of SMAD family activity and TGF-ß receptor protein expression in the monocyte expression profiles of patients with familial hyperlipidaemia (FH). The effect of endoglin on endothelial cell proliferation, migration, and apoptosis was examined by disrupting the endoglin gene in human umbilical vein endothelial cells (HUVECs) and validated by western blotting. The related genes and pathways regulated by endoglin were obtained by analysing the sequencing data. Results Research has shown that interference with endoglin can promote the proliferation and migration and significantly inhibit the apoptosis of vascular endothelial cells. Interference with endoglin particularly encourages the expression of VEGFB in vascular endothelial cells. Conclusion The endoglin gene in vascular endothelial cells regulates the PI3K-Akt, Wnt, TNF, and cellular metabolism pathways by activating the SMAD pathway. RAB26, MR1, CCL2, SLC29A4, IBTK, VEGFB, and GOLGA8B play critical roles. Endoglin interacts closely with 11 proteins such as CCL2 and SEPRINE1, which participate in the vital pathway of plaque formation. Interference with endoglin can alter the course of coronary atherosclerosis.
Collapse
Affiliation(s)
- Haibin Chen
- Department of Cardiovascular, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xincun Road, Putuo District, 200065, Shanghai, China
| | - Yiping Wang
- Department of Cardiovascular, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xincun Road, Putuo District, 200065, Shanghai, China
| | - Bing Sun
- Department of Cardiovascular, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xincun Road, Putuo District, 200065, Shanghai, China
| | - Xunxia Bao
- School of Life Science, Anhui Medical University, Hefei, 230022, China
| | - Yu Tang
- Department of Cardiovascular, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xincun Road, Putuo District, 200065, Shanghai, China
| | - Feifei Huang
- Department of Cardiovascular, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xincun Road, Putuo District, 200065, Shanghai, China
| | - Sibo Zhu
- Department of Epidemiology, School of Public Health, State Key Laboratory of Genetic Engineering and Human Phenome Institute, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| | - Jiahong Xu
- Department of Cardiovascular, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xincun Road, Putuo District, 200065, Shanghai, China.
| |
Collapse
|
8
|
Choi GC, Kim S, Rahman MM, Oh JH, Cho YS, Shin HJ. Entelon (vitis vinifera seed extract) reduces degenerative changes in bovine pericardium valve leaflet in a dog intravascular implant model. PLoS One 2021; 16:e0235454. [PMID: 33661896 PMCID: PMC7932063 DOI: 10.1371/journal.pone.0235454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 02/10/2021] [Indexed: 11/25/2022] Open
Abstract
Background and aims Inflammation and calcification are major factors responsible for degeneration of bioprosthetic valve and other substitute heart valve implantations. The objective of this study was to evaluate the anti-inflammatory and anti-calcification effects of Entelon150® (consisting of grape-seed extract) in a beagle dog model of intravascular bovine pericardium implantation. Methods In total, 8 healthy male beagle dogs were implanted with a bovine pericardium bilaterally in the external jugular veins and divided into two groups. Animals in the Entelon150® group (n = 4) were treated with 150 mg of Entelon150® twice daily for six weeks after surgery. The negative control (NC) group (n = 4) was treated with 5 ml of saline using the same method. After six weeks, we measured the calcium content, performed histological examination, and performed molecular analysis. Results The calcium content of implanted tissue in the Entelon150® group (0.56±0.14 mg/g) was significantly lower than that in the NC group (1.48±0.57 mg/g) (p < 0.05). Histopathological examination showed that infiltration of chronic inflammatory cells, such as fibroblasts and macrophages, occurred around the graft in all groups; however, the inflammation level of the implanted tissue in the Entelon150® group was s lower than that in the NC group. Both immunohistochemical and western blot analyses revealed that bone morphogenetic protein 2 expression was significantly attenuated in the Entelon150® group. Conclusions Our results indicate that Entelon150® significantly attenuates post-implantation inflammation and degenerative calcification of the bovine pericardium in dogs. Therefore, Entelon150® may increase the longevity of the bovine pericardium after intravascular implantation.
Collapse
Affiliation(s)
- Gab-Chol Choi
- Animal Medical Center W, Mapo-gu, Seoul, Korea
- Department of Veterinary Surgery, College of Veterinary Medicine, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Sokho Kim
- KNOTUS Co., Ltd., Research Center, Incheon, Korea
| | | | - Ji Hyun Oh
- Department of Thoracic and Cardiovascular Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Yun Seok Cho
- Department of Thoracic and Cardiovascular Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Hong Ju Shin
- Department of Thoracic and Cardiovascular Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
- * E-mail:
| |
Collapse
|
9
|
Ma H, Caldwell AS, Azagarsamy MA, Gonzalez Rodriguez A, Anseth KS. Bioorthogonal click chemistries enable simultaneous spatial patterning of multiple proteins to probe synergistic protein effects on fibroblast function. Biomaterials 2020; 255:120205. [PMID: 32574845 PMCID: PMC7396286 DOI: 10.1016/j.biomaterials.2020.120205] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 12/18/2022]
Abstract
Three biorthogonal click reactions, a photoinitiated thiol-yne reaction, an azide-alkyne cycloaddition, and a methyltetrazine-transcyclooctene Diels Alder, were used to independently control the presentation of several bioactive proteins to valvular interstitial cells (VICs) in hydrogel scaffolds. Tethered fibroblast growth factor (FGF-2) was found to suppress myofibroblast activation (from 48 ± 7% to 17 ± 6%) and promote proliferation (from 10 ± 2% to 54 ± 3%) at a concentration of 10 ng/mL. In the presence of the pro-fibrotic cytokine transforming growth factor-beta (TGF-β1), FGF-2 could protect the VIC fibroblast phenotype, even at much higher concentrations of TGF-β1 than that of FGF-2. With respect to the fibrocalcific VIC phenotype, TGF-β1 and bone-morphogenic protein-2 (BMP-2) were found to synergistically promote calcific nodule formation (a five-fold increase in nodules compared to TGF-β1 or BMP-2 alone). Exploiting the orthogonal click reactions, FGF-2, TGF-β1 and BMP-2 combinations were patterned into distinct regions on a hydrogel to control VIC activation and nodule formation. Cellular crosstalk between separate regions of the same scaffold was affected by the size of each region as well as the interfacial area between different regions. Collectively, these results demonstrate the versatility and robustness of a photoinitiated thiol-yne reaction to template pendant functionalities that allow for the bioconjugation of multiple proteins. This approach maintains protein bioactivity, providing an in vitro platform capable of achieving a better understanding of the complex mechanisms involved in tissue fibrosis.
