1
|
Cui Y, Tan C, Zhang W, Jiang P, Sun J, Mei F. Establishment of Mouse Models of Abdominal Aortic Aneurysm. Angiology 2024:33197241284848. [PMID: 39268808 DOI: 10.1177/00033197241284848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a chronic vascular disease that commonly affects elderly individuals but has recently increased in younger populations. As the aneurysm grows, it can cause compression symptoms such as abdominal pain, rupture, and bleeding, which are absent in the early stages. Once an AAA ruptures and causes bleeding, the mortality rate is alarmingly high. Currently, the pathogenesis for AAA is unknown, and therapeutic options are limited, necessitating improvement in treatment efficacy. An essential research method for studying the processes and potential treatment of AAA is establishing animal models using mice. The present study provides a detailed overview of the widely used AAA mouse animal models and their construction strategies, advantages, disadvantages, scope of applications, and prospects.
Collapse
Affiliation(s)
- Yongpan Cui
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Chengpeng Tan
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Wuming Zhang
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Peng Jiang
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Jianfeng Sun
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Fei Mei
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| |
Collapse
|
2
|
Di Gregoli K, Atkinson G, Williams H, George SJ, Johnson JL. Pharmacological Inhibition of MMP-12 Exerts Protective Effects on Angiotensin II-Induced Abdominal Aortic Aneurysms in Apolipoprotein E-Deficient Mice. Int J Mol Sci 2024; 25:5809. [PMID: 38891996 PMCID: PMC11172660 DOI: 10.3390/ijms25115809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Human abdominal aortic aneurysms (AAAs) are characterized by increased activity of matrix metalloproteinases (MMP), including MMP-12, alongside macrophage accumulation and elastin degradation, in conjunction with superimposed atherosclerosis. Previous genetic ablation studies have proposed contradictory roles for MMP-12 in AAA development. In this study, we aimed to elucidate if pharmacological inhibition of MMP-12 activity with a phosphinic peptide inhibitor protects from AAA formation and progression in angiotensin (Ang) II-infused Apoe-/- mice. Complimentary studies were conducted in a human ex vivo model of early aneurysm development. Administration of an MMP-12 inhibitor (RXP470.1) protected hypercholesterolemia Apoe-/- mice from Ang II-induced AAA formation and rupture-related death, associated with diminished medial thinning and elastin fragmentation alongside increased collagen deposition. Proteomic analyses confirmed a beneficial effect of MMP-12 inhibition on extracellular matrix remodeling proteins combined with inflammatory pathways. Furthermore, RXP470.1 treatment of mice with pre-existing AAAs exerted beneficial effects as observed through suppressed aortic dilation and rupture, medial thinning, and elastin destruction. Our findings indicate that pharmacological inhibition of MMP-12 activity retards AAA progression and improves survival in mice providing proof-of-concept evidence to motivate translational work for MMP-12 inhibitor therapy in humans.
Collapse
Affiliation(s)
| | | | | | | | - Jason L. Johnson
- Laboratory of Cardiovascular Pathology, Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK; (K.D.G.); (G.A.); (H.W.); (S.J.G.)
