1
|
Zhang B, Hou M, Huang J, Liu Y, Yang C, Lin J. Pax6 regulates neuronal migration and cell proliferation via interacting with Wnt3a during cortical development. Sci Rep 2025; 15:4726. [PMID: 39922861 PMCID: PMC11807113 DOI: 10.1038/s41598-025-88662-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 01/29/2025] [Indexed: 02/10/2025] Open
Abstract
The paired box 6 (Pax6) gene encodes a highly conserved transcription factor, involved in the development of eyes, brain, and endocrine glands. Homozygous loss of Pax6 resulted in neonatal death in mice, plus loss of eyes and malformation of cerebral cortex. In patients with heterozygous Pax6 mutations, a reduction in thickness of the frontoparietal cortex was detected, which was also observed in small eye mice. In this study, we found that Pax6 overexpression increased the cortical thickness, especially in the intermediate zone of the cortex, which conflicts with the report of Manuel et al. Pax6 overexpression appears to detain neurons in the intermediate zone while promoting cell proliferation. It is worth noting that the impact of Pax6 overexpression on cortical thickness and neuronal migration was temporal, explaining the differences with other reports. We postulated that the alteration of Pax6 isoform ratio by autoregulation might be responsible for this. JASPAR analysis together with the results of qPCR, Western blot, CUT&Tag, and rescue experiments revealed that Pax6 regulates neuronal migration and cell proliferation by indirectly mediating Wnt3a expression. Therefore, we propose that Pax6 participates in corticogenesis via interaction with Wnt3a in regulating neuronal migration and cell proliferation.
Collapse
Affiliation(s)
- Bichao Zhang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China
- Henan International Joint Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Meihua Hou
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Henan International Joint Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jiayan Huang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Henan International Joint Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yunfei Liu
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Henan International Joint Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Ciqing Yang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China
- Henan International Joint Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Juntang Lin
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China.
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China.
- Henan International Joint Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
2
|
Wechselberger C, Messner B, Bernhard D. The Role of Trace Elements in Cardiovascular Diseases. TOXICS 2023; 11:956. [PMID: 38133357 PMCID: PMC10747024 DOI: 10.3390/toxics11120956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/06/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Essential trace elements play an important role in human physiology and are associated with various functions regulating cellular metabolism. Non-essential trace elements, on the other hand, often have well-documented toxicities that are dangerous for the initiation and development of diseases due to their widespread occurrence in the environment and their accumulation in living organisms. Non-essential trace elements are therefore regarded as serious environmental hazards that are harmful to health even in low concentrations. Many representatives of these elements are present as pollutants in our environment, and many people may be exposed to significant amounts of these substances over the course of their lives. Among the most common non-essential trace elements are heavy metals, which are also associated with acute poisoning in humans. When these elements accumulate in the body over years of chronic exposure, they often cause severe health damage in a variety of tissues and organs. In this review article, the role of selected essential and non-essential trace elements and their role in the development of exemplary pathophysiological processes in the cardiovascular system will be examined in more detail.
