1
|
Brückner A, Brandtner A, Rieck S, Matthey M, Geisen C, Fels B, Stei M, Kusche-Vihrog K, Fleischmann BK, Wenzel D. Site-specific genetic and functional signatures of aortic endothelial cells at aneurysm predilection sites in healthy and AngII ApoE -/- mice. Angiogenesis 2024; 27:719-738. [PMID: 38965173 PMCID: PMC11564227 DOI: 10.1007/s10456-024-09933-9] [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: 12/18/2023] [Accepted: 06/16/2024] [Indexed: 07/06/2024]
Abstract
Aortic aneurysm is characterized by a pathological dilation at specific predilection sites of the vessel and potentially results in life-threatening vascular rupture. Herein, we established a modified "Häutchen method" for the local isolation of endothelial cells (ECs) from mouse aorta to analyze their spatial heterogeneity and potential role in site-specific disease development. When we compared ECs from aneurysm predilection sites of healthy mice with adjacent control segments we found regulation of genes related to extracellular matrix remodeling, angiogenesis and inflammation, all pathways playing a critical role in aneurysm development. We also detected enhanced cortical stiffness of the endothelium at these sites. Gene expression of ECs from aneurysms of the AngII ApoE-/- model when compared to sham animals mimicked expression patterns from predilection sites of healthy animals. Thus, this work highlights a striking genetic and functional regional heterogeneity in aortic ECs of healthy mice, which defines the location of aortic aneurysm formation in disease.
Collapse
Affiliation(s)
- Alexander Brückner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Adrian Brandtner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Sarah Rieck
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Michaela Matthey
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Caroline Geisen
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Benedikt Fels
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Marta Stei
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Kristina Kusche-Vihrog
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Bernd K Fleischmann
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Daniela Wenzel
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany.
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany.
| |
Collapse
|
2
|
Millar JK, Salmon M, Nasser E, Malik S, Kolli P, Lu G, Pinteaux E, Hawkins RB, Ailawadi G. Endothelial to mesenchymal transition in the interleukin-1 pathway during aortic aneurysm formation. J Thorac Cardiovasc Surg 2024; 167:e146-e158. [PMID: 37951532 PMCID: PMC11029391 DOI: 10.1016/j.jtcvs.2023.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 10/30/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
OBJECTIVE Endothelial to mesenchymal transition may represent a key link between inflammatory stress and endothelial dysfunction seen in aortic aneurysm disease. Endothelial to mesenchymal transition is regulated by interleukin-1β, and previous work has demonstrated an essential role of interleukin-1 signaling in experimental aortic aneurysm models. We hypothesize that endothelial to mesenchymal transition is present in murine aortic aneurysms, and loss of interleukin-1 signaling attenuates this process. METHODS Murine aortic aneurysms were created in novel CDH5-Cre lineage tracking mice by treating the intact aorta with peri-adventitial elastase. Endothelial to mesenchymal transition transcription factors as well as endothelial and mesenchymal cell markers were analyzed via immunohistochemistry and immunofluorescence (n = 10/group). To determine the role of interleukin-1 signaling, endothelial-specific interleukin-1 receptor 1 knockout and wild-type mice (n = 10/group) were treated with elastase. Additionally, C57/BL6 mice were treated with the interleukin-1 receptor 1 antagonist Anakinra (n = 7) or vehicle (n = 8). RESULTS Elastase treatment yielded greater aortic dilation compared with controls (elastase 97.0% ± 34.0%; control 5.3% ± 4.8%; P < .001). Genetic deletion of interleukin-1 receptor 1 attenuated aortic dilation (control 126.7% ± 38.7%; interleukin-1 receptor 1 knockout 35.2% ± 14.7%; P < .001), as did pharmacologic inhibition of interleukin-1 receptor 1 with Anakinra (vehicle 146.3% ± 30.1%; Anakinra 63.5% ± 23.3%; P < .001). Elastase treatment resulted in upregulation of endothelial to mesenchymal transition transcription factors (Snail, Slug, Twist, ZNF) and mesenchymal cell markers (S100, alpha smooth muscle actin) and loss of endothelial cell markers (vascular endothelial cadherin, endothelial nitric oxide synthase, von Willebrand factor). These changes were attenuated by interleukin-1 receptor 1 knockout and Anakinra treatment. CONCLUSIONS Endothelial to mesenchymal transition occurs in aortic aneurysm disease and is attenuated by loss of interleukin-1 signaling. Endothelial dysfunction through endothelial to mesenchymal transition represents a new and novel pathway in understanding aortic aneurysm disease and may be a potential target for future treatment.
Collapse
Affiliation(s)
- Jessica K Millar
- Department of Surgery, University of Michigan, Ann Arbor, Mich; Department of Cardiac Surgery, University of Michigan, Ann Arbor, Mich
| | - Morgan Salmon
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Mich
| | | | | | | | - Guanyi Lu
- Department of Surgery, University of Florida, Gainesville, Fla
| | - Emmanuel Pinteaux
- Division of Neuroscience, University of Manchester, Manchester, United Kingdom
| | - Robert B Hawkins
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Mich
| | - Gorav Ailawadi
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
3
|
Tarraf SA, de Souza RB, Herrick A, Pereira LV, Bellini C. The Fbn1 gene variant governs passive ascending aortic mechanics in the mgΔ lpn mouse model of Marfan syndrome when superimposed to perlecan haploinsufficiency. Front Cardiovasc Med 2024; 11:1319164. [PMID: 38545339 PMCID: PMC10965555 DOI: 10.3389/fcvm.2024.1319164] [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: 10/11/2023] [Accepted: 02/12/2024] [Indexed: 11/11/2024] Open
Abstract
Introduction Ascending thoracic aortic aneurysms arise from pathological tissue remodeling that leads to abnormal wall dilation and increases the risk of fatal dissection/rupture. Large variability in disease manifestations across family members who carry a causative genetic variant for thoracic aortic aneurysms suggests that genetic modifiers may exacerbate clinical outcomes. Decreased perlecan expression in the aorta of mgΔlpn mice with severe Marfan syndrome phenotype advocates for exploring perlecan-encoding Hspg2 as a candidate modifier gene. Methods To determine the effect of concurrent Hspg2 and Fbn1 mutations on the progression of thoracic aortopathy, we characterized the microstructure and passive mechanical response of the ascending thoracic aorta in female mice of four genetic backgrounds: wild-type, heterozygous with a mutation in the Fbn1 gene (mgΔlpn), heterozygous with a mutation in the Hspg2 gene (Hspg2+/-), and double mutants carrying both the Fbn1 and Hspg2 variants (dMut). Results Elastic fiber fragmentation and medial disarray progress from the internal elastic lamina outward as the ascending thoracic aorta dilates in mgΔlpn and dMut mice. Concurrent increase in total collagen content relative to elastin reduces energy storage capacity and cyclic distensibility of aortic tissues from mice that carry the Fbn1 variant. Inherent circumferential tissue stiffening strongly correlates with the severity of aortic dilatation in mgΔlpn and dMut mice. Perlecan haploinsufficiency superimposed to the mgΔlpn mutation curbs the viability of dMut mice, increases the occurrence of aortic enlargement, and reduces the axial stretch in aortic tissues. Discussion Overall, our findings show that dMut mice are more vulnerable than mgΔlpn mice without an Hspg2 mutation, yet later endpoints and additional structural and functional readouts are needed to identify causative mechanisms.
