1
|
Yao Q, The E, Nedumaran B, Zhai Y, Ao L, Fullerton DA, Meng X. Cathepsin D elevates the fibrocalcific activity in human aortic valve cells through the ERK1/2-Sox9 pathway. Front Cardiovasc Med 2024; 11:1410862. [PMID: 39380629 PMCID: PMC11458440 DOI: 10.3389/fcvm.2024.1410862] [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: 04/01/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
Background Human Aortic valve interstitial cells (AVICs) from calcific aortic valve disease (CAVD)-affected valves exhibit elevated fibrocalcific activity although the underlying mechanism remains incompletely understood. This study aimed to identify endogenous factors that promote aortic valve fibrocalcification. Methods and results Proteomic analysis found increased cathepsin D levels in AVICs from CAVD-affected valves compared to AVICs from normal valves, and this finding was validated by immunoblotting. ELISA assay identified exacerbated release of cathepsin D by AVICs of diseased valves. Recombinant human cathepsin D upregulated the expression of runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP), collagen I and collagen IV in human AVICs, resulting in the deposition of calcium and collagen. Blocking of the ERK1/2-Sox9 signaling pathway markedly reduced the pro-fibrocalcific effect of cathepsin D. Moreover, normal AVICs express and release greater levels of cathepsin D when exposed to soluble matrilin 2. Knockdown of cathepsin D attenuated the fibrocalcific response induced by soluble matrilin 2. Conclusion AVICs of diseased aortic valves produce and release greater levels of cathepsin D that exerts a pro-fibrocalcific effect on AVICs through the ERK1/2-Sox9 pathway. Soluble matrilin 2 up-regulates cathepsin D to elevate AVIC fibrocalcific activity. Over-expression of cathepsin D in the aortic valve may enhance the pathobiological activities in AVICs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xianzhong Meng
- Department of Surgery, University of Colorado, Denver, CO, United States
| |
Collapse
|
2
|
Deng XS, Meng X, Fullerton D, Stone M, Iguidbashian J, Jaggers J. Complement Cross Talks With H-K-ATPase to Upregulate Runx2 in Human Aortic Valve Interstitial Cells. J Surg Res 2023; 286:118-126. [PMID: 36822134 PMCID: PMC10120867 DOI: 10.1016/j.jss.2022.12.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/16/2022] [Accepted: 12/25/2022] [Indexed: 02/23/2023]
Abstract
INTRODUCTION Calcific aortic valve disease (CAVD) is a slowly progressive fibro-calcific valve leaflet disorder. The underlying pathophysiology is complex and not yet well understood. Complement is known to play a role in the pathogenesis of CAVD by upregulating Runx2 to induce profibrogenic change in human aortic valve interstitial cells (AVICs). Furthermore, H-K-ATPase has independently been shown to induce tissue calcification. Therefore, we hypothesized that complement cross talks with H-K-ATPase to upregulate Runx2 in human AVICs. MATERIALS AND METHODS Human AVICs were isolated from normal and calcified aortic valves. Cells were treated with a variation of complement, H-K-ATPase, or ERK1/2 inhibitors. H-K-ATPase and its association with complement in AVICs were investigated by reverse transcriptase-polymerase chain reaction, immunofluorescence, and Western blot. RESULTS Calcified human AVICs expressed significantly higher H-K-ATPase level than normal human AVICs. Presence of complement C3 with H-K-ATPase is found in AVICs after complement treatment. Complement induced both H-K-ATPase and Runx2 expression in AVICs, which was associated with increased phosphorylation of ERK1/2 and its downstream molecule p-70 S6. Pharmacological inhibition of either H-K-ATPase or Erk1/2 abolished complement-induced Runx2 expression. CONCLUSIONS These findings indicate that complement cross talks with H-K-ATPase to upregulate Runx2 in human AVICs by activation of ERK1/2 signaling pathways. The study revealed the potential role of H-K-ATPase in the pathogenesis of CAVD and therapeutically targeting either complement system or H-K-ATPase may limit the development of CAVD.
