1
|
Feng Y, Xue Y, Feng X, Li Z, Ren L, Guo W, Hou Y, Shu T, Zhang W, Yang Y, Zhou Y, Song K, Xiong J, Liu B, Wang J, Zhao H. Phosphodiesterase 5 inactivation in vascular smooth muscle cells aggravates aortic aneurysm and dissection. J Pathol 2025; 266:144-159. [PMID: 40145405 DOI: 10.1002/path.6411] [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: 05/08/2024] [Revised: 12/16/2024] [Accepted: 01/30/2025] [Indexed: 03/28/2025]
Abstract
Aortic aneurysm and dissection (AAD) are vascular disorders with high mortality. Previous evidence has suggested an elevated risk of AAD associated with the use of phosphodiesterase 5A (PDE5A) inhibitors. PDE5A, a cGMP-hydrolyzing enzyme, is enriched in vascular smooth muscle cells (SMCs), but the role of SMC-specific PDE5A in the pathogenesis of AAD is still unclear. In this study, PDE5A expression in human and mouse aortic tissues was analyzed by single-cell RNA sequencing (scRNA-seq), western blotting, immunofluorescence, and immunohistochemistry staining. SMC-specific PDE5A knockout (PDE5ASMC-/-) and PDE5A-overexpressing (PDE5ASMC-OE) mice were constructed and utilized, along with an AAD mouse model induced by a high-fat diet and angiotensin II (Ang II) infusion. In vivo imaging and histological analyses were performed to assess aortic pathologies. PDE5A expression was reduced in human and mouse AAD aortic tissues, primarily in SMCs. Pharmacological inhibition or genetic knockout of PDE5A in SMCs exacerbated aortic wall dilatation and elastin fiber degradation, increasing AAD incidence. In contrast, the AAD phenotype was rescued in challenged PDE5ASMC-OE mice. Mechanistically, PDE5A expression influenced myosin light chain (MLC) phosphorylation, a key regulator of SMC contractility. In AAD tissues from PDE5ASMC-/- mice, increased cGMP-dependent protein kinase (PKG) activation and decreased MLC phosphorylation indicate enhanced aortic relaxation. In conclusion, our findings suggest that PDE5A downregulation or inhibition plays a causative role in exacerbating AAD likely by potentiating cGMP/PKG-mediated aortic SMC relaxation. Our findings highlight the need for caution in the clinical use of PDE5 inhibitors in patients at risk of aortic diseases. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
MESH Headings
- Animals
- Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 5/genetics
- Humans
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/drug effects
- Aortic Dissection/pathology
- Aortic Dissection/enzymology
- Aortic Dissection/genetics
- Aortic Dissection/chemically induced
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- Aortic Aneurysm/pathology
- Aortic Aneurysm/enzymology
- Aortic Aneurysm/genetics
- Mice, Knockout
- Mice
- Disease Models, Animal
- Male
- Phosphodiesterase 5 Inhibitors/pharmacology
- Mice, Inbred C57BL
- Angiotensin II
- Aorta/pathology
- Aorta/enzymology
- Female
Collapse
Affiliation(s)
- Yuyao Feng
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, PR China
| | - Yunfei Xue
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Xiaohang Feng
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Zhiwei Li
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Luxia Ren
- Department of Vascular and Endovascular Surgery, Chinese PLA General Hospital, Beijing, PR China
| | - Wenjun Guo
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Yangfeng Hou
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Ting Shu
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
| | - Wensi Zhang
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Yang Yang
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Yitian Zhou
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Kai Song
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Jiang Xiong
- Department of Vascular and Endovascular Surgery, Chinese PLA General Hospital, Beijing, PR China
| | - Bao Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, PR China
| | - Jing Wang
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
| | - Hongmei Zhao
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Beijing, PR China
| |
Collapse
|
2
|
Kimura K, Motoyama E, Kanki S, Asano K, Sips P, Sheikh MAA, Clarin MTRDC, Raja E, Takeda M, Ishii R, Murata K, Deleeuw V, Muiño Mosquera L, De Backer J, Mizuno S, Sakai LY, Nakamura T, Yanagisawa H. Novel Aortic Dissection Model Links Endothelial Dysfunction and Immune Infiltration. Circ Res 2025. [PMID: 40365676 DOI: 10.1161/circresaha.125.326230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Aortic dissection (AD) is the separation of medial layers of the aorta and is a major cause of death in patients with connective tissue disorders such as Marfan syndrome. However, molecular triggers instigating AD, its temporospatial progression, and how vascular cells in each vessel layer interact and participate in the pathological process remain incompletely understood. To unravel the underlying molecular mechanisms of AD, we generated a spontaneous AD mouse model. METHODS We incorporated a novel missense variant (p.G234D) in FBN1, the gene for fibrillin-1, identified in a patient with nonsyndromic familial AD into mice using the CRISPR/Cas9 system. We performed molecular pathological analyses of the aortic lesions by histology, immunofluorescence staining, electron microscopy, synchrotron-based imaging, and single-cell RNA sequencing. Biochemical analysis was performed to examine the binding capacity of mutant human FBN1G234D protein to LTBPs (latent TGFβ [transforming growth factor-beta] binding proteins), and signaling pathways in the mutant aortic wall were examined by the Western blot analysis. RESULTS Fifty percent of the Fbn1G234D/G234D mutant mice died within 5 weeks of age from multiple intimomedial tears that expanded longitudinally and progressed to aortic rupture accompanied by massive immune cell infiltration. Fbn1G234D/G234D endothelial cells exhibited altered mechanosensing with loss of parallel alignment to blood flow and upregulation of VCAM-1 and ICAM-1 as early as 1 week of age. Single-cell RNA sequencing, validated by immunostaining, revealed a cluster of monocyte/macrophage predominantly in the intima at 3 weeks of age before the dissection, and the second cluster of macrophages increased during the progression of intimomedial tears, exhibiting strong CCR2+ and both M1- and M2-like features. Consistently, upregulation of MMP2/9 was observed. Biochemically, FBN1G234D lost the ability to bind to LTBP-1, -2, and -4, resulting in the downregulation of TGFβ signaling in the aortic wall. CONCLUSIONS We show that interactions involving endothelial cells and macrophages/monocytes in the intima, where the ECM (extracellular matrix) microenvironment contains reduced TGFβ signaling, contribute to the initiation of AD. Our novel AD mouse model provides a unique opportunity to identify target molecules involved in the intimomedial tears that can be utilized for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Kenichi Kimura
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
| | - Eri Motoyama
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
| | - Sachiko Kanki
- Thoracic and Cardiovascular Surgery, Osaka Medical and Pharmaceutical University, Japan (S.K.)
| | - Keiichi Asano
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
| | - Patrick Sips
- Department of Biomolecular Medicine, Ghent University, Belgium (P.S., V.D.)
| | - Md Al Amin Sheikh
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
- School of Integrative and Global Major, University of Tsukuba, Japan. (M.A.A.S., M.T.R.D.C.C.)
| | - Maria Thea Rane Dela Cruz Clarin
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
- School of Integrative and Global Major, University of Tsukuba, Japan. (M.A.A.S., M.T.R.D.C.C.)
- National Institute for Material Science, Tsukuba, Japan (M.T.R.D.C.C.)
| | - Erna Raja
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
| | - Mariko Takeda
- Department of Pharmacology, Kansai Medical University, Osaka, Japan (M.T., T.N.)
| | - Ryutaro Ishii
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
- Faculty of Medicine, University of Tsukuba, Japan. (R.I., H.Y.)
| | - Kazuya Murata
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Japan. (K.M., S.M.)
| | - Violette Deleeuw
- Department of Biomolecular Medicine, Ghent University, Belgium (P.S., V.D.)
| | - Laura Muiño Mosquera
- Department of Pediatric Cardiology and Center for Medical Genetics, Ghent University Hospital, Belgium. (L.M.M.)
| | - Julie De Backer
- Department of Cardiology and Center for Medical Genetics, Ghent University Hospital, Belgium. (J.D.B.)
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Japan. (K.M., S.M.)
| | - Lynn Y Sakai
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland (L.Y.S.)
| | - Tomoyuki Nakamura
- Department of Pharmacology, Kansai Medical University, Osaka, Japan (M.T., T.N.)
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
- Faculty of Medicine, University of Tsukuba, Japan. (R.I., H.Y.)
| |
Collapse
|
3
|
Deng J, Tian L, Chi H, Chen L, Wang J, Xue Y, Zhao Q, Zheng N, Dong J, Li J, Guo W, Xiao C, Yang M. Establishment of a murine chronic proximal thoracic aortic aneurysm model by combining periaortic elastase application with oral BAPN administration. Eur J Pharmacol 2025; 999:177678. [PMID: 40320110 DOI: 10.1016/j.ejphar.2025.177678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 05/09/2025]
Abstract
This study aimed to develop a chronic proximal thoracic aortic aneurysm (PTAA) model by combining periaortic elastase application with oral administration of 3-aminopropionitrile fumarate salt (BAPN) after surgery. Sixty 8-week-old C57BL/6J male mice were divided into four groups: Sham, Sham + BAPN, Elastase, and Elastase + BAPN. High-resolution micro-ultrasound was performed on days 7, 14, 21, 28, 56, and 90 post-operation to measure aortic diameter. Histopathological, transcriptomic, and bioinformatics analyses were conducted to assess the model's relevance to human PTAA. The operative mortality rate was 10 % (6/60). During follow-up, 4 animals in the elastase + BAPN group and 1 in the elastase group died from aortic rupture. Significant continuous dilation of the proximal thoracic aorta was observed only in the elastase + BAPN group, with average dilation rates of 116.60 %, 178.99 %, and 231.90 % on days 28, 56, and 90, respectively, compared to 66.46 %, 61.13 %, and 68.73 % in the elastase group. Histopathology revealed greater aortic wall thickening, collagen deposition, MMP2 expression, elastin degradation, smooth muscle cell loss, calcification, and immune cell infiltration in the elastase + BAPN group. Transcriptomic analysis identified 3039 differentially expressed genes, enriched in immune and inflammation-related pathways. Weighted gene co-expression network analysis showed significant overlap in the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment results between human and murine PTAA-related gene modules which were most positively correlated with PTAA diameters. This study establishes a chronic PTAA model that mimics key features of human disease, providing a valuable tool for investigating PTAA mechanisms and developing new therapies.
Collapse
Affiliation(s)
- Jianqing Deng
- Department of Cardiovascular Surgery, Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, No.6 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Lei Tian
- General Surgery Department, The Sixth Medical Center of the People's Liberation Army General Hospital, No.6 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Haitao Chi
- Department of Cardiovascular Surgery, Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, No.6 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Lei Chen
- Department of Cardiovascular Surgery, Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, No.6 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Junhui Wang
- Department of Cardiovascular Surgery, Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, No.6 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Yan Xue
- Department of Cardiovascular Surgery, Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, No.6 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Qiang Zhao
- Department of Cardiovascular Surgery, Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, No.6 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Nan Zheng
- Department of Cardiovascular Surgery, Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, No.6 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Jie Dong
- Department of Cardiovascular Surgery, Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, No.6 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Jiaying Li
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, No. 100 West 4th Ring Road Middle, Fengtai District, Beijing, 100071, China
| | - Wei Guo
- Department of Vascular and Endovascular Surgery, The First Medical Center of PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Cangsong Xiao
- Department of Cardiovascular Surgery, Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, No.6 Fucheng Road, Haidian District, Beijing, 100048, China.
| | - Ming Yang
- Department of Cardiovascular Surgery, Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, No.6 Fucheng Road, Haidian District, Beijing, 100048, China.
| |
Collapse
|
4
|
Rolf-Pissarczyk M, Schussnig R, Fries TP, Fleischmann D, Elefteriades JA, Humphrey JD, Holzapfel GA. Mechanisms of aortic dissection: From pathological changes to experimental and in silico models. PROGRESS IN MATERIALS SCIENCE 2025; 150:101363. [PMID: 39830801 PMCID: PMC11737592 DOI: 10.1016/j.pmatsci.2024.101363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Aortic dissection continues to be responsible for significant morbidity and mortality, although recent advances in medical data assimilation and in experimental and in silico models have improved our understanding of the initiation and progression of the accumulation of blood within the aortic wall. Hence, there remains a pressing necessity for innovative and enhanced models to more accurately characterize the associated pathological changes. Early on, experimental models were employed to uncover mechanisms in aortic dissection, such as hemodynamic changes and alterations in wall microstructure, and to assess the efficacy of medical implants. While experimental models were once the only option available, more recently they are also being used to validate in silico models. Based on an improved understanding of the deteriorated microstructure of the aortic wall, numerous multiscale material models have been proposed in recent decades to study the state of stress in dissected aortas, including the changes associated with damage and failure. Furthermore, when integrated with accessible patient-derived medical data, in silico models prove to be an invaluable tool for identifying correlations between hemodynamics, wall stresses, or thrombus formation in the deteriorated aortic wall. They are also advantageous for model-guided design of medical implants with the aim of evaluating the deployment and migration of implants in patients. Nonetheless, the utility of in silico models depends largely on patient-derived medical data, such as chosen boundary conditions or tissue properties. In this review article, our objective is to provide a thorough summary of medical data elucidating the pathological alterations associated with this disease. Concurrently, we aim to assess experimental models, as well as multiscale material and patient data-informed in silico models, that investigate various aspects of aortic dissection. In conclusion, we present a discourse on future perspectives, encompassing aspects of disease modeling, numerical challenges, and clinical applications, with a particular focus on aortic dissection. The aspiration is to inspire future studies, deepen our comprehension of the disease, and ultimately shape clinical care and treatment decisions.
Collapse
Affiliation(s)
| | - Richard Schussnig
- High-Performance Scientific Computing, University of Augsburg, Germany
- Institute of Structural Analysis, Graz University of Technology, Austria
| | - Thomas-Peter Fries
- Institute of Structural Analysis, Graz University of Technology, Austria
| | - Dominik Fleischmann
- 3D and Quantitative Imaging Laboratory, Department of Radiology, Stanford University, USA
| | | | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, USA
| | - Gerhard A. Holzapfel
- Institute of Biomechanics, Graz University of Technology, Austria
- Department of Structural Engineering, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
5
|
Sun W, Yu C, Guo J, Wang H, Dou S, Zhang Y, Zheng J, Gao Y. Angiotensin II promotes intramural hematoma of aorta in juvenile mice at early stage. Ultrastruct Pathol 2025; 49:148-157. [PMID: 40103374 DOI: 10.1080/01913123.2025.2474447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
OBJECTIVES Intramural hematoma (IMH) is a serious aortic condition characterized by the presence of a contained hematoma within the aortic media. However, the animal model with a high incidence of IMH was lacking, and the specific pathological characteristics of IMH have not been thoroughly characterized. METHODS AND RESULTS We conducted an experimental study using 4-week-old male, 4-week-old female, and 8-week-old male C57BL/6J mice. These mice were subjected to angiotensin II infusion at a rate of 1000 ng/kg/min for a period of 4 days. In situ imaging was performed, and aorta was harvested and serially sectioned. Histological staining and immunostaining techniques were employed, and the subcellular structure was examined using transmission electron microscopy. Our findings revealed that 4-week-old male mice exhibited a higher susceptibility to angiotensin II-induced IMH, characterized by more circumferential appearances and larger affected areas. Furthermore, IMH was more likely to occur in the upper segment of the descending aorta rather than the lower segment. Within the IMH, older fibrinous thrombus was predominantly observed near the adventitia, while younger red thrombus was more prevalent near the lumen. Additionally, platelet activation and degranulation were observed, along with fibrin cross-linking and thrombus organization, indicating a potential relationship between platelet activation and the progression of IMH. CONCLUSION Our study demonstrated that angiotensin II infusion promoted the development of IMH during the early stages, especially in juvenile mice. Furthermore, the presence of platelet activation and thrombus organization suggested their potential involvement in the progression of IMH.