Collapse
Affiliation(s)
- Samar A. Tarraf
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | | | - Ashley Herrick
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Lygia V. Pereira
- Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo, Brazil
| | - Chiara Bellini
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| |
Collapse
|
4
|
Kucher AN, Koroleva IA, Nazarenko MS. Pathogenetic Significance of Long Non-Coding RNAs in the Development of Thoracic and Abdominal Aortic Aneurysms. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:130-147. [PMID: 38467550 DOI: 10.1134/s0006297924010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 03/13/2024]
Abstract
Aortic aneurysm (AA) is a life-threatening condition with a high prevalence and risk of severe complications. The aim of this review was to summarize the data on the role of long non-coding RNAs (lncRNAs) in the development of AAs of various location. Within less than a decade of studies on the role of lncRNAs in AA, using experimental and bioinformatic approaches, scientists have obtained the data confirming the involvement of these molecules in metabolic pathways and pathogenetic mechanisms critical for the aneurysm development. Regardless of the location of pathological process (thoracic or abdominal aorta), AA was found to be associated with changes in the expression of various lncRNAs in the tissue of the affected vessels. The consistency of changes in the expression level of lncRNA, mRNA and microRNA in aortic tissues during AA development has been recordedand regulatory networks implicated in the AA pathogenesis in which lncRNAs act as competing endogenous RNAs (ceRNA networks) have been identified. It was found that the same lncRNA can be involved in different ceRNA networks and regulate different biochemical and cellular events; on the other hand, the same pathological process can be controlled by different lncRNAs. Despite some similarities in pathogenesis and overlapping of involved lncRNAs, the ceRNA networks described for abdominal and thoracic AA are different. Interactions between lncRNAs and other molecules, including those participating in epigenetic processes, have also been identified as potentially relevant to the AA pathogenesis. The expression levels of some lncRNAs were found to correlate with clinically significant aortic features and biochemical parameters. Identification of regulatory RNAs functionally significant in the aneurysm development is important for clarification of disease pathogenesis and will provide a basis for early diagnostics and development of new preventive and therapeutic drugs.
Collapse
Affiliation(s)
- Aksana N Kucher
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Iuliia A Koroleva
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Maria S Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia.
| |
Collapse
|
5
|
Yang K, Cui S, Wang J, Xu T, Du H, Yue H, Ye H, Guo J, Zhang J, Li P, Guo Y, Pan C, Pang J, Wang J, Yu X, Zhang C, Liu Z, Chen Y, Xu F. Early Progression of Abdominal Aortic Aneurysm is Decelerated by Improved Endothelial Barrier Function via ALDH2-LIN28B-ELK3 Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302231. [PMID: 37822152 PMCID: PMC10646281 DOI: 10.1002/advs.202302231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/25/2023] [Indexed: 10/13/2023]
Abstract
The involvement of endothelial barrier function in abdominal aortic aneurysm (AAA) and its upstream regulators remains unknown. Single-cell RNA sequencing shows that disrupted endothelial focal junction is an early (3 days) and persistent (28 days) event during Angiotensin II (Ang II)-induced AAA progression. Consistently, mRNA sequencing on human aortic dissection tissues confirmed downregulated expression of endothelial barrier-related genes. Aldehyde dehydrogenase 2 (ALDH2), a negative regulator of AAA, is found to be upregulated in the intimal media of AAA samples, leading to testing its role in early-stage AAA. ALDH2 knockdown/knockout specifically in endothelial cells (ECs) significantly increases expression of EC barrier markers related to focal adhesion and tight junction, restores endothelial barrier integrity, and suppresses early aortic dilation of AAA (7 and 14 days post-Ang II). Mechanically, ELK3 acts as an ALDH2 downstream regulator for endothelial barrier function preservation. At the molecular level, ALDH2 directly binds to LIN28B, a regulator of ELK3 mRNA stability, hindering LIN28B binding to ELK3 mRNA, thereby depressing ELK3 expression and impairing endothelial barrier function. Therefore, preserving vascular endothelial barrier integrity via ALDH2-specific knockdown in ECs holds therapeutic potential in the early management of AAAs.
Collapse
|
6
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
7
|
Yang X, Xu C, Yao F, Ding Q, Liu H, Luo C, Wang D, Huang J, Li Z, Shen Y, Yang W, Li Z, Yu F, Fu Y, Wang L, Ma Q, Zhu J, Xu F, Cong X, Kong W. Targeting endothelial tight junctions to predict and protect thoracic aortic aneurysm and dissection. Eur Heart J 2023; 44:1248-1261. [PMID: 36638776 DOI: 10.1093/eurheartj/ehac823] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 11/16/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
AIMS Whether changes in endothelial tight junctions (TJs) lead to the formation of thoracic aortic aneurysm and dissection (TAAD) and serve as an early indicator and therapeutic target remains elusive. METHODS AND RESULTS Single-cell RNA sequencing analysis showed aberrant endothelial TJ expressions in the thoracic aortas of patients with TAAD. In a β-aminopropionitrile (BAPN)-induced TAAD mouse model, endothelial TJ function was disrupted in the thoracic aortas at an early stage (5 and 10 days) as observed by a vascular permeability assay, while the intercellular distribution of crucial TJ components was significantly decreased by en face staining. For the non-invasive detection of endothelial TJ function, two dextrans of molecular weights 4 and 70 kDa were conjugated with the magnetic resonance imaging (MRI) contrast agent Gd-DOTA to synthesize FITC-dextran-DOTA-Gd and rhodamine B-dextran-DOTA-Gd. MRI images showed that both probes accumulated in the thoracic aortas of the BAPN-fed mice. Particularly, the mice with increased accumulated signals from 5 to 10 days developed TAAD at 14 days, whereas the mice with similar signals between the two time points did not. Furthermore, the protease-activated receptor 2 inhibitor AT-1001, which seals TJs, alleviated the BAPN-induced impairment of endothelial TJ function and expression and subsequently reduced TAAD incidence. Notably, endothelial-targeted ZO-1 conditional knockout increased TAAD incidence. Mechanistically, vascular inflammation and edema were observed in the thoracic aortas of the BAPN-fed mice, whereas these phenomena were attenuated by AT-1001. CONCLUSION The disruption of endothelial TJ function is an early event prior to TAAD formation, herein serving as a potential indicator and a promising target for TAAD.