Collapse
Affiliation(s)
- Hao Ma
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO, 80303, USA; BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80303, USA
| | - Alexander S Caldwell
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO, 80303, USA; BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80303, USA
| | - Malar A Azagarsamy
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO, 80303, USA; BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80303, USA
| | - Andrea Gonzalez Rodriguez
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO, 80303, USA; BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80303, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO, 80303, USA; BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80303, USA.
| |
Collapse
|
10
|
Barth M, Selig JI, Klose S, Schomakers A, Kiene LS, Raschke S, Boeken U, Akhyari P, Fischer JW, Lichtenberg A. Degenerative aortic valve disease and diabetes: Implications for a link between proteoglycans and diabetic disorders in the aortic valve. Diab Vasc Dis Res 2019; 16:254-269. [PMID: 30563371 DOI: 10.1177/1479164118817922] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Degenerative aortic valve disease in combination with diabetes is an increasing burden worldwide. There is growing evidence that particularly small leucine-rich proteoglycans are involved in the development of degenerative aortic valve disease. Nevertheless, the role of these molecules in this disease in the course of diabetes has not been elucidated in detail and previous studies remain controversial. Therefore, the aim of this study is to broaden the knowledge about small leucine-rich proteoglycans in degenerative aortic valve disease and the influence of diabetes and hyperglycaemia on aortic valves and valvular interstitial cells is examined. Analyses were performed using reverse-transcription polymerase chain reaction, Western blot, enzyme-linked immunosorbent assay, (immuno)histology and colorimetric assays. We could show that biglycan, but not decorin and lumican, is upregulated in degenerated human aortic valve cusps. Subgroup analysis reveals that upregulation of biglycan is stage-dependent. In vivo, loss of biglycan leads to stage-dependent calcification and also to migratory effects on interstitial cells within the extracellular matrix. In late stages of degenerative aortic valve disease, diabetes increases the expression of biglycan in aortic valves. In vitro, the combinations of hyperglycaemic with pro-degenerative conditions lead to an upregulation of biglycan. In conclusion, biglycan represents a potential link between degenerative aortic valve disease and diabetes.
Collapse
Affiliation(s)
- Mareike Barth
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Jessica I Selig
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Svenja Klose
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Antje Schomakers
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Lena S Kiene
- 2 Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Silja Raschke
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Udo Boeken
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Payam Akhyari
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Jens W Fischer
- 2 Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Artur Lichtenberg
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
11
|
Stephens EH, Han J, Trawick EA, Di Martino ES, Akkiraju H, Brown LM, Connell JP, Grande-Allen KJ, Vunjak-Novakovic G, Takayama H. Left-Ventricular Assist Device Impact on Aortic Valve Mechanics, Proteomics and Ultrastructure. Ann Thorac Surg 2017; 105:572-580. [PMID: 29223417 DOI: 10.1016/j.athoracsur.2017.08.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/24/2017] [Accepted: 08/07/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Aortic regurgitation is a prevalent, detrimental complication of left ventricular assist devices (LVADs). The altered hemodynamics of LVADs results in aortic valves (AVs) having distinct mechanical stimulation. Our hypothesis was that the altered AV hemodynamics modulates the valve cells and matrix, resulting in changes in valvular mechanical properties that then can lead to regurgitation. METHODS AVs were collected from 16 LVAD and 6 non-LVAD patients at time of heart transplant. Standard demographic and preoperative data were collected and comparisons between the two groups were calculated using standard statistical methods. Samples were analyzed using biaxial mechanical tensile testing, mass spectrometry-based proteomics, and transmission electron microscopy to assess ultrastructure. RESULTS The maximum circumferential leaflet strain in LVAD patients was less than in non-LVAD patients (0.35 ± 0.10MPa versus 0.52 ± 0.18 MPa, p = 0.03) with a trend of reduced radial strain (p = 0.06) and a tendency for the radial strain to decrease with increasing LVAD duration (p = 0.063). Numerous proteins associated with actin and myosin, immune signaling and oxidative stress, and transforming growth factor β were increased in LVAD patients. Ultrastructural analysis showed a trend of increased fiber diameter in LVAD patients (46.2 ± 7.2 nm versus 45.1 ± 6.9 nm, p = 0.10), but no difference in fiber density. CONCLUSIONS AVs in LVAD patients showed decreased compliance and increased expression of numerous proteins related to valve activation and injury compared to non-LVAD patients. Further knowledge of AV changes leading to regurgitation in LVAD patients and the pathways by which they occur may provide an opportunity for interventions to prevent and/or reverse this detrimental complication.
Collapse
Affiliation(s)
- Elizabeth H Stephens
- Division of Cardiac, Thoracic and Vascular Surgery, Columbia University Medical Center, New York, New York.
| | - Jiho Han
- Division of Cardiac, Thoracic and Vascular Surgery, Columbia University Medical Center, New York, New York
| | - Emma A Trawick
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Elena S Di Martino
- Schulich School of Engineering and Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Hemanth Akkiraju
- Quantitative Proteomics and Metabolomics Center and Department of Biological Sciences, Columbia University, New York, New York
| | - Lewis M Brown
- Quantitative Proteomics and Metabolomics Center and Department of Biological Sciences, Columbia University, New York, New York
| | | | | | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, New York; Department of Medicine, Columbia University Medical Center, New York, New York
| | - Hiroo Takayama
- Division of Cardiac, Thoracic and Vascular Surgery, Columbia University Medical Center, New York, New York
| |
Collapse
|
12
|
Wu MY, Li CJ, Hou MF, Chu PY. New Insights into the Role of Inflammation in the Pathogenesis of Atherosclerosis. Int J Mol Sci 2017; 18:2034. [PMID: 28937652 PMCID: PMC5666716 DOI: 10.3390/ijms18102034] [Citation(s) in RCA: 268] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipids, smooth muscle cell proliferation, cell apoptosis, necrosis, fibrosis, and local inflammation. Immune and inflammatory responses have significant effects on every phase of atherosclerosis, and increasing evidence shows that immunity plays a more important role in atherosclerosis by tightly regulating its progression. Therefore, understanding the relationship between immune responses and the atherosclerotic microenvironment is extremely important. This article reviews existing knowledge regarding the pathogenesis of immune responses in the atherosclerotic microenvironment, and the immune mechanisms involved in atherosclerosis formation and activation.