Collapse
Affiliation(s)
- Weiliang Sun
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Changan Yu
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Jing Guo
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Huina Wang
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Shurui Dou
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Yuting Zhang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Jingang Zheng
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Yanxiang Gao
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
6
|
Sumi T, Kugo H, Higashihara M, Moriyama T, Zaima N. Food components and abdominal aortic aneurysm. Biosci Biotechnol Biochem 2025; 89:152-160. [PMID: 39375828 DOI: 10.1093/bbb/zbae141] [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: 08/28/2024] [Accepted: 09/28/2024] [Indexed: 10/09/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a disease in which the abdominal aorta expands irreversibly and ruptures. At present, no preventive methods are available for this disease. Among potential risk factors, certain foods are considered to play important roles in the development of AAA. Epidemiological studies suggest a close relationship between AAA and dietary habits. Experimental studies have clarified potential suppressive or progressive food components for AAA. In this review, a summary of studies related to nutritional science in the fields of AAA and/or aortic degeneration are provided.
Collapse
Affiliation(s)
- Tomoko Sumi
- Graduate School of Agriculture, Kindai University, Nara City, Nara, Japan
| | - Hirona Kugo
- Graduate School of Agriculture, Kindai University, Nara City, Nara, Japan
| | - Mayo Higashihara
- Graduate School of Agriculture, Kindai University, Nara City, Nara, Japan
| | - Tatsuya Moriyama
- Graduate School of Agriculture, Kindai University, Nara City, Nara, Japan
- Agricultural Technology and Innovation Research Institute, Kindai University, Nara, Japan
| | - Nobuhiro Zaima
- Graduate School of Agriculture, Kindai University, Nara City, Nara, Japan
- Agricultural Technology and Innovation Research Institute, Kindai University, Nara, Japan
| |
Collapse
|
7
|
Alloisio M, Siika A, Freiholtz D, Franco-Cereceda A, Roy J, Björck HM, Gasser TC. Fracture properties of porcine versus human thoracic aortas from tricuspid/bicuspid aortic valve patients via symmetry-constraint Compact Tension testing. Sci Rep 2025; 15:667. [PMID: 39753641 PMCID: PMC11699116 DOI: 10.1038/s41598-024-83233-6] [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: 10/17/2024] [Accepted: 12/12/2024] [Indexed: 01/06/2025] Open
Abstract
Aneurysm rupture is a life-threatening event, yet its underlying mechanisms remain largely unclear. This study investigated the fracture properties of the thoracic aneurysmatic aorta (TAA) using the symmetry-constraint Compact Tension (symconCT) test and compared results to native and enzymatic-treated porcine aortas' tests. With age, the aortic stiffness increased, and tissues ruptured at lower fracture energy [Formula: see text]. Patients with bicuspid aortic valves were more sensitive to age, had stronger aortas and required more [Formula: see text] than tricuspid valves individuals (peak load: axial loading 4.42 ± 1.56 N vs 2.51 ± 1.60 N; circumferential loading 5.76 ± 2.43 N vs 4.82 ± 1.49 N. Fracture energy: axial loading 1.92 ± 0.60 kJ m-2 vs 0.74 ± 0.50 kJ m-2; circumferential loading 2.12 ± 2.39 kJ m-2 vs 1.47 ± 0.91 kJ m-2). Collagen content partly explained the variability in [Formula: see text], especially in bicuspid cases. Besides the primary crack, TAAs and enzymatic-treated porcine aortas displayed diffuse and shear-dominated dissection and tearing. As human tissue tests resembled enzymatic-treated porcine aortas, microstructural degeneration, including elastin loss and collagen degeneration, seems to be the main cause of TAA wall weakening. Additionally, a tortuous crack developing during the symconCT test reflected intact fracture toughening mechanisms and might characterize a healthier aorta.
Collapse
Affiliation(s)
- Marta Alloisio
- Department of Engineering Mechanics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Antti Siika
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - David Freiholtz
- Section of Cardiothoracic Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Division of Cardiology, Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Anders Franco-Cereceda
- Section of Cardiothoracic Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Joy Roy
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Vascular Surgery, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Hanna M Björck
- Division of Cardiology, Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Solna, Stockholm, Sweden
| | - T Christian Gasser
- Department of Engineering Mechanics, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
8
|
Zhang ZB, Cheng YW, Xu L, Li JQ, Pan X, Zhu M, Chen XH, Sun AJ, Lin JR, Gao PJ. Activation of β3-adrenergic receptor by mirabegron prevents aortic dissection/aneurysm by promoting lymphangiogenesis in perivascular adipose tissue. Cardiovasc Res 2024; 120:2307-2319. [PMID: 39288197 DOI: 10.1093/cvr/cvae213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/06/2024] [Accepted: 07/23/2024] [Indexed: 09/19/2024] Open
Abstract
AIMS β3-Adrenergic receptor (β3-AR) is essential for cardiovascular homoeostasis through regulating adipose tissue function. Perivascular adipose tissue (PVAT) has been implicated in the pathogenesis of aortic dissection and aneurysm (AD/AA). Here, we aim to investigate β3-AR activation-mediated PVAT function in AD/AA. METHODS AND RESULTS Aortas from patients with thoracic aortic dissection (TAD) were collected to detect β3-AR expression in PVAT. ApoE-/- and β-aminopropionitrile monofumarate (BAPN)-treated C57BL/6 mice were induced with Angiotensin II (AngII) to simulate AD/AA and subsequently received either placebo or mirabegron, a β3-AR agonist. The results demonstrated an up-regulation of β3-AR in PVAT of TAD patients and AD/AA mice. Moreover, activation of β3-AR by mirabegron significantly prevented AngII-induced AD/AA formation in mice. RNA-sequencing analysis of adipocytes from PVAT revealed a notable increase of the lymphangiogenic factor, vascular endothelial growth factor C (VEGF-C), in mirabegron-treated mice. Consistently, enhanced lymphangiogenesis was found in PVAT with mirabegron treatment. Mechanistically, the number of CD4+/CD8+ T cells and CD11c+ cells was reduced in PVAT but increased in adjacent draining lymph nodes of mirabegron-treated mice, indicating the improved draining and clearance of inflammatory cells in PVAT by lymphangiogenesis. Importantly, adipocyte-specific VEGF-C knockdown by the adeno-associated virus system restrained lymphangiogenesis and exacerbated inflammatory cell infiltration in PVAT, which ultimately abolished the protection of mirabegron on AD/AA. In addition, the conditional medium derived from mirabegron-treated adipocytes activated the proliferation and tube formation of LECs, which was abrogated by the silencing of VEGF-C in adipocytes. CONCLUSION Our findings illustrated the therapeutic potential of β3-AR activation by mirabegron on AD/AA, which promoted lymphangiogenesis by increasing adipocyte-derived VEGF-C and, therefore, ameliorated PVAT inflammation.
Collapse
MESH Headings
- Animals
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/prevention & control
- Aortic Dissection/chemically induced
- Aortic Dissection/physiopathology
- Aortic Dissection/genetics
- Lymphangiogenesis/drug effects
- Adrenergic beta-3 Receptor Agonists/pharmacology
- Mice, Inbred C57BL
- Receptors, Adrenergic, beta-3/metabolism
- Receptors, Adrenergic, beta-3/genetics
- Acetanilides/pharmacology
- Vascular Endothelial Growth Factor C/metabolism
- Vascular Endothelial Growth Factor C/genetics
- Humans
- Male
- Disease Models, Animal
- Adipose Tissue/metabolism
- Adipose Tissue/drug effects
- Adipose Tissue/pathology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/prevention & control
- Aortic Aneurysm, Thoracic/physiopathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/chemically induced
- Aortic Aneurysm, Thoracic/drug therapy
- Mice, Knockout, ApoE
- Thiazoles/pharmacology
- Signal Transduction
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aorta, Thoracic/drug effects
- Angiotensin II
- Adipocytes/metabolism
- Adipocytes/pathology
- Adipocytes/drug effects
- Cells, Cultured
- Mice
- Lymphatic Vessels/metabolism
- Lymphatic Vessels/drug effects
- Lymphatic Vessels/pathology
- Lymphatic Vessels/physiopathology
- Female
- Middle Aged
Collapse
Affiliation(s)
- Ze-Bei Zhang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Yu-Wen Cheng
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian Xu
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Jia-Qi Li
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Xin Pan
- Department of Gerontology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Zhu
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Xiao-Hui Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Ai-Jun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Jing-Rong Lin
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| |
Collapse
|
9
|
Wu H, Li Z, Yang L, He L, Liu H, Yang S, Xu Q, Li Y, Li W, Li Y, Gong Z, Shen Y, Yang X, Huang J, Yu F, Li L, Zhu J, Sun L, Fu Y, Kong W. ANK Deficiency-Mediated Cytosolic Citrate Accumulation Promotes Aortic Aneurysm. Circ Res 2024; 135:1175-1192. [PMID: 39513269 DOI: 10.1161/circresaha.124.325152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Disturbed metabolism and transport of citrate play significant roles in various pathologies. However, vascular citrate regulation and its potential role in aortic aneurysm (AA) development remain poorly understood. METHODS Untargeted metabolomics by mass spectrometry was applied to identify upregulated metabolites of the tricarboxylic acid cycle in AA tissues of mice. To investigate the role of citrate and its transporter ANK (progressive ankylosis protein) in AA development, vascular smooth muscle cell (VSMC)-specific Ank-knockout mice were used in both Ang II (angiotensin II)- and CaPO4-induced AA models. RESULTS Citrate was abnormally increased in both human and murine aneurysmal tissues, which was associated with downregulation of ANK, a citrate membrane transporter, in VSMCs. The knockout of Ank in VSMCs promoted AA formation in both Ang II- and CaPO4-induced AA models, while its overexpression inhibited the development of aneurysms. Mechanistically, ANK deficiency in VSMCs caused abnormal cytosolic accumulation of citrate, which was cleaved into acetyl coenzyme A and thus intensified histone acetylation at H3K23, H3K27, and H4K5. Cleavage under target and tagmentation analysis further identified that ANK deficiency-induced histone acetylation activated the transcription of inflammatory genes in VSMCs and thus promoted a citrate-related proinflammatory VSMC phenotype during aneurysm diseases. Accordingly, suppressing citrate cleavage to acetyl coenzyme A downregulated inflammatory gene expression in VSMCs and restricted ANK deficiency-aggravated AA formation. CONCLUSIONS Our studies define the pathogenic role of ANK deficiency-induced cytosolic citrate accumulation in AA pathogenesis and an undescribed citrate-related proinflammatory VSMC phenotype. Targeting ANK-mediated citrate transport may emerge as a novel diagnostic and therapeutic strategy in AA.
Collapse
MESH Headings
- Animals
- Mice
- Citric Acid/metabolism
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice, Knockout
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/genetics
- Aortic Aneurysm/pathology
- Aortic Aneurysm/etiology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Inbred C57BL
- Cytosol/metabolism
- Male
- Cells, Cultured
- Acetylation
- Acetyl Coenzyme A/metabolism
- Disease Models, Animal
- Histones/metabolism
Collapse
Affiliation(s)
- Hao Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China (L.Y.)
| | - Lin He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Hao Liu
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, China (H.L., Q.X., J.Z.)
| | - Shiyu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Qinfeng Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yanjie Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wenqiang Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yiran Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
- Hwamei College of Life and Health Sciences, Zhejiang Wanli University, Ningbo, China (Z.G.)
| | - Yicong Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xueyuan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiaqi Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Li Li
- Department of Pathology, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.L.)
| | - Junming Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Luyang Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | | |
Collapse
|
10
|
Liao M, Zou S, Wu J, Bai J, Liu Y, Zhi K, Qu L. METTL3-mediated m6A modification of NORAD inhibits the ferroptosis of vascular smooth muscle cells to attenuate the aortic dissection progression in an YTHDF2-dependent manner. Mol Cell Biochem 2024; 479:3471-3487. [PMID: 38383916 DOI: 10.1007/s11010-024-04930-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/05/2024] [Indexed: 02/23/2024]
Abstract
Ferroptosis of vascular smooth muscle cells (VSMCs) is related to the incidence of aortic dissection (AD). Long non-coding RNA (lncRNA) NORAD plays a crucial role in the progression of various diseases. The present study aimed to investigate the effects of NORAD on the ferroptosis of VSMCs and the molecular mechanisms. The expression of NORAD, HUR, and GPX4 was detected using quantitative real-time PCR (qPCR) or western blot. Ferroptosis was evaluated by detecting lactate dehydrogenase (LDH) activity, lipid reactive oxygen species (ROS), malonaldehyde (MDA) content, L-Glutathione (GSH) level, Fe2+ content, and ferroptosis-related protein levels. The molecular mechanism was assessed using RNA pull-down, RNA-binding protein immunoprecipitation (RIP), and luciferase reporter assay. The histology of aortic tissues was assessed using H&E, elastic Verhoeff-Van Gieson (EVG), and Masson staining assays. The data indicated that NORAD was downregulated in patients with AD and AngII-treated VSMCs. Overexpression of NORAD promoted VSMC growth and inhibited the ferroptosis induced by AngII. Mechanistically, NORAD interacted with HUR, which promoted GPX4 mRNA stability and elevated GPX4 levels. Knockdown of GPX4 abrogated the effects of NORAD on cell growth and ferroptosis of AngII-treated VSMCs. Moreover, METTL3 promoted m6A methylation of NORAD in an YTHDF2-dependent manner. In addition, NORAD attenuated AAD symptoms, incidence, histopathology, inflammation, and ferroptosis in AAD mice. In conclusion, METTL3-mediated NORAD inhibited ferroptosis of VSMCs via the HUR/GPX4 axis and decelerated AAD progression, suggesting that NORAD may be an AD therapeutic target.