Collapse
Affiliation(s)
- Xin-Sheng Deng
- Cardiothoracic Surgery, University of Colorado, Children's Hospital Colorado, Aurora, Colorado; Cardiothoracic Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Xianzhong Meng
- Cardiothoracic Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - David Fullerton
- Cardiothoracic Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Matthew Stone
- Cardiothoracic Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - John Iguidbashian
- Cardiothoracic Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - James Jaggers
- Cardiothoracic Surgery, University of Colorado, Children's Hospital Colorado, Aurora, Colorado; Cardiothoracic Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
3
|
Song L, Wang Y, Feng Y, Peng H, Wang C, Duan J, Liu K, Shen X, Gu W, Qi Y, Jin S, Pang L. Bioinformatics-Based Identification of CircRNA-MicroRNA-mRNA Network for Calcific Aortic Valve Disease. Genet Res (Camb) 2023; 2023:8194338. [PMID: 37234568 PMCID: PMC10208756 DOI: 10.1155/2023/8194338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 02/13/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Background Calcific aortic valve disease (CAVD) is the most common native valve disease. Valvular interstitial cell (VIC) osteogenic differentiation and valvular endothelial cell (VEC) dysfunction are key steps in CAVD progression. Circular RNA (circRNAs) is involved in regulating osteogenic differentiation with mesenchymal cells and is associated with multiple disease progression, but the function of circRNAs in CAVD remains unknown. Here, we aimed to investigate the effect and potential significance of circRNA-miRNA-mRNA networks in CAVD. Methods Two mRNA datasets, one miRNA dataset, and one circRNA dataset of CAVD downloaded from GEO were used to identify DE-circRNAs, DE-miRNAs, and DE-mRNAs. Based on the online website prediction function, the common mRNAs (FmRNAs) for constructing circRNA-miRNA-mRNA networks were identified. GO and KEGG enrichment analyses were performed on FmRNAs. In addition, hub genes were identified by PPI networks. Based on the expression of each data set, the circRNA-miRNA-hub gene network was constructed by Cytoscape (version 3.6.1). Results 32 DE-circRNAs, 206 DE-miRNAs, and 2170 DE-mRNAs were identified. Fifty-nine FmRNAs were obtained by intersection. The KEGG pathway analysis of FmRNAs was enriched in pathways in cancer, JAK-STAT signaling pathway, cell cycle, and MAPK signaling pathway. Meanwhile, transcription, nucleolus, and protein homodimerization activity were significantly enriched in GO analysis. Eight hub genes were identified based on the PPI network. Three possible regulatory networks in CAVD disease were obtained based on the biological functions of circRNAs including: hsa_circ_0026817-hsa-miR-211-5p-CACNA1C, hsa_circ_0007215-hsa-miR-1252-5p-MECP2, and hsa_circ_0007215-hsa-miR-1343-3p- RBL1. Conclusion The present bionformatics analysis suggests the functional effect for the circRNA-miRNA-mRNA network in CAVD pathogenesis and provides new targets for therapeutics.
Collapse
Affiliation(s)
- Linghong Song
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Yubing Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Yufei Feng
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Hao Peng
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Chengyan Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Juncang Duan
- Department of Cardiology, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| | - Kejian Liu
- Department of Cardiology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xihua Shen
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Australia
| | - Yan Qi
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Department of Pathology, Central People's Hospital of Zhanjiang and Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Shan Jin
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Lijuan Pang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Department of Pathology, Central People's Hospital of Zhanjiang and Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
4
|
Xiao F, Pan H, Yang D, Wang R, Wu B, Shao Y, Zhou B. Identification of TNFα-mediated inflammation as potential pathological marker and therapeutic target for calcification progress of congenital bicuspid aortic valve. Eur J Pharmacol 2023; 951:175783. [PMID: 37172927 DOI: 10.1016/j.ejphar.2023.175783] [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: 02/27/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUD Congenital bicuspid aortic valve (cBAV) develops calcification and stenotic obstruction early compared with degenerative tricuspid aortic valve (dTAV), which requires surgical intervention. Here we report a comparative study of patients with cBAV or dTAV to identify risk factors associated with the rapid development of calcified bicuspid valves. METHODS A total of 69 aortic valves (24 dTAV and 45 cBAV) were collected at the time of surgical aortic valve replacement for comparative clinical characteristics. Ten samples were randomly selected from each group for histology, pathology, and inflammatory factors expression and comparison analyses. OM-induced calcification in porcine aortic valve interstitial cell cultures were prepared for illustrating the underlying molecular mechanisms about calcification progress of cBAV and dTAV. RESULTS We found that cBAV patients have increased cases of aortic valve stenosis compared with dTAV patients. Histopathological examinations revealed increased collagens deposition, neovascularization and infiltrations by inflammatory cells, especially T-lymphocytes and macrophages. We identified that tumor necrosis factor α (TNFα) and its regulated inflammatory cytokines are upregulated in cBAV. Further in vitro study indicated that TNFα-NFκB and TNFα-GSK3β pathway accelerate aortic valve interstitial cells calcification, while inhibition of TNFα significantly delays this process. CONCLUSION The finding of intensified TNFα-mediated inflammation in the pathological cBAV advocates the inhibition of TNFα as a potential treatment for patients with cBAV by alleviating the progress of inflammation-induced valve damage and calcification.
Collapse
Affiliation(s)
- Feng Xiao
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China.