Collapse
Affiliation(s)
- Christian Wechselberger
- Division of Pathophysiology, Institute of Physiology and Pathophysiology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Barbara Messner
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria;
| | - David Bernhard
- Division of Pathophysiology, Institute of Physiology and Pathophysiology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
- Clinical Research Institute for Cardiovascular and Metabolic Diseases, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
| |
Collapse
|
3
|
Liu H, Lu P, He S, Luo Y, Fang Y, Benkaci S, Wu B, Wang Y, Zhou B. β-Catenin regulates endocardial cushion growth by suppressing p21. Life Sci Alliance 2023; 6:e202302163. [PMID: 37385754 PMCID: PMC10310929 DOI: 10.26508/lsa.202302163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023] Open
Abstract
Endocardial cushion formation is essential for heart valve development and heart chamber separation. Abnormal endocardial cushion formation often causes congenital heart defects. β-Catenin is known to be essential for endocardial cushion formation; however, the underlying cellular and molecular mechanisms remain incompletely understood. Here, we show that endothelial-specific deletion of β-catenin in mice resulted in formation of hypoplastic endocardial cushions due to reduced cell proliferation and impaired cell migration. By using a β-catenin DM allele in which the transcriptional function of β-catenin is selectively disrupted, we further reveal that β-catenin regulated cell proliferation and migration through its transcriptional and non-transcriptional function, respectively. At the molecular level, loss of β-catenin resulted in increased expression of cell cycle inhibitor p21 in cushion endocardial and mesenchymal cells in vivo. In vitro rescue experiments with HUVECs and pig aortic valve interstitial cells confirmed that β-catenin promoted cell proliferation by suppressing p21. In addition, one savvy negative observation is that β-catenin was dispensable for endocardial-to-mesenchymal fate change. Taken together, our findings demonstrate that β-catenin is essential for cell proliferation and migration but dispensable for endocardial cells to gain mesenchymal fate during endocardial cushion formation. Mechanistically, β-catenin promotes cell proliferation by suppressing p21. These findings inform the potential role of β-catenin in the etiology of congenital heart defects.
Collapse
Affiliation(s)
- Huahua Liu
- Department of Cardiology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shan He
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Yuru Luo
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Yuan Fang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Sonia Benkaci
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yidong Wang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences; Department of Cardiology, First Affiliated Hospital; Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Bin Zhou
- Departments of Genetics, Pediatrics (Pediatric Genetic Medicine), and Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, The Einstein Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
4
|
Yang R, Tang Y, Chen X, Yang Y. Telocytes-derived extracellular vesicles alleviate aortic valve calcification by carrying miR-30b. ESC Heart Fail 2021; 8:3935-3946. [PMID: 34165260 PMCID: PMC8497371 DOI: 10.1002/ehf2.13460] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/13/2021] [Accepted: 05/23/2021] [Indexed: 12/28/2022] Open
Abstract
AIMS Calcific aortic valve disease (CAVD) is frequent in the elderly. Telocytes (TCs) are implicated in intercellular communication by releasing extracellular vesicles (EVs). This study investigated the role of TC-EVs in aortic valve calcification. METHODS AND RESULTS TCs were obtained and identified using enzymolysis method and flow cytometry. EVs were isolated from TCs using differential high-speed centrifugation method and identified using transmission electron microscope, western blot, and qNano analysis. The mouse model of CAVD was established. The changes of aortic valve activity-related indicators were analysed by ultrasound, and the expressions of TC markers CD34 and vimentin in mouse valve tissues were detected using RT-qPCR and western blot. The model mice were injected with TC-derived EVs. The expressions of Runx2, osteocalcin, and caspase-3 were detected using RT-qPCR and western blot. The calcification model of valvular interstitial cells (VICs) was established. TC-EVs were co-cultured with calcified VICs, and calcium deposition was detected using alizarin red S staining. miR-30b expression in calcified valvular tissues and cells was detected after EV treatment. miR-30b expression in TCs was knocked