| |
Collapse
|
3
|
Hof A, Guthoff H, Ahdab M, Landerer M, Schäkel J, Niehues J, Schorscher M, Zimmermann O, Winkels H, von Stein P, Geißen S, Baldus S, Adam M, Mollenhauer M, Mehrkens D. Vascular Ultrasound for In Vivo Assessment of Arterial Pathologies in a Murine Model of Atherosclerosis and Aortic Aneurysm. Int J Mol Sci 2023; 24:15261. [PMID: 37894941 PMCID: PMC10607567 DOI: 10.3390/ijms242015261] [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: 09/18/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Vascular diseases like atherosclerosis and abdominal aortic aneurysm (AAA) are common pathologies in the western world, promoting various potentially fatal conditions. Here, we evaluate high-resolution (HR) ultrasound in mouse models of atherosclerosis and AAA as a useful tool for noninvasive monitoring of early vascular changes in vivo. We used Apolipoprotein E-deficient (ApoE-/-) mice as an atherosclerosis model and induced AAA development by the implementation of Angiotensin II-releasing osmotic minipumps. HR ultrasound of the carotid artery or the abdominal aorta was performed to monitor vascular remodeling in vivo. Images were analyzed by speckle tracking algorithms and correlated to histological analyses and subsequent automated collagen quantification. Consistent changes were observed via ultrasound in both models: Global radial strain (GRS) was notably reduced in the AAA model (23.8 ± 2.8% vs. 12.5 ± 2.5%, p = 0.01) and in the atherosclerotic mice (20.6 ± 1.3% vs. 15.8 ± 0.9%, p = 0.02). In mice with AAA, vessel distensibility was significantly reduced, whereas intima-media thickness was increased in atherosclerotic mice. The area and collagen content of the tunica media were increased in diseased arteries of both models as measured by automated image analysis of Picrosirius Red-stained aortic sections. Correlation analysis revealed a strong correlation of multiple parameters, predicting early vascular damage in HR ultrasound and histological examinations. In conclusion, our findings underscore the potential of HR ultrasound in effectively tracing early alterations in arterial wall properties in murine models of atherosclerosis and AAA.
Collapse
Affiliation(s)
- Alexander Hof
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50937 Cologne, Germany
| | - Henning Guthoff
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50937 Cologne, Germany
| | - Maysam Ahdab
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
| | - Max Landerer
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
| | - Jasper Schäkel
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50937 Cologne, Germany
| | - Jana Niehues
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
| | - Maximilian Schorscher
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50937 Cologne, Germany
| | - Oscar Zimmermann
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50937 Cologne, Germany
| | - Holger Winkels
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50937 Cologne, Germany
| | - Philipp von Stein
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
| | - Simon Geißen
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50937 Cologne, Germany
| | - Stephan Baldus
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50937 Cologne, Germany
| | - Matti Adam
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50937 Cologne, Germany
| | - Martin Mollenhauer
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50937 Cologne, Germany
| | - Dennis Mehrkens
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, Clinic III for Internal Medicine, University Hospital Cologne, 50937 Cologne, Germany; (A.H.); (M.A.); (M.L.); (J.S.); (J.N.); (M.S.); (O.Z.); (H.W.); (P.v.S.); (S.G.); (S.B.); (M.A.); (M.M.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
4
|
Atkinson G, Bianco R, Di Gregoli K, Johnson JL. The contribution of matrix metalloproteinases and their inhibitors to the development, progression, and rupture of abdominal aortic aneurysms. Front Cardiovasc Med 2023; 10:1248561. [PMID: 37799778 PMCID: PMC10549934 DOI: 10.3389/fcvm.2023.1248561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/07/2023] [Indexed: 10/07/2023] Open
Abstract
Abdominal aortic aneurysms (AAAs) account for up to 8% of deaths in men aged 65 years and over and 2.2% of women. Patients with AAAs often have atherosclerosis, and intimal atherosclerosis is generally present in AAAs. Accordingly, AAAs are considered a form of atherosclerosis and are frequently referred to as atherosclerotic aneurysms. Pathological observations advocate inflammatory cell infiltration alongside adverse extracellular matrix degradation as key contributing factors to the formation of human atherosclerotic AAAs. Therefore, macrophage production of proteolytic enzymes is deemed responsible for the damaging loss of ECM proteins, especially elastin and fibrillar collagens, which characterise AAA progression and rupture. Matrix metalloproteinases (MMPs) and their regulation by tissue inhibitors metalloproteinases (TIMPs) can orchestrate not only ECM remodelling, but also moderate the proliferation, migration, and apoptosis of resident aortic cells, alongside the recruitment and subsequent behaviour of inflammatory cells. Accordingly, MMPs are thought to play a central regulatory role in the development, progression, and eventual rupture of abdominal aortic aneurysms (AAAs). Together, clinical and animal studies have shed light on the complex and often diverse effects MMPs and TIMPs impart during the development of AAAs. This dichotomy is underlined from evidence utilising broad-spectrum MMP inhibition in animal models and clinical trials which have failed to provide consistent protection from AAA progression, although more encouraging results have been observed through deployment of selective inhibitors. This review provides a summary of the supporting evidence connecting the contribution of individual MMPs to AAA development, progression, and eventual rupture. Topics discussed include structural, functional, and cell-specific diversity of MMP members; evidence from animal models of AAA and comparisons with findings in humans; the dual role of MMPs and the requirement to selectively target individual MMPs; and the advances in identifying aberrant MMP activity. As evidenced, our developing understanding of the multifaceted roles individual MMPs perform during the progression and rupture of AAAs, should motivate clinical trials assessing the therapeutic potential of selective MMP inhibitors, which could restrict AAA-related morbidity and mortality worldwide.