| | - Haotian Pan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Di Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Ruxing Wang
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Bingruo Wu
- Departments of Genetics, Pediatrics and Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Yongfeng Shao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Bin Zhou
- Departments of Genetics, Pediatrics and Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| |
Collapse
|
5
|
Wu L, Huang K, Li Q, Wu H, Gao Y, Xu X, Liu X, Han L. Crosstalk between myofibroblasts and macrophages: A regulative factor of valvular fibrosis in calcific aortic valve disease. Cell Biol Int 2023; 47:754-767. [PMID: 36542640 DOI: 10.1002/cbin.11980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
Inflammation and fibrosis are highly correlated with the progression of calcific aortic valve disease (CAVD). As one of the differentiated forms of valvular interstitial cells, myofibroblasts play a critical role in CAVD's development as do macrophages. Although numerous studies have been conducted on them separately, their communication and interaction remain unclear. We used porcine aortic valves to isolate valve interstitial cells (VICs). VICs were induced to differentiate into myofibroblasts by transforming growth factor-β1 (TGF-β1). After successful activation was determined, the myofibroblast-conditioned medium (CM) was collected and used to act on RAW264.7, a macrophage cell line. A migration and adhesion assay estimated the recruitment capability of myofibroblasts on macrophages. We used flow cytometry, quantitative polymerase chain reaction (qPCR), and Western blot analysis to investigate myofibroblasts' polarity promotion function in macrophages. Finally, we used macrophage-CM on VICs to explore the differentiation induction function of polarized macrophages. Myofibroblast marker alpha-smooth muscle actin and M2 macrophage marker CD163 were detected as upregulated in CAVD patients, and their expression has a certain correlation. The Smad3/HA/CD44 axis activated the differentiation of myofibroblasts by Western blot. The myofibroblast-CM can promote chemotaxis and adhesion of macrophages through protein kinase B/chemokine (C-C motif) ligand5 and Smad3/HA/CD44, respectively. Hyaluronic acid (HA) inside the myofibroblast-CM stimulates macrophages to polarize into M2 macrophages. In turn, M2 macrophage-CM has the promotive ability to activate myofibroblasts but fails to induce the osteoblast differentiation of VICs directly. The crosstalk between myofibroblasts and macrophages causes the excessive activation of myofibroblasts. This positive feedback loop may play a vital role in CAVD progression.
Collapse
Affiliation(s)
- Lujia Wu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Kai Huang
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qin Li
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hao Wu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuan Gao
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiangyang Xu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaohong Liu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lin Han
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
6
|
Qiu M, Lu Y, Li J, Gu J, Ji Y, Shao Y, Kong X, Sun W. Interaction of SOX5 with SOX9 promotes warfarin-induced aortic valve interstitial cell calcification by repressing transcriptional activation of LRP6. J Mol Cell Cardiol 2021; 162:81-96. [PMID: 34520801 DOI: 10.1016/j.yjmcc.2021.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 09/05/2021] [Accepted: 09/08/2021] [Indexed: 11/18/2022]
Abstract
Calcific aortic valve disease (CAVD) is an important health burden due to its increasing prevalence and lack of available approaches. Osteogenic transdifferentiation of aortic valve interstitial cells (AVICs) contributes to valve calcification. SRY-related HMG-box transcription factor 5 (SOX5) is essential for cartilage development. Whether SOX5 is involved in AVIC calcification has not been determined. This study aimed to explore the role of SOX5 in warfarin-induced AVIC calcification. Immunostaining showed decreased SOX5 in human calcific AV and warfarin induced mouse calcific AV tissues compared with human noncalcific AV and control mouse AV tissues. In calcific human AVICs (hAVICs) and porcine AVICS (pAVICs), both knockdown and overexpression of SOX5 inhibited calcium deposition and osteogenic marker gene expression. Protein expression assays and ChIP assays showed that overexpression of SOX5 led to increased recruitment of SOX5 to the SOX9 promoter and resulted in increased mRNA and protein expression of SOX9. Coimmunoprecipitation and immunofluorescence showed that SOX5 binds to SOX9 with its HMG domain in nucleus. Blue Native PAGE showed overexpression of SOX5 led to multimeric complex formation of SOX5 and resulted in decreased binding of SOX5 to SOX9 similar to the results of knockdown of SOX5. Further ChIP and western blotting assays showed that both knockdown and overexpression of SOX5 resulted in SOX9 initiating transcription of anti-calcific gene LRP6 in warfarin-treated pAVICs. Knockdown of LRP6 rescues the anti-calcification effect of SOX5 overexpression. We found that both loss and gain of function of SOX5 lead to the same phenotype: decreased warfarin induced calcification. The stoichiometry of SOX5 is crucial for cooperation with SOX9, SOX9 nuclear localization and subsequent binding of SOX9 to LRP6 promoter. These results suggest that SOX5 is a potential target for the development of anti-calcification therapy.
Collapse
Affiliation(s)
- Ming Qiu
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing 210009, PR China; Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China
| | - Yan Lu
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China
| | - Junhan Li
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China
| | - Jia Gu
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China
| | - Yue Ji
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China
| | - Yongfeng Shao
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China
| | - Xiangqing Kong
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing 210009, PR China; Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing 211166, China.
| | - Wei Sun
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China.