Collapse
Affiliation(s)
- Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Chia-Jung Li
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Ming-Feng Hou
- Department of Surgery, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Surgery, Kaohsiung Municipal Hsiao Kang Hospital, Kaohsiung 807, Taiwan.
- Division of Breast Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Pei-Yi Chu
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei 242, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| |
Collapse
|
13
|
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: 225] [Impact Index Per Article: 28.1] [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
|
14
|
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
|
15
|
Entstehung und Progression der Aortenklappendegeneration. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2017. [DOI: 10.1007/s00398-016-0086-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Schoen FJ, Gotlieb AI. Heart valve health, disease, replacement, and repair: a 25-year cardiovascular pathology perspective. Cardiovasc Pathol 2016; 25:341-352. [PMID: 27242130 DOI: 10.1016/j.carpath.2016.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/04/2016] [Accepted: 05/05/2016] [Indexed: 01/24/2023] Open
Abstract
The past several decades have witnessed major advances in the understanding of the structure, function, and biology of native valves and the pathobiology and clinical management of valvular heart disease. These improvements have enabled earlier and more precise diagnosis, assessment of the proper timing of surgical and interventional procedures, improved prosthetic and biologic valve replacements and repairs, recognition of postoperative complications and their management, and the introduction of minimally invasive approaches that have enabled definitive and durable treatment for patients who were previously considered inoperable. This review summarizes the current state of our understanding of the mechanisms of heart valve health and disease arrived at through innovative research on the cell and molecular biology of valves, clinical and pathological features of the most frequent intrinsic structural diseases that affect the valves, and the status and pathological considerations in the technological advances in valvular surgery and interventions. The contributions of many cardiovascular pathologists and other scientists, engineers, and clinicians are emphasized, and potentially fruitful areas for research are highlighted.
Collapse
Affiliation(s)
- Frederick J Schoen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115; Pathology and Health Sciences and Technology (HST), Harvard Medical School, 75 Francis Street, Boston, MA 02115.
| | - Avrum I Gotlieb
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Laboratory Medicine Program, University Health Network, Medical Sciences Building, 1 King's College Circle, Rm. 6275A, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
17
|
Hecht E, Freise C, Websky KV, Nasser H, Kretzschmar N, Stawowy P, Hocher B, Querfeld U. The matrix metalloproteinases 2 and 9 initiate uraemic vascular calcifications. Nephrol Dial Transplant 2015; 31:789-97. [PMID: 26333546 DOI: 10.1093/ndt/gfv321] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/30/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The matrix metalloproteinases (MMP) MMP-2 and MMP-9 are physiological regulators of vascular remodelling. Their dysregulation could contribute to vascular calcification. We examined the role of the MMP-2 and MMP-9 in uraemic vascular calcification in vivo and in vitro. METHODS The impact of pharmacological MMP inhibition on the development of media calcifications was explored in an aggressive animal model of uraemic calcification. In addition, the selective effects of addition and inhibition, respectively, of MMP-2 and MMP-9 on calcium-/phosphate-induced calcifications were studied in a murine cell line of vascular smooth muscle cells (VSMCs). RESULTS High-dose calcitriol treatment of uraemic rats given a high phosphate diet induced massive calcifications, apoptosis and increased gene expressions of MMP-2, MMP-9 and of osteogenic transcription factors and proteins in aortic VSMC. The MMP inhibitor doxycycline prevented the VSMC transdifferentiation to osteoblastic cells, suppressed transcription of mediators of matrix remodelling and almost completely blocked aortic calcifications while further increasing apoptosis. Similarly, specific inhibitors of either MMP-2 or -9, or of both gelatinases (Ro28-2653) and a selective knockdown of MMP-2/-9 mRNA expression blocked calcification of murine VSMC induced by calcification medium (CM). In contrast to MMP inhibition, recombinant MMP-2 or MMP-9 enhanced CM-induced calcifications and the secretion of gelatinases. CONCLUSIONS These data indicate that both gelatinases provide essential signals for phenotypic VSMC conversion, matrix remodelling and the initiation of vascular calcification. Their inhibition seems a promising strategy in the prevention of vascular calcifications.