Collapse
Affiliation(s)
| | | | | | - Jason L. Johnson
- Laboratory of Cardiovascular Pathology, Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
5
|
Wang X, Ma J, Lin D, Dong X, Wu J, Bai Y, Zhang D, Gao J. The risk factors of postoperative hypoxemia in patients with Stanford type A acute aortic dissection. Medicine (Baltimore) 2023; 102:e34704. [PMID: 37603505 PMCID: PMC10443739 DOI: 10.1097/md.0000000000034704] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/20/2023] [Indexed: 08/23/2023] Open
Abstract
Hypoxemia is one of the most common complications in patients after Stanford type A acute aortic dissection surgery. The aim of this study was to investigate the association of circulating ANG II level with postoperative hypoxemia and to identify the risk factors for postoperative hypoxemia in Stanford type A acute aortic dissection patients. In this study, 88 patients who underwent Stanford type A acute aortic dissection surgery were enrolled. Postoperative hypoxemia is defined by the oxygenation index (OI). Perioperative clinical data were collected and the serum ANG II and sACE2 levels were measured. The differences in the basic characteristics, intraoperative details, biochemical parameters, laboratory test data and clinical outcomes were compared between the hypoxemia group and the non-hypoxemia group by univariate analysis. Multivariate logistic regression analysis was performed on the variables with P < .1 in univariate analysis or that were considered clinically important to identify risk factors for postoperative hypoxemia. Twenty-five patients (28.4%) were considered to have postoperative hypoxemia (OI ≤ 200 mm Hg). The ANG II concentration remained a risk factor associated with postoperative hypoxemia [OR = 1.018, 95% CI (1.003-1.034), P = .022]. The other risk factors remaining in the logistic regression model were BMI [OR = 1.417, 95% CI (1.159-1.733), P = .001] and cTnI [OR = 1.003, 95% CI (1.000-1.005), P = .032]. Elevated levels of ANG II, BMI and cTnI are risk factors for postoperative hypoxemia in patients with Stanford type A acute aortic dissection.
Collapse
Affiliation(s)
- Xu’an Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jun Ma
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Duomao Lin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiuhua Dong
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jinjing Wu
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yang Bai
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Dongni Zhang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Junwei Gao
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
De Moudt S, Hendrickx JO, De Meyer GRY, Martinet W, Fransen P. Disparate biomechanical properties of the aorta in non-aneurysmal and aneurysmal mice treated with angiotensin II. Physiol Rep 2022; 10:e15410. [PMID: 36117398 PMCID: PMC9483617 DOI: 10.14814/phy2.15410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 06/15/2023] Open
Abstract
In vivo angiotensin II (AngII)-treatment is a widely used experimental model to induce cardiovascular disease and results in a high likelihood of abdominal aorta aneurysm (AAA) formation. This involves progressive and irreversible focal dilation of the abdominal aorta and induces adverse aortic connective tissue remodeling contributing to aortic wall stiffening through inflammation, elastin degradation, and collagen restructuring. Hence, the present study aimed to investigate how AAA formation in AngII-treated mice affects aortic function and biomechanics. To this end, C57Bl/6J mice were treated with AngII (1000 ng/[kg.min]) or PBS infusion for 28 days. Peripheral blood pressure, echocardiography, and aortic pulse wave velocity were measured in vivo. Thoracic aorta rings were studied ex vivo in organ chambers, while aortic vascular smooth muscle cell (VSMC) phenotype was investigated histologically. We confirmed peripheral hypertension, cardiac hypertrophy, aortic stiffening, and increased VSMC proliferation and migration after AngII-treatment. Abdominal aorta aneurysm formation was observed in 8/13 AngII-treated mice. Ex vivo thoracic aortic rings of both aneurysmal and non-aneurysmal AngII-treated mice showed high isobaric aortic stiffness, endothelial dysfunction, heightened α1 -adrenergic contractility, and altered VSMC contractile calcium signaling. However, aortic biomechanics were differently affected, with heightened α1 -adrenoreceptor mediated aortic stiffening in non-aneurysmal mice, whereas contraction-dependent stiffening was impaired in aneurysmal mice. In conclusion, although aneurysmal and non-aneurysmal 4-week AngII-treated mice displayed similar changes in aortic physiology, aortic biomechanics were dissimilarly affected.