| |
Collapse
|
7
|
Lima VSS, Mariano DOC, Vigerelli H, Janussi SC, Baptista TVL, Claudino MA, Pimenta DC, Sciani JM. Effects of Kynurenic Acid on the Rat Aorta Ischemia-Reperfusion Model: Pharmacological Characterization and Proteomic Profiling. Molecules 2021; 26:2845. [PMID: 34064778 PMCID: PMC8150825 DOI: 10.3390/molecules26102845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/28/2022] Open
Abstract
Kynurenic acid (KYNA) is derived from tryptophan, formed by the kynurenic pathway. KYNA is being widely studied as a biomarker for neurological and cardiovascular diseases, as it is found in ischemic conditions as a protective agent; however, little is known about its effect after ischemia-reperfusion in the vascular system. We induced ischemia for 30 min followed by 5 min reperfusion (I/R) in the rat aorta for KYNA evaluation using functional assays combined with proteomics. KYNA recovered the exacerbated contraction induced by phenylephrine and relaxation induced by acetylcholine or sodium nitroprussiate in the I/R aorta, with vessel responses returning to values observed without I/R. The functional recovery can be related to the antioxidant activity of KYNA, which may be acting on the endothelium-injury prevention, especially during reperfusion, and to proteins that regulate neurotransmission and cell repair/growth, expressed after the KYNA treatment. These proteins interacted in a network, confirming a protein profile expression for endothelium and neuron repair after I/R. Thus, the KYNA treatment had the ability to recover the functionality of injured ischemic-reperfusion aorta, by tissue repairing and control of neurotransmitter release, which reinforces its role in the post-ischemic condition, and can be useful in the treatment of such disease.
Collapse
Affiliation(s)
- Viviane Soares Souza Lima
- Laboratório Multidisciplinar em Pesquisa, Universidade São Francisco, 12916-900 Bragança Paulista, Brazil; (V.S.S.L.); (S.C.J.); (T.V.L.B.); (M.A.C.)
| | | | - Hugo Vigerelli
- Laboratório de Genética, Instituto Butantan, 05503-900 São Paulo, Brazil;
| | - Sabrina Cardoso Janussi
- Laboratório Multidisciplinar em Pesquisa, Universidade São Francisco, 12916-900 Bragança Paulista, Brazil; (V.S.S.L.); (S.C.J.); (T.V.L.B.); (M.A.C.)
| | - Thayz Vanalli Lima Baptista
- Laboratório Multidisciplinar em Pesquisa, Universidade São Francisco, 12916-900 Bragança Paulista, Brazil; (V.S.S.L.); (S.C.J.); (T.V.L.B.); (M.A.C.)
| | - Mário Angelo Claudino
- Laboratório Multidisciplinar em Pesquisa, Universidade São Francisco, 12916-900 Bragança Paulista, Brazil; (V.S.S.L.); (S.C.J.); (T.V.L.B.); (M.A.C.)
| | - Daniel Carvalho Pimenta
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, 05503-900 São Paulo, Brazil; (D.O.C.M.); (D.C.P.)
| | - Juliana Mozer Sciani
- Laboratório Multidisciplinar em Pesquisa, Universidade São Francisco, 12916-900 Bragança Paulista, Brazil; (V.S.S.L.); (S.C.J.); (T.V.L.B.); (M.A.C.)
| |
Collapse
|
8
|
Deng XS, Meng X, Fullerton D, Stone M, Jaggers J. Complement Upregulates Runx-2 to Induce Profibrogenic Change in Aortic Valve Interstitial Cells. Ann Thorac Surg 2021; 112:1962-1972. [PMID: 33545156 DOI: 10.1016/j.athoracsur.2020.12.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/06/2020] [Accepted: 12/14/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Calcium accumulation and fibrotic activities are principal mechanisms for calcific aortic valve disease (CAVD). Active complement products are observed in human stenotic aortic valves. Runt-related transcription factor 2 (Runx-2) is involved in tissue calcification. We hypothesized that complement upregulates Runx-2 to induce profibrogenic change in human aortic valve interstitial cells (AVICs). METHODS AVICs were isolated from 6 normal and 6 CAVD donor valves. Cells were treated with complement cocktails. Profibrogenic activities and associated signaling molecules were analyzed by Western blot assay and collagen staining. RESULTS Complement time and dose dependently enhanced profibrogenic activities in AVICs, and complement exposure also induced total collagen deposition in AVICs. Complement-induced profibrogenic responses were associated with increased Runx-2 expression and phosphorylation of extracellular signal-regulated kinase 1 and 2 (ERK1/2). Genetic silencing of Runx-2 decreased both matrix metalloproteinase 9 (MMP-9) and collagen I levels. Pharmacological inhibition of ERK1/2 decreased complement-mediated MMP-9, collagen I, and Runx-2 expression as well as total collagen deposition in human AVICs. Further, treating AVICs with heat-deactivated complement resulted in reduced MMP-9, collagen I, and Runx-2 levels compared with active complement treatment. CONCLUSIONS Complement induced profibrogenic activities in AVICs by activation of ERK1/2-mediated Runx-2 signaling pathways. This study demonstrates a potential role for complement-mediated CAVD pathogenesis, establishing a possible therapeutic target to limit CAVD progression.