Collapse
Affiliation(s)
- Eva Hecht
- Center for Cardiovascular Research, Charité-University Medicine, Campus Mitte, Berlin, Germany Department of Pediatric Nephrology, Charité-University Medicine, Campus Virchow Clinic, Berlin, Germany
| | - Christian Freise
- Center for Cardiovascular Research, Charité-University Medicine, Campus Mitte, Berlin, Germany Department of Pediatric Nephrology, Charité-University Medicine, Campus Virchow Clinic, Berlin, Germany
| | - Karoline V Websky
- Institute of Nutritional Science, University of Potsdam, Potsdam-Rehbrücke, Germany
| | - Hamoud Nasser
- Center for Cardiovascular Research, Charité-University Medicine, Campus Mitte, Berlin, Germany Department of Pediatric Nephrology, Charité-University Medicine, Campus Virchow Clinic, Berlin, Germany
| | - Nadja Kretzschmar
- Center for Cardiovascular Research, Charité-University Medicine, Campus Mitte, Berlin, Germany Department of Pediatric Nephrology, Charité-University Medicine, Campus Virchow Clinic, Berlin, Germany
| | - Philipp Stawowy
- Center for Cardiovascular Research, Charité-University Medicine, Campus Mitte, Berlin, Germany Department of Medicine and Cardiology, German Heart Institute, Berlin, Germany
| | - Berthold Hocher
- Institute of Nutritional Science, University of Potsdam, Potsdam-Rehbrücke, Germany
| | - Uwe Querfeld
- Center for Cardiovascular Research, Charité-University Medicine, Campus Mitte, Berlin, Germany Department of Pediatric Nephrology, Charité-University Medicine, Campus Virchow Clinic, Berlin, Germany
| |
Collapse
|
18
|
Latif N, Sarathchandra P, Chester AH, Yacoub MH. Expression of smooth muscle cell markers and co-activators in calcified aortic valves. Eur Heart J 2015; 36:1335-45. [PMID: 24419809 DOI: 10.1093/eurheartj/eht547] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 10/24/2013] [Indexed: 01/06/2023] Open
Abstract
AIMS Similar risk factors and mediators are involved in calcific aortic stenosis (CAS) and atherosclerosis. Since normal valves harbour a low percentage of smooth muscle cells (SMCs), we hypothesize that the SMC phenotype participates in the pathogenesis of CAS. METHOD AND RESULTS We analysed 12 normal and 22 calcified aortic valves for SMC markers and the expression of co-activators of SMC gene expression, myocardin and myocardin-related transcription factors (MRTF-A/B). Transforming growth factor β (TGFβ1) was used to upregulate SMC markers and co-activators in valve interstitial cells (VICs) and transmission electron microscopy (TEM) was used to detect the presence of SMC in atypical regions of the valve leaflets. Smooth muscle cell markers and co-activators, myocardin, MRTF-A, and MRTF-B, demonstrated an increased incidence and aberrant expression around calcified nodules in all 22 calcified valves as well as in surface and microvessel endothelial cells. Smooth muscle cell markers and MRTF-A were significantly increased in calcified valves. Transforming growth factor β1 (TGFβ1) (10 ng/mL) was able to significantly upregulate the expression of some SMC markers and MRTF-A in VICs. Transmission electron microscopy of the fibrosa layer of calcified valves demonstrated the presence of bundles of SMCs and smooth muscle-derived foam cells. CONCLUSION Smooth muscle cell markers and co-activators, myocardin and MRTFs, were aberrantly expressed in calcified valves. Transforming growth factor β1 was able to significantly upregulate SMC markers and MRTF-A in VICs. Transmission electron microscopy unequivocally identified the presence of SMCs in calcified regions of valve leaflets. These findings provide evidence that the SMC phenotype plays a role in the development of CAS.
Collapse
Affiliation(s)
- Najma Latif
- Heart Science Centre, Imperial College, Harefield Hospital, Harefield, Middlesex UB9 6JH, UK QCRC, Qatar Cardiovascular Research Centre, Qatar Foundation, Qatar
| | - Padmini Sarathchandra
- Heart Science Centre, Imperial College, Harefield Hospital, Harefield, Middlesex UB9 6JH, UK QCRC, Qatar Cardiovascular Research Centre, Qatar Foundation, Qatar
| | - Adrian H Chester
- Heart Science Centre, Imperial College, Harefield Hospital, Harefield, Middlesex UB9 6JH, UK QCRC, Qatar Cardiovascular Research Centre, Qatar Foundation, Qatar
| | - Magdi H Yacoub
- Heart Science Centre, Imperial College, Harefield Hospital, Harefield, Middlesex UB9 6JH, UK QCRC, Qatar Cardiovascular Research Centre, Qatar Foundation, Qatar
| |
Collapse
|
19
|
Regulation of valve endothelial cell vasculogenic network architectures with ROCK and Rac inhibitors. Microvasc Res 2015; 98:108-18. [PMID: 25660064 PMCID: PMC4974942 DOI: 10.1016/j.mvr.2015.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 01/03/2015] [Accepted: 01/26/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The age- and disease-dependent presence of microvessels within heart valves is an understudied characteristic of these tissues. Neovascularization involves endothelial cell (EC) migration and cytoskeletal reorientation, which are heavily regulated by the Rho family of GTPases. Given that valve ECs demonstrate unique mesenchymal transdifferentiation and cytoskeletal mechanoresponsiveness, compared to vascular ECs, this study quantified the effect of inhibiting two members of the Rho family on vasculogenic network formation by valve ECs. APPROACH AND RESULTS A tubule-like structure vasculogenesis assay (assessing lacunarity, junction density, and vessel density) was performed with porcine aortic valve ECs treated with small molecule inhibitors of Rho-associated serine-threonine protein kinase (ROCK), Y-27632, or the Rac1 inhibitor, NSC-23766. Actin coordination, cell number, and cell migration were assessed through immunocytochemistry, MTT assay, and scratch wound healing assay. ROCK inhibition reduced network lacunarity and interrupted proper cell-cell adhesion and actin coordination. Rac1 inhibition increased lacunarity and delayed actin-mediated network formation. ROCK inhibition alone significantly inhibited migration, whereas both ROCK and Rac1 inhibition significantly reduced cell number over time compared to controls. Compared to a vascular EC line, the valve ECs generated a network with larger total vessel length, but a less smooth appearance. CONCLUSIONS Both ROCK and Rac1 inhibition interfered with key processes in vascular network formation by valve ECs. This is the first report of manipulation of valve EC vasculogenic organization in response to small molecule inhibitors. Further study is warranted to comprehend this facet of valvular cell biology and pathology and how it differs from vascular biology.
Collapse
|
20
|
Lerman DA, Prasad S, Alotti N. Calcific Aortic Valve Disease: Molecular Mechanisms and Therapeutic Approaches. Eur Cardiol 2015; 10:108-112. [PMID: 27274771 PMCID: PMC4888946 DOI: 10.15420/ecr.2015.10.2.108] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 10/28/2015] [Indexed: 01/28/2023] Open
Abstract
Calcification occurs in atherosclerotic vascular lesions and In the aortic valve. Calcific aortic valve disease (CAVD) is a slow, progressive disorder that ranges from mild valve thickening without obstruction of blood flow, termed aortic sclerosis, to severe calcification with impaired leaflet motion, termed aortic stenosis. In the past, this process was thought to be 'degenerative' because of time-dependent wear and tear of the leaflets, with passive calcium deposition. The presence of osteoblasts in atherosclerotic vascular lesions and in CAVD implies that calcification is an active, regulated process akin to atherosclerosis, with lipoprotein deposition and chronic inflammation. If calcification is active, via pro-osteogenic pathways, one might expect that development and progression of calcification could be inhibited. The overlap in the clinical factors associated with calcific valve disease and atherosclerosis provides further support for a shared disease mechanism. In our recent research we used an in vitro porcine valve interstitial cell model to study spontaneous calcification and potential promoters and inhibitors. Using this model, we found that denosumab, a human monoclonal antibody targeting the receptor activator of nuclear factor-κB ligand may, at a working concentration of 50 μg/mL, inhibit induced calcium deposition to basal levels.