Collapse
Affiliation(s)
- Xueyuan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Chen Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Fang Yao
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China.,Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing 100037, China
| | - Qianhui Ding
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Hao Liu
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Engineering Research Center of Vascular Prostheses, Beijing 100029, China
| | - Congcong Luo
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Engineering Research Center of Vascular Prostheses, Beijing 100029, China
| | - Daidai Wang
- Department of Emergency, Peking University Third Hospital, Beijing 100191, China
| | - Jiaqi Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Yicong Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Weijie Yang
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zhuofan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Li Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China.,Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing 100037, China
| | - Qingbian Ma
- Department of Emergency, Peking University Third Hospital, Beijing 100191, China
| | - Junming Zhu
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Engineering Research Center of Vascular Prostheses, Beijing 100029, China
| | - Fujian Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xin Cong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| |
Collapse
|
8
|
Nguyen TAV, Lino CA, Hang HT, Alves JV, Thang BQ, Shin SJ, Sugiyama K, Matsunaga H, Takeyama H, Yamashiro Y, Yanagisawa H. Protective Role of Endothelial Fibulin-4 in Valvulo-Arterial Integrity. J Am Heart Assoc 2022; 12:e026942. [PMID: 36565192 PMCID: PMC9973605 DOI: 10.1161/jaha.122.026942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Homeostasis of the vessel wall is cooperatively maintained by endothelial cells (ECs), smooth muscle cells, and adventitial fibroblasts. The genetic deletion of fibulin-4 (Fbln4) in smooth muscle cells (SMKO) leads to the formation of thoracic aortic aneurysms with the disruption of elastic fibers. Although Fbln4 is expressed in the entire vessel wall, its function in ECs and relevance to the maintenance of valvulo-arterial integrity are not fully understood. Methods and Results Gene silencing of FBLN4 was conducted on human aortic ECs to evaluate morphological changes and gene expression profile. Fbln4 double knockout (DKO) mice in ECs and smooth muscle cells were generated and subjected to histological analysis, echocardiography, Western blotting, RNA sequencing, and immunostaining. An evaluation of the thoracic aortic aneurysm phenotype and screening of altered signaling pathways were performed. Knockdown of FBLN4 in human aortic ECs induced mesenchymal cell-like changes with the upregulation of mesenchymal genes, including TAGLN and MYL9. DKO mice showed the exacerbation of thoracic aortic aneurysms when compared with those of SMKO and upregulated Thbs1, a mechanical stress-responsive molecule, throughout the aorta. DKO mice also showed progressive aortic valve thickening with collagen deposition from postnatal day 14, as well as turbulent flow in the ascending aorta. Furthermore, RNA sequencing and immunostaining of the aortic valve revealed the upregulation of genes involved in endothelial-to-mesenchymal transition, inflammatory response, and tissue fibrosis in DKO valves and the presence of activated valve interstitial cells. Conclusions The current study uncovers the pivotal role of endothelial fibulin-4 in the maintenance of valvulo-arterial integrity, which influences thoracic aortic aneurysm progression.
Collapse
Affiliation(s)
- Tram Anh Vu Nguyen
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Ph.D. Program in Human Biology, School of Integrative and Global MajorsUniversity of TsukubaIbarakiJapan
| | - Caroline Antunes Lino
- Department of AnatomyUniversity of Sao Paulo, Institute of Biomedical SciencesSao PauloBrazil
| | - Huynh Thuy Hang
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Graduate School of Comprehensive Human SciencesUniversity of TsukubaIbarakiJapan
| | - Juliano Vilela Alves
- Department of PharmacologyUniversity of Sao Paulo, Ribeirao Preto Medical SchoolRibeirao PretoBrazil
| | - Bui Quoc Thang
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Deputy Head of Scientific Research Department‐ Training center, Cho Ray hospitalHo Chi Minh CityVietnam
| | - Seung Jae Shin
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Graduate School of Life and Environmental SciencesUniversity of TsukubaIbarakiJapan
| | - Kaori Sugiyama
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda UniversityTokyoJapan
| | - Hiroko Matsunaga
- Research organization for Nano and Life InnovationWaseda UniversityTokyoJapan
| | - Haruko Takeyama
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda UniversityTokyoJapan,Research organization for Nano and Life InnovationWaseda UniversityTokyoJapan,Department of Life Science and Medical BioscienceWaseda UniversityTokyoJapan,Computational Bio Big‐Data Open Innovation LaboratoryAIST‐Waseda UniversityTokyoJapan
| | - Yoshito Yamashiro
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Present address:
Department of Advanced Medical TechnologiesNational Cerebral and Cardiovascular Center Research InstituteOsaka564‐8565Japan
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Faculty of MedicineUniversity of TsukubaIbarakiJapan
| |
Collapse
|
9
|
Lim S, Kim SW, Kim IK, Song BW, Lee S. Organ-on-a-chip: Its use in cardiovascular research. Clin Hemorheol Microcirc 2022; 83:315-339. [DOI: 10.3233/ch-221428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Organ-on-a-chip (OOAC) has attracted great attention during the last decade as a revolutionary alternative to conventional animal models. This cutting-edge technology has also brought constructive changes to the field of cardiovascular research. The cardiovascular system, especially the heart as a well-protected vital organ, is virtually impossible to replicate in vitro with conventional approaches. This made scientists assume that they needed to use animal models for cardiovascular research. However, the frequent failure of animal models to correctly reflect the native cardiovascular system necessitated a search for alternative platforms for preclinical studies. Hence, as a promising alternative to conventional animal models, OOAC technology is being actively developed and tested in a wide range of biomedical fields, including cardiovascular research. Therefore, in this review, the current literature on the use of OOACs for cardiovascular research is presented with a focus on the basis for using OOACs, and what has been specifically achieved by using OOACs is also discussed. By providing an overview of the current status of OOACs in cardiovascular research and its future perspectives, we hope that this review can help to develop better and optimized research strategies for cardiovascular diseases (CVDs) as well as identify novel applications of OOACs in the near future.
Collapse
Affiliation(s)
- Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Il-Kwon Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Byeong-Wook Song
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| |
Collapse
|
10
|
Rombouts KB, van Merrienboer TAR, Ket JCF, Bogunovic N, van der Velden J, Yeung KK. The role of vascular smooth muscle cells in the development of aortic aneurysms and dissections. Eur J Clin Invest 2022; 52:e13697. [PMID: 34698377 PMCID: PMC9285394 DOI: 10.1111/eci.13697] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/12/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Aortic aneurysms (AA) are pathological dilations of the aorta, associated with an overall mortality rate up to 90% in case of rupture. In addition to dilation, the aortic layers can separate by a tear within the layers, defined as aortic dissections (AD). Vascular smooth muscle cells (vSMC) are the predominant cell type within the aortic wall and dysregulation of vSMC functions contributes to AA and AD development and progression. However, since the exact underlying mechanism is poorly understood, finding potential therapeutic targets for AA and AD is challenging and surgery remains the only treatment option. METHODS In this review, we summarize current knowledge about vSMC functions within the aortic wall and give an overview of how vSMC functions are altered in AA and AD pathogenesis, organized per anatomical location (abdominal or thoracic aorta). RESULTS Important functions of vSMC in healthy or diseased conditions are apoptosis, phenotypic switch, extracellular matrix regeneration and degradation, proliferation and contractility. Stressors within the aortic wall, including inflammatory cell infiltration and (epi)genetic changes, modulate vSMC functions and cause disturbance of processes within vSMC, such as changes in TGF-β signalling and regulatory RNA expression. CONCLUSION This review underscores a central role of vSMC dysfunction in abdominal and thoracic AA and AD development and progression. Further research focused on vSMC dysfunction in the aortic wall is necessary to find potential targets for noninvasive AA and AD treatment options.