Collapse
Affiliation(s)
- Xin-Sheng Deng
- Cardiothoracic Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Xianzhong Meng
- Cardiothoracic Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - David Fullerton
- Cardiothoracic Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Matthew Stone
- Cardiothoracic Surgery, Children's Hospital Colorado, University of Colorado, Aurora, Colorado
| | - James Jaggers
- Cardiothoracic Surgery, Children's Hospital Colorado, University of Colorado, Aurora, Colorado.
| |
Collapse
|
9
|
Jarrett MJ, Houk AK, McCuistion PE, Weyant MJ, Reece TB, Meng X, Fullerton DA. Wnt Signaling Mediates Pro-Fibrogenic Activity in Human Aortic Valve Interstitial Cells. Ann Thorac Surg 2020; 112:519-525. [PMID: 33189669 DOI: 10.1016/j.athoracsur.2020.08.068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 07/27/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Proinflammatory activation of toll-like receptor-4 (TLR4) drives phenotypic changes in aortic valve interstitial cells (AVICs) and produces a fibrogenic phenotype that mediates valvular fibrosis and contributes to aortic stenosis. Prior work identified upregulated Wnt signaling in AVICs taken from valves affected by aortic stenosis. Our purpose was to determine the contribution of Wnt signaling to TLR4-dependent fibrogenic activity in isolated human AVICs. METHODS Human AVICs were isolated from hearts explanted for cardiac transplantation (N = 4). To test whether Wnt signaling contributed to TLR4-dependent fibrogenic activity, AVICs were treated with Wnt inhibitor (Dkk1) prior to TLR4 activation (LPS) and fibrogenic markers assessed. To determine the mediator of TLR4-to-Wnt signaling, expression of the key Wnt ligand, Wnt3a, was assessed after TLR4 activation and neutralizing antibodies confirmed the identity of the mediator. Fibrogenic activity was assessed after AVICs were treated with recombinant Wnt3a. Statistics were by analysis of variance (P < .05). RESULTS TLR4 activation upregulated in vitro collagen deposition, type IV collagen and MMP2 expression, and Dkk1 inhibited these responses (P < .05). Expression of Wnt3a was upregulated after TLR4 activation (P < .05). Anti-Wnt3a neutralizing antibodies abrogated TLR4-dependent type IV collagen and MMP2 expression (P < .05). Wnt3a upregulated type IV collagen and MMP2 expression independent of TLR4 activation (P < .05). CONCLUSIONS This study found that TLR4-dependent fibrogenic activity was mediated through Wnt signaling. The mediator of profibrogenic TLR4-to-Wnt signaling was a key Wnt ligand, Wnt3a. The abrogation of TLR4-induced fibrogenic activity in human AVICs by Wnt blockade illustrates a potential therapeutic role for Wnt inhibition in treatment and/or prevention of aortic stenosis.
Collapse
Affiliation(s)
- Michael J Jarrett
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado.
| | - Anna K Houk
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Peyton E McCuistion
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Michael J Weyant
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - T Brett Reece
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Xianzhong Meng
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - David A Fullerton
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
10
|
Amadio P, Zarà M, Sandrini L, Ieraci A, Barbieri SS. Depression and Cardiovascular Disease: The Viewpoint of Platelets. Int J Mol Sci 2020; 21:E7560. [PMID: 33066277 PMCID: PMC7589256 DOI: 10.3390/ijms21207560] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Depression is a major cause of morbidity and low quality of life among patients with cardiovascular disease (CVD), and it is now considered as an independent risk factor for major adverse cardiovascular events. Increasing evidence indicates not only that depression worsens the prognosis of cardiac events, but also that a cross-vulnerability between the two conditions occurs. Among the several mechanisms proposed to explain this interplay, platelet activation is the more attractive, seeing platelets as potential mirror of the brain function. In this review, we dissected the mechanisms linking depression and CVD highlighting the critical role of platelet behavior during depression as trigger of cardiovascular complication. In particular, we will discuss the relationship between depression and molecules involved in the CVD (e.g., catecholamines, adipokines, lipids, reactive oxygen species, and chemokines), emphasizing their impact on platelet activation and related mechanisms.
Collapse
Affiliation(s)
- Patrizia Amadio
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| | - Marta Zarà
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| | - Leonardo Sandrini
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| | - Alessandro Ieraci
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy;
| | - Silvia Stella Barbieri
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| |
Collapse
|
11
|
Jenke A, Kistner J, Saradar S, Chekhoeva A, Yazdanyar M, Bergmann AK, Rötepohl MV, Lichtenberg A, Akhyari P. Transforming growth factor-β1 promotes fibrosis but attenuates calcification of valvular tissue applied as a three-dimensional calcific aortic valve disease model. Am J Physiol Heart Circ Physiol 2020; 319:H1123-H1141. [PMID: 32986963 DOI: 10.1152/ajpheart.00651.