down and then EVs were extracted and co-cultured with calcified VICs. The target of miR-30b was predicted through bioinformatics website and verified using dual-luciferase assay. The levels of Wnt/β-catenin pathway-related proteins were detected. ApoE-/- mice fed with a high-fat diet showed decreased aortic valve orifice area, increased aortic transvalvular pressure difference and velocity, reduced left ventricular ejection fraction, decreased CD34 and vimentin, and increased caspase-3, Runx2, and osteocalcin. The levels of apoptosis- and osteogenesis- related proteins were inhibited after EV treatment. TC-EVs reduced calcium deposition and osteogenic proteins in calcified VICs. EVs could be absorbed by VICs. miR-30b expression was promoted in calcified valvular tissues and cells after EV treatment. Knockdown of miR-30b weakened the inhibitory effects of TC-EVs on calcium deposition and osteogenic proteins. miR-30b targeted Runx2. EV treatment inhibited the Wnt/β-catenin pathway, and knockdown of miR-30b in TCs attenuated the inhibitory effect of TC-EVs on the Wnt/β-catenin pathway. CONCLUSION TC-EVs played a protective role in aortic valve calcification via the miR-30b/Runx2/Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Rong Yang
- Department of Rheumatology, The Affiliated Zhongda Hospital, Southeast University, Nanjing, China
| | - Yihu Tang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Xiaowen Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Venardos N, Gergen AK, Jarrett M, Weyant MJ, Reece TB, Meng X, Fullerton DA. Warfarin Induces Calcification of the Aortic Valve via ERK1/2 and β-Catenin Signaling. Ann Thorac Surg 2021; 113:824-835. [PMID: 33901456 PMCID: PMC10109508 DOI: 10.1016/j.athoracsur.2021.03.099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Recent clinical evidence suggests an association between warfarin use and calcification of the aortic valve. We sought to determine the effect of warfarin on aortic valve interstitial cell (AVIC) osteogenic protein expression and the signaling pathways by which this effect is mediated. METHODS Human AVICs were isolated from normal aortic valves of patients undergoing cardiac transplantation while diseased AVICs were isolated from patients undergoing aortic valve replacement for aortic stenosis. AVICs were treated with various anticoagulants and osteogenic protein expression was evaluated using immunoblotting. Phosphorylation of LRP6 and ERK1/2 was evaluated following treatment with warfarin. AVICs were pretreated with LRP6 inhibitor dkk1 and ERK1/2 inhibitor PD98059 followed by treatment with warfarin and osteogenic protein expression was evaluated. RESULTS Warfarin, but not heparin or dabigatran, significantly increased Runx-2 and Osx expression in both normal and diseased human AVICs. Upregulation of β-catenin protein expression and nuclear translocation occurred in diseased AVICs, but not normal AVICs after warfarin treatment. Warfarin induced phosphorylation of LRP6 in diseased AVICs only, and phosphorylation of ERK1/2 in both normal and diseased AVICs. LRP6 inhibition attenuated warfarin-induced Runx-2 expression in diseased AVICs. ERK1/2 inhibition attenuated warfarin-induced Runx-2 expression in both normal and diseased AVICs. CONCLUSIONS Warfarin induces osteogenic activity in normal and diseased isolated human AVICs. This effect is mediated by ERK1/2 in both diseased and normal AVICs, but in diseased AVICs, β-catenin signaling also plays a role. These results implicate the role of warfarin in aortic valve calcification and highlight potential mechanisms for warfarin-induced aortic stenosis.
Collapse
Affiliation(s)
- Neil Venardos
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, CO, USA
| | - Anna K Gergen
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, CO, USA.
| | - Michael Jarrett
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, CO, USA
| | - Michael J Weyant
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, CO, USA
| | - T Brett Reece
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, CO, USA
| | - Xianzhong Meng
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, CO, USA
| | - David A Fullerton
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, CO, USA
| |
Collapse
|
6
|
Li Y, Du L, Cheng S, Guo J, Zhu S, Wang Y, Gao H. Hypoxia exacerbates cardiomyocyte injury via upregulation of Wnt3a and inhibition of Sirt3. Cytokine 2020; 136:155237. [DOI: 10.1016/j.cyto.2020.155237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/17/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022]
|
7
|
Zheng J, Yu J, Yang M, Tang L. Gefitinib suppresses cervical cancer progression by inhibiting cell cycle progression and epithelial-mesenchymal transition. Exp Ther Med 2019; 18:1823-1830. [PMID: 31410143 DOI: 10.3892/etm.2019.7754] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 03/26/2019] [Indexed: 11/06/2022] Open