Collapse
Affiliation(s)
- Karlijn B Rombouts
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Tara A R van Merrienboer
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | | | - Natalija Bogunovic
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands.,Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Dawson A, Wang Y, Li Y, LeMaire SA, Shen YH. New Technologies With Increased Precision Improve Understanding of Endothelial Cell Heterogeneity in Cardiovascular Health and Disease. Front Cell Dev Biol 2021; 9:679995. [PMID: 34513826 PMCID: PMC8430032 DOI: 10.3389/fcell.2021.679995] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/17/2021] [Indexed: 01/08/2023] Open
Abstract
Endothelial cells (ECs) are vital for blood vessel integrity and have roles in maintaining normal vascular function, healing after injury, and vascular dysfunction. Extensive phenotypic heterogeneity has been observed among ECs of different types of blood vessels in the normal and diseased vascular wall. Although ECs with different phenotypes can share common functions, each has unique features that may dictate a fine-tuned role in vascular health and disease. Recent studies performed with single-cell technology have generated powerful information that has significantly improved our understanding of EC biology. Here, we summarize a variety of EC types, states, and phenotypes recently identified by using new, increasingly precise techniques in transcriptome analysis.
Collapse
Affiliation(s)
- Ashley Dawson
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Yidan Wang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Scott A. LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, United States
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, United States
| |
Collapse
|
12
|
Varela-López E, del Valle-Mondragón L, Castrejón-Téllez V, Pérez-Torres I, Arenas AP, Rojas FM, Guarner-Lans V, Vargas-González A, Pastelín-Hernández G, Torres-Narváez JC. Role of the Transient Receptor Potential Vanilloid Type 1 (TRPV1) in the Regulation of Nitric Oxide Release in Wistar Rat Aorta. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8531975. [PMID: 34394835 PMCID: PMC8355966 DOI: 10.1155/2021/8531975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 11/18/2022]
Abstract
The potential transient vanilloid receptor type 1 (TRPV1) plays important functional roles in the vascular system. In the present study, we explored the role of the TRPV1 in the production of nitric oxide (NO), biopterines (BH4 and BH2), cyclic guanosine monophosphate (cGMP), malondialdehyde (MDA), phosphodiesterase-3 (PDE-3), total antioxidant capacity (TAC), and calcitonin gene-related peptide (CGRP) in the rat aorta. Wistar rats were divided into four groups: (1) control, (2) capsaicin (CS, 20 mg/kg), (3) capsazepine (CZ, 24 mg/kg), and (4) CZ + CS. Treatments were applied daily for 4 days before removing the thoracic aortas for testing of aortic tissue and endothelial cells. TRPV1 activation produced increases in BH4 14%, cGMP 25%, NO 29%, and TAC 59.2% in comparison to the controls. BH2 and MDA increased with CZ. CGRP shows a tendency to decrease with CZ. The analysis by immunocytochemistry confirmed that the TRPV1 is present in aortic endothelial cells. Aortic endothelial cells were obtained from healthy rats and cultured to directly explore the effects of CS and CZ. The activation of the TRPV1 (CS 30 μM) produced increases in BH4 17%, NO 36.6%, TAC 56.3%, and CGRP 65%, when compared to controls. BH2 decreased with CZ + CS. CS effects were diminished by CZ in cells and in the tissue. We conclude that the TRPV1 is a structure present in the membrane of aortic endothelial cells and that it participates in the production of NO. The importance of the TRPV1 should be considered in vascular reactivity studies.
Collapse
Affiliation(s)
- Elvira Varela-López
- Laboratorio de Cardiología Translacional, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, 14080 Tlalpan, Ciudad de México, Mexico
| | - Leonardo del Valle-Mondragón
- Departamento de Farmacología “Dr. Rafael Méndez Martínez”, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, 14080 Tlalpan, Ciudad de México, Mexico
| | - Vicente Castrejón-Téllez
- Departamento de Fisiología, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, 14080 Tlalpan, Ciudad de México, Mexico
| | - Israel Pérez-Torres
- Departamento of Biomedicina Cardiovascular, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, 14080 Tlalpan, Ciudad de México, Mexico
| | - Araceli Páez Arenas
- Laboratorio de Cardiología Translacional, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, 14080 Tlalpan, Ciudad de México, Mexico
| | - Felipe Massó Rojas
- Laboratorio de Cardiología Translacional, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, 14080 Tlalpan, Ciudad de México, Mexico
| | - Verónica Guarner-Lans
- Departamento de Fisiología, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, 14080 Tlalpan, Ciudad de México, Mexico
| | - Alvaro Vargas-González
- Departamento de Fisiología, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, 14080 Tlalpan, Ciudad de México, Mexico
| | - Gustavo Pastelín-Hernández
- Departamento de Farmacología “Dr. Rafael Méndez Martínez”, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, 14080 Tlalpan, Ciudad de México, Mexico
| | - Juan Carlos Torres-Narváez
- Departamento de Farmacología “Dr. Rafael Méndez Martínez”, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, 14080 Tlalpan, Ciudad de México, Mexico
| |
Collapse
|
13
|
Martin‐Blazquez A, Heredero A, Aldamiz‐Echevarria G, Martin‐Lorenzo M, Alvarez‐Llamas G. Non-syndromic thoracic aortic aneurysm: cellular and molecular insights. J Pathol 2021; 254:229-238. [PMID: 33885146 PMCID: PMC8251829 DOI: 10.1002/path.5683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/25/2021] [Accepted: 04/13/2021] [Indexed: 01/16/2023]
Abstract
Thoracic aortic aneurysm (TAA) develops silently and asymptomatically and is a major cause of mortality. TAA prevalence is greatly underestimated, it is usually diagnosed incidentally, and its treatment consists mainly of prophylactic surgery based on the aortic diameter. The lack of effective drugs and biological markers to identify and stratify TAAs by risk before visible symptoms results from scant knowledge of its pathophysiological mechanisms. Here we integrate the structural impairment affecting non-syndromic non-familial TAA with the main cellular and molecular changes described so far and consider how these changes are interconnected through specific pathways. The ultimate goal is to define much-needed novel markers of TAA, and so the potential of previously identified molecules to aid in early diagnosis/prognosis is also discussed. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Angeles Heredero
- Department of Cardiac SurgeryFundación Jiménez Díaz, UAMMadridSpain
| | | | | | - Gloria Alvarez‐Llamas
- Department of ImmunologyIIS‐Fundación Jiménez Díaz, UAMMadridSpain
- REDInRENMadridSpain
| |
Collapse
|
14
|
Portelli SS, Hambly BD, Jeremy RW, Robertson EN. Oxidative stress in genetically triggered thoracic aortic aneurysm: role in pathogenesis and therapeutic opportunities. Redox Rep 2021; 26:45-52. [PMID: 33715602 PMCID: PMC7971305 DOI: 10.1080/13510002.2021.1899473] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: The primary objective of this review was to explore the contribution of oxidative stress to the pathogenesis of genetically-triggered thoracic aortic aneurysm (TAA). Genetically-triggered TAAs manifest substantial variability in onset, progression, and risk of aortic dissection, posing a significant clinical management challenge. There is a need for non-invasive biomarkers that predict the natural course of TAA and therapeutics that prevent aneurysm progression. Methods: An online systematic search was conducted within PubMed, MEDLINE, Scopus and ScienceDirect databases using keywords including: oxidative stress, ROS, nitrosative stress, genetically triggered thoracic aortic aneurysm, aortic dilatation, aortic dissection, Marfan syndrome, Bicuspid Aortic Valve, familial TAAD, Loeys Dietz syndrome, and Ehlers Danlos syndrome. Results: There is extensive evidence of oxidative stress and ROS imbalance in genetically triggered TAA. Sources of ROS imbalance are variable but include dysregulation of redox mediators leading to either insufficient ROS removal or increased ROS production. Therapeutic exploitation of redox mediators is being explored in other cardiovascular conditions, with potential application to TAA warranting further investigation. Conclusion: Oxidative stress occurs in genetically triggered TAA, but the precise contribution of ROS to pathogenesis remains incompletely understood. Further research is required to define causative pathological relationships in order to develop therapeutic options.