Collapse
Affiliation(s)
- Mingfang Liao
- Department of Vascular & Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Sili Zou
- Department of Vascular & Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Jianjin Wu
- Department of Vascular & Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Jun Bai
- Department of Vascular & Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Yandong Liu
- Department of Vascular & Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Kangkang Zhi
- Department of Vascular & Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Lefeng Qu
- Department of Vascular & Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China.
| |
Collapse
|
11
|
Nguyen TLL, Van Nguyen D, Jin Y, Kim L, Heo KS. Potential effects of a human milk oligosaccharide 6'-sialyllactose on angiotensin II-induced aortic aneurysm via p90RSK/TGF-β/SMAD2 signaling pathway. Arch Pharm Res 2024; 47:854-869. [PMID: 39463210 DOI: 10.1007/s12272-024-01515-z] [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: 07/07/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
The aberrant phenotypic transformation of vascular smooth muscle cells (VSMCs) is a key factor in the formation of aortic aneurysm (AA). This study aimed to explore the effects of 6'-sialyllactose (6'-SL), a human milk oligosaccharide, on angiotensin II (Ang II)-induced VSMC dysfunction and AA formation both in vitro and in vivo. An AA model was established in male C57BL/6 mice challenged with Ang II via osmotic pumps and a lysyl oxidase inhibitor, β-aminopropionitrile (BAPN), in drinking water. The mice were administered with 6'-SL, FMK (a p90RSK inhibitor), or losartan (as a positive control). In vitro, VSMCs were pretreated with 6'-SL before Ang II stimulation. We found that p90RSK inhibition abolished Ang II/BAPN-induced thoracic AA and abdominal AA formation. Treatment with 100 mg/kg 6'-SL significantly attenuated Ang II/BAPN-induced aortic dilatation. 6'-SL attenuated Ang II-induced collagen deposition, calcification, and immune cell accumulation. Consistently, 6'-SL downregulated p-p90RSK, p90RSK, and p-SMAD2, and mitigated VSMC contractility loss, as indicated by α-SMA expression in vivo. Interestingly, Ang II-induced transforming growth factor-beta (TGF-β) signaling pathway was suppressed by p90RSK inhibition in VSMCs. 6'-SL treatment significantly reduced TGF-β/SMAD2 targets, including dedifferentiation markers such as osteopontin and vimentin, and elastin degradation factors MMP2 and MMP9. Overexpression of p90RSK in VSMCs enhanced TGF-β and abrogated the effects of 6'-SL. Furthermore, 6'-SL co-treatment abolished high phosphate-induced calcification in vitro via p90RSK/TGF-β signaling pathway. Altogether, our findings suggest that 6'-SL could be a potential therapeutic candidate for protecting against Ang II-induced AA formation by inhibiting the p90RSK/TGF-β/SMAD2 signaling pathway.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Mice, Inbred C57BL
- Male
- Mice
- Aortic Aneurysm/chemically induced
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/prevention & control
- Aortic Aneurysm/pathology
- Aortic Aneurysm/drug therapy
- Signal Transduction/drug effects
- Transforming Growth Factor beta/metabolism
- Humans
- Smad2 Protein/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors
- Oligosaccharides/pharmacology
- Cells, Cultured
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Disease Models, Animal
Collapse
Affiliation(s)
- Thuy Le Lam Nguyen
- College of Pharmacy, Chungnam National University, Daejeon, 34134, South Korea
| | - Dung Van Nguyen
- College of Pharmacy, Chungnam National University, Daejeon, 34134, South Korea
| | - Yujin Jin
- College of Pharmacy, Chungnam National University, Daejeon, 34134, South Korea
| | - Lila Kim
- NeuraGene Inc., 17 Techno 2-Ro, Yuseong-Gu, Daejeon, 34025, South Korea
| | - Kyung-Sun Heo
- College of Pharmacy, Chungnam National University, Daejeon, 34134, South Korea.
| |
Collapse
|
12
|
Bäumler K, Phillips EH, Grande Gutiérrez N, Fleischmann D, Marsden AL, Goergen CJ. Longitudinal investigation of aortic dissection in mice with computational fluid dynamics. Comput Methods Biomech Biomed Engin 2024; 27:2161-2174. [PMID: 37897230 DOI: 10.1080/10255842.2023.2274281] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/16/2023] [Accepted: 09/30/2023] [Indexed: 10/29/2023]
Abstract
Predicting late adverse events in aortic dissections is challenging. One commonly observed risk factor is partial thrombosis of the false lumen. In this study we investigated false lumen thrombus progression over 7 days in four mice with angiotensin II-induced aortic dissection. We performed computational fluid dynamic simulations with subject-specific boundary conditions from velocity and pressure measurements. We investigated endothelial cell activation potential, mean velocity, thrombus formation potential, and other hemodynamic factors. Our findings support the hypothesis that flow stagnation is the predominant hemodynamic factor driving a large thrombus ratio in false lumina, particularly those with a single fenestration.
Collapse
Affiliation(s)
| | - Evan H Phillips
- Weldon School of Biomedical Engineering, Purdue University, IN, USA
- Department of Pharmaceutical Sciences, University of IL at Chicago, IL, USA
| | | | | | - Alison L Marsden
- Department of Bioengineering, Stanford University, CA, USA
- Department of Pediatrics (Cardiology), Stanford University, CA, USA
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, IN, USA
| |
Collapse
|
13
|
Golledge J, Lu HS, Curci JA. Small AAAs: Recommendations for Rodent Model Research for the Identification of Novel Therapeutics. Arterioscler Thromb Vasc Biol 2024; 44:1467-1473. [PMID: 38924435 PMCID: PMC11384288 DOI: 10.1161/atvbaha.124.320823] [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: 03/04/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
CLINICAL PROBLEM Most abdominal aortic aneurysms (AAAs) are small with low rupture risk (<1%/y) when diagnosed but slowly expand to ≥55 mm and undergo surgical repair. Patients and clinicians require medications to limit AAA growth and rupture, but drugs effective in animal models have not translated to patients. RECOMMENDATIONS FOR INCREASING TRANSLATION FROM MOUSE MODELS Use models that simulate human AAA tissue pathology, growth patterns, and rupture; focus on the clinically relevant outcomes of growth and rupture; design studies with the rigor required of human clinical trials; monitor AAA growth using reproducible ultrasound; and perform studies in both males and females. SUMMARY OF STRENGTHS AND WEAKNESSES OF MOUSE MODELS The aortic adventitial elastase oral β-aminopropionitrile model has many strengths including simulating human AAA pathology and modeling prolonged aneurysm growth. The Ang II (angiotensin II) model performed less well as it better simulates acute aortic syndrome than AAA. The elastase plus TGFβ (transforming growth factor-β) blocking antibody model displays a high rupture rate, making prolonged monitoring of AAA growth not feasible. The elastase perfusion and calcium chloride models both display limited AAA growth.
Collapse
Affiliation(s)
- Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia
- The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, Queensland, Australia
- The Australian Institute of Tropical Health and Medicine, Townsville, Queensland, Australia
| | - Hong S. Lu
- Saha Cardiovascular Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - John A. Curci
- Department of Vascular Surgery, Vanderbilt Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Section of Vascular Surgery, Department of Surgery, Tennessee Valley Health System, VA Medical Center, Nashville, Tennessee, United States
| |
Collapse
|
14
|
Hassab AH, Hur DJ, Vallabhajosyula P, Tellides G, Assi R. Intimomedial tears of the aorta heal by smooth muscle cell-mediated fibrosis without atherosclerosis. JCI Insight 2024; 9:e172437. [PMID: 38592807 PMCID: PMC11141924 DOI: 10.1172/jci.insight.172437] [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: 05/19/2023] [Accepted: 04/03/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUNDDisease of the aorta varies from atherosclerosis to aneurysms, with complications including rupture, dissection, and poorly characterized limited tears. We studied limited tears without any mural hematoma, termed intimomedial tears, to gain insight into aortic vulnerability to excessive wall stresses. Our premise is that minimal injuries in aortas with sufficient medial resilience to prevent tear progression correspond to initial mechanisms leading to complete structural failure in aortas with significantly compromised medial resilience.METHODSIntimomedial tears were macroscopically identified in 9 of 108 ascending aortas after surgery and analyzed by histology and immunofluorescence confocal microscopy.RESULTSNonhemorrhagic, nonatheromatous tears correlated with advanced aneurysmal disease and most lacked distinctive symptoms or radiological signs. Tears traversed the intima and part of the subjacent media, while the resultant defects were partially or completely filled with neointima characterized by differentiated smooth muscle cells, scattered leukocytes, dense fibrosis, and absent elastic laminae despite tropoelastin synthesis. Healed lesions contained organized fibrin at tear edges without evidence of plasma and erythrocyte extravasation or lipid accumulation.CONCLUSIONThese findings suggest a multiphasic model of aortic wall failure in which primary lesions of intimomedial tears either heal if the media is sufficiently resilient or progress as dissection or rupture by medial delamination and tear completion, respectively. Moreover, mural incorporation of thrombus and cellular responses to injury, two historically important concepts in atheroma pathogenesis, contribute to vessel wall repair with adequate conduit function, but even together are not sufficient to induce atherosclerosis.FUNDINGNIH (R01-HL146723, R01-HL168473) and Yale Department of Surgery.
Collapse
Affiliation(s)
| | - David J. Hur
- Department of Internal Medicine (Cardiovascular Medicine), Yale School of Medicine, New Haven, Connecticut, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | | | - George Tellides
- Department of Surgery (Cardiac), and
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
- Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Roland Assi
- Department of Surgery (Cardiac), and
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
- Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
15
|
Shu T, Zhou Y, Yan C. The perspective of cAMP/cGMP signaling and cyclic nucleotide phosphodiesterases in aortic aneurysm and dissection. Vascul Pharmacol 2024; 154:107278. [PMID: 38262506 PMCID: PMC10939884 DOI: 10.1016/j.vph.2024.107278] [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: 12/13/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/25/2024]
Abstract
Aortic aneurysm (AA) and dissection (AD) are aortic diseases caused primarily by medial layer degeneration and perivascular inflammation. They are lethal when the rupture happens. Vascular smooth muscle cells (SMCs) play critical roles in the pathogenesis of medial degeneration, characterized by SMC loss and elastin fiber degradation. Many molecular pathways, including cyclic nucleotide signaling, have been reported in regulating vascular SMC functions, matrix remodeling, and vascular structure integrity. Intracellular cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) are second messengers that mediate intracellular signaling transduction through activating effectors, such as protein kinase A (PKA) and PKG, respectively. cAMP and cGMP are synthesized by adenylyl cyclase (AC) and guanylyl cyclase (GC), respectively, and degraded by cyclic nucleotide phosphodiesterases (PDEs). In this review, we will discuss the roles and mechanisms of cAMP/cGMP signaling and PDEs in AA/AD formation and progression and the potential of PDE inhibitors in AA/AD, whether they are beneficial or detrimental. We also performed database analysis and summarized the results showing PDEs with significant expression changes under AA/AD, which should provide rationales for future research on PDEs in AA/AD.
Collapse
Affiliation(s)
- Ting Shu
- Aab Cardiovascular Research Institute, School of Medicine and Dentistry, University of Rochester, New York, United States
| | - Yitian Zhou
- Peking Union Medical College, MD Program, Beijing, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, School of Medicine and Dentistry, University of Rochester, New York, United States.
| |
Collapse
|
16
|
Ibrahim N, Bleichert S, Klopf J, Kurzreiter G, Hayden H, Knöbl V, Artner T, Krall M, Stiglbauer-Tscholakoff A, Oehler R, Petzelbauer P, Busch A, Bailey MA, Eilenberg W, Neumayer C, Brostjan C. Reducing Abdominal Aortic Aneurysm Progression by Blocking Neutrophil Extracellular Traps Depends on Thrombus Formation. JACC Basic Transl Sci 2024; 9:342-360. [PMID: 38559632 PMCID: PMC10978405 DOI: 10.1016/j.jacbts.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 10/02/2023] [Accepted: 11/01/2023] [Indexed: 04/04/2024]
Abstract
Neutrophil extracellular traps (NETs) are implicated in the pathogenesis of abdominal aortic aneurysm (AAA), located in adventitia and intraluminal thrombus. We compared the therapeutic potential of targeting upstream or downstream effector molecules of NET formation in 2 murine AAA models based on angiotensin II or peri-adventitial elastase application. In both models, NETs were detected in formed aneurysms at treatment start. Although NET inhibitors failed in the elastase model, they prevented progression of angiotensin II-induced aneurysms with thrombus, which resembles established human disease (including thrombus development). Blockade of upstream NET mediators was more effective than interference with downstream NET molecules.
Collapse
Affiliation(s)
- Nahla Ibrahim
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Sonja Bleichert
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Johannes Klopf
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Gabriel Kurzreiter
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Hubert Hayden
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Viktoria Knöbl
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Tyler Artner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Moritz Krall
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Alexander Stiglbauer-Tscholakoff
- Division of Cardiovascular and Interventional Radiology, Division of Molecular and Gender Imaging, Department of Biomedical Imaging and Image Guided Therapy, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Rudolf Oehler
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Peter Petzelbauer
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Albert Busch
- Department for Visceral, Thoracic and Vascular Surgery, Technical University of Dresden and University Hospital Carl-Gustav Carus, Dresden, Germany
| | - Marc A. Bailey
- Leeds Institute for Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom
- Leeds Vascular Institute, Leeds General Infirmary, Leeds, United Kingdom
| | - Wolf Eilenberg
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| | - Christine Brostjan
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna and University Hospital Vienna, Vienna, Austria
| |
Collapse
|
17
|
Gadanec LK, McSweeney KR, Kubatka P, Caprnda M, Gaspar L, Prosecky R, Dragasek J, Kruzliak P, Apostolopoulos V, Zulli A. Angiotensin II constricts mouse iliac arteries: possible mechanism for aortic aneurysms. Mol Cell Biochem 2024; 479:233-242. [PMID: 37027096 DOI: 10.1007/s11010-023-04724-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/27/2023] [Indexed: 04/08/2023]
Abstract
Abdominal aortic aneurysms (AAA) result from maladaptive remodeling of the vascular wall and reduces structural integrity. Angiotensin II (AngII) infusion has become a standard laboratory model for studying AAA initiation and progression. We determined the different vasoactive responses of various mouse arteries to Ang II. Ex vivo isometric tension analysis was conducted on 18-week-old male C57BL/6 mice (n = 4) brachiocephalic arteries (BC), iliac arteries (IL), and abdominal (AA) and thoracic aorta (TA). Arterial rings were mounted between organ hooks, gently stretched and an AngII dose response was performed. Rings were placed in 4% paraformaldehyde for immunohistochemistry analysis to quantify peptide expression of angiotensin type 1 (AT1R) and 2 receptors (AT2R) in the endothelium, media, and adventitia. Results from this study demonstrated vasoconstriction responses in IL were significantly higher at all AngII doses when compared to BC, and TA and AA responses (maximum constriction-IL: 68.64 ± 5.47% vs. BC: 1.96 ± 1.00%; TA: 3.13 ± 0.16% and AA: 2.75 ± 1.77%, p < 0.0001). Expression of AT1R was highest in the endothelium of IL (p < 0.05) and in the media and (p < 0.05) adventitia (p < 0.05) of AA. In contrast, AT2R expression was highest in endothelium (p < 0.05), media (p < 0.01, p < 0.05) and adventitia of TA. These results suggest that mouse arteries display different vasoactive responses to AngII, and the exaggerated response in IL arteries may play a role during AAA development.