Collapse
Affiliation(s)
- Daniel Alejandro Lerman
- Royal Infirmary Hospital of Edinburgh (NHS Lothian), The University of Edinburgh, United Kingdom
| | - Sai Prasad
- Royal Infirmary Hospital of Edinburgh (NHS Lothian), The University of Edinburgh, United Kingdom
| | | |
Collapse
|
21
|
Vizzardi E, D'Aloia A, Sciatti E, Bonadei I, Gelsomino S, Lorusso R, Metra M. Echocardiographic evaluation of aortic atheromas in patients with aortic stenosis. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2015; 34:105-110. [PMID: 25542945 DOI: 10.7863/ultra.34.1.105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
OBJECTIVES The association of aortic atheromas in patients with isolated aortic stenosis has recently been acknowledged, probably because the pathogenic mechanisms are similar. Therefore, this study evaluated the extent and severity of thoracic aortic atheromas in patients with different grades of aortic stenosis using transesophageal echocardiography. METHODS We retrospectively evaluated transesophageal echocardiographic examinations of 686 consecutive patients with a diagnosis of aortic stenosis. The prevalence and morphologic characteristics of atheromas in 3 segments of the thoracic aorta were assessed. Plaque thickness was measured at each segment, and the thickest plaque was used to establish severity. Atheromas were graded as mild, moderate, or severe according to plaque thickness (<2, 2-4, or >4 mm, respectively). Aortic stenosis was graded as mild, moderate, or severe on the basis of the gradient and anatomic aortic valve area (>1.5, 1.0-1.5, or <1.0 cm(2)). RESULTS A total of 382 patients were men, and 304 were women (mean age ± SD, 74 ± 15 years); 86% of the patients had aortic atheromas. The severe stenosis group had a significantly higher rate of atheromas (95% versus 40%; P < .001) than the mild stenosis group, with more complex atheromas (52% versus 22%; P< .001). There was no significant difference in the atheroma grades between the severe and moderate stenosis groups, but moderate cases had more moderate and severe atheromas than mild cases (45% and 15% versus 19% and 3%; P < .01). CONCLUSIONS This study showed a correlation in the extent of aortic atheromas across several degrees of aortic stenosis. Patients with moderate and severe stenosis had more extensive atherosclerotic atheromas than those with mild stenosis.
Collapse
Affiliation(s)
- Enrico Vizzardi
- From the Section of Cardiovascular Diseases, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy (E.V., A.D., E.S., I.B., M.M.); Heart and Vessels Department, Careggi Hospital, Florence, Italy (S.G.); and Experimental Cardiac Surgery Laboratory, Cardiac Surgery Unit, Civil Hospital, Brescia, Italy (R.L.).
| | - Antonio D'Aloia
- From the Section of Cardiovascular Diseases, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy (E.V., A.D., E.S., I.B., M.M.); Heart and Vessels Department, Careggi Hospital, Florence, Italy (S.G.); and Experimental Cardiac Surgery Laboratory, Cardiac Surgery Unit, Civil Hospital, Brescia, Italy (R.L.)
| | - Edoardo Sciatti
- From the Section of Cardiovascular Diseases, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy (E.V., A.D., E.S., I.B., M.M.); Heart and Vessels Department, Careggi Hospital, Florence, Italy (S.G.); and Experimental Cardiac Surgery Laboratory, Cardiac Surgery Unit, Civil Hospital, Brescia, Italy (R.L.)
| | - Ivano Bonadei
- From the Section of Cardiovascular Diseases, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy (E.V., A.D., E.S., I.B., M.M.); Heart and Vessels Department, Careggi Hospital, Florence, Italy (S.G.); and Experimental Cardiac Surgery Laboratory, Cardiac Surgery Unit, Civil Hospital, Brescia, Italy (R.L.)
| | - Sandro Gelsomino
- From the Section of Cardiovascular Diseases, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy (E.V., A.D., E.S., I.B., M.M.); Heart and Vessels Department, Careggi Hospital, Florence, Italy (S.G.); and Experimental Cardiac Surgery Laboratory, Cardiac Surgery Unit, Civil Hospital, Brescia, Italy (R.L.)
| | - Roberto Lorusso
- From the Section of Cardiovascular Diseases, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy (E.V., A.D., E.S., I.B., M.M.); Heart and Vessels Department, Careggi Hospital, Florence, Italy (S.G.); and Experimental Cardiac Surgery Laboratory, Cardiac Surgery Unit, Civil Hospital, Brescia, Italy (R.L.)
| | - Marco Metra
- From the Section of Cardiovascular Diseases, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy (E.V., A.D., E.S., I.B., M.M.); Heart and Vessels Department, Careggi Hospital, Florence, Italy (S.G.); and Experimental Cardiac Surgery Laboratory, Cardiac Surgery Unit, Civil Hospital, Brescia, Italy (R.L.)
| |
Collapse
|
22
|
Abstract
Background—
Although a previous study has suggested that a genetic variant in the
LPA
region was associated with the presence of aortic valve stenosis (AVS), no prospective study has suggested a role for lipoprotein(a) levels in the pathophysiology of AVS. Our objective was to determine whether lipoprotein(a) levels and a common genetic variant that is strongly associated with lipoprotein(a) levels are associated with an increased risk of developing AVS.