Collapse
Affiliation(s)
- Stefanie S Portelli
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Brett D Hambly
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Richmond W Jeremy
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, Australia
| | - Elizabeth N Robertson
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, Australia.,Cardiology Department, Royal Prince Alfred Hospital, Sydney, Australia
| |
Collapse
|
15
|
Dong L, Zhang H, Quan Y, Jin P, Yang J, Fan X, Fan J, Li M, Gong Y, Wang Y. Establishment and characterization of human induced pluripotent stem cell line (WMUi020-A) from a patient with bicuspid aortic valve aortopathy. Stem Cell Res 2021; 53:102260. [PMID: 33631418 DOI: 10.1016/j.scr.2021.102260] [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: 01/28/2021] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 11/25/2022] Open
Abstract
A human induced pluripotent stem cell (hiPSC) line (WMUi020-A) was generated from the aortic smooth muscle cells of a 56-year-old donor with bicuspid aortic valve and ascending aortic aneurysm. Episomal vector-mediated Non-integration iPSC reprogramming was used for this iPSC line generation. The established iPSC line highly expressed pluripotency markers with three germ-layer differentiation potential in vitro, as well as a normal karyotype. We further found that this iPSC line has a potential mutation of ROBO4 (c.161 T>C, p.Q54R), which may be useful for the disease modeling of bicuspid aortic valve aortopathy.
Collapse
Affiliation(s)
- Li Dong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Huan Zhang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Yingyi Quan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Peifeng Jin
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Jiwen Yang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Ming Li
- Cardiac Regeneration Research Institute, School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Yongyu Wang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Cardiac Regeneration Research Institute, School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.
| |
Collapse
|
16
|
Creamer TJ, Bramel EE, MacFarlane EG. Insights on the Pathogenesis of Aneurysm through the Study of Hereditary Aortopathies. Genes (Basel) 2021; 12:183. [PMID: 33514025 PMCID: PMC7912671 DOI: 10.3390/genes12020183] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Thoracic aortic aneurysms (TAA) are permanent and localized dilations of the aorta that predispose patients to a life-threatening risk of aortic dissection or rupture. The identification of pathogenic variants that cause hereditary forms of TAA has delineated fundamental molecular processes required to maintain aortic homeostasis. Vascular smooth muscle cells (VSMCs) elaborate and remodel the extracellular matrix (ECM) in response to mechanical and biochemical cues from their environment. Causal variants for hereditary forms of aneurysm compromise the function of gene products involved in the transmission or interpretation of these signals, initiating processes that eventually lead to degeneration and mechanical failure of the vessel. These include mutations that interfere with transduction of stimuli from the matrix to the actin-myosin cytoskeleton through integrins, and those that impair signaling pathways activated by transforming growth factor-β (TGF-β). In this review, we summarize the features of the healthy aortic wall, the major pathways involved in the modulation of VSMC phenotypes, and the basic molecular functions impaired by TAA-associated mutations. We also discuss how the heterogeneity and balance of adaptive and maladaptive responses to the initial genetic insult might contribute to disease.
Collapse
Affiliation(s)
- Tyler J. Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily E. Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Predoctoral Training in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
17
|
Boezio GL, Bensimon-Brito A, Piesker J, Guenther S, Helker CS, Stainier DY. Endothelial TGF-β signaling instructs smooth muscle cell development in the cardiac outflow tract. eLife 2020; 9:57603. [PMID: 32990594 PMCID: PMC7524555 DOI: 10.7554/elife.57603] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022] Open
Abstract
The development of the cardiac outflow tract (OFT), which connects the heart to the great arteries, relies on a complex crosstalk between endothelial (ECs) and smooth muscle (SMCs) cells. Defects in OFT development can lead to severe malformations, including aortic aneurysms, which are frequently associated with impaired TGF-β signaling. To better understand the role of TGF-β signaling in OFT formation, we generated zebrafish lacking the TGF-β receptor Alk5 and found a strikingly specific dilation of the OFT: alk5-/- OFTs exhibit increased EC numbers as well as extracellular matrix (ECM) and SMC disorganization. Surprisingly, endothelial-specific alk5 overexpression in alk5-/- rescues the EC, ECM, and SMC defects. Transcriptomic analyses reveal downregulation of the ECM gene fibulin-5, which when overexpressed in ECs ameliorates OFT morphology and function. These findings reveal a new requirement for endothelial TGF-β signaling in OFT morphogenesis and suggest an important role for the endothelium in the etiology of aortic malformations.
Collapse
Affiliation(s)
- Giulia Lm Boezio
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Anabela Bensimon-Brito
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Janett Piesker
- Scientific Service Group Microscopy, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Guenther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Christian Sm Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Didier Yr Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
18
|
Martínez-Micaelo N, Ligero C, Antequera-González B, Junza A, Yanes O, Alegret JM. Plasma Metabolomic Profiling Associates Bicuspid Aortic Valve Disease and Ascending Aortic Dilation with a Decrease in Antioxidant Capacity. J Clin Med 2020; 9:jcm9072215. [PMID: 32668689 PMCID: PMC7408840 DOI: 10.3390/jcm9072215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/17/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The bicuspid aortic valve (BAV) is the most common cardiac congenital disease and is associated with an increased risk of developing ascending aorta dilation; which can have fatal consequences. Currently; no established risk biomarkers exist to facilitate the diagnosis and prognosis of BAV. METHODS Using an untargeted metabolomic approach; we identified the levels of metabolites in plasma samples and compared them depending on the bicuspid or tricuspid morphology of the aortic valve. Including those patients with ascending aortic dilation and/or aortic stenosis (n = 212), we analyzed the role possibly played by alpha-Tocopherol in BAV disease; considering its association with the pathophysiological characteristics of BAV and biomarkers related to inflammation, oxidative stress and endothelial damage, as well as characteristics related to alpha-Tocopherol functionality and metabolism. RESULTS We found that BAV patients; especially those with ascending aortic dilation; presented lower antioxidant capacity; as determined by decreased plasma levels of alpha-Tocopherol; paraoxonase 1 and high-density lipoprotein (HDL), as well as increased levels of C-reactive protein (CRP; a biomarker of inflammation) and endothelial microparticles (EMPs; an endothelial damage biomarker). By applying random forest analyses; we evaluated the significant screening capacity of alpha-Tocopherol; CRP and EMPs to classify patients depending on the morphology of the aortic valve. DISCUSSION Our findings support the role of decreased antioxidant capacity; increased inflammation and endothelial damage in the pathogenesis of BAV and the progression of aortic dilation. Moreover; determining the plasma levels of alpha-Tocopherol; CRP and EMPs could improve BAV diagnosis in large populations.
Collapse
Affiliation(s)
- Neus Martínez-Micaelo
- Grup de Recerca Cardiovascular, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, 43201 Reus, Spain; (C.L.); (B.A.-G.)