Abstract
Cervical cancer (CC) is the second most common malignant cancer among women. Gefitinib was one of the first-generation epidermal growth factor receptor-tyrosine kinase inhibitors in clinical trials. However, the underlying mechanism of gefitinib in regulating CC progression remains unknown. In the current study, two CC cell lines, HeLa and Siha, were used to investigate the effects of gefitinib. Cell counting kit-8 assays demonstrated that treatment with gefitinib exerted strong cytotoxicity in HeLa and Siha cells. Flow cytometry was used to examine cell cycle progression and apoptosis. Treatment with gefitinib enhanced the number of cells in the G0/G1 phase and increased apoptosis in HeLa and Siha cells. Furthermore, treatment with gefitinib decreased the protein expression level of Bcl-2 and increased the protein expression level of Bax. Taken together, these results suggest that gefitinib may suppress CC cell proliferation and induce cell cycle arrest and apoptosis. The current study also demonstrated that treatment with gefitinib suppressed epithelial-mesenchymal transition (EMT) as the expression level of the epithelial marker, E-cadherin was increased, while the expression level of the mesenchymal marker, vimentin was decreased. The current study demonstrated that treatment with gefitinib decreased the protein expression levels of phosphorylated-GSK3β and β-catenin, which suggests that gefitinib may be a potential novel therapeutic strategy in CC by suppressing the Wnt/β-catenin signaling pathway and EMT to inhibit tumor metastasis in CC cells. In conclusion, gefitinib may suppress the EMT process during cell invasion and induce cell apoptosis and cell cycle arrest via inhibition of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Jianyun Zheng
- Department of Pathology, The First Affiliated Hospital of Xi'an Medical University, School of General Medicine, Xi'an Medical University, Xi'an, Shaanxi 710077, P.R. China
| | - Jianxin Yu
- Department of Laboratory, Central Hospital of Shanxian County, Heze, Shandong 274399, P.R. China
| | - Min Yang
- Department of Gynaecology and Obstetrics, Puyang Oil Field General Hospital, Puyang, Henan 457001, P.R. China
| | - Li Tang
- Department of Gynaecology and Obstetrics, Puyang Oil Field General Hospital, Puyang, Henan 457001, P.R. China
| |
Collapse
|
8
|
Development of calcific aortic valve disease: Do we know enough for new clinical trials? J Mol Cell Cardiol 2019; 132:189-209. [PMID: 31136747 DOI: 10.1016/j.yjmcc.2019.05.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/11/2019] [Accepted: 05/19/2019] [Indexed: 12/19/2022]
Abstract
Calcific aortic valve disease (CAVD), previously thought to represent a passive degeneration of the valvular extracellular matrix (VECM), is now regarded as an intricate multistage disorder with sequential yet intertangled and interacting underlying processes. Endothelial dysfunction and injury, initiated by disturbed blood flow and metabolic disorders, lead to the deposition of low-density lipoprotein cholesterol in the VECM further provoking macrophage infiltration, oxidative stress, and release of pro-inflammatory cytokines. Such changes in the valvular homeostasis induce differentiation of normally quiescent valvular interstitial cells (VICs) into synthetically active myofibroblasts producing excessive quantities of the VECM and proteins responsible for its remodeling. As a result of constantly ongoing degradation and re-deposition, VECM becomes disorganised and rigid, additionally potentiating myofibroblastic differentiation of VICs and worsening adaptation of the valve to the blood flow. Moreover, disrupted and excessively vascularised VECM is susceptible to the dystrophic calcification caused by calcium and phosphate precipitating on damaged collagen fibers and concurrently accompanied by osteogenic differentiation of VICs. Being combined, passive calcification and biomineralisation synergistically induce ossification of the aortic valve ultimately resulting in its mechanical incompetence requiring surgical replacement. Unfortunately, multiple attempts have failed to find an efficient conservative treatment of CAVD; however, therapeutic regimens and clinical settings have also been far from the optimal. In this review, we focused on interactions and transitions between aforementioned mechanisms demarcating ascending stages of CAVD, suggesting a predisposing condition (bicuspid aortic valve) and drug combination (lipid-lowering drugs combined with angiotensin II antagonists and cytokine inhibitors) for the further testing in both preclinical and clinical trials.