Collapse
Affiliation(s)
- Sofie De Moudt
- Laboratory of PhysiopharmacologyUniversity of AntwerpAntwerpBelgium
| | | | | | - Wim Martinet
- Laboratory of PhysiopharmacologyUniversity of AntwerpAntwerpBelgium
| | - Paul Fransen
- Laboratory of PhysiopharmacologyUniversity of AntwerpAntwerpBelgium
| |
Collapse
|
7
|
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: 1.5] [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
|
8
|
Weng Y, Lou J, Bao Y, Cai C, Zhu K, Du C, Chen X, Tang L. Single-Cell RNA Sequencing Technology Revealed the Pivotal Role of Fibroblast Heterogeneity in Angiotensin II-Induced Abdominal Aortic Aneurysms. DNA Cell Biol 2022; 41:498-520. [PMID: 35451888 DOI: 10.1089/dna.2021.0923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The mechanism of abdominal aortic aneurysm (AAA) has not been fully elucidated. In this study, we aimed to map the cellular heterogeneity, molecular alteration, and functional transformation of angiotensin (Ang) II-induced AAA in mice based on single-cell RNA sequencing (sc-RNA seq) technology. sc-RNA seq was performed on suprarenal abdominal aorta tissue from male Apoe-/- C57BL/6 mice of Ang II-induced AAA and shame models to determine the heterogeneity and phenotypic transformation of all cells. Immunohistochemistry was used to determine the pathophysiological characteristics of AAA. The single-cell trajectory was performed to predict the differentiation of fibroblasts. Finally ligand-receptor analysis was used to evaluate intercellular communication between fibroblasts and smooth muscle cells (SMCs). More than 27,000 cells were isolated and 25 clusters representing 8 types of cells were identified, including fibroblasts, macrophages, endothelial cells, SMCs, T lymphocytes, B lymphocytes, granulocytes, and natural killer cells. During AAA progression, the function and phenotype of different type cells altered separately, including activation of inflammatory cells, alternations of macrophage polarization, phenotypic transformation of vascular smooth muscle cells, and endothelial to mesenchymal transformation. The alterations of fibroblasts were the most conspicuous. Single-cell trajectory revealed the critical reprogramming genes of fibroblasts mainly enriched in regulation of immune system. Finally, the ligand-receptor analysis confirmed that disorder of collagen metabolism led by fibroblasts was one of the most prominent characteristics of Ang II-induced AAA. Our study revealed the cellular heterogeneity of Ang II-induced AAA. Fibroblasts may play a critical role in Ang II-induced AAA progression according to multiple biological functions, including immune regulation and extracellular matrix metabolic balance. Our study may provide us with a different perspective on the etiology and pathogenesis of AAA.