Methods and Results—
Serum lipoprotein(a) levels were measured in 17 553 participants of the European Prospective Investigation into Cancer (EPIC)-Norfolk study. Among these study participants, 118 developed AVS during a mean follow-up of 11.7 years. The rs10455872 genetic variant in
LPA
was genotyped in 14 735 study participants, who simultaneously had lipoprotein(a) level measurements, and in a replication study of 379 patients with echocardiography-confirmed AVS and 404 controls. In EPIC-Norfolk, compared with participants in the bottom lipoprotein(a) tertile, those in the top lipoprotein(a) tertile had a higher risk of AVS (hazard ratio, 1.57; 95% confidence interval, 1.02–2.42) after adjusting for age, sex, and smoking. Compared with rs10455872 AA homozygotes, carriers of 1 or 2 G alleles were at increased risk of AVS (hazard ratio, 1.78; 95% confidence interval, 1.11–2.87, versus hazard ratio, 4.83; 95% confidence interval, 1.77–13.20, respectively). In the replication study, the genetic variant rs10455872 also showed a positive association with AVS (odds ratio, 1.57; 95% confidence interval, 1.10–2.26).
Conclusions—
Patients with high lipoprotein(a) levels are at increased risk for AVS. The rs10455872 variant, which is associated with higher lipoprotein(a) levels, is also associated with increased risk of AVS, suggesting that this association may be causal.
Collapse
|
23
|
Lee WR, Kim KH, An HJ, Park YY, Kim KS, Lee CK, Min BK, Park KK. Effects of chimeric decoy oligodeoxynucleotide in the regulation of transcription factors NF-κB and Sp1 in an animal model of atherosclerosis. Basic Clin Pharmacol Toxicol 2013; 112:236-243. [PMID: 23107157 DOI: 10.1111/bcpt.12029] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 10/09/2012] [Indexed: 12/21/2022]
Abstract
Atherosclerosis is a multifactorial and progressive disease in which the inflammatory reaction and inflammation-related factors play important roles at all stages. Modulation of NF-κB and Sp1 expression may be important targets for the prevention and treatment of atherosclerotic vascular disease. To develop a novel therapeutic approach in atherosclerosis, we examined the simultaneous suppression of the transcription factors NF-κB and Sp1 which regulate inflammation. We employed chimeric decoy oligodeoxynucleotide (ODN) containing the consensus of NF-κB and Sp1-binding sites to suppress these transcription factors simultaneously and to test chimeric decoy for anti-atherogenic effects in an atherogenic diet-induced atherosclerotic mouse model with inflammatory stimulation. C57BL/6 mice were fed with an atherogenic diet (15% fat, 1.25% cholesterol and 0.5% cholic acid) for 12 weeks to induce atherosclerosis; lipopolysaccharide (LPS, 2 mg/kg) was intraperitoneally injected in the first week of study to simulate underlying infectious burden during development of atherosclerosis. Decoy ODNs were injected into tail vein at 2, 4, 6, 8, 10 and 12 weeks after only three LPS injections in mice fed the atherogenic diet. Chimeric decoy ODN alleviated atherosclerotic changes and reduced serum cholesterol and inflammatory cytokines. In accordance with these results, the expressions of atherosclerotic markers were inhibited by chimeric decoy ODN. Chimeric decoy ODN modulates multiple pathogenic aspects of an atherogenic diet-induced atherosclerosis with inflammatory stimulation: hypercholesterolaemia and inflammation. Therefore, this study demonstrates the efficacy of chimeric decoy ODN on atherosclerosis with immunological complication.
Collapse
Affiliation(s)
- Woo-Ram Lee
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Transforming growth factor-β and abdominal aortic aneurysms. Cardiovasc Pathol 2013; 22:126-32. [DOI: 10.1016/j.carpath.2012.07.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Revised: 07/22/2012] [Accepted: 07/30/2012] [Indexed: 12/22/2022] Open
|
25
|
Riley G, Syeda F, Kirchhof P, Fabritz L. An introduction to murine models of atrial fibrillation. Front Physiol 2012; 3:296. [PMID: 22934047 PMCID: PMC3429067 DOI: 10.3389/fphys.2012.00296] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 07/08/2012] [Indexed: 01/28/2023] Open
Abstract
Understanding the mechanism of re-entrant arrhythmias in the past 30 years has allowed the development of almost curative therapies for many rhythm disturbances. The complex, polymorphic arrhythmias of atrial fibrillation (AF) and sudden death are, unfortunately, not yet well understood, and hence still in need of adequate therapy. AF contributes markedly to morbidity and mortality in aging Western populations. In the past decade, many genetically altered murine models have been described and characterized. Here, we review genetically altered murine models of AF; powerful tools that will enable a better understanding of the mechanisms of AF and the assessment of novel therapeutic interventions.
Collapse
Affiliation(s)
- Genna Riley
- Centre for Cardiovascular Sciences, School of Clinical and Experimental Medicine, University of Birmingham Birmingham, UK
| | | | | | | |
Collapse
|
26
|
McCoy CM, Nicholas DQ, Masters KS. Sex-related differences in gene expression by porcine aortic valvular interstitial cells. PLoS One 2012; 7:e39980. [PMID: 22808080 PMCID: PMC3393722 DOI: 10.1371/journal.pone.0039980] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 05/30/2012] [Indexed: 12/22/2022] Open
Abstract
While many large-scale risk factors for calcific aortic valve disease (CAVD) have been identified, the molecular etiology and subsequent pathogenesis of CAVD have yet to be fully understood. Specifically, it is unclear what biological phenomena underlie the significantly higher occurrence of CAVD in the male population. We hypothesized the existence of intrinsic, cellular-scale differences between male and female valvular interstitial cells (VICs) that contribute to male sex being a risk factor for CAVD. Differences in gene expression profiles between healthy male and female porcine VICs were investigated via microarray analysis. Mean expression values of each probe set in the male samples were compared to the female samples, and biological processes were analyzed for overrepresentation using Gene Ontology term enrichment analysis. There were 183 genes identified as significantly (fold change>2; P<0.05) different in male versus female aortic valve leaflets. Within this significant gene list there were 298 overrepresented biological processes, several of which are relevant to pathways identified in CAVD pathogenesis. In particular, pathway analysis indicated that cellular proliferation, apoptosis, migration, ossification, angiogenesis, inflammation, and extracellular matrix reorganization were all significantly represented in the data set. These gene expression findings also translated into functional differences in VIC behavior in the in vitro environment, as sex-related differences in proliferation and apoptosis were confirmed in VIC populations cultured in vitro. These data suggest that a sex-related propensity for CAVD exists on the cellular level in healthy subjects, a phenomenon that could have significant clinical implications. These findings also strongly support discontinuing the use of mixed-sex VIC cultures, thereby changing the current standard in the field.