Collapse
|
9
|
Wang W, Zhao Y, Yao S, Cui X, Pan W, Huang W, Gao J, Dong T, Zhang S. Nigericin Inhibits Epithelial Ovarian Cancer Metastasis by Suppressing the Cell Cycle and Epithelial-Mesenchymal Transition. BIOCHEMISTRY (MOSCOW) 2017; 82:933-941. [PMID: 28941461 DOI: 10.1134/s0006297917080089] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epithelial ovarian cancer (EOC) has the highest mortality among various types of gynecological malignancies. Most patients die of metastasis and recurrence due to cisplatin resistance. Thus, it is urgent to develop novel therapies to cure this disease. CCK-8 assay showed that nigericin exhibited strong cytotoxicity on A2780 and SKOV3 cell lines. Flow cytometry indicated that nigericin could induce cell cycle arrest at G0/G1 phase and promote cell apoptosis. Boyden chamber assay revealed that nigericin could inhibit migration and invasion in a dose-dependent manner by suppressing epithelial-mesenchymal transition (EMT) in EOC cells. These effects were mediated, at least partly, by the Wnt/β-catenin signaling pathway. Our results demonstrated that nigericin could inhibit EMT during cell invasion and metastasis through the canonical Wnt/β-catenin signaling pathway. Nigericin may prove to be a novel therapeutic strategy that is effective in patients with metastatic EOC.
Collapse
Affiliation(s)
- Wen Wang
- Qilu Hospital, Shandong University, Department of Obstetrics and Gynecology, Jinan, Shandong, 250012, P.R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
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
|
11
|
Human interstitial cellular model in therapeutics of heart valve calcification. Amino Acids 2017; 49:1981-1997. [DOI: 10.1007/s00726-017-2432-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/27/2017] [Indexed: 12/27/2022]
|
12
|
Tandon I, Razavi A, Ravishankar P, Walker A, Sturdivant NM, Lam NT, Wolchok JC, Balachandran K. Valve interstitial cell shape modulates cell contractility independent of cell phenotype. J Biomech 2016; 49:3289-3297. [DOI: 10.1016/j.jbiomech.2016.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 01/08/2023]
|
13
|
Lim J, Ehsanipour A, Hsu JJ, Lu J, Pedego T, Wu A, Walthers CM, Demer LL, Seidlits SK, Tintut Y. Inflammation Drives Retraction, Stiffening, and Nodule Formation via Cytoskeletal Machinery in a Three-Dimensional Culture Model of Aortic Stenosis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2378-89. [PMID: 27392969 DOI: 10.1016/j.ajpath.2016.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 04/27/2016] [Accepted: 05/05/2016] [Indexed: 12/26/2022]
Abstract
In calcific aortic valve disease, the valve cusps undergo retraction, stiffening, and nodular calcification. The inflammatory cytokine, tumor necrosis factor (TNF)-α, contributes to valve disease progression; however, the mechanisms of its actions on cusp retraction and stiffening are unclear. We investigated effects of TNF-α on murine aortic valvular interstitial cells (VICs) within three-dimensional, free-floating, compliant, collagen hydrogels, simulating their natural substrate and biomechanics. TNF-α increased retraction (percentage of diameter), stiffness, and formation of macroscopic, nodular structures with calcification in the VIC-laden hydrogels. The effects of TNF-α were attenuated by blebbistatin inhibition of myosin II-mediated cytoskeletal contraction. Inhibition of actin polymerization with cytochalasin-D, but not inhibition of Rho kinase with Y27632, blocked TNF-α-induced retraction in three-dimensional VIC hydrogels, suggesting that actin stress fibers mediate TNF-α-induced effects. In the hydrogels, inhibitors of NF-κB blocked TNF-α-induced retraction, whereas simultaneous inhibition of c-Jun N-terminal kinase was required to block TNF-α-induced stiffness. TNF-α also significantly increased collagen deposition, as visualized by Masson's trichrome staining, and up-regulated mRNA expression of discoidin domain receptor tyrosine kinase 2, fibronectin, and α-smooth muscle actin. In human aortic valves, calcified cusps were stiffer and had more collagen deposition than noncalcified cusps. These findings suggest that inflammation, through stimulation of cytoskeletal contractile activity, may be responsible for valvular cusp retraction, stiffening, and formation of calcified nodules.