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Calcific aortic valve disease (CAVD) is characterized by valvular fibrosis and calcification and driven by differentiating valvular interstitial cells (VICs). Expression data from patient biopsies suggest that transforming growth factor (TGF)-β1 is implicated in CAVD pathogenesis. However, CAVD models using isolated VICs failed to deliver clear evidence on the role of TGF-β1. Thus, employing cultures of aortic valve leaflets, we investigated effects of TGF-β1 in a tissue-based three-dimensional (3-D) CAVD model. We found that TGF-β1 induced phosphorylation of Mothers against decapentaplegic homolog (SMAD) 3 and expression of SMAD7, indicating effective downstream signal transduction in valvular tissue. Thus, TGF-β1 increased VIC contents of rough endoplasmic reticulum, Golgi, and secretory vesicles as well as tissue levels of RNA and protein. In addition, TGF-β1 raised expression of proliferation marker cyclin D1, attenuated VIC apoptosis, and upregulated VIC density. Moreover, TGF-β1 intensified myofibroblastic VIC differentiation as evidenced by increased α-smooth muscle actin and collagen type I along with diminished vimentin expression. In contrast, TGF-β1 attenuated phosphorylation of SMAD1/5/8 and upregulation of β-catenin while inhibiting osteoblastic VIC differentiation as revealed by downregulation of osteocalcin expression, alkaline phosphatase activity, and extracellular matrix incorporation of hydroxyapatite. Collectively, these effects resulted in blocking of valvular tissue calcification and associated disintegration of collagen fibers. Instead, TGF-β1 induced development of fibrosis. Overall, in a tissue-based 3-D CAVD model, TGF-β1 intensifies expressional and proliferative activation along with myofibroblastic differentiation of VICs, thus triggering dominant fibrosis. Simultaneously, by inhibiting SMAD1/5/8 activation and canonical Wnt/β-catenin signaling, TGF-β1 attenuates osteoblastic VIC differentiation, thus blocking valvular tissue calcification. These findings question a general phase-independent CAVD-promoting role of TGF-β1.NEW & NOTEWORTHY Employing aortic valve leaflets as a tissue-based three-dimensional disease model, our study investigates the role of transforming growth factor (TGF)-β1 in calcific aortic valve disease pathogenesis. We find that, by activating Mothers against decapentaplegic homolog 3, TGF-β1 intensifies expressional and proliferative activation along with myofibroblastic differentiation of valvular interstitial cells, thus triggering dominant fibrosis. Simultaneously, by inhibiting activation of Mothers against decapentaplegic homolog 1/5/8 and canonical Wnt/β-catenin signaling, TGF-β1 attenuates apoptosis and osteoblastic differentiation of valvular interstitial cells, thus blocking valvular tissue calcification. These findings question a general phase-independent calcific aortic valve disease-promoting role of TGF-β1.
Collapse
Affiliation(s)
- Alexander Jenke
- Department of Cardiac Surgery, Düsseldorf University Hospital, Düsseldorf, Germany.,Research Group Experimental Surgery, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Kistner
- Department of Cardiac Surgery, Düsseldorf University Hospital, Düsseldorf, Germany.,Research Group Experimental Surgery, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sarah Saradar
- Department of Cardiac Surgery, Düsseldorf University Hospital, Düsseldorf, Germany.,Research Group Experimental Surgery, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Agunda Chekhoeva
- Department of Cardiac Surgery, Düsseldorf University Hospital, Düsseldorf, Germany.,Research Group Experimental Surgery, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mariam Yazdanyar
- Department of Cardiac Surgery, Düsseldorf University Hospital, Düsseldorf, Germany.,Research Group Experimental Surgery, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ann Kathrin Bergmann
- Core Facility for Electron Microscopy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Melanie Vera Rötepohl
- Department of Cardiac Surgery, Düsseldorf University Hospital, Düsseldorf, Germany.,Research Group Experimental Surgery, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Artur Lichtenberg
- Department of Cardiac Surgery, Düsseldorf University Hospital, Düsseldorf, Germany.,Research Group Experimental Surgery, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Payam Akhyari
- Department of Cardiac Surgery, Düsseldorf University Hospital, Düsseldorf, Germany.,Research Group Experimental Surgery, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
12
|
Li BB, Chen ZY, Jiang N, Guo S, Yang JQ, Chai SB, Yan HF, Sun PM, Hu G, Zhang T, Xu BX, Sun HW, Zhou JL, Yang HM, Cui Y. Simulated microgravity significantly altered metabolism in epidermal stem cells. In Vitro Cell Dev Biol Anim 2020; 56:200-212. [PMID: 32198676 PMCID: PMC7186248 DOI: 10.1007/s11626-020-00435-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/08/2020] [Indexed: 12/23/2022]
Abstract
Simulated microgravity can significantly affect various cell types and multiple systems of the human body, such as cardiovascular system, skeletal muscle system, and immune system, and is known to cause anemia and loss of electrolyte and fluids. Epidermal stem cells (EpSCs) were cultured in a rotary cell culture system (RCCS) bioreactor to simulate microgravity. The metabolites of EpSCs were identified by liquid chromatography-mass spectrometry (LC-MS). Compared with normal gravity (NG) group, a total of 57 different metabolites of EpSCs were identified (P < 0.05, VIP > 1), including lipids and lipid-like molecules (51 molecules), amino acids (5 molecules), nucleosides, nucleotides, and analogues (1 molecule). According to the partial least squares discriminant analysis (PLS-DA) score plot, a VIP > 1 and P < 0.05 were obtained for the 57 different metabolites, of which 23 molecules were significantly downregulated and 34 were significantly upregulated in simulated microgravity (SMG) group. These results showed that SMG has a significant impact on different pathways, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis indicated that multiple pathways were involved, mainly the amino acid metabolism pathway, lipid metabolism pathway, membrane transport pathway, and cell growth and death pathways. Thus, the metabolic profile of EpSCs was changed under SMG. Exploring the metabolic profile of EpSCs would be helpful to further understand the growth characteristics of EpSCs under SMG, which will provide a new approach to explore the metabolomics mechanism of stress injury and repair trauma under SMG.