Collapse
Affiliation(s)
- Laura Kate Gadanec
- Institute of Health and Sport, Victoria University, Werribee Camous, Melbourne, VIC, 3030, Australia.
| | - Kristen Renee McSweeney
- Institute of Health and Sport, Victoria University, Werribee Camous, Melbourne, VIC, 3030, Australia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | - Ludovit Gaspar
- Faculty of Health Sciences, University of Ss. Cyril and Methodius in Trnava, Trnava, Slovakia
| | - Robert Prosecky
- 2nd Department of Internal Medicine, Faculty of Medicine, Masaryk University and St. Anne'S University Hospital, Brno, Czech Republic
- International Clinical Research Centre, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Jozef Dragasek
- Faculty of Medicine, Pavol Jozef Safarik University and University Hospital, Kosice, Slovakia
| | - Peter Kruzliak
- 2nd Department of Surgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic.
| | - Vasso Apostolopoulos
- Institute of Health and Sport, Victoria University, Werribee Camous, Melbourne, VIC, 3030, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Sunshine Hospital, Melbourne, VIC, 3021, Australia
| | - Anthony Zulli
- Institute of Health and Sport, Victoria University, Werribee Camous, Melbourne, VIC, 3030, Australia.
| |
Collapse
|
18
|
Loste A, Clément M, Delbosc S, Guedj K, Sénémaud J, Gaston AT, Morvan M, Even G, Gautier G, Eggel A, Arock M, Procopio E, Deschildre C, Louedec L, Michel JB, Deschamps L, Castier Y, Coscas R, Alsac JM, Launay P, Caligiuri G, Nicoletti A, Le Borgne M. Involvement of an IgE/Mast cell/B cell amplification loop in abdominal aortic aneurysm progression. PLoS One 2023; 18:e0295408. [PMID: 38055674 DOI: 10.1371/journal.pone.0295408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 11/21/2023] [Indexed: 12/08/2023] Open
Abstract
AIMS IgE type immunoglobulins and their specific effector cells, mast cells (MCs), are associated with abdominal aortic aneurysm (AAA) progression. In parallel, immunoglobulin-producing B cells, organised in tertiary lymphoid organs (TLOs) within the aortic wall, have also been linked to aneurysmal progression. We aimed at investigating the potential role and mechanism linking local MCs, TLO B cells, and IgE production in aneurysmal progression. METHODS AND RESULTS Through histological assays conducted on human surgical samples from AAA patients, we uncovered that activated MCs were enriched at sites of unhealed haematomas, due to subclinical aortic wall fissuring, in close proximity to adventitial IgE+ TLO B cells. Remarkably, in vitro the IgEs deriving from these samples enhanced MC production of IL-4, a cytokine which favors IgE class-switching and production by B cells. Finally, the role of MCs in aneurysmal progression was further analysed in vivo in ApoE-/- mice subjected to angiotensin II infusion aneurysm model, through MC-specific depletion after the establishment of dissecting aneurysms. MC-specific depletion improved intramural haematoma healing and reduced aneurysmal progression. CONCLUSIONS Our data suggest that MC located close to aortic wall fissures are activated by adventitial TLO B cell-produced IgEs and participate to their own activation by providing support for further IgE synthesis through IL-4 production. By preventing prompt repair of aortic subclinical fissures, such a runaway MC activation loop could precipitate aneurysmal progression, suggesting that MC-targeting treatments may represent an interesting adjunctive therapy for reducing AAA progression.
Collapse
Affiliation(s)
- Alexia Loste
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Marc Clément
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Sandrine Delbosc
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Kevin Guedj
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Jean Sénémaud
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
- Department of Vascular and Thoracic Surgery, AP-HP, Bichat Hospital, Université Paris Cité, Paris, France
| | - Anh-Thu Gaston
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Marion Morvan
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Guillaume Even
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Grégory Gautier
- DHU FIRE, Paris, France
- INSERM UMRS 1149, Centre de Recherche sur l'Inflammation (CRI), Université Paris Cité, Paris, France
| | - Alexander Eggel
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Michel Arock
- Department of Biology and CNRS UMR8113, Ecole Normale Supérieure de Paris-Saclay, Saclay, France
| | - Emanuele Procopio
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Catherine Deschildre
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Liliane Louedec
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Jean-Baptiste Michel
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Lydia Deschamps
- Department of Pathology, AP-HP, Bichat Hospital, Université Paris Cité, Paris, France
| | - Yves Castier
- INSERM UMRS 1149, Centre de Recherche sur l'Inflammation (CRI), Université Paris Cité, Paris, France
| | - Raphaël Coscas
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- Department of Vascular Surgery, AP-HP, Ambroise Paré University Hospital, Université Paris Cité, Boulogne-Billancourt, France
| | - Jean-Marc Alsac
- Department of Vascular Surgery, AP-HP, Hôpital Européen Georges Pompidou, Université Paris Cité, Paris, France
| | - Pierre Launay
- DHU FIRE, Paris, France
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Giuseppina Caligiuri
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
- Department of Cardiology, AP-HP, Bichat Hospital, Université Paris Cité, Paris, France
| | - Antonino Nicoletti
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| | - Marie Le Borgne
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
- DHU FIRE, Paris, France
| |
Collapse
|
19
|
Alloisio M, Gasser TC. Fracture of the porcine aorta. Part 2: FEM modelling and inverse parameter identification. Acta Biomater 2023:S1742-7061(23)00345-8. [PMID: 37422007 DOI: 10.1016/j.actbio.2023.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/10/2023]
Abstract
The mechanics of vascular tissue, particularly its fracture properties, are crucial in the onset and progression of vascular diseases. Vascular tissue properties are complex, and the identification of fracture mechanical properties relies on robust and efficient numerical tools. In this study, we propose a parameter identification pipeline to extract tissue properties from force-displacement and digital image correlation (DIC) data. The data has been acquired by symconCT testing porcine aorta wall specimens. Vascular tissue is modelled as a non-linear viscoelastic isotropic solid, and an isotropic cohesive zone model describes tissue fracture. The model closely replicated the experimental observations and identified the fracture energies of 1.57±0.82 kJ m-2 and 0.96±0.34 kJ m-2 for rupturing the porcine aortic media along the axial and circumferential directions, respectively. The identified strength was always below 350 kPa, a value significantly lower than identified through classical protocols, such as simple tension, and sheds new light on the resilience of the aorta. Further refinements to the model, such as considering rate effects in the fracture process zone and tissue anisotropy, could have improved the simulation results. STATEMENT OF SIGNIFICANCE: This paper identified porcine aorta's biomechanical properties using data acquired through a previously developed experimental protocol, the symmetry-constraint compact tension test. An implicit finite element method model mimicked the test, and a two-step approach identified the material's elastic and fracture properties directly from force-displacement curves and digital image correlation-based strain measurements. Our findings show a lower strength of the abdominal aorta as compared to the literature, which may have significant implications for the clinical evaluation of the risk of aortic rupture.
Collapse
Affiliation(s)
- Marta Alloisio
- Solid Mechanics, Department of Engineering Mechanics, KTH Royal Institute of Technology, Sweden
| | - T Christian Gasser
- Solid Mechanics, Department of Engineering Mechanics, KTH Royal Institute of Technology, Sweden.
| |
Collapse
|
20
|
Alloisio M, Chatziefraimidou M, Roy J, Christian Gasser T. Fracture of the porcine aorta. Part 1: symconCT fracture testing and DIC. Acta Biomater 2023:S1742-7061(23)00347-1. [PMID: 37355178 DOI: 10.1016/j.actbio.2023.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
Tissue failure and damage are inherent parts of vascular diseases and tightly linked to clinical events. Additionally, experimental set-ups designed to study classical engineering materials are suboptimal in the exploration of vessel wall fracture properties. The classical Compact Tension (CT) test was augmented to enable stable fracture propagation, resulting in the symmetry-constraint Compact Tension (symconCT) test, a suitable set-up for fracture testing of vascular tissue. The test was combined with Digital Image Correlation (DIC) to study tissue fracture in 45 porcine aorta specimens. Test specimens were loaded in axial and circumferential directions in a physiological solution at 37∘ C. Loading the aortic vessel wall in the axial direction resulted in mode I tissue failure and a fracture path aligned with the circumferential vessel direction. Circumferential loading resulted in mode I-dominated failure with multiple deflections of the fracture path. The aorta ruptured at a principal Green-Lagrange strain of approximately 0.7, and strain rate peaks that develop ahead of the crack tip reached nearly 400 times the strain rate on average over the test specimen. It required approximately 70% more external work to fracture the aorta by circumferential than axial load; normalised with the fracture surface, similar energy levels are, however, observed. The symconCT test resulted in a stable fracture propagation, which, combined with DIC, provided a set-up for the in-depth analysis of vascular tissue failure. The high strain rates ahead of the crack tip indicate the significance of rate effects in the constitutive description of vascular tissue fracture. STATEMENT OF SIGNIFICANCE: This paper represents a significant step forward in understanding the fracture properties of porcine aorta. Inspired by the Compact Tension test, we developed an ad hoc experimental protocol to investigate stable crack propagation in soft materials, providing new insights into the mechanical processes that lead to the rupture of vascular tissue. The set-up enables the assessment of strains and strain rates ahead of the crack tip, and our findings could improve the clinical risk assessment of vascular pathologies as well as optimize medical device design.
Collapse
Affiliation(s)
- Marta Alloisio
- Solid Mechanics, Department of Engineering Mechanics, KTH Royal Institute of Technology, Sweden
| | - Marina Chatziefraimidou
- Solid Mechanics, Department of Engineering Mechanics, KTH Royal Institute of Technology, Sweden
| | - Joy Roy
- Department of Molecular Medicine and Surgery, KI Karolinska Institute, Sweden
| | - T Christian Gasser
- Solid Mechanics, Department of Engineering Mechanics, KTH Royal Institute of Technology, Sweden.
| |
Collapse
|
21
|
Qiu C, Li Y, Xiao L, Zhang J, Guo S, Zhang P, Li R, Gong K. A novel rabbit model of abdominal aortic aneurysm: Construction and evaluation. Heliyon 2023; 9:e17279. [PMID: 37389075 PMCID: PMC10300360 DOI: 10.1016/j.heliyon.2023.e17279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 07/01/2023] Open
Abstract
Prior research has indicated that animal models of abdominal aortic aneurysm (AAA) utilizing porcine pancreatic elastase (PPE) exhibit a perfusion duration of 30 min, and extended perfusion durations are associated with elevated mortality rates. Similarly, the AAA model, which relies solely on balloon dilation (BD), is limited by the occurrence of self-healing aneurysms. Consequently, we constructed a novel AAA model by PPE combined with balloon expansion to shorten the modeling time and improve the modeling success rate. The findings indicated that 5 min was the optimal BD time for rabbits, 3 min BD was ineffective for aneurysm formation, and 10 min BD had a high mortality rate. The model, constructed in combination with PPE and 5 min BD, exhibited a 100% model formation rate and a 244.7% ± 9.83% dilation rate. HE staining exhibited that severe disruption of the inner, middle, and outer membranes of the abdominal aorta, with a marked decrease in smooth muscle cells and elastase, and a marked increase in fibroblasts of the middle membrane, and many infiltrating inflammatory cells were seen in all three layers, especially in the middle membrane. EVG staining displayed that the elastic fibers of the abdominal aortic wall were fractured and degraded, and lost their normal wavy appearance. The protein expression of inflammatory factor (IL-1β, IL-6 and TNF-α) as well as extracellular matrix components (MMP-2 and MMP-9) were significantly increased compared to PPE and 5 min BD alone. In conclusion, PPE combined with BD allows the establishment of a novel AAA model that closely mimics human AAA in terms of histomorphology, inflammatory cell infiltration, and vascular stromal destruction. This model provides an ideal animal model for understanding the pathogenesis of AAA.
Collapse
Affiliation(s)
- Changtao Qiu
- Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
- Department of General Surgery, The First People’s Hospital of Yunnan Province Kunming, Kunming, 650032, Yunnan, China
| | - Yuejin Li
- Department of General Surgery, The First People’s Hospital of Yunnan Province Kunming, Kunming, 650032, Yunnan, China
| | - Le Xiao
- Department of General Surgery, The First People’s Hospital of Yunnan Province Kunming, Kunming, 650032, Yunnan, China
| | - Jian Zhang
- Department of General Surgery, The First People’s Hospital of Yunnan Province Kunming, Kunming, 650032, Yunnan, China
| | - Shikui Guo
- Department of General Surgery, The First People’s Hospital of Yunnan Province Kunming, Kunming, 650032, Yunnan, China
| | - Peng Zhang
- Department of General Surgery, The First People’s Hospital of Yunnan Province Kunming, Kunming, 650032, Yunnan, China
| | - Ruoxi Li
- Department of General Surgery, The First People’s Hospital of Yunnan Province Kunming, Kunming, 650032, Yunnan, China
| | - Kunmei Gong
- Department of General Surgery, The First People’s Hospital of Yunnan Province Kunming, Kunming, 650032, Yunnan, China
| |
Collapse
|
22
|
Gasser TC, Miller C, Polzer S, Roy J. A quarter of a century biomechanical rupture risk assessment of abdominal aortic aneurysms. Achievements, clinical relevance, and ongoing developments. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2023; 39:e3587. [PMID: 35347895 DOI: 10.1002/cnm.3587] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/28/2022] [Accepted: 03/03/2022] [Indexed: 05/12/2023]
Abstract
Abdominal aortic aneurysm (AAA) disease, the local enlargement of the infrarenal aorta, is a serious condition that causes many deaths, especially in men exceeding 65 years of age. Over the past quarter of a century, computational biomechanical models have been developed towards the assessment of AAA risk of rupture, technology that is now on the verge of being integrated within the clinical decision-making process. The modeling of AAA requires a holistic understanding of the clinical problem, in order to set appropriate modeling assumptions and to draw sound conclusions from the simulation results. In this article we summarize and critically discuss the proposed modeling approaches and report the outcome of clinical validation studies for a number of biomechanics-based rupture risk indices. Whilst most of the aspects concerning computational mechanics have already been settled, it is the exploration of the failure properties of the AAA wall and the acquisition of robust input data for simulations that has the greatest potential for the further improvement of this technology.
Collapse
Affiliation(s)
- T Christian Gasser
- Department of Engineering Mechanics, KTH Royal Institute of Technology, Stockholm, Sweden
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Christopher Miller
- Department of Engineering Mechanics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Stanislav Polzer
- Department of Applied Mechanics, VSB-Technical University of Ostrava, Ostrava-Poruba, Czech Republic
| | - Joy Roy
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
23
|
Staal AHJ, Cortenbach KRG, Gorris MAJ, van der Woude LL, Srinivas M, Heijmen RH, Geuzebroek GSC, Grewal N, Hebeda KM, de Vries IJM, DeRuiter MC, van Kimmenade RRJ. Adventitial adaptive immune cells are associated with ascending aortic dilatation in patients with a bicuspid aortic valve. Front Cardiovasc Med 2023; 10:1127685. [PMID: 37057097 PMCID: PMC10086356 DOI: 10.3389/fcvm.2023.1127685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/14/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundBicuspid aortic valve (BAV) is associated with ascending aorta aneurysms and dissections. Presently, genetic factors and pathological flow patterns are considered responsible for aneurysm formation in BAV while the exact role of inflammatory processes remains unknown.MethodsIn order to objectify inflammation, we employ a highly sensitive, quantitative immunohistochemistry approach. Whole slides of dissected, dilated and non-dilated ascending aortas from BAV patients were quantitatively analyzed.ResultsDilated aortas show a 4-fold increase of lymphocytes and a 25-fold increase in B lymphocytes in the adventitia compared to non-dilated aortas. Tertiary lymphoid structures with B cell follicles and helper T cell expansion were identified in dilated and dissected aortas. Dilated aortas were associated with an increase in M1-like macrophages in the aorta media, in contrast the number of M2-like macrophages did not change significantly.ConclusionThis study finds unexpected large numbers of immune cells in dilating aortas of BAV patients. These findings raise the question whether immune cells in BAV aortopathy are innocent bystanders or contribute to the deterioration of the aortic wall.