Collapse
Affiliation(s)
- Jina Lim
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, California
| | - Arshia Ehsanipour
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | - Jeffrey J Hsu
- Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Jinxiu Lu
- Department of Physiology, University of California, Los Angeles, Los Angeles, California
| | - Taylor Pedego
- Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Alexander Wu
- Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Chris M Walthers
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | - Linda L Demer
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California; Department of Medicine, University of California, Los Angeles, Los Angeles, California; Department of Physiology, University of California, Los Angeles, Los Angeles, California
| | - Stephanie K Seidlits
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | - Yin Tintut
- Department of Medicine, University of California, Los Angeles, Los Angeles, California; Department of Physiology, University of California, Los Angeles, Los Angeles, California; Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
14
|
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
|
15
|
Laguna-Fernandez A, Carracedo M, Jeanson G, Nagy E, Eriksson P, Caligiuri G, Franco-Cereceda A, Bäck M. Iron alters valvular interstitial cell function and is associated with calcification in aortic stenosis. Eur Heart J 2016; 37:3532-3535. [PMID: 27091952 PMCID: PMC5216198 DOI: 10.1093/eurheartj/ehw122] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/06/2016] [Accepted: 03/02/2016] [Indexed: 11/16/2022] Open
Abstract
Aims Aortic valve stenosis (AS) is the most common valvulopathy and is characterized by inflammation, extracellular matrix (ECM) remodelling and calcification, causing a narrowing of the valve and the consequential obstruction of the cardiac outflow. Although intraleaflet haemorrhage is associated with AS progression, the mechanisms involved are not known. The aims of this study were to identify valvular iron in relation to pathological changes associated with AS and the effects on valvular interstitial cells (VIC) in terms of iron uptake and iron-induced responses. Methods and results Valvular iron accumulation was detected by Perls' staining on aortic valve sections and shown to increase with the extent of calcification. Furthermore, qRT–PCR analysis revealed that iron-containing valve regions exhibited increased expression of genes involved in ECM remodelling and calcification. In addition, we demonstrate that iron transporters are regulated by pathways with major impact on AS and that VIC can take up and accumulate iron, which resulted in increased proliferation and decreased elastin production. Conclusion Iron, which may accumulate in the aortic valve by means of intraleaflet haemorrhages, can be taken up by VIC in a pro-inflammatory environment and actively contribute to VIC proliferation, ECM remodelling and calcification. These findings suggest a possible mechanism through which iron uptake by VIC may favour AS progression.