- Correspondence: (N.M.-M.); (J.M.A.); Tel.: +34-977310300 (N.M.-M.); Fax: +34-977315144 (N.M.-M.)
| | - Carme Ligero
- Grup de Recerca Cardiovascular, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, 43201 Reus, Spain; (C.L.); (B.A.-G.)
- Servei de Cardiologia, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Borja Antequera-González
- Grup de Recerca Cardiovascular, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, 43201 Reus, Spain; (C.L.); (B.A.-G.)
| | - Alexandra Junza
- Metabolomics Platform, Institut d’Investigació Sanitària Pere Virgili (IISPV), Department of Electronic Engineering, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.J.); (O.Y.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Oscar Yanes
- Metabolomics Platform, Institut d’Investigació Sanitària Pere Virgili (IISPV), Department of Electronic Engineering, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.J.); (O.Y.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Josep M. Alegret
- Grup de Recerca Cardiovascular, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, 43201 Reus, Spain; (C.L.); (B.A.-G.)
- Servei de Cardiologia, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, 43201 Reus, Spain
- Correspondence: (N.M.-M.); (J.M.A.); Tel.: +34-977310300 (N.M.-M.); Fax: +34-977315144 (N.M.-M.)
| |
Collapse
|
19
|
Bons LR, Geenen LW, van den Hoven AT, Dik WA, van den Bosch AE, Duijnhouwer AL, Siebelink HMJ, Budde RPJ, Boersma E, Wessels MW, van de Laar IMBH, DeRuiter MC, Goumans MJ, Loeys BL, Roos-Hesselink JW. Blood biomarkers in patients with bicuspid aortic valve disease. J Cardiol 2020; 76:287-294. [PMID: 32265086 DOI: 10.1016/j.jjcc.2020.02.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/10/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Patients with a bicuspid aortic valve (BAV) are at risk of developing valve deterioration and aortic dilatation. We aimed to investigate whether blood biomarkers are associated with disease stage in patients with BAV. METHODS Serum levels of high sensitivity C-reactive protein (hsCRP), high sensitivity troponin T (hsTnT), N-terminal pro-B-type natriuretic peptide (NT-proBNP), and total transforming growth factor-beta 1 (TGF-ß1) were measured in adult BAV patients with valve dysfunction or aortic pathology. Age-matched general population controls were included for TGFß-1 measurements. Correlation analyses and multivariable linear regression were used to determine the association between (2log-transformed) biomarker levels and aortic valve regurgitation, aortic valve stenosis, aortic dilatation, or left ventricular function. RESULTS hsCRP and hsTnT were measured in the total group of 183 patients (median age 34 years, 25th-75th percentile 23-46), NT-proBNP in 162 patients, and TGF-ß1 beta in 108 patients. Elevated levels of NT-proBNP were found in 20% of the BAV patients, elevated hsTnT in 6%, and elevated hsCRP in 7%. Higher hsTnT levels were independently associated with aortic regurgitation [odds ratio per doubling (OR2log) 1.34, 95% CI 1.01;1.76] and higher NT-proBNP levels with aortic valve maximal velocity (ß2log 0.17, 95%CI 0.07;0.28) and aortic regurgitation (OR2log 1.41, 95%CI 1.11;1.79). Both BAV patients with (9.9 ± 2.7 ng/mL) and without aortic dilatation (10.4 ± 2.9 ng/mL) showed lower TGF-ß1 levels compared to general population controls (n = 85, 11.8 ± 3.2 ng/mL). CONCLUSIONS Higher NT-proBNP and hsTNT levels were associated with aortic valve disease in BAV patients. TGF-ß1 levels were lower in BAV patients than in the general population, and not related to aortic dilatation. Longitudinal data are needed to further investigate the prognostic value of biomarkers in these patients.
Collapse
Affiliation(s)
- Lidia R Bons
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Laurie W Geenen
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Allard T van den Hoven
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Willem A Dik
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Annemien E van den Bosch
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | | | - Ricardo P J Budde
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eric Boersma
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Clinical Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marja W Wessels
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ingrid M B H van de Laar
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marco C DeRuiter
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Laboratory for Cardiovascular Cell Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Bart L Loeys
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Jolien W Roos-Hesselink
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
20
|
Malashicheva A, Kostina A, Kostareva A, Irtyuga O, Gordeev M, Uspensky V. Notch signaling in the pathogenesis of thoracic aortic aneurysms: A bridge between embryonic and adult states. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165631. [PMID: 31816439 DOI: 10.1016/j.bbadis.2019.165631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/23/2019] [Accepted: 12/02/2019] [Indexed: 12/17/2022]
Abstract
Aneurysms of the thoracic aorta are a "silent killer" with no evident clinical signs until the fatal outcome. Molecular and genetic bases of thoracic aortic aneurysms mainly include transforming growth factor beta signaling, smooth muscle contractile units and metabolism genes, and extracellular matrix genes. In recent studies, a role of Notch signaling, among other pathways, has emerged in disease pathogenesis. Notch is a highly conserved signaling pathway that regulates the development and differentiation of many types of tissues and influences major cellular processes such as cell proliferation, differentiation and apoptosis. Mutations in several Notch signaling components have been associated with a number of heart defects, demonstrating an essential role of Notch signaling both in cardiovascular system development and its maintenance during postnatal life. This review discusses the role of Notch signaling in the pathogenesis of thoracic aortic aneurysms considering development and maintenance of the aortic root and how developmental regulations by Notch signaling may influence thoracic aortic aneurysms.
Collapse
Affiliation(s)
- Anna Malashicheva
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia; Institute of Cytology, Russian Academy of Sciences, Tikhoretskiy, 4, 194064 Saint Petersburg, Russia; Saint Petersburg State University, Department of Embryology, Universitetskaya nab., 7/9, 199034, Saint Petersburg, Russia.
| | - Aleksandra Kostina
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia; Institute of Cytology, Russian Academy of Sciences, Tikhoretskiy, 4, 194064 Saint Petersburg, Russia
| | - Anna Kostareva
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| | - Olga Irtyuga
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| | - Mikhail Gordeev
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| | - Vladimir Uspensky
- Almazov National Medical Research Centre, Akkuratova, 2, 197341 Saint Petersburg, Russia
| |
Collapse
|
21
|
Wu ZY, Trenner M, Boon RA, Spin JM, Maegdefessel L. Long noncoding RNAs in key cellular processes involved in aortic aneurysms. Atherosclerosis 2019; 292:112-118. [PMID: 31785492 PMCID: PMC6949864 DOI: 10.1016/j.atherosclerosis.2019.11.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/05/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022]
Abstract
Aortic aneurysm (AA) is a complex and dangerous vascular disease, featuring progressive and irreversible vessel dilatation. AA is typically detected either by screening, or identified incidentally through imaging studies. To date, no effective pharmacological therapies have been identified for clinical AA management, and either endovascular repair or open surgery remains the only option capable of preventing aneurysm rupture. In recent years, multiple research groups have endeavored to both identify noncoding RNAs and to clarify their function in vascular diseases, including aneurysmal pathologies. Notably, the molecular roles of noncoding RNAs in AA development appear to vary significantly between thoracic aortic aneurysms (TAAs) and abdominal aortic aneurysms (AAAs). Some microRNAs (miRNA - a non-coding RNA subspecies) appear to contribute to AA pathophysiology, with some showing major potential for use as biomarkers or as therapeutic targets. Studies of long noncoding RNAs (lncRNAs) are more limited, and their specific contributions to disease development and progression largely remain unexplored. This review aims to summarize and discuss the most current data on lncRNAs and their mediation of AA pathophysiology. This current review covers studies that have identified long non-coding RNAs in aortic aneurysm development and progression. We separately discuss transcripts and mechanisms of importance to thoracic as well as abdominal aortic aneurysms. Functional data on lncRNAs being identified are highlighted. Some have been studied in human as well as experimental models of the disease pathology.