Collapse
Affiliation(s)
- Bin-Bin Li
- Department of General Surgery, PLA 306 Clinical Hospital of Anhui Medical University, Beijing, 230000 China
| | - Zheng-Yang Chen
- Department of General Surgery, PLA 306 Teaching Hospital of Peking University Health Science Center, Beijing, 100101 China
| | - Nan Jiang
- The Center for Hepatopancreatobiliary Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218 China
| | - Song Guo
- Department of General Surgery, PLA 306 Teaching Hospital of Peking University Health Science Center, Beijing, 100101 China
| | - Jia-Qi Yang
- Department of General Surgery, PLA 306 Teaching Hospital of Peking University Health Science Center, Beijing, 100101 China
| | - Shao-Bin Chai
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Hong-Feng Yan
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Pei-Ming Sun
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Gang Hu
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Tao Zhang
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Bing-Xin Xu
- Medicine and Experimental Research Center, 306 Hospital of PLA, Beijing, 100101 China
| | - Hong-Wei Sun
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Jin-Lian Zhou
- Department of Pathology, 306 Hospital of PLA, Beijing, 100101 China
| | - He-Ming Yang
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| | - Yan Cui
- Department of General Surgery, 306 Hospital of PLA, Beijing, 100101 China
| |
Collapse
|
13
|
Yao Q, The E, Ao L, Zhai Y, Osterholt MK, Fullerton DA, Meng X. TLR4 Stimulation Promotes Human AVIC Fibrogenic Activity through Upregulation of Neurotrophin 3 Production. Int J Mol Sci 2020; 21:ijms21041276. [PMID: 32074942 PMCID: PMC7072994 DOI: 10.3390/ijms21041276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is a chronic inflammatory disease that manifests as progressive valvular fibrosis and calcification. An inflammatory milieu in valvular tissue promotes fibrosis and calcification. Aortic valve interstitial cell (AVIC) proliferation and the over-production of the extracellular matrix (ECM) proteins contribute to valvular thickening. However, the mechanism underlying elevated AVIC fibrogenic activity remains unclear. Recently, we observed that AVICs from diseased aortic valves express higher levels of neurotrophin 3 (NT3) and that NT3 exerts pro-osteogenic and pro-fibrogenic effects on human AVICs. HYPOTHESIS Pro-inflammatory stimuli upregulate NT3 production in AVICs to promote fibrogenic activity in human aortic valves. METHODS AND RESULTS AVICs were isolated from normal human aortic valves and were treated with lipopolysaccharide (LPS, 0.20 µg/mL). LPS induced TLR4-dependent NT3 production. This effect of LPS was abolished by inhibition of the Akt and extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) pathways. The stimulation of TLR4 in human AVICs with LPS resulted in a greater proliferation rate and an upregulated production of matrix metallopeptidases-9 (MMP-9) and collagen III, as well as augmented collagen deposition. Recombinant NT3 promoted AVIC proliferation in a tropomyosin receptor kinase (Trk)-dependent fashion. The neutralization of NT3 or the inhibition of Trk suppressed LPS-induced AVIC fibrogenic activity. CONCLUSIONS The stimulation of TLR4 in human AVICs upregulates NT3 expression and promotes cell proliferation and collagen deposition. The NT3-Trk cascade plays a critical role in the TLR4-mediated elevation of fibrogenic activity in human AVICs. Upregulated NT3 production by endogenous TLR4 activators may contribute to aortic valve fibrosis associated with CAVD progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xianzhong Meng
- Correspondence: ; Tel.: +303-724-6303; Fax: +303-724-6330
| |
Collapse
|
14
|
Jarrett MJ, Yao Q, Venardos N, Weyant MJ, Reece TB, Meng X, Fullerton DA. Simvastatin down-regulates osteogenic response in cultured human aortic valve interstitial cells. J Thorac Cardiovasc Surg 2019; 161:e261-e271. [PMID: 31924353 DOI: 10.1016/j.jtcvs.2019.10.081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/28/2019] [Accepted: 10/11/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Aortic valve interstitial cells have been implicated in the pathogenesis of aortic stenosis. In response to proinflammatory stimuli, aortic valve interstitial cells undergo an osteogenic phenotypic change. The purpose of this study was to determine whether the anti-inflammatory effects of statins prevent osteogenic activity in cultured aortic valve interstitial cells. METHODS Human aortic valve interstitial cells were isolated from hearts explanted for cardiac transplantation. To test whether simvastatin down-regulates TLR4-induced osteogenic response, aortic valve interstitial cells were treated with simvastatin with and without TLR4 agonist lipopolysaccharide (LPS), and osteogenic markers were measured. Simvastatin's influence on in vitro calcium deposition was assessed by alizarin red staining. Knockdown of postreceptor signaling proteins (MyD88 and TRIF) was performed to determine which of 2 TLR4-associated pathways mediates the osteogenic response. Expression levels of TLR4-induced nuclear factor kappa light chain enhancer of activated B cells (NF-κB) and TLR4 expression were assessed after treatment with simvastatin. Statistical testing was done by analysis of variance (P < .05). RESULTS Simvastatin decreased LPS-induced ALP and Runx2 expression and inhibited in vitro calcium deposition in aortic valve interstitial cells. Knockdown of MyD88 and TRIF attenuated the osteogenic response. Simvastatin attenuated TLR4-dependent NF-κB signaling and down-regulated TLR4 levels. CONCLUSIONS Simvastatin prevented TLR4-induced osteogenic phenotypic changes in isolated aortic valve interstitial cells via down-regulation of TLR4 and inhibition of NF-κB signaling. These data offer mechanistic insight into a possible therapeutic role for simvastatin in the prevention of aortic stenosis.