Collapse
Affiliation(s)
- Alexander H. J. Staal
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Kimberley R. G. Cortenbach
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mark A. J. Gorris
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, Netherlands
| | - Lieke L. van der Woude
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Pathology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, Netherlands
| | - Robin H. Heijmen
- Department of Cardiothoracic Surgery, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Nimrat Grewal
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Konnie M. Hebeda
- Department of Pathology, Radboud University Medical Center, Nijmegen, Netherlands
| | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marco C. DeRuiter
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Roland R. J. van Kimmenade
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
- *Correspondence: Roland R. J. van Kimmenade,
| |
Collapse
|
24
|
Deng J, Li D, Zhang X, Lu W, Rong D, Wang X, Sun G, Jia S, Zhang H, Jia X, Guo W. Murine model of elastase-induced proximal thoracic aortic aneurysm through a midline incision in the anterior neck. Front Cardiovasc Med 2023; 10:953514. [PMID: 36815017 PMCID: PMC9939838 DOI: 10.3389/fcvm.2023.953514] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 01/03/2023] [Indexed: 02/09/2023] Open
Abstract
Objective This study was performed to develop a murine model of elastase-induced proximal thoracic aortic aneurysms (PTAAs). Methods The ascending thoracic aorta and aortic arch of adult C57BL/6J male mice were exposed through a midline incision in the anterior neck, followed by peri-adventitial elastase or saline application. The maximal ascending thoracic aorta diameter was measured with high-resolution micro-ultrasound. Twenty-eight days after the operation, the aortas were harvested and analyzed by histopathological examination and qualitative polymerase chain reaction to determine the basic characteristics of the aneurysmal lesions. Results Fourteen days after the operation, the dilation rate (mean ± standard error) in the 10-min elastase application group (n = 10, 71.44 ± 10.45%) or 5-min application group (n = 9, 42.67 ± 3.72%) were significantly higher than that in the saline application group (n = 9, 7.37 ± 0.94%, P < 0.001 for both). Histopathological examination revealed aortic wall thickening, degradation of elastin fibers, loss of smooth muscle cells, more vasa vasorum, enhanced extracellular matrix degradation, augmented collagen synthesis, upregulated apoptosis and proliferation capacity of smooth muscle cells, and increased macrophages and CD4+ T cells infiltration in the PTAA lesions. Qualitative analyses indicated higher expression of the proinflammatory markers, matrix metalloproteinase-2 and -9 as well as Collagen III, Collagen I in the PTAAs than in the controls. Conclusion We established a novel in vivo mouse model of PTAAs through a midline incision in the anterior neck by peri-adventitial application of elastase. This model may facilitate research into the pathogenesis of PTAA formation and the treatment strategy for this devastating disease.
Collapse
Affiliation(s)
- Jianqing Deng
- Department of Vascular and Endovascular Surgery, The First Medical Center of PLA General Hospital, Beijing, 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, China,Department of Cardiovascular Surgery, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Dandan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuelin Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weihang Lu
- Department of Vascular and Endovascular Surgery, The First Medical Center of PLA General Hospital, Beijing, China
| | - Dan Rong
- Department of Vascular and Endovascular Surgery, The First Medical Center of PLA General Hospital, Beijing, China
| | - Xinhao Wang
- Department of Vascular and Endovascular Surgery, The First Medical Center of PLA General Hospital, Beijing, China
| | - Guoyi Sun
- Department of Vascular and Endovascular Surgery, The First Medical Center of PLA General Hospital, Beijing, China
| | - Senhao Jia
- Department of Vascular and Endovascular Surgery, The First Medical Center of PLA General Hospital, Beijing, China
| | - Hongpeng Zhang
- Department of Vascular and Endovascular Surgery, The First Medical Center of PLA General Hospital, Beijing, China
| | - Xin Jia
- Department of Vascular and Endovascular Surgery, The First Medical Center of PLA General Hospital, Beijing, China
| | - Wei Guo
- Department of Vascular and Endovascular Surgery, The First Medical Center of PLA General Hospital, Beijing, China,*Correspondence: Wei Guo,
| |
Collapse
|
25
|
Targeted Inhibition of Matrix Metalloproteinase-8 Prevents Aortic Dissection in a Murine Model. Cells 2022; 11:cells11203218. [PMID: 36291087 PMCID: PMC9600539 DOI: 10.3390/cells11203218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/30/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
Aortic dissection (AD) is a lethal aortic pathology without effective medical treatments since the underlying pathological mechanisms responsible for AD remain elusive. Matrix metalloproteinase-8 (MMP8) has been previously identified as a key player in atherosclerosis and arterial remodeling. However, the functional role of MMP8 in AD remains largely unknown. Here, we report that an increased level of MMP8 was observed in 3-aminopropionitrile fumarate (BAPN)-induced murine AD. AD incidence and aortic elastin fragmentation were markedly reduced in MMP8-knockout mice. Importantly, pharmacologic inhibition of MMP8 significantly reduced the AD incidence and aortic elastin fragmentation. We observed less inflammatory cell accumulation, a lower level of aortic inflammation, and decreased smooth muscle cell (SMC) apoptosis in MMP8-knockout mice. In line with our previous observation that MMP8 cleaves Ang I to generate Ang II, BAPN-treated MMP8-knockout mice had increased levels of Ang I, but decreased levels of Ang II and lower blood pressure. Additionally, we observed a decreased expression level of vascular cell adhesion molecule-1 (VCAM1) and a reduced level of reactive oxygen species (ROS) in MMP8-knockout aortas. Mechanistically, our data show that the Ang II/VCAM1 signal axis is responsible for MMP8-mediated inflammatory cell invasion and transendothelial migration, while MMP8-mediated SMC inflammation and apoptosis are attributed to Ang II/ROS signaling. Finally, we observed higher levels of aortic and serum MMP8 in patients with AD. We therefore provide new insights into the molecular mechanisms underlying AD and identify MMP8 as a potential therapeutic target for this life-threatening aortic disease.
Collapse
|
26
|
Ren J, Wu L, Wu J, Tang X, Lv Y, Wang W, Li F, Yang D, Liu C, Zheng Y. The molecular mechanism of Ang II induced-AAA models based on proteomics analysis in ApoE -/- and CD57BL/6J mice. J Proteomics 2022; 268:104702. [PMID: 35988846 DOI: 10.1016/j.jprot.2022.104702] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/19/2022]
Abstract
Apolipoprotein knockout (ApoE-/-) and CD57BL/6J mouse models of angiotensin II (Ang II)-induced abdominal aortic aneurysm (AAA) are commonly used in AAA research. However, the similarities and differences in the molecular mechanisms of AAA in these two genotypes have not been reported. In our study, we analyzed proteomics data from ApoE-/- and CD57BL/6J mouse models of Ang II-induced AAA and control mice by LC-MS/MS. Gene set enrichment analysis (GSEA) of differentially abundance proteins (DAPs) in the ApoE-/- or CD57BL/6J mouse groups was performed in R software, and infiltration of immune cells in groups was assessed. DAP that showed the same trend in abundance in ApoE-/- and CD57BL/6J mice (S-DAP) were identified and subjected to GO enrichment, KEGG pathway, and connectivity map (CMap) analyses. The protein-protein interaction (PPI) network of the S-DAP was drawn, the key S-DAP were identified by MCODE, and the transcription factors (TFs) of crucial S-DAP were predicted by iRegulon in Cytoscape. Male ApoE-/- and CD57BL/6J mouse models of Ang II-induced AAA are commonly used in AAA research, and extracellular matrix organization is associated with AAA in both of these models. However, there are some differences between the mechanisms underlying AAA in these two genotypes, and these differences need to be considered when studying AAA and selecting models. SIGNIFICANCE: Our research provided the first insight into the similarity and differential mechanisms of Ang II infused AAA models using ApoE-/- and CD57BL/6J mice. This study might provide the some advises for the selection of Ang II infused AAA models for further AAA researches.
Collapse
Affiliation(s)
- Jinrui Ren
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Vascular Surgery, State Key Laboratory of Complex Severe andRare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Lianglin Wu
- Department of Vascular Surgery, State Key Laboratory of Complex Severe andRare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jianqiang Wu
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyue Tang
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanze Lv
- Department of Vascular Surgery, State Key Laboratory of Complex Severe andRare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wei Wang
- Department of Vascular Surgery, State Key Laboratory of Complex Severe andRare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Fangda Li
- Department of Vascular Surgery, State Key Laboratory of Complex Severe andRare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Dan Yang
- Department of Computational Biology and Bioinformatics, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changzheng Liu
- National Health Commission of the People's Republic of China (NHC), Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuehong Zheng
- Department of Vascular Surgery, State Key Laboratory of Complex Severe andRare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
27
|
Yin L, Kent EW, Wang B. Progress in murine models of ruptured abdominal aortic aneurysm. Front Cardiovasc Med 2022; 9:950018. [PMID: 36035911 PMCID: PMC9411998 DOI: 10.3389/fcvm.2022.950018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/27/2022] [Indexed: 02/03/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a focal dilation of the aorta that is prevalent in aged populations. The progressive and unpredictable expansion of AAA could result in aneurysmal rupture, which is associated with ~80% mortality. Due to the expanded screening efforts and progress in diagnostic tools, an ever-increasing amount of asymptomatic AAA patients are being identified yet without a cure to stop the rampant aortic expansion. A key barrier that hinders the development of effective AAA treatment is our incomplete understanding of the cellular and molecular basis of its pathogenesis and progression into rupture. Animal models provide invaluable mechanistic insights into AAA pathophysiology. However, there is no single experimental model that completely recapitulate the complex biology behind AAA, and different AAA-inducing methodologies are associated with distinct disease course and rupture rate. In this review article, we summarize the established murine models of ruptured AAA and discuss their respective strengths and utilities.
Collapse
Affiliation(s)
| | | | - Bowen Wang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
28
|
Morgan S, Lee LH, Halu A, Nicolau JS, Higashi H, Ha AH, Wen JR, Daugherty A, Libby P, Cameron SJ, Mix D, Aikawa E, Owens AP, Singh SA, Aikawa M. Identifying novel mechanisms of abdominal aortic aneurysm via unbiased proteomics and systems biology. Front Cardiovasc Med 2022; 9:889994. [PMID: 35990960 PMCID: PMC9382335 DOI: 10.3389/fcvm.2022.889994] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Background Abdominal aortic aneurysm (AAA), characterized by a continued expansion of the aorta, leads to rupture if not surgically repaired. Mice aid the study of disease progression and its underlying mechanisms since sequential studies of aneurysm development are not feasible in humans. The present study used unbiased proteomics and systems biology to understand the molecular relationship between the mouse models of AAA and the human disease. Methods and results Aortic tissues of developing and established aneurysms produced by either angiotensin II (AngII) infusion in Apoe -/- and Ldlr -/- mice or intraluminal elastase incubation in wildtype C57BL/6J mice were examined. Aortas were dissected free and separated into eight anatomical segments for proteomics in comparison to their appropriate controls. High-dimensional proteome cluster analyses identified site-specific protein signatures in the suprarenal segment for AngII-infused mice (159 for Apoe -/- and 158 for Ldlr -/-) and the infrarenal segment for elastase-incubated mice (173). Network analysis revealed a predominance of inflammatory and coagulation factors in developing aneurysms, and a predominance of fibrosis-related pathways in established aneurysms for both models. To further substantiate our discovery platform, proteomics was performed on human infrarenal aortic aneurysm tissues as well as aortic tissue collected from age-matched controls. Protein processing and inflammatory pathways, particularly neutrophil-associated inflammation, dominated the proteome of the human aneurysm abdominal tissue. Aneurysmal tissue from both mouse and human had inflammation, coagulation, and protein processing signatures, but differed in the prevalence of neutrophil-associated pathways, and erythrocyte and oxidative stress-dominated networks in the human aneurysms. Conclusions Identifying changes unique to each mouse model will help to contextualize model-specific findings. Focusing on shared proteins between mouse experimental models or between mouse and human tissues may help to better understand the mechanisms for AAA and establish molecular bases for novel therapies.
Collapse
Affiliation(s)
- Stephanie Morgan
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Lang Ho Lee
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Arda Halu
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Jessica S. Nicolau
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Hideyuki Higashi
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Anna H. Ha
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Jennifer R. Wen
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Alan Daugherty
- Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
| | - Peter Libby
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Scott J. Cameron
- Department of Cardiovascular Medicine, Section of Vascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Doran Mix
- Division of Vascular Surgery, Department of Surgery, University of Rochester School of Medicine, Rochester, NY, United States
| | - Elena Aikawa
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - A. Phillip Owens
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Sasha A. Singh
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
29
|
Li Z, Cong X, Kong W. Matricellular proteins: Potential biomarkers and mechanistic factors in aortic aneurysms. J Mol Cell Cardiol 2022; 169:41-56. [DOI: 10.1016/j.yjmcc.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/30/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
|
30
|
Weiss D, Long AS, Tellides G, Avril S, Humphrey JD, Bersi MR. Evolving Mural Defects, Dilatation, and Biomechanical Dysfunction in Angiotensin II-Induced Thoracic Aortopathies. Arterioscler Thromb Vasc Biol 2022; 42:973-986. [PMID: 35770665 PMCID: PMC9339505 DOI: 10.1161/atvbaha.122.317394] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Thoracic aortopathy associates with extracellular matrix remodeling and altered biomechanical properties. We sought to quantify the natural history of thoracic aortopathy in a common mouse model and to correlate measures of wall remodeling such as aortic dilatation or localized mural defects with evolving microstructural composition and biomechanical properties of the wall. METHODS We combined a high-resolution multimodality imaging approach (panoramic digital image correlation and optical coherence tomography) with histopathologic examinations and biaxial mechanical testing to correlate spatially, for the first time, macroscopic mural defects and medial degeneration within the ascending aorta with local changes in aortic wall composition and mechanical properties. RESULTS Findings revealed strong correlations between local decreases in elastic energy storage and increases in circumferential material stiffness with increasing proximal aortic diameter and especially mural defect size. Mural defects tended to exhibit a pronounced biomechanical dysfunction that is driven by an altered organization of collagen and elastic fibers. CONCLUSIONS While aneurysmal dilatation is often observed within particular segments of the aorta, dissection and rupture initiate as highly localized mechanical failures. We show that wall composition and material properties are compromised in regions of local mural defects, which further increases the dilatation and overall structural vulnerability of the wall. Identification of therapies focused on promoting robust collagen accumulation may protect the wall from these vulnerabilities and limit the incidence of dissection and rupture.