Collapse
Affiliation(s)
- Chloe M. McCoy
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Dylan Q. Nicholas
- Department of Mechanical Engineering, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Kristyn S. Masters
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, United States of America
| |
Collapse
|
27
|
Kim SJ, Park JH, Kim KH, Lee WR, Pak SC, Han SM, Park KK. The Protective Effect of Apamin on LPS/Fat-Induced Atherosclerotic Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2012; 2012:305454. [PMID: 22645626 PMCID: PMC3357006 DOI: 10.1155/2012/305454] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 01/30/2012] [Accepted: 02/05/2012] [Indexed: 11/18/2022]
Abstract
Apamin, a peptide component of bee venom (BV), has anti-inflammatory properties. However, the molecular mechanisms by which apamin prevents atherosclerosis are not fully understood. We examined the effect of apamin on atherosclerotic mice. Atherosclerotic mice received intraperitoneal (ip) injections of lipopolysaccharide (LPS, 2 mg/kg) to induce atherosclerotic change and were fed an atherogenic diet for 12 weeks. Apamin (0.05 mg/kg) was administered by ip injection. LPS-induced THP-1-derived macrophage inflammation treated with apamin reduced expression of tumor necrosis factor (TNF)-α, vascular cell adhesion molecule (VCAM)-1, and intracellular cell adhesion molecule (ICAM)-1, as well as the nuclear factor kappa B (NF-κB) signaling pathway. Apamin decreased the formation of atherosclerotic lesions as assessed by hematoxylin and elastic staining. Treatment with apamin reduced lipids, Ca(2+) levels, and TNF-α in the serum from atherosclerotic mice. Further, apamin significantly attenuated expression of VCAM-1, ICAM-1, TGF-β1, and fibronectin in the descending aorta from atherosclerotic mice. These results indicate that apamin plays an important role in monocyte/macrophage inflammatory processing and may be of potential value for preventing atherosclerosis.
Collapse
Affiliation(s)
- Soo-Jung Kim
- Department of Pathology, School of Medicine, Catholic University of Daegu, 3056-6 Daemyung 4-Dong, Nam-Gu, Daegu 705-718, Republic of Korea
| | - Ji-Hyun Park
- Department of Pathology, School of Medicine, Catholic University of Daegu, 3056-6 Daemyung 4-Dong, Nam-Gu, Daegu 705-718, Republic of Korea
| | - Kyung-Hyun Kim
- Department of Pathology, School of Medicine, Catholic University of Daegu, 3056-6 Daemyung 4-Dong, Nam-Gu, Daegu 705-718, Republic of Korea
| | - Woo-Ram Lee
- Department of Pathology, School of Medicine, Catholic University of Daegu, 3056-6 Daemyung 4-Dong, Nam-Gu, Daegu 705-718, Republic of Korea
| | - Sok Cheon Pak
- School of Biomedical Sciences, Charles Sturt University, Bathurst, NSW 2795, Australia
| | - Sang-Mi Han
- Department of Agricultural Biology, National Academy of Agricultural Science, Suwon 441-100, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Catholic University of Daegu, 3056-6 Daemyung 4-Dong, Nam-Gu, Daegu 705-718, Republic of Korea
| |
Collapse
|
28
|
Alexopoulos A, Kaoukis A, Papadaki H, Pyrgakis V. Pathophysiologic mechanisms of calcific aortic stenosis. Ther Adv Cardiovasc Dis 2012; 6:71-80. [DOI: 10.1177/1753944712439337] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Calcific aortic stenosis (CAS) comprises the leading indication for valve replacement in the Western world. Until recently, progressive calcification was considered to be a passive process. Emerging evidence, however, suggests that degenerative aortic stenosis constitutes an active process involving stimulation of several pathophysiologic pathways such as inflammation and osteogenesis. In addition, CAS and atherosclerosis share common features regarding histopathology of lesions. These novel data raise a new perspective on the prevention and treatment of disease. The current article reviews the most important pathophysiologic mechanisms of senile aortic stenosis.
Collapse
Affiliation(s)
- Alexandros Alexopoulos
- Department of Cardiology, Athens General Hospital, Mesogeion Avenue, 154, 15669 Athens, Greece
| | - Andreas Kaoukis
- Department of Cardiology, General Hospital of Athens ‘G. Gennimatas’, Greece, Athens, Greece
| | - Helen Papadaki
- Department of Anatomy, School of Medicine, University of Patras, Greece, Rio, Patras, Greece
| | - Vlassios Pyrgakis
- Department of Cardiology, General Hospital of Athens ‘G. Gennimatas’, Greece, Athens, Greece
| |
Collapse
|
29
|
Doetschman T, Barnett JV, Runyan RB, Camenisch TD, Heimark RL, Granzier HL, Conway SJ, Azhar M. Transforming growth factor beta signaling in adult cardiovascular diseases and repair. Cell Tissue Res 2011; 347:203-23. [PMID: 21953136 DOI: 10.1007/s00441-011-1241-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 09/02/2011] [Indexed: 01/15/2023]
Abstract
The majority of children with congenital heart disease now live into adulthood due to the remarkable surgical and medical advances that have taken place over the past half century. Because of this, adults now represent the largest age group with adult cardiovascular diseases. It includes patients with heart diseases that were not detected or not treated during childhood, those whose defects were surgically corrected but now need revision due to maladaptive responses to the procedure, those with exercise problems and those with age-related degenerative diseases. Because adult cardiovascular diseases in this population are relatively new, they are not well understood. It is therefore necessary to understand the molecular and physiological pathways involved if we are to improve treatments. Since there is a developmental basis to adult cardiovascular disease, transforming growth factor beta (TGFβ) signaling pathways that are essential for proper cardiovascular development may also play critical roles in the homeostatic, repair and stress response processes involved in adult cardiovascular diseases. Consequently, we have chosen to summarize the current information on a subset of TGFβ ligand and receptor genes and related effector genes that, when dysregulated, are known to lead to cardiovascular diseases and adult cardiovascular deficiencies and/or pathologies. A better understanding of the TGFβ signaling network in cardiovascular disease and repair will impact genetic and physiologic investigations of cardiovascular diseases in elderly patients and lead to an improvement in clinical interventions.