Collapse
Affiliation(s)
- Yingzheng Weng
- Department of Cardiology, Zhejiang Hospital, Hangzhou, China
| | - Jiangjie Lou
- Department of Cardiology, Zhejiang Hospital, Hangzhou, China
| | - Yizong Bao
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Changhong Cai
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, China
| | - Kefu Zhu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, China
| | - Changqing Du
- Department of Cardiology, Zhejiang Hospital, Hangzhou, China.,Department of Medicine, The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaofeng Chen
- Department of Cardiology, Taizhou Hospital, Wenzhou Medical University, Taizhou, China.,Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, China.,Department of Medicine, The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
9
|
Rastogi V, Stefens SJM, Houwaart J, Verhagen HJM, de Bruin JL, van der Pluijm I, Essers J. Molecular Imaging of Aortic Aneurysm and Its Translational Power for Clinical Risk Assessment. Front Med (Lausanne) 2022; 9:814123. [PMID: 35492343 PMCID: PMC9051391 DOI: 10.3389/fmed.2022.814123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/21/2022] [Indexed: 01/03/2023] Open
Abstract
Aortic aneurysms (AAs) are dilations of the aorta, that are often fatal upon rupture. Diagnostic radiological techniques such as ultrasound (US), magnetic resonance imaging (MRI), and computed tomography (CT) are currently used in clinical practice for early diagnosis as well as clinical follow-up for preemptive surgery of AA and prevention of rupture. However, the contemporary imaging-based risk prediction of aneurysm enlargement or life-threatening aneurysm-rupture remains limited as these are restricted to visual parameters which fail to provide a personalized risk assessment. Therefore, new insights into early diagnostic approaches to detect AA and therefore to prevent aneurysm-rupture are crucial. Multiple new techniques are developed to obtain a more accurate understanding of the biological processes and pathological alterations at a (micro)structural and molecular level of aortic degeneration. Advanced anatomical imaging combined with molecular imaging, such as molecular MRI, or positron emission tomography (PET)/CT provides novel diagnostic approaches for in vivo visualization of targeted biomarkers. This will aid in the understanding of aortic aneurysm disease pathogenesis and insight into the pathways involved, and will thus facilitate early diagnostic analysis of aneurysmal disease. In this study, we reviewed these molecular imaging modalities and their association with aneurysm growth and/or rupture risk and their limitations. Furthermore, we outline recent pre-clinical and clinical developments in molecular imaging of AA and provide future perspectives based on the advancements made within the field. Within the vastness of pre-clinical markers that have been studied in mice, molecular imaging targets such as elastin/collagen, albumin, matrix metalloproteinases and immune cells demonstrate promising results regarding rupture risk assessment within the pre-clinical setting. Subsequently, these markers hold potential as a future diagnosticum of clinical AA assessment. However currently, clinical translation of molecular imaging is still at the onset. Future human trials are required to assess the effectivity of potentially viable molecular markers with various imaging modalities for clinical rupture risk assessment.
Collapse
Affiliation(s)
- Vinamr Rastogi
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Sanne J. M. Stefens
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Judith Houwaart
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Hence J. M. Verhagen
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jorg L. de Bruin
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ingrid van der Pluijm
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jeroen Essers
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Radiation Oncology, Erasmus University Medical Center, Rotterdam, Netherlands
- *Correspondence: Jeroen Essers
| |
Collapse
|
10
|
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: 32] [Impact Index Per Article: 16.0] [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
|
11
|
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: 1.5] [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
|
12
|
Zhu E, Yuan C, Hu S, Liao Y, Li B, Zhou Y, Zhou W. Injection of Matrix Metalloproteinase-9 Leads to Ventricular Remodeling. DISEASE MARKERS 2022; 2022:1659771. [PMID: 36193497 PMCID: PMC9526576 DOI: 10.1155/2022/1659771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/06/2022] [Indexed: 12/08/2022]
Abstract