Collapse
Affiliation(s)
- Dar Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Aaron S. Long
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - George Tellides
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| | - Stéphane Avril
- Mines Saint-Etienne, University of Lyon, University Jean Monnet, INSERM, Saint-Etienne, France
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| | - Matthew R. Bersi
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
31
|
Ibrahim N, Bleichert S, Klopf J, Kurzreiter G, Knöbl V, Hayden H, Busch A, Stiglbauer-Tscholakoff A, Eilenberg W, Neumayer C, Bailey MA, Brostjan C. 3D Ultrasound Measurements Are Highly Sensitive to Monitor Formation and Progression of Abdominal Aortic Aneurysms in Mouse Models. Front Cardiovasc Med 2022; 9:944180. [PMID: 35903666 PMCID: PMC9314770 DOI: 10.3389/fcvm.2022.944180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Background Available mouse models for abdominal aortic aneurysms (AAAs) differ substantially in the applied triggers, associated pathomechanisms and rate of vessel expansion. While maximum aortic diameter (determined after aneurysm excision or by 2D ultrasound) is commonly applied to document aneurysm development, we evaluated the sensitivity and reproducibility of 3D ultrasound to monitor aneurysm growth in four distinct mouse models of AAA. Methods The models included angiotensin-II infusion in ApoE deficient mice, topical elastase application on aortas in C57BL/6J mice (with or without oral administration of β-aminoproprionitrile) and intraluminal elastase perfusion in C57BL/6J mice. AAA development was monitored using semi-automated 3D ultrasound for aortic volume calculation over 12 mm length and assessment of maximum aortic diameter. Results While the models differed substantially in the time course of aneurysm development, 3D ultrasound measurements (volume and diameter) proved highly reproducible with concordance correlation coefficients > 0.93 and variations below 9% between two independent observers. Except for the elastase perfusion model where aorta expansion was lowest and best detected by diameter increase, all other models showed high sensitivity of absolute volume and diameter measurements in monitoring AAA formation and progression by 3D ultrasound. When compared to standard 2D ultrasound, the 3D derived parameters generally reached the highest effect size. Conclusion This study has yielded novel information on the robustness and limitations of semi-automated 3D ultrasound analysis and provided the first direct comparison of aortic volume increase over time in four widely applied mouse models of AAA. While 3D ultrasound generally proved highly sensitive in detecting early AAA formation, the 3D based volume analysis was found inferior to maximum diameter assessment in the elastase perfusion model where the extent of inflicted local injury is determined by individual anatomical features.
Collapse
Affiliation(s)
- Nahla Ibrahim
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Sonja Bleichert
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Johannes Klopf
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Gabriel Kurzreiter
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Viktoria Knöbl
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Hubert Hayden
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Albert Busch
- Department for Visceral, Thoracic and Vascular Surgery, Technical University of Dresden, University Hospital Carl-Gustav Carus, Dresden, Germany
| | - Alexander Stiglbauer-Tscholakoff
- Division of Cardiovascular and Interventional Radiology, Division of Molecular and Gender Imaging, Department of Biomedical Imaging and Image Guided Therapy, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Wolf Eilenberg
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Marc A. Bailey
- School of Medicine, Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
- Leeds Vascular Institute, Leeds General Infirmary, Leeds, United Kingdom
| | - Christine Brostjan
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| |
Collapse
|
32
|
Tsuruda T, Yamashita A, Otsu M, Koide M, Nakamichi Y, Sekita-Hatakeyama Y, Hatakeyama K, Funamoto T, Chosa E, Asada Y, Udagawa N, Kato J, Kitamura K. Angiotensin II Induces Aortic Rupture and Dissection in Osteoprotegerin-Deficient Mice. J Am Heart Assoc 2022; 11:e025336. [PMID: 35411794 PMCID: PMC9238451 DOI: 10.1161/jaha.122.025336] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background The biological mechanism of action for osteoprotegerin, a soluble decoy receptor for the receptor activator of nuclear factor‐kappa B ligand in the vascular structure, has not been elucidated. The study aim was to determine if osteoprotegerin affects aortic structural integrity in angiotensin II (Ang II)‐induced hypertension. Methods and Results Mortality was higher (P<0.0001 by log‐rank test) in 8‐week‐old male homozygotes of osteoprotegerin gene‐knockout mice given subcutaneous administration of Ang II for 28 days, with an incidence of 21% fatal aortic rupture and 23% aortic dissection, than in age‐matched wild‐type mice. Ang II‐infused aorta of wild‐type mice showed that osteoprotegerin immunoreactivity was present with proteoglycan. The absence of osteoprotegerin was associated with decreased medial and adventitial thickness and increased numbers of elastin breaks as well as with increased periostin expression and soluble receptor activator of nuclear factor‐kappa B ligand concentrations. PEGylated human recombinant osteoprotegerin administration decreased all‐cause mortality (P<0.001 by log‐rank test), the incidence of fatal aortic rupture (P=0.08), and aortic dissection (P<0.001) with decreasing numbers of elastin breaks, periostin expressions, and soluble receptor activator of nuclear factor‐kappa B ligand concentrations in Ang II‐infused osteoprotegerin gene‐knockout mice. Conclusions These data suggest that osteoprotegerin protects against aortic rupture and dissection in Ang II‐induced hypertension by inhibiting receptor activator of nuclear factor‐kappa B ligand activity and periostin expression.
Collapse
Affiliation(s)
- Toshihiro Tsuruda
- Division of Internal Medicine, Cardiovascular Medicine and Nephrology Faculty of Medicine University of Miyazaki Japan
| | - Atsushi Yamashita
- Department of Pathology Faculty of Medicine University of Miyazaki Japan
| | - Misa Otsu
- Division of Internal Medicine, Cardiovascular Medicine and Nephrology Faculty of Medicine University of Miyazaki Japan
| | - Masanori Koide
- Institute for Oral Science Matsumoto Dental University Nagano Japan
| | - Yuko Nakamichi
- Institute for Oral Science Matsumoto Dental University Nagano Japan
| | | | - Kinta Hatakeyama
- Department of Pathology National Cerebral and Cardiovascular Center Osaka Japan
| | - Taro Funamoto
- Division of Orthopedic Surgery Department of Medicine of Sensory and Motor Organs Faculty of Medicine University of Miyazaki Japan
| | - Etsuo Chosa
- Division of Orthopedic Surgery Department of Medicine of Sensory and Motor Organs Faculty of Medicine University of Miyazaki Japan
| | - Yujiro Asada
- Department of Pathology Faculty of Medicine University of Miyazaki Japan
| | - Nobuyuki Udagawa
- Department of Biochemistry Matsumoto Dental University Nagano Japan
| | - Johji Kato
- Frontier Science Research Center University of Miyazaki Japan
| | - Kazuo Kitamura
- Frontier Science Research Center University of Miyazaki Japan
| |
Collapse
|
33
|
Van Hoose PM, Yang L, Kraemer M, Ubele M, Morris AJ, Smyth SS. Lipid phosphate phosphatase 3 in smooth muscle cells regulates angiotensin II-induced abdominal aortic aneurysm formation. Sci Rep 2022; 12:5664. [PMID: 35383201 PMCID: PMC8983654 DOI: 10.1038/s41598-022-08422-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/10/2021] [Indexed: 01/28/2023] Open
Abstract
Genetic variants that regulate lipid phosphate phosphatase 3 (LPP3) expression are risk factors for the development of atherosclerotic cardiovascular disease. LPP3 is dynamically upregulated in the context of vascular inflammation with particularly heightened expression in smooth muscle cells (SMC), however, the impact of LPP3 on vascular pathology is not fully understood. We investigated the role of LPP3 and lysophospholipid signaling in a well-defined model of pathologic aortic injury and observed Angiotensin II (Ang II) increases expression of PLPP3 in SMCs through nuclear factor kappa B (NF-κB) signaling Plpp3 global reduction (Plpp3+/-) or SMC-specific deletion (SM22-Δ) protects hyperlipidemic mice from AngII-mediated aneurysm formation. LPP3 expression regulates SMC differentiation state and lowering LPP3 levels promotes a fibroblast-like phenotype. Decreased inactivation of bioactive lysophosphatidic acid (LPA) in settings of LPP3 deficiency may underlie these phenotypes because deletion of LPA receptor 4 in mice promotes early aortic dilation and rupture in response to AngII. LPP3 expression and LPA signaling influence SMC and vessel wall responses that are important for aortic dissection and aneurysm formation. These findings could have important implications for therapeutics targeting LPA metabolism and signaling in ongoing clinical trials.
Collapse
Affiliation(s)
- Patrick M Van Hoose
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA
| | - Liping Yang
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA
| | - Maria Kraemer
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA
| | - Margo Ubele
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA
| | - Andrew J Morris
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA
- Lexington Veterans Affair Medical Center, Lexington, KY, USA
| | - Susan S Smyth
- Gill Heart and Vascular Institute, University of Kentucky, 741 South Limestone BBSRB, Rm: B347, Lexington, KY, 40536-0509, USA.
- Lexington Veterans Affair Medical Center, Lexington, KY, USA.
| |
Collapse
|
34
|
Oller J, Gabandé-Rodríguez E, Roldan-Montero R, Ruiz-Rodríguez MJ, Redondo JM, Martín-Ventura JL, Mittelbrunn M. Rewiring Vascular Metabolism Prevents Sudden Death due to Aortic Ruptures-Brief Report. Arterioscler Thromb Vasc Biol 2022; 42:462-469. [PMID: 35196876 DOI: 10.1161/atvbaha.121.317346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The goal of this study was to determine whether boosting mitochondrial respiration prevents the development of fatal aortic ruptures triggered by atherosclerosis and hypertension. METHODS Ang-II (angiotensin-II) was infused in ApoE (Apolipoprotein E)-deficient mice fed with a western diet to induce acute aortic aneurysms and lethal ruptures. RESULTS We found decreased mitochondrial respiration and mitochondrial proteins in vascular smooth muscle cells from murine and human aortic aneurysms. Boosting NAD levels with nicotinamide riboside reduced the development of aortic aneurysms and sudden death by aortic ruptures. CONCLUSIONS Targetable vascular metabolism is a new clinical strategy to prevent fatal aortic ruptures and sudden death in patients with aortic aneurysms.
Collapse
Affiliation(s)
- Jorge Oller
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain (J.O., E.G.-R., M.M.).,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain (J.O., E.G.-R., M.M.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain (J.O., R.R.-M. M.J.R.-R., J.M.R., J.L.M.-V.)
| | - Enrique Gabandé-Rodríguez
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain (J.O., E.G.-R., M.M.).,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain (J.O., E.G.-R., M.M.)
| | - Raquel Roldan-Montero
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain (J.O., R.R.-M. M.J.R.-R., J.M.R., J.L.M.-V.).,Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain (R.R.-M., J.L.M.-V.)
| | - María Jesús Ruiz-Rodríguez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain (J.O., R.R.-M. M.J.R.-R., J.M.R., J.L.M.-V.).,Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (M.J.R.-R., J.M.R.)
| | - Juan Miguel Redondo
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain (J.O., R.R.-M. M.J.R.-R., J.M.R., J.L.M.-V.).,Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (M.J.R.-R., J.M.R.)
| | - José Luís Martín-Ventura
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain (J.O., R.R.-M. M.J.R.-R., J.M.R., J.L.M.-V.).,Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain (R.R.-M., J.L.M.-V.)
| | - María Mittelbrunn
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain (J.O., E.G.-R., M.M.).,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain (J.O., E.G.-R., M.M.)
| |
Collapse
|
35
|
Sawada H, Lu HS, Cassis LA, Daugherty A. Twenty Years of Studying AngII (Angiotensin II)-Induced Abdominal Aortic Pathologies in Mice: Continuing Questions and Challenges to Provide Insight Into the Human Disease. Arterioscler Thromb Vasc Biol 2022; 42:277-288. [PMID: 35045728 PMCID: PMC8866209 DOI: 10.1161/atvbaha.121.317058] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AngII (angiotensin II) infusion in mice has been used to provide mechanistic insight into human abdominal aortic aneurysms for over 2 decades. This is a technically facile animal model that recapitulates multiple facets of the human disease. Although numerous publications have reported abdominal aortic aneurysms with AngII infusion in mice, there remain many fundamental unanswered questions such as uniformity of describing the pathological characteristics and which cell type is stimulated by AngII to promote abdominal aortic aneurysms. Extrapolation of the findings to provide insight into the human disease has been hindered by the preponderance of studies designed to determine the effects on initiation of abdominal aortic aneurysms, rather than a more clinically relevant scenario of determining efficacy on the established disease. The purpose of this review is to enhance understanding of AngII-induced abdominal aortic pathologies in mice, thereby providing greater insight into the human disease.
Collapse
Affiliation(s)
- Hisashi Sawada
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY,Saha Aortic Center, University of Kentucky, Lexington, KY,Department of Physiology, University of Kentucky, Lexington, KY
| | - Hong S. Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY,Saha Aortic Center, University of Kentucky, Lexington, KY,Department of Physiology, University of Kentucky, Lexington, KY
| | - Lisa A. Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY,Saha Aortic Center, University of Kentucky, Lexington, KY,Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
36
|
Imaging Techniques for Aortic Aneurysms and Dissections in Mice: Comparisons of Ex Vivo, In Situ, and Ultrasound Approaches. Biomolecules 2022; 12:biom12020339. [PMID: 35204838 PMCID: PMC8869425 DOI: 10.3390/biom12020339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 01/04/2023] Open
Abstract
Aortic aneurysms and dissections are life-threatening conditions that have a high risk for lethal bleeding and organ malperfusion. Many studies have investigated the molecular basis of these diseases using mouse models. In mice, ex vivo, in situ, and ultrasound imaging are major approaches to evaluate aortic diameters, a common parameter to determine the severity of aortic aneurysms. However, accurate evaluations of aortic dimensions by these imaging approaches could be challenging due to pathological features of aortic aneurysms. Currently, there is no standardized mode to assess aortic dissections in mice. It is important to understand the characteristics of each approach for reliable evaluation of aortic dilatations. In this review, we summarize imaging techniques used for aortic visualization in recent mouse studies and discuss their pros and cons. We also provide suggestions to facilitate the visualization of mouse aortas.