Collapse
Affiliation(s)
- Michael J Jarrett
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colo
| | - Qingzhou Yao
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colo
| | - Neil Venardos
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colo
| | - Michael J Weyant
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colo
| | - T Brett Reece
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colo
| | - Xianzhong Meng
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colo
| | - David A Fullerton
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colo.
| |
Collapse
|
15
|
Charitakis E, Karlsson LO, Papageorgiou JM, Walfridsson U, Carlhäll CJ. Echocardiographic and Biochemical Factors Predicting Arrhythmia Recurrence After Catheter Ablation of Atrial Fibrillation-An Observational Study. Front Physiol 2019; 10:1215. [PMID: 31632285 PMCID: PMC6783634 DOI: 10.3389/fphys.2019.01215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/06/2019] [Indexed: 11/13/2022] Open
Abstract
Background: RFA is a well-established treatment for symptomatic patients with AF. However, the success rate of a single procedure is low. We aimed to investigate the association between the risk of recurrence of atrial fibrillation (AF) after a single radiofrequency ablation (RFA) procedure and cardiac neurohormonal function, left atrial (LA) mechanical function as well as proteins related to inflammation, fibrosis, and apoptosis. Methods and Results: We studied 189 patients undergoing RFA between January 2012 and April 2014, with a follow-up period of 12 months. A logistic regression analysis was performed to investigate the association between pre-ablation LA emptying fraction (LAEF), MR-proANP, Caspase-8 (CASP8), Neurotrophin-3 (NT3), and the risk for recurrence of AF after a single RFA procedure. 119 (63.0%) patients had a recurrence during a mean follow-up of 402 ± 73 days. An increased risk of recurrence was associated with: Elevated MR-proANP (fourth quartile vs. first quartile: HR, 2.80 (95% CI, 1.14–6.90]; P = 0.025); Low LAEF (fourth quartile vs. first quartile: hazard ratio [HR], 2.41 [95% CI, 1.01–5.79]; P = 0.045); Elevated CASP8 (fourth quartile vs. first quartile: HR 12.198 95% CI 2.216–67.129; P = 0.004); Elevated NT-3 (fourth quartile vs. first quartile: HR 7.485 95% CI 1.353–41.402; P = 0.021). In a receiver operating characteristic curve analysis, the combination of MR-proANP, CASP8, and NT3 produced an area under the curve of 0.819; CI 95% (0.710–0.928). Conclusions: Patients with better LA mechanical function and lower levels of atrial neurohormones as well as of proteins related to fibrosis and apoptosis, have a better outcome after an RFA procedure. Unique identifier: No. NCT01553045 (https://clinicaltrials.gov/ct2/show/NCT01553045?term=NCT01553045&rank=1).
Collapse
Affiliation(s)
- Emmanouil Charitakis
- Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Lars O Karlsson
- Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Joanna-Maria Papageorgiou
- Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Ulla Walfridsson
- Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Carl-Johan Carlhäll
- Division of Cardiovascular Medicine and CMIV, Linköping University, Linköping, Sweden.,Department of Clinical Physiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
16
|
The role of neurotrophins in psychopathology and cardiovascular diseases: psychosomatic connections. J Neural Transm (Vienna) 2019; 126:265-278. [PMID: 30767081 PMCID: PMC6449302 DOI: 10.1007/s00702-019-01973-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/16/2019] [Indexed: 12/18/2022]
Abstract
Cardiovascular (CV) diseases and mood disorders are common public health problems worldwide. Their connections are widely studied, and the role of neurotrophins (NTs) is already supposed in both conditions. However, data in the literature of clinical aspects are sometimes controversial and no reviews are available describing possible associations between CV risk and mood disorders based on NTs. The mostly studied NT is brain-derived neurotrophic factor (BDNF). Decreased level of BDNF is observed in depression and its connection to hypertension has also been demonstrated with affecting the arterial baroreceptors, renin–angiotensin system and endothelial nitric oxide synthase. BDNF was also found to be the predictor of CV outcome in different patient populations. Other types of human NT-s, such as nerve growth factor, neurotrophin 3 and neurotrophin 4 also seem to have both psychopathological and CV connections. Our aim was to overview the present knowledge in this area, demonstrating a new aspect of the associations between mood disorders and CV diseases through the mediation of NTs. These findings might enlighten new psychosomatic connections and suggest new therapeutic targets that are beneficial both in respect of mood disorders and CV pathology.
Collapse
|