Collapse
|
30
|
Li X, Johnson KR, Bryant M, Elkahloun AG, Amar M, Remaley AT, De Silva R, Hallenbeck JM, Quandt JA. Intranasal delivery of E-selectin reduces atherosclerosis in ApoE-/- mice. PLoS One 2011; 6:e20620. [PMID: 21701687 PMCID: PMC3119064 DOI: 10.1371/journal.pone.0020620] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 05/09/2011] [Indexed: 11/20/2022] Open
Abstract
Mucosal tolerance to E-selectin prevents stroke and protects against ischemic brain damage in experimental models of stroke studying healthy animals or spontaneously hypertensive stroke-prone rats. A reduction in inflammation and neural damage was associated with immunomodulatory or “tolerogenic” responses to E-selectin. The purpose of the current study on ApoE deficient mice is to assess the capacity of this stroke prevention innovation to influence atherosclerosis, a major underlying cause for ischemic strokes; human E-selectin is being translated as a potential clinical prevention strategy for secondary stroke. Female ApoE−/− mice received intranasal delivery of E-selectin prior to (pre-tolerization) or simultaneously with initiation of a high-fat diet. After 7 weeks on the high-fat diet, lipid lesions in the aorta, serum triglycerides, and total cholesterol were assessed as markers of atherosclerosis development. We also assessed E-selectin-specific antibodies and cytokine responses, in addition to inflammatory responses that included macrophage infiltration of the aorta and altered gene expression profiles of aortic mRNA. Intranasal delivery of E-selectin prior to initiation of high-fat chow decreased atherosclerosis, serum total cholesterol, and expression of the leucocyte chemoattractant CCL21 that is typically upregulated in atherosclerotic lesions of ApoE−/− mice. This response was associated with the induction of E-selectin specific cells producing the immunomodulatory cytokine IL-10 and immunosuppressive antibody isotypes. Intranasal administration of E-selectin generates E-selectin specific immune responses that are immunosuppressive in nature and can ameliorate atherosclerosis, a major risk factor for ischemic stroke. These results provide additional preclinical support for the potential of induction of mucosal tolerance to E-selectin to prevent stroke.
Collapse
Affiliation(s)
- Xinhui Li
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kory R. Johnson
- Bioinformatics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mark Bryant
- Division of Veterinary Resources, Office of Research Support, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Abdel G. Elkahloun
- Division of Intramural Research Programs Microarray Core Facility, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Marcelo Amar
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alan T. Remaley
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ranil De Silva
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John M. Hallenbeck
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (JAQ); (JMH)
| | - Jacqueline A. Quandt
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (JAQ); (JMH)
| |
Collapse
|
31
|
MacGrogan D, Luna-Zurita L, de la Pompa JL. Notch signaling in cardiac valve development and disease. ACTA ACUST UNITED AC 2011; 91:449-59. [PMID: 21563298 DOI: 10.1002/bdra.20815] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 01/20/2011] [Accepted: 02/23/2011] [Indexed: 12/22/2022]
Abstract
The Notch pathway is an intercellular signaling mechanism involved in multiple cell-to-cell communication processes that regulate cell fate specification, differentiation, and tissue patterning during embryogenesis and adulthood. Functional studies in the mouse have shown that a Hey-Bmp2 regulatory circuit restricts Bmp2 expression to presumptive valve myocardium (atrioventricular canal and outflow tract). Likewise, a Notch-Hey-Bmp2 axis represses Bmp2 in the endocardium. During cardiac valve formation, endocardial Notch signaling activates the epithelial-mesenchyme transition (EMT) that will give rise to the cardiac valve primordia. During this process, Notch integrates with myocardially derived signals (Bmp2 or Bmp4) to promote, via Snail1/2 activation a complete, invasive EMT in presumptive valve tissue. In humans, mutations in Notch signaling components are associated with several congenital disorders involving malformed valves, aortic arch, and defective chamber septation. Data suggest that the same embryonic Notch-Hey-Bmp2 regulatory axis is active in the adult valve. This review examines the experimental evidence supporting a role for Notch in heart valve development and homeostasis, and how altered Notch signaling may lead to valve disease in the newborn and adult.
Collapse
Affiliation(s)
- Donal MacGrogan
- Laboratorio de Señalizacion Intercelular, Dpto. de Biología del Desarrollo Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares, Melchor Fernández Almagro 3, Madrid, Spain
| | | | | |
Collapse
|
32
|
The response to valve injury. A paradigm to understand the pathogenesis of heart valve disease. Cardiovasc Pathol 2011; 20:183-90. [DOI: 10.1016/j.carpath.2010.09.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 08/23/2010] [Accepted: 09/09/2010] [Indexed: 12/21/2022] Open
|
33
|
Abstract
The Notch-signaling pathway is involved in multiple processes during vertebrate cardiac development. Cardiomyocyte differentiation, patterning of the different cardiac regions, valve development, ventricular trabeculation, and outflow tract development have all been shown to depend on the activity of specific Notch-signaling elements. From these studies, it becomes obvious that Notch regulates in a cell autonomous or non-cell autonomous manner different signaling pathways, pointing to a role for Notch as a signal coordinator during cardiogenesis. While most of the research has concentrated on Notch signaling in the myocardium, the importance of Notch activity in the cardiac endothelium (endocardium) must not be overlooked. Endocardial Notch activity is crucial for valve and ventricular trabeculae development, two processes that illustrate the role of Notch as a signal coordinator. The importance of Notch signaling in human disease is evident from the discovery that many mutations in components of this pathway segregate in several inherited and acquired disorders. This reflects the fundamental roles that Notch performs during cardiac ontogeny. This review examines the experimental evidence supporting a role for Notch in cardiac development and adult heart homeostasis, and how dysregulated Notch signaling may lead to cardiac disease in the newborn and in the adult.
Collapse
|