Collapse
|
37
|
He C, Jiang B, Wang M, Ren P, Murtada SI, Caulk AW, Li G, Qin L, Assi R, Lovoulos CJ, Schwartz MA, Humphrey JD, Tellides G. mTOR inhibition prevents angiotensin II-induced aortic rupture and pseudoaneurysm but promotes dissection in Apoe-deficient mice. JCI Insight 2022; 7:155815. [PMID: 35132962 PMCID: PMC8855820 DOI: 10.1172/jci.insight.155815] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/23/2021] [Indexed: 01/04/2023] Open
Abstract
Aortic dissection and rupture are triggered by decreased vascular wall strength and/or increased mechanical loads. We investigated the role of mTOR signaling in aortopathy using a well-described model of angiotensin II–induced dissection, aneurysm, or rupture of the suprarenal abdominal aorta in Apoe-deficient mice. Although not widely appreciated, nonlethal hemorrhagic lesions present as pseudoaneurysms without significant dissection in this model. Angiotensin II–induced aortic tears result in free rupture, contained rupture with subadventitial hematoma (forming pseudoaneurysms), dilatation, or healing, while the media invariably thickens regardless of mural tears. Medial thickening results from smooth muscle cell hypertrophy and extracellular matrix accumulation, including matricellular proteins. Angiotensin II activates mTOR signaling in vascular wall cells, and inhibition of mTOR signaling by rapamycin prevents aortic rupture but promotes dissection. Decreased aortic rupture correlates with decreased inflammation and metalloproteinase expression, whereas extensive dissection correlates with induction of matricellular proteins that modulate adhesion of vascular cells. Thus, mTOR activation in vascular wall cells determines whether aortic tears progress to dissection or rupture. Previous mechanistic studies of aortic aneurysm and dissection by angiotensin II in Apoe-deficient mice should be reinterpreted as clinically relevant to pseudoaneurysms, and mTOR inhibition for aortic disease should be explored with caution.
Collapse
Affiliation(s)
- Changshun He
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Bo Jiang
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Mo Wang
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Pengwei Ren
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Sae-Il Murtada
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA
| | - Alexander W Caulk
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA
| | - Guangxin Li
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Lingfeng Qin
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Roland Assi
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA.,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Constantinos J Lovoulos
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA.,Department of Surgery, Frank H. Netter MD School of Medicine, Quinnipiac University, North Haven, Connecticut, USA
| | - Martin A Schwartz
- Department of Medicine (Cardiology).,Department of Cell Biology, and.,Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA
| | - George Tellides
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA.,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
38
|
Wortmann M, Klotz R, Kalkum E, Dihlmann S, Böckler D, Peters AS. Inflammasome Targeted Therapy as Novel Treatment Option for Aortic Aneurysms and Dissections: A Systematic Review of the Preclinical Evidence. Front Cardiovasc Med 2022; 8:805150. [PMID: 35127865 PMCID: PMC8811141 DOI: 10.3389/fcvm.2021.805150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/28/2021] [Indexed: 12/09/2022] Open
Abstract
Both aortic aneurysm and dissection are life threatening pathologies. In the lack of a conservative medical treatment, the only therapy consists of modifying cardiovascular risk factors and either surgical or endovascular treatment. Like many other cardiovascular diseases, in particular atherosclerosis, aortic aneurysm and dissection have a strong inflammatory phenotype. Inflammasomes are part of the innate immune system. Upon stimulation they form multi protein complexes resulting mainly in activation of interleukin-1β and other cytokines. Considering the gathering evidence, that inflammasomes are decisively involved in the emergence and progression of aortic diseases, inflammasome targeted therapy provides a promising new treatment approach. A systematic review following the PRISMA guidelines on the current preclinical data regarding the potential role of inflammasome targeted drug therapy as novel treatment option for aortic aneurysms and dissections was performed. Included were all rodent models of aortic disease (aortic aneurysm and dissection) evaluating a drug therapy with direct or indirect inhibition of inflammasomes and a suitable control group with the use of the same aortic model without the inflammasome targeted therapy. Primary and secondary outcomes were incidence of aortic disease, aortic rupture, aortic related death, and the maximum aortic diameter. The literature search of MEDLINE (via PubMed), the Web of Science, EMBASE and the Cochrane Central Registry of Registered Trials (CENTRAL) resulted in 8,137 hits. Of these, four studies met the inclusion criteria and were therefore eligible for data analysis. In all of them, targeting of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome effectively reduced the incidence of aortic disease and aortic rupture, and additionally reduced destruction of the aortic wall. Treatment strategies aiming at other inflammasomes could not be identified. In conclusion, inflammasome targeted therapies, more precisely targeting the NLRP3 inflammasome, have shown promising results in rodent models and deserve further investigation in preclinical research to potentially translate them into clinical research for the treatment of human patients with aortic disease. Regarding other inflammasomes, more preclinical research is needed to investigate their role in the pathophysiology of aortic disease. Protocol Registration: PROSPERO 2021 CRD42021279893, https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021279893
Collapse
Affiliation(s)
- Markus Wortmann
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, Heidelberg, Germany
- *Correspondence: Markus Wortmann
| | - Rosa Klotz
- Study Center of the German Surgical Society (SDGC), University of Heidelberg, Heidelberg, Germany
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Eva Kalkum
- Study Center of the German Surgical Society (SDGC), University of Heidelberg, Heidelberg, Germany
| | - Susanne Dihlmann
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Dittmar Böckler
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas S. Peters
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
39
|
Berman AG, Romary DJ, Kerr KE, Gorazd NE, Wigand MM, Patnaik SS, Finol EA, Cox AD, Goergen CJ. Experimental aortic aneurysm severity and growth depend on topical elastase concentration and lysyl oxidase inhibition. Sci Rep 2022; 12:99. [PMID: 34997075 PMCID: PMC8742076 DOI: 10.1038/s41598-021-04089-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 12/15/2021] [Indexed: 11/23/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) formation and expansion is highly complex and multifactorial, and the improvement of animal models is an important step to enhance our understanding of AAA pathophysiology. In this study, we explore our ability to influence aneurysm growth in a topical elastase plus β-Aminopropionitrile (BAPN) mouse model by varying elastase concentration and by altering the cross-linking capability of the tissue. To do so, we assess both chronic and acute effects of elastase concentration using volumetric ultrasound. Our results suggest that the applied elastase concentration affects initial elastin degradation, as well as long-term vessel expansion. Additionally, we assessed the effects of BAPN by (1) removing it to restore the cross-linking capability of tissue after aneurysm formation and (2) adding it to animals with stable aneurysms to interrupt cross-linking. These results demonstrate that, even after aneurysm formation, lysyl oxidase inhibition remains necessary for continued expansion. Removing BAPN reduces the aneurysm growth rate to near zero, resulting in a stable aneurysm. In contrast, adding BAPN causes a stable aneurysm to expand. Altogether, these results demonstrate the ability of elastase concentration and BAPN to modulate aneurysm growth rate and severity. The findings open several new areas of investigation in a murine model that mimics many aspects of human AAA.
Collapse
Affiliation(s)
- Alycia G Berman
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN, 47907, USA
| | - Daniel J Romary
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN, 47907, USA
| | - Katherine E Kerr
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN, 47907, USA
| | - Natalyn E Gorazd
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN, 47907, USA
| | - Morgan M Wigand
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN, 47907, USA
| | - Sourav S Patnaik
- Department of Mechanical Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Ender A Finol
- Department of Mechanical Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Abigail D Cox
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN, 47907, USA.
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
40
|
Weiss D, Latorre M, Rego BV, Cavinato C, Tanski BJ, Berman AG, Goergen CJ, Humphrey JD. Biomechanical consequences of compromised elastic fiber integrity and matrix cross-linking on abdominal aortic aneurysmal enlargement. Acta Biomater 2021; 134:422-434. [PMID: 34332103 DOI: 10.1016/j.actbio.2021.07.059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/07/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022]
Abstract
Abdominal aortic aneurysms (AAAs) are characterized histopathologically by compromised elastic fiber integrity, lost smooth muscle cells or their function, and remodeled collagen. We used a recently introduced mouse model of AAAs that combines enzymatic degradation of elastic fibers and blocking of lysyl oxidase, and thus matrix cross-linking, to study progressive dilatation of the infrarenal abdominal aorta, including development of intraluminal thrombus. We quantified changes in biomaterial properties and biomechanical functionality within the aneurysmal segment as a function of time of enlargement and degree of thrombosis. Towards this end, we combined multi-modality imaging with state-of-the art biomechanical testing and histology to quantify regional heterogeneities for the first time and we used a computational model of arterial growth and remodeling to test multiple hypotheses, suggested by the data, regarding the degree of lost elastin, accumulation of glycosaminoglycans, and rates of collagen turnover. We found that standard histopathological findings can be misleading, while combining advanced experimental and computational methods revealed that glycosaminoglycan accumulation is pathologic, not adaptive, and that heightened collagen deposition is ineffective if not cross-linked. In conclusion, loss of elastic fiber integrity can be a strong initiator of aortic aneurysms, but it is the rate and effectiveness of fibrillar collagen remodeling that dictates enlargement. STATEMENT OF SIGNIFICANCE: Precise mechanisms by which abdominal aortic aneurysms enlarge remain unclear, but a recent elastase plus β-aminopropionitrile mouse model provides new insight into disease progression. As in the human condition, the aortic degeneration and adverse remodeling are highly heterogeneous in this model. Our multi-modality experiments quantify and contrast the heterogeneities in geometry and biomaterial properties, and our computational modeling shows that standard histopathology can be misleading. Neither accumulating glycosaminoglycans nor frustrated collagen synthesis slow disease progression, thus highlighting the importance of stimulating adaptive collagen remodeling to limit lesion enlargement.
Collapse
Affiliation(s)
- D Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - M Latorre
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - B V Rego
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - C Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - B J Tanski
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - A G Berman
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - C J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - J D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
41
|
Transglutaminase 2 moderates the expansion of mouse abdominal aortic aneurysms. JVS Vasc Sci 2021; 2:95-109. [PMID: 34617062 PMCID: PMC8489235 DOI: 10.1016/j.jvssci.2021.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/01/2021] [Indexed: 11/23/2022] Open
Abstract
Objective Previously published work has indicated that transcripts encoding transglutaminase 2 (TG2) increase markedly in a rat model of abdominal aortic aneurysm. This study determines whether TG2 and the related TG, factor XIII-A (FXIII-A), protect against aortic aneurysm development in mice. Methods C57BL/6J wild-type, Tgm2 -/- knockout, F13a1 -/- knockout, and Tgm2 -/- /F13a1 -/- double knockout mice were subjected to laparotomy and periaortic application of CaCl2. Results Tgm2 -/- mice showed slightly greater aortic dilatation at 6 weeks after treatment when compared with wild type. However, vessels from Tgm2 -/- mice, but not wild-type mice, continued to dilate up to 6 months after injury and by 24 weeks, a greater number of Tgm2 -/- mice had developed aneurysms (16/17 vs 10/19; P = .008). Laparotomy resulted in a high death rate in F13a1 -/- knockout mice, more frequently from cardiac complications than from hemorrhage, but among F13a1 -/- mice that survived for 6 weeks after CaCl2 treatment, abdominal aortic aneurysm diameter was unaltered relative to wild-type mice. Laparotomy resulted in a higher death rate among Tgm2 -/- /F13a1 -/- double knockout mice, owing to an increased frequency of delayed bleeding. Surprisingly, Tgm2 -/- /F13a1 -/- double knockout mice showed a trend toward decreased dilatation of the aorta 6 weeks after injury, and this finding was replicated in Tgm2 -/- /F13a1 -/- mice subjected to carotid artery injury. Levels of transcripts encoding TG2 were not increased in the aortas of injured wild-type or F13a1 -/- knockout mice relative to uninjured mice, although changes in the levels of other transcripts accorded with previous descriptions of the CaCl2 aneurysm model in mice. Conclusions Knockout of Tgm2, but not F13a1 exacerbates aortic dilatation, suggesting that TG2 confers protection. However, levels of TG2 messenger RNA are not acutely elevated after injury. FXIII-A plays a role in preventing postoperative damage after laparotomy, confirming previous reports that it prevents distal organ damage after trauma. TG2 promotes wound healing after surgery and, in its absence, the bleeding diathesis associated with FXIII-A deficiency is further exposed.
Collapse
|
42
|
Wu XW, Li G, Cheng XB, Wang M, Wang LL, Wang HH, Yang JY, Hu XJ. Association of Angiotensin II Type 1 Receptor Agonistic Autoantibodies With Outcomes in Patients With Acute Aortic Dissection. JAMA Netw Open 2021; 4:e2127587. [PMID: 34596673 PMCID: PMC8486983 DOI: 10.1001/jamanetworkopen.2021.27587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
IMPORTANCE Angiotensin II is significantly associated with the pathogenesis of acute aortic dissection. Angiotensin II type 1 receptor agonistic autoantibodies (AT1-AAs) can mimic the effect of angiotensin II. OBJECTIVE To investigate the association between AT1-AAs and all-cause and cause-specific mortality risk in patients with acute aortic dissection. DESIGN, SETTING, AND PARTICIPANTS A total of 662 patients with clinically suspected aortic dissection from 3 medical centers in Wuhan, China, were enrolled in this cohort study from August 1, 2014, to July 31, 2016. Of these, 315 patients were included in the 3-year follow-up study. Follow-up was mainly performed via telephone interviews and outpatient clinic visits. Data analysis was conducted from March 1 to May 31, 2020. MAIN OUTCOMES AND MEASURES The primary outcomes of interest were all-cause mortality, death due to aortic dissection, and late aortic-related adverse events. RESULTS The full study cohort included 315 patients with AAD (mean [SD] age, 56.2 [12.7] years; 230 men [73.0%]). Ninety-two patients (29.2%) were positive for AT1-AAs. The mortality of AT1-AA-positive patients was significantly higher than that of AT1-AA-negative patients (40 [43.5%] vs 37 [16.6%]; P < .001). The mortality risk in AT1-AA-positive patients was always significantly higher than that in AT1-AA-negative patients in patients with both type A and type B dissection. Multivariable analysis showed that the risk of AT1-AA-positive patients for type A dissection was significantly higher than that of AT1-AA-negative patients (odds ratio [OR], 1.88; 95% CI, 1.12-3.13; P = .02). The Cox proportional hazards regression model showed a significant increase of all-cause mortality risk (OR, 2.27; 95% CI, 1.44-3.57; P < .001) and late aortic-related adverse events (OR, 1.58; 95% CI, 1.06-2.36; P = .03) among AT1-AA-positive patients during the follow-up period compared with AT1-AA-negative patients. CONCLUSIONS AND RELEVANCE This cohort study first detected AT1-AAs in patients with acute aortic dissection. The presence of AT1-AAs was associated with significantly higher all-cause and cause-specific mortality during a follow-up period of 3 years. The antibodies may be a risk factor for aortic dissection.
Collapse
Affiliation(s)
- Xiao-wei Wu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Li
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| | - Xiao-bin Cheng
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Min Wang
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Liu-lin Wang
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Hai-hao Wang
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-ye Yang
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing-jian Hu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Wortmann M, Peters AS, Erhart P, Körfer D, Böckler D, Dihlmann S. Inflammasomes in the Pathophysiology of Aortic Disease. Cells 2021; 10:cells10092433. [PMID: 34572082 PMCID: PMC8468335 DOI: 10.3390/cells10092433] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/26/2021] [Accepted: 08/29/2021] [Indexed: 12/27/2022] Open
Abstract
Aortic diseases comprise aneurysms, dissections, and several other pathologies. In general, aging is associated with a slow but progressive dilation of the aorta, along with increased stiffness and pulse pressure. The progression of aortic disease is characterized by subclinical development or acute presentation. Recent evidence suggests that inflammation participates causally in different clinical manifestations of aortic diseases. As of yet, diagnostic imaging and surveillance is mainly based on ultrasonography, computed tomography (CT), and magnetic resonance imaging (MRI). Little medical therapy is available so far to prevent or treat the majority of aortic diseases. Endovascular therapy by the introduction of covered stentgrafts provides the main treatment option, although open surgery and implantation of synthetic grafts remain necessary in many situations. Because of the risks associated with surgery, there is a need for identification of pharmaceutical targets interfering with the pathophysiology of aortic remodeling. The participation of innate immunity and inflammasome activation in different cell types is common in aortic diseases. This review will thus focus on inflammasome activities in vascular cells of different chronic and acute aortic diseases and discuss their role in development and progression. We will also identify research gaps and suggest promising therapeutic targets, which may be used for future medical interventions.