Collapse
Affiliation(s)
- Zhi-Yuan Wu
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany
| | - Matthias Trenner
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Reinier A Boon
- Department of Physiology, VU University Medical Center Amsterdam, Netherlands; Institute for Cardiovascular Regeneration, University Frankfurt, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Germany
| | - Joshua M Spin
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany; Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
22
|
Endothelial Colony Forming Cells as an Autologous Model to Study Endothelial Dysfunction in Patients with a Bicuspid Aortic Valve. Int J Mol Sci 2019; 20:ijms20133251. [PMID: 31269711 PMCID: PMC6651394 DOI: 10.3390/ijms20133251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 02/07/2023] Open
Abstract
Bicuspid aortic valve (BAV), the most common congenital heart defect, is associated with an increased prevalence of aortic dilation, aortic rupture and aortic valve calcification. Endothelial cells (ECs) play a major role in vessel wall integrity. Little is known regarding EC function in BAV patients due to lack of patient derived primary ECs. Endothelial colony forming cells (ECFCs) have been reported to be a valid surrogate model for several cardiovascular pathologies, thereby facilitating an in vitro system to assess patient-specific endothelial dysfunction. Therefore, the aim of this study was to investigate cellular functions in ECFCs isolated from BAV patients. Outgrowth and proliferation of ECFCs from patients with BAV (n = 34) and controls with a tricuspid aortic valve (TAV, n = 10) were determined and related to patient characteristics. Interestingly, we were only able to generate ECFCs from TAV and BAV patients without aortic dilation, and failed to isolate ECFC colonies from patients with a dilated aorta. Analyzing EC function showed that while proliferation, cell size and endothelial-to-mesenchymal transition were similar in TAV and BAV ECFCs, migration and the wound healing capacity of BAV ECFCs is significantly higher compared to TAV ECFCs. Furthermore, calcification is blunted in BAV compared to TAV ECFCs. Our results reveal ECs dysfunction in BAV patients and future research is required to unravel the underlying mechanisms and to further validate ECFCs as a patient-specific in vitro model for BAV.
Collapse
|
23
|
Pasipoularides A. Clinical-pathological correlations of BAV and the attendant thoracic aortopathies. Part 2: Pluridisciplinary perspective on their genetic and molecular origins. J Mol Cell Cardiol 2019; 133:233-246. [PMID: 31175858 DOI: 10.1016/j.yjmcc.2019.05.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/10/2019] [Accepted: 05/27/2019] [Indexed: 12/30/2022]
Abstract
Bicuspid aortic valve (BAV) arises during valvulogenesis when 2 leaflets/cusps of the aortic valve (AOV) are fused together. Its clinical manifestations pertain to faulty AOV function, the associated aortopathy, and other complications surveyed in Part 1 of the present bipartite-series. Part 2 examines mainly genetic and epigenetic causes of BAV and BAV-associated aortopathies (BAVAs) and disease syndromes (BAVD). Part 1 explored the heterogeneity among subsets of patients with BAV and BAVA/BAVD, and investigated abnormal fluid dynamic stress and strain patterns sustained by the cusps. Specific BAV morphologies engender systolic outflow asymmetries, associated with abnormal aortic regional wall-shear-stress distributions and the expression/localization of BAVAs. Understanding fluid dynamic factors besides the developmental mechanisms and underlying genetics governing these congenital anomalies is necessary to explain patient predisposition to aortopathy and phenotypic heterogeneity. BAV aortopathy entails complex/multifactorial pathophysiology, involving alterations in genetics, epigenetics, hemodynamics, and in cellular and molecular pathways. There is always an interdependence between organismic developmental signals and genes-no systemic signals, no gene-expression; no active gene, no next step. An apposite signal induces the expression of the next developmental gene, which needs be expressed to trigger the next signal, and so on. Hence, embryonic, then post-partum, AOV and thoracic aortic development comprise cascades of developmental genes and their regulation. Interdependencies between them arise, entailing reciprocal/cyclical mutual interactions and adaptive feedback loops, by which developmental morphogenetic processes self-correct responding to environmental inputs/reactions. This Survey can serve as a reference point and driver for further pluridisciplinary BAV/BAVD studies and their clinical translation.
Collapse
Affiliation(s)
- Ares Pasipoularides
- Duke/NSF Center for Emerging Cardiovascular Technologies, Emeritus Faculty of Surgery and of Biomedical Engineering, Duke University School of Medicine and Graduate School, Durham, NC, USA.
| |
Collapse
|
24
|
Messner B, Bernhard D. Bicuspid aortic valve-associated aortopathy: Where do we stand? J Mol Cell Cardiol 2019; 133:76-85. [PMID: 31152748 DOI: 10.1016/j.yjmcc.2019.05.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/23/2019] [Accepted: 05/28/2019] [Indexed: 01/30/2023]
Abstract
Herein we summarize the current knowledge on the bicuspid aortic valve (BAV)-associated aortopathy regarding clinical presentation and disease sub-classification, genetic background, hemodynamics, histopathology, cells and signaling, animal models, and biomarkers. Despite enormous efforts in research in all of the above areas, important issues remain unknown: (i) what is the ontogenetic basis of BAV development? (ii) how can we explain the diversity of BAV and associated aortopathy phenotypes? (iii) what are the signaling processes in aortopathy pathogenesis and how can we interfere with these processes? Despite undoubtedly great progress that has been made in the understanding of BAV-associated aortopathy, so far researchers have put together a heap of Lego bricks, but at present it is unclear if the bricks are compatible, how they fit together, and which parts are missing to build the true model of the BAV aorta. A joint approach is needed to accelerate research progress.
Collapse
Affiliation(s)
- Barbara Messner
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - David Bernhard
- Center for Medical Research, Medical Faculty, Johannes Kepler University Linz, Linz, Austria.
| |
Collapse
|
25
|
Is there a role for autophagy in ascending aortopathy associated with tricuspid or bicuspid aortic valve? Clin Sci (Lond) 2019; 133:805-819. [PMID: 30991346 DOI: 10.1042/cs20181092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/27/2019] [Accepted: 03/18/2019] [Indexed: 01/04/2023]
Abstract
Autophagy is a conserved process by which cytoplasmatic elements are sequestered in vesicles and degraded after their fusion with lysosomes, thus recycling the precursor molecules. The autophagy-mediated removal of redundant/harmful/damaged organelles and biomolecules plays not only a replenishing function, but protects against stressful conditions through an adaptive mechanism. Autophagy, known to play a role in several pathological conditions, is now gaining increasing attention also in the perspective of the identification of the pathogenetic mechanisms at the basis of ascending thoracic aortic aneurysm (TAA), a localized or diffused dilatation of the aorta with an abnormal widening greater than 50 percent of the vessel's normal diameter. TAA is less frequent than abdominal aortic aneurysm (AAA), but is encountered with a higher percentage in patients with congenital heart disease or known genetic syndromes. Several biological aspects of TAA pathophysiology remain to be elucitated and therapeutic needs are still widely unmet. One of the most controversial and epidemiologically important forms of TAA is that associated with the congenital bicuspid malformation of the aortic valve (BAV). Dysregulated autophagy in response, for example, to wall shear stress alterations, has been demonstrated to affect the phenotype of vascular cells relevant to aortopathy, with potential consequences on signaling, remodeling, and angiogenesis. The most recent findings and hypotheses concerning the multiple aspects of autophagy and of its dysregulation are summarized, both in general and in the context of the different vascular cell types and of TAA progression, with particular reference to BAV-related aortopathy.