Collapse
Affiliation(s)
| | - Miguel Carracedo
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gregoire Jeanson
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Edit Nagy
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Per Eriksson
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Magnus Bäck
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden .,Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
16
|
Chen K, Fu Q, Li D, Wu Y, Sun S, Zhang X. Wnt3a suppresses Pseudomonas aeruginosa-induced inflammation and promotes bacterial killing in macrophages. Mol Med Rep 2016; 13:2439-46. [PMID: 26846714 PMCID: PMC4768980 DOI: 10.3892/mmr.2016.4869] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 01/11/2016] [Indexed: 12/31/2022] Open
Abstract
Pseudomonas aeruginosa (PA) is a common Gram‑negative bacterium and can cause serious infections, including hospital‑acquired pneumonia, suppurative bacterial keratitis and acute burn wound infection. The pathogenesis of PA infections is closely associated with excessive inflammatory responses and bacterial virulence factors. Wingless‑type MMTV integration site family, member 3A (Wnt3a), an upstream mediator in the canonical Wnt signaling pathway, has been implicated as a regulator of inflammation. However, its role in PA‑induced inflammation and bacterial clearance remains unknown. In the present study, the efficacy of Wnt3a conditioned media (Wnt3a‑CM) was assessed using western blotting and immunofluorescence, which showed that β‑catenin, a downstream molecule of Wnt3a, was upregulated and translocated to the nucleus following exposure to 50% Wnt3a‑CM for 6 h. To explore the role of Wnt3a in PA‑induced inflammation, the mRNA levels of pro‑inflammatory cytokines and apoptosis in macrophages were measured using reverse transcription‑quantitative polymerase chain reaction and flow cytometry, respectively. This indicated that Wnt3a suppressed inflammation by reducing the production of pro‑inflammatory cytokines and by promoting apoptosis in macrophages. Furthermore, the mechanism of macrophage‑mediated bacterial killing was investigated, and the results indicated that Wnt3a enhanced macrophage‑mediated intracellular bacterial killing via the induction of the production of cathelicidin‑related antimicrobial peptide and β‑defensins 1. Taken together, the current study explored the role of Wnt3a in inflammation and bacterial invasion, which may provide an improved understanding of host resistance to PA infection.
Collapse
Affiliation(s)
- Kang Chen
- Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat‑Sen University, Zhongshan, Guangdong 528403, P.R. China
| | - Qiang Fu
- Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat‑Sen University, Zhongshan, Guangdong 528403, P.R. China
| | - Dandan Li
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yongjian Wu
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Shijun Sun
- Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat‑Sen University, Zhongshan, Guangdong 528403, P.R. China
| | - Xiumin Zhang
- Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat‑Sen University, Zhongshan, Guangdong 528403, P.R. China
| |
Collapse
|
17
|
Wong FF, Ho ML, Yamagami M, Lam MT, Grande-Allen KJ, Suh J. Effective Gene Delivery to Valvular Interstitial Cells Using Adeno-Associated Virus Serotypes 2 and 3. Tissue Eng Part C Methods 2015; 21:808-15. [DOI: 10.1089/ten.tec.2014.0493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Fergus F. Wong
- Department of Bioengineering, Rice University, Houston, Texas
| | - Michelle L. Ho
- Department of Bioengineering, Rice University, Houston, Texas
| | - Momona Yamagami
- Department of Bioengineering, Rice University, Houston, Texas
| | - Michael T. Lam
- Department of Bioengineering, Rice University, Houston, Texas
| | | | - Junghae Suh
- Department of Bioengineering, Rice University, Houston, Texas
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, Texas
| |
Collapse
|
18
|
Thalji NM, Hagler MA, Zhang H, Casaclang-Verzosa G, Nair AA, Suri RM, Miller JD. Nonbiased Molecular Screening Identifies Novel Molecular Regulators of Fibrogenic and Proliferative Signaling in Myxomatous Mitral Valve Disease. ACTA ACUST UNITED AC 2015; 8:516-28. [PMID: 25814644 DOI: 10.1161/circgenetics.114.000921] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 03/12/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pathological processes underlying myxomatous mitral valve degeneration (MMVD) remain poorly understood. We sought to identify novel mechanisms contributing to the development of this condition. METHODS AND RESULTS Microarrays were used to measure gene expression in 11 myxomatous and 11 nonmyxomatous human mitral valves. Differential gene expression (thresholds P<0.05; fold-change >1.5) and pathway activation (Ingenuity) were confirmed using quantitative reverse transcriptase polymerase chain reaction and immunohistochemistry. Contributions of bone morphogenetic protein 4 and transforming growth factor (TGF)-β2 to differential gene expression were evaluated in vitro. Contributions of angiotensin II to differential pathway activation were examined in mice in vivo. A total of 2602 genes were differentially expressed between myxomatous and nonmyxomatous valves. Canonical TGF-β signaling was increased in MMVD because of increased ligand expression and derepression of SMA mothers against decapentaplegic 2/3 signaling and was confirmed with quantitative reverse transcriptase polymerase chain reaction and immunohistochemistry. Myxomatous valves demonstrated activation of canonical bone morphogenetic protein and Wnt/β-catenin signaling and upregulation of their common target runt-related transcription factor 2. Our data set provided transcriptional and immunohistochemical evidence for activated immune cell infiltration. In vitro treatment of mitral valve interstitial cells with TGF-β2 increased β-catenin signaling at mRNA and protein levels, suggesting interactions between TGF-β2 and Wnt signaling. In vivo infusion of mice with angiotensin II recaptured several changes in signaling pathways characteristic of human MMVD. CONCLUSIONS These data support a new disease framework whereby activation of TGF-β2, bone morphogenetic protein 4, Wnt/β-catenin, or immune signaling plays major roles in the pathogenesis of MMVD. We propose these pathways act in a context-dependent manner to drive phenotypic changes that fundamentally differ from those observed in aortic valve disease and open novel avenues guiding future research into the pathogenesis of MMVD.