Collapse
|
44
|
Logghe G, Trachet B, Segers P, De Backer J, Mulorz J, Dueppers P, Vermassen F, Schelzig H, Van Herzeele I, Wagenhäuser MU. Outflow Through Aortic Side Branches Drives False Lumen Patency in Type B Aortic Dissection. Front Cardiovasc Med 2021; 8:710603. [PMID: 34485410 PMCID: PMC8414589 DOI: 10.3389/fcvm.2021.710603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 07/02/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: Thoracic endovascular aortic repair (TEVAR) for type B aortic dissection (TBAD) aims to induce false lumen (FL) thrombosis by sealing intimal tears between the true (TL) and the FL, and blocking the inflow into the FL. Incomplete thrombosis of the FL is correlated with poor clinical outcome. We hypothesize that the number of major and minor branches arising from the FL affects FL patency and may negatively influence TEVAR induced FL thrombosis. Methods: Computed tomography (CT)-scans from 89 patients diagnosed with TBAD [best medical treatment (BMT) n = 52, TEVAR n = 37] from two high-volume vascular surgery centers were analyzed retrospectively. Analysis included evaluation of the FL patency status, the number, location and size of intimal tears, and the presence of minor and major side branches originating from the FL. Multiple regression analysis was conducted to evaluate obtained parameters as predictors for FL thrombosis status. Results: In univariate analysis, the strongest correlation for FL patency was found for the number of major (R = 0.79) and minor (R = 0.86) side branches originating from the FL. When applying a multiple linear regression model, the number of major (normalized beta 0.37; P < 0.001) and minor (normalized beta 0.41; P < 0.01) side branches arising from the FL were valid predictors for the axial length of the patent and non-patent FL, and additionally determined the length of the patent FL at 12-month follow-up in patients that underwent TEVAR. Conclusions: Our data suggest that the number of minor side branches that originate from the FL in TBAD is an important determinant of FL patency, to a greater degree than previously assumed.
Collapse
Affiliation(s)
- Gerlinde Logghe
- Institute for Biomedical Engineering and Technology, Ghent University, Ghent, Belgium
| | - Bram Trachet
- Institute for Biomedical Engineering and Technology, Ghent University, Ghent, Belgium
| | - Patrick Segers
- Institute for Biomedical Engineering and Technology, Ghent University, Ghent, Belgium
| | - Julie De Backer
- Department of Cardiology and Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Joscha Mulorz
- Department of Vascular- and Endovascular Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Philip Dueppers
- Department of Vascular- and Endovascular Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.,Department of Vascular Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Frank Vermassen
- Department of Thoracic and Vascular Surgery, Ghent University Hospital, Ghent, Belgium
| | - Hubert Schelzig
- Department of Vascular- and Endovascular Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Isabelle Van Herzeele
- Department of Thoracic and Vascular Surgery, Ghent University Hospital, Ghent, Belgium
| | - Markus U Wagenhäuser
- Department of Vascular- and Endovascular Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
45
|
Gäbel G, Northoff BH, Balboa A, Becirovic-Agic M, Petri M, Busch A, Maegdefessel L, Mahlmann A, Ludwig S, Teupser D, de Waard V, Golledge J, Wanhainen A, Wågsäter D, Holdt LM, Lindeman JHN. Parallel Murine and Human Aortic Wall Genomics Reveals Metabolic Reprogramming as Key Driver of Abdominal Aortic Aneurysm Progression. J Am Heart Assoc 2021; 10:e020231. [PMID: 34420357 PMCID: PMC8649280 DOI: 10.1161/jaha.120.020231] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background While numerous interventions effectively interfered with abdominal aortic aneurysm (AAA) formation/progression in preclinical models, none of the successes translated into clinical success. Hence, a systematic exploration of parallel and divergent processes in clinical AAA disease and its 2 primary models (the porcine pancreatic elastase and angiotensin-II infusion [AngII] murine model) was performed to identify mechanisms relevant for aneurysm disease. Methods and Results This study combines Movat staining and pathway analysis for histological and genomic comparisons between clinical disease and its models. The impact of a notable genomic signal for metabolic reprogramming was tested in a rescue trial (AngII model) evaluating the impact of 1-(4-pyridinyl)-3-(2-quinolinyl)-2-propen-1-one (PFK15)-mediated interference with main glycolytic switch PFKFB3. Histological evaluation characterized the AngII model as a dissection model that is accompanied by adventitial fibrosis. The porcine pancreatic elastase model showed a transient inflammatory response and aortic dilatation, followed by stabilization and fibrosis. Normalization of the genomic responses at day 14 confirmed the self-limiting nature of the porcine pancreatic elastase model. Clear parallel genomic responses with activated adaptive immune responses, and particularly strong signals for metabolic switching were observed in human AAA and the AngII model. Rescue intervention with the glycolysis inhibitor PFK15 in the AngII model showed that interference with the glycolytic switching quenches aneurysm formation. Conclusions Despite clear morphological contrasts, remarkable genomic parallels exist for clinical AAA disease and the AngII model. The metabolic response appears causatively involved in AAA progression and provides a novel therapeutic target. The clear transient genomic response classifies the porcine pancreatic elastase model as a disease initiation model.
Collapse
Affiliation(s)
- Gabor Gäbel
- Department of Vascular Surgery HELIOS Klinikum Krefeld Krefeld Germany
| | - Bernd H Northoff
- Institute of Laboratory Medicine Ludwig-Maximilians-University Munich Munich Germany
| | - Amanda Balboa
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| | | | - Marcelo Petri
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| | - Albert Busch
- Department of Vascular and Endovascular Surgery Technical University Munich Munich Germany
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery Technical University Munich Munich Germany
| | - Adrian Mahlmann
- University Centre for Vascular Medicine University Hospital Carl Gustav CarusTechnical University Dresden Dresden Germany
| | - Stefan Ludwig
- University Centre for Vascular Medicine University Hospital Carl Gustav CarusTechnical University Dresden Dresden Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine Ludwig-Maximilians-University Munich Munich Germany
| | - Vivian de Waard
- Department Medical Biochemistry Amsterdam University Medical CentersAmsterdam Cardiovascular SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease College of Medicine and Dentistry James Cook University Townsville Qld. Australia
| | - Anders Wanhainen
- Department of Surgical Sciences Section of Vascular Surgery Uppsala University Uppsala Sweden
| | - Dick Wågsäter
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| | - Lesca M Holdt
- Institute of Laboratory Medicine Ludwig-Maximilians-University Munich Munich Germany
| | - Jan H N Lindeman
- Department of Vascular Surgery Leiden University Medical Center (LUMC) Leiden The Netherlands
| |
Collapse
|
46
|
Mochida A, Mita T, Azuma K, Osonoi Y, Masuyama A, Nakajima K, Goto H, Nishida Y, Miyatsuka T, Mitsumata M, Watada H. Defective autophagy in vascular smooth muscle cells enhances the healing of abdominal aortic aneurysm. Physiol Rep 2021; 9:e15000. [PMID: 34491001 PMCID: PMC8422599 DOI: 10.14814/phy2.15000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/02/2021] [Accepted: 07/18/2021] [Indexed: 11/24/2022] Open
Abstract
Autophagy is an evolutionarily conserved cellular catabolic process essential for cell homeostasis, and thus its failure is associated with several diseases. While autophagy has been reported to play a role in vascular smooth muscle cells (SMCs) in vascular disorders, its precise role in the pathogenesis of abdominal aortic aneurysm (AAA) has not yet been elucidated. In this study, we investigated the role of SMC autophagy in AAA formation. As a mouse model of AAA, we used control apolipoprotein E-deficient (apoeKO) mice and Atg7cKO (SMC-specific Atg7-deficient mice):apoeKO mice administered angiotensin II for 4 weeks. Intriguingly, Kaplan-Meier curves showed that the survival rates of Atg7cKO:apoeKO mice were significantly higher than those of apoeKO mice. The hematoma area in AAA of Atg7cKO:apoeKO mice was smaller than in apoeKO mice despite the lack of a significant difference in AAA incidence between the two groups. Furthermore, the amount of granulomatous tissues was significantly larger and the collagen-positive area within AAA was significantly larger in Atg7cKO:apoeKO mice than in apoeKO mice. In accordance with these findings, SMCs cultured from Atg7cKO mice showed increased expression of collagens, independent of angiotensin II action. Taken together, our data suggest that defective autophagy in SMCs elicits AAA healing that may underlie the better survival rate under dyslipidemia and angiotensin II infusion.
Collapse
MESH Headings
- Angiotensin II/administration & dosage
- Angiotensin II/toxicity
- Animals
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/pathology
- Autophagy/drug effects
- Autophagy/physiology
- Cells, Cultured
- Infusion Pumps, Implantable
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
Collapse
Affiliation(s)
- Akihiro Mochida
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Tomoya Mita
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
- Center for Therapeutic Innovations in DiabetesJuntendo University Graduate School of MedicineTokyoJapan
- Center for Identification of Diabetic Therapeutic TargetsJuntendo University Graduate School of MedicineTokyoJapan
| | - Kosuke Azuma
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Yusuke Osonoi
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Atsushi Masuyama
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Kenichi Nakajima
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Hiromasa Goto
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Yuya Nishida
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Takeshi Miyatsuka
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Masako Mitsumata
- Division of PathologyDepartment of Clinical LaboratoryYamanashi Kosei HospitalYamanashi cityYamanashiJapan
| | - Hirotaka Watada
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
- Center for Therapeutic Innovations in DiabetesJuntendo University Graduate School of MedicineTokyoJapan
- Center for Identification of Diabetic Therapeutic TargetsJuntendo University Graduate School of MedicineTokyoJapan
| |
Collapse
|
47
|
Current pharmacological management of aortic aneurysm. J Cardiovasc Pharmacol 2021; 78:211-220. [PMID: 33990514 DOI: 10.1097/fjc.0000000000001054] [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: 02/11/2021] [Accepted: 04/23/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Aortic aneurysm (AA) remains one of the primary causes of death worldwide. Of the major treatments, prophylactic operative repair is used for AA to avoid potential aortic dissection (AD) or rupture. To halt the development of AA and alleviate its progression into AD, pharmacological treatment has been investigated for years. Currently, β-adrenergic blocking agents, losartan, irbesartan, angiotensin-converting-enzyme inhibitors, statins, antiplatelet agents, doxycycline, and metformin have been investigated as potential candidates for preventing AA progression. However, the paradox between preclinical successes and clinical failures still exists, with no medical therapy currently available for ideally negating the disease progression. This review describes the current drugs used for pharmacological management of AA and their individual potential mechanisms. Preclinical models for drug screening and evaluation are also discussed to gain a better understanding of the underlying pathophysiology and ultimately find new therapeutic targets for AA.
Collapse
|
48
|
Brunet J, Pierrat B, Badel P. Review of Current Advances in the Mechanical Description and Quantification of Aortic Dissection Mechanisms. IEEE Rev Biomed Eng 2021; 14:240-255. [PMID: 31905148 DOI: 10.1109/rbme.2019.2950140] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Aortic dissection is a life-threatening event associated with a very poor outcome. A number of complex phenomena are involved in the initiation and propagation of the disease. Advances in the comprehension of the mechanisms leading to dissection have been made these last decades, thanks to improvements in imaging and experimental techniques. However, the micro-mechanics involved in triggering such rupture events remains poorly described and understood. It constitutes the primary focus of the present review. Towards the goal of detailing the dissection phenomenon, different experimental and modeling methods were used to investigate aortic dissection, and to understand the underlying phenomena involved. In the last ten years, research has tended to focus on the influence of microstructure on initiation and propagation of the dissection, leading to a number of multiscale models being developed. This review brings together all these materials in an attempt to identify main advances and remaining questions.
Collapse
|
49
|
Kopacz A, Klóska D, Werner E, Hajduk K, Grochot-Przęczek A, Józkowicz A, Piechota-Polańczyk A. A Dual Role of Heme Oxygenase-1 in Angiotensin II-Induced Abdominal Aortic Aneurysm in the Normolipidemic Mice. Cells 2021; 10:cells10010163. [PMID: 33467682 PMCID: PMC7830394 DOI: 10.3390/cells10010163] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 11/16/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) bears a high risk of rupture and sudden death of the patient. The pathogenic mechanisms of AAA remain elusive, and surgical intervention represents the only treatment option. Heme oxygenase-1 (HO-1), a heme degrading enzyme, is induced in AAA, both in mice and humans. HO-1 was reported to mitigate AAA development in an angiotensin II (AngII)-induced model of AAA in hyperlipidemic ApoE-/- mice. Since the role of hyperlipidaemia in the pathogenesis of AAA remains controversial, we aimed to evaluate the significance of HO-1 in the development and progression of AAA in normolipidemic animals. The experiments were performed in HO-1-deficient mice and their wild-type counterparts. We demonstrated in non-hypercholesterolemic mice that the high-dose of AngII leads to the efficient formation of AAA, which is attenuated by HO-1 deficiency. Yet, if formed, they are significantly more prone to rupture upon HO-1 shortage. Differential susceptibility to AAA formation does not rely on enhanced inflammatory response or oxidative stress. AAA-resistant mice are characterized by an increase in regulators of aortic remodeling and angiotensin receptor-2 expression, significant medial thickening, and delayed blood pressure elevation in response to AngII. To conclude, we unveil a dual role of HO-1 deficiency in AAA in normolipidemic mice, where it protects against AAA development, but exacerbates the state of formed AAA.
Collapse
Affiliation(s)
- Aleksandra Kopacz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
| | - Damian Klóska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
| | - Ewa Werner
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
- Department of Animal Reproduction, Anatomy and Genomic, Faculty of Animal Science, University of Agriculture, 30-059 Krakow, Poland
| | - Karolina Hajduk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
| | - Anna Grochot-Przęczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
| | - Aleksandra Piechota-Polańczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-392 Krakow, Poland; (A.K.); (D.K.); (E.W.); (K.H.); (A.G.-P.); (A.J.)
- Correspondence:
| |
Collapse
|
50
|
Meng J, Liu HL, Ma D, Wang HY, Peng Y, Wang HL. Upregulation of aurora kinase A promotes vascular smooth muscle cell proliferation and migration by activating the GSK-3β/β-catenin pathway in aortic-dissecting aneurysms. Life Sci 2020; 262:118491. [DOI: 10.1016/j.lfs.2020.118491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/29/2022]
|