Collapse
|
26
|
Forte A, Balistreri CR, De Feo M, Della Corte A, Hellstrand P, Persson L, Nilsson BO. Polyamines and microbiota in bicuspid and tricuspid aortic valve aortopathy. J Mol Cell Cardiol 2019; 129:179-187. [PMID: 30825483 DOI: 10.1016/j.yjmcc.2019.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023]
Abstract
Polyamines are small aliphatic cationic molecules synthesized via a highly regulated pathway and involved in general molecular and cellular phenomena. Both mammalian cells and microorganisms synthesize polyamines, and both sources may contribute to the presence of polyamines in the circulation. The dominant location for microorganisms within the body is the gut. Accordingly, the gut microbiota probably synthesizes most of the polyamines in the circulation in addition to those produced by the mammalian host cells. Polyamines are mandatory for cellular growth and proliferation. Established evidence suggests that the polyamine spermidine prolongs lifespan and improves cardiovascular health in animal models and humans through both local mechanisms, involving improved cardiomyocyte function, and systemic mechanisms, including increased NO bioavailability and reduced systemic inflammation. Higher levels of polyamines have been detected in non-dilated aorta of patients affected by bicuspid aortic valve congenital malformation, an aortopathy associated with an increased risk for thoracic ascending aorta aneurysm. In this review, we discuss metabolism of polyamines and their potential effects on vascular smooth muscle and endothelial cell function in vascular pathology of the thoracic ascending aorta associated with bicuspid or tricuspid aortic valve.
Collapse
Affiliation(s)
- Amalia Forte
- Department of Translational Medical Sciences, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Carmela Rita Balistreri
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Marisa De Feo
- Department of Translational Medical Sciences, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Alessandro Della Corte
- Department of Translational Medical Sciences, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Per Hellstrand
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Lo Persson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Bengt-Olof Nilsson
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
27
|
Editorial commentary: Another notch for bicuspid aortic valve aortopathy? Trends Cardiovasc Med 2018; 29:69-70. [PMID: 30017228 DOI: 10.1016/j.tcm.2018.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 06/28/2018] [Indexed: 01/04/2023]
|
28
|
Defective NOTCH signaling drives increased vascular smooth muscle cell apoptosis and contractile differentiation in bicuspid aortic valve aortopathy: A review of the evidence and future directions. Trends Cardiovasc Med 2018; 29:61-68. [PMID: 30621852 DOI: 10.1016/j.tcm.2018.06.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/01/2018] [Accepted: 06/08/2018] [Indexed: 12/23/2022]
Abstract
Bicuspid aortic valve (BAV) disease remains the most common congenital cardiac disease and is associated with an increased risk of potentially fatal aortopathy including aortic aneurysm and dissection. Mutations in the NOTCH1 gene are one of only a few genetic anomalies identified in BAV disease; however evidence for defective NOTCH signaling, and its involvement in the characteristic histological changes of VSMC apoptosis and differentiation in ascending aortae of BAV patients is lacking. This review scrutinizes the evidence for the interactions of NOTCH signaling, cellular differentiation and apoptosis in the context of aortic VSMCs and provides focus for future research efforts in the diagnosis of BAV aortopathy and prevention of catastrophic complications through NOTCH signaling manipulation.
Collapse
|
29
|
Sophocleous F, Milano EG, Pontecorboli G, Chivasso P, Caputo M, Rajakaruna C, Bucciarelli-Ducci C, Emanueli C, Biglino G. Enlightening the Association between Bicuspid Aortic Valve and Aortopathy. J Cardiovasc Dev Dis 2018; 5:E21. [PMID: 29671812 PMCID: PMC6023468 DOI: 10.3390/jcdd5020021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/16/2018] [Accepted: 04/16/2018] [Indexed: 12/11/2022] Open
Abstract
Bicuspid aortic valve (BAV) patients have an increased incidence of developing aortic dilation. Despite its importance, the pathogenesis of aortopathy in BAV is still largely undetermined. Nowadays, intense focus falls both on BAV morphology and progression of valvular dysfunction and on the development of aortic dilation. However, less is known about the relationship between aortic valve morphology and aortic dilation. A better understanding of the molecular pathways involved in the homeostasis of the aortic wall, including the extracellular matrix, the plasticity of the vascular smooth cells, TGFβ signaling, and epigenetic dysregulation, is key to enlighten the mechanisms underpinning BAV-aortopathy development and progression. To date, there are two main theories on this subject, i.e., the genetic and the hemodynamic theory, with an ongoing debate over the pathogenesis of BAV-aortopathy. Furthermore, the lack of early detection biomarkers leads to challenges in the management of patients affected by BAV-aortopathy. Here, we critically review the current knowledge on the driving mechanisms of BAV-aortopathy together with the current clinical management and lack of available biomarkers allowing for early detection and better treatment optimization.
Collapse
Affiliation(s)
- Froso Sophocleous
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol BS2 89HW, UK.
| | - Elena Giulia Milano
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol BS2 89HW, UK.
- Department of Medicine, Division of Cardiology, University of Verona, 37100 Verona, Italy.
| | - Giulia Pontecorboli
- Structural Interventional Cardiology Division, Department of Experimental and Clinical Medicine, University of Florence, 50100 Florence, Italy.
| | - Pierpaolo Chivasso
- Cardiac Surgery, University Hospitals Bristol, NHS Foundation Trust, Bristol BS2 8HW, UK.
| | - Massimo Caputo
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol BS2 89HW, UK.
- Cardiac Surgery, University Hospitals Bristol, NHS Foundation Trust, Bristol BS2 8HW, UK.
| | - Cha Rajakaruna
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol BS2 89HW, UK.
- Cardiac Surgery, University Hospitals Bristol, NHS Foundation Trust, Bristol BS2 8HW, UK.
| | - Chiara Bucciarelli-Ducci
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol BS2 89HW, UK.
- Cardiac Surgery, University Hospitals Bristol, NHS Foundation Trust, Bristol BS2 8HW, UK.
| | - Costanza Emanueli
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol BS2 89HW, UK.
- Cardiac Surgery, University Hospitals Bristol, NHS Foundation Trust, Bristol BS2 8HW, UK.
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK.
| | - Giovanni Biglino
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol BS2 89HW, UK.
- Cardiorespiratory Unit, Great Ormond Street Hospital for Children, NHS Foundation Trust, London WC1N 3JH, UK.
| |
Collapse
|