Collapse
Affiliation(s)
- Nassir M Thalji
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
| | - Michael A Hagler
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
| | - Heyu Zhang
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
| | - Grace Casaclang-Verzosa
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
| | - Asha A Nair
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
| | - Rakesh M Suri
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN.
| | - Jordan D Miller
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
19
|
Wirrig EE, Gomez MV, Hinton RB, Yutzey KE. COX2 inhibition reduces aortic valve calcification in vivo. Arterioscler Thromb Vasc Biol 2015; 35:938-47. [PMID: 25722432 DOI: 10.1161/atvbaha.114.305159] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Calcific aortic valve disease (CAVD) is a significant cause of morbidity and mortality, which affects ≈1% of the US population and is characterized by calcific nodule formation and stenosis of the valve. Klotho-deficient mice were used to study the molecular mechanisms of CAVD as they develop robust aortic valve (AoV) calcification. Through microarray analysis of AoV tissues from klotho-deficient and wild-type mice, increased expression of the gene encoding cyclooxygenase 2 (COX2; Ptgs2) was found. COX2 activity contributes to bone differentiation and homeostasis, thus the contribution of COX2 activity to AoV calcification was assessed. APPROACH AND RESULTS In klotho-deficient mice, COX2 expression is increased throughout regions of valve calcification and is induced in the valvular interstitial cells before calcification formation. Similarly, COX2 expression is increased in human diseased AoVs. Treatment of cultured porcine aortic valvular interstitial cells with osteogenic media induces bone marker gene expression and calcification in vitro, which is blocked by inhibition of COX2 activity. In vivo, genetic loss of function of COX2 cyclooxygenase activity partially rescues AoV calcification in klotho-deficient mice. Moreover, pharmacological inhibition of COX2 activity in klotho-deficient mice via celecoxib-containing diet reduces AoV calcification and blocks osteogenic gene expression. CONCLUSIONS COX2 expression is upregulated in CAVD, and its activity contributes to osteogenic gene induction and valve calcification in vitro and in vivo.
Collapse
Affiliation(s)
- Elaine E Wirrig
- From The Heart Institute, Cincinnati Children's Hospital Medical Center, OH
| | - M Victoria Gomez
- From The Heart Institute, Cincinnati Children's Hospital Medical Center, OH
| | - Robert B Hinton
- From The Heart Institute, Cincinnati Children's Hospital Medical Center, OH
| | - Katherine E Yutzey
- From The Heart Institute, Cincinnati Children's Hospital Medical Center, OH.
| |
Collapse
|
20
|
Abstract
The aortic valve is highly responsive to cyclical and continuous mechanical forces, at the macroscopic and cellular levels. In this report, we delineate mechanokinetics (effects of mechanical inputs on the cells) and mechanodynamics (effects of cells and pathologic processes on the mechanics) of the aortic valve, with a particular focus on how mechanical inputs synergize with the inflammatory cytokine and other biomolecular signaling to contribute to the process of aortic valve calcification.
Collapse
|