OBJECTIVE Previous studies have found that some ventricular remodeling is accompanied by increased matrix metalloproteinase-9 (MMP-9) in vivo, and MMP-9 inhibitors can reduce ventricular remodeling. However, there is still no direct evidence that MMP-9 causes ventricular remodeling. In this study, MMP-9 was injected into rats to observe whether MMP-9 caused ventricular remodeling, thereby providing direct evidence of MMP-9-induced ventricular remodeling. METHODS Forty-eight eight-week-old male Wistar rats were randomly divided, by weight, into control, low-, medium-, and high-dose MMP-9 groups and were administered normal saline or recombinant rat MMP-9 0.7, 1.4, or 2.1 ng/g, respectively, via intraperitoneal injection, twice per week. On the 28th day, six rats were randomly selected from each group (Stage I). The remaining rats continued receiving injections until the 56th day (Stage II). Echocardiography was performed to observe cardiac structure and function, and the left ventricular mass index (LVWI) was calculated. Myocardial pathological changes and the collagen volume fraction (CVF) were observed by HE and VG staining in myocardial tissue. MMP-9 levels in serum were tested using ELISA. Myocardial MMP-9 levels were measured using Western blots, and the myocardial expression levels of MMP-9 mRNA were assessed using RT-PCR. RESULTS During Stage I, serum MMP-9 and myocardial MMP-9 mRNA levels are increased; hypertrophic cardiomyocytes, disorderly arrangement of fibers, and endochylema dissolution are observed in the medium- and high-dose groups. The left ventricular weight index (LVWI) and myocardial MMP-9 increased, and the collagen volume fraction (CVF) reduced in the high-dose group. In Stage II, the left ventricular end-diastolic volume (LVEDV) and diameter (LVIDd) are higher, and CVF decreased in the medium- and high-dose groups. Myocardial pathological lesions intensified. Serum MMP-9 in the model groups and myocardial MMP-9 in the medium- and high-dose groups are increased. CONCLUSIONS Injection of MMP-9 can lead to ventricular remodeling.
Collapse
Affiliation(s)
- Enzheng Zhu
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Province, 510080, China
| | - Congcong Yuan
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Province, 510080, China
| | - Simiao Hu
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Province, 510080, China
| | - Yiling Liao
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Province, 510080, China
| | - Bowei Li
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Province, 510080, China
| | - Yuliang Zhou
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Province, 510080, China
| | - Wanxing Zhou
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Province, 510080, China
- Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Province, 510080, China
| |
Collapse
|
13
|
Waduud MA, Kandavelu P, Reay M, Paradine K, Scott DJA, Bailey MA. High-Frequency Three-Dimensional Lumen Volume Ultrasound Is a Sensitive Method to Detect Early Aneurysmal Change in Elastase-Induced Murine Abdominal Aortic Aneurysm. AORTA 2021; 9:215-220. [PMID: 34963161 PMCID: PMC8714318 DOI: 10.1055/s-0041-1731404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Objective
The aim of this study was to investigate the reproducibility of anterior–posterior diameter (APd
max
) and three-dimensional lumen volume (3DLV) measurements of abdominal aortic aneurysms (AAA) in a classical murine AAA model. We also compared the magnitude of change in the aortic size detected with each method of assessment.
Methods
Periadventitial application of porcine pancreatic elastase (PPE AAA) or sham surgery was performed in two cohorts of mice. Cohort 1 was used to assess for observer variability with the APd
max
and 3DLV measurements. Cohort 2 highlighted the relationship between APd
max
and 3DLV and changes in AAA detected.
Results
There was no significant observer variability detected with APd
max
measurement. Similarly, no significant intraobserver variability was evident with 3DLV; however, a small but significant interobserver difference was present. APd
max
and 3DLV measurements of PPE AAA significantly correlated. However, changes in the AAA morphology were detected earlier with 3DLV.
Conclusion
APd
max
and 3DLV are both reliable methods for measuring an AAA. Both these methods correlate with each other. However, changes in AAA morphology were detected earlier with 3DLV, which is important to detect subtle but important changes to aortic geometry in a laboratory setting. 3DLV measurement of AAA is a simple, reproducible, and comprehensive method for assessing changes in disease morphology.
Collapse
Affiliation(s)
- Mohammed A. Waduud
- Leeds Institute for Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom
- Leeds Vascular Institute, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Parkavi Kandavelu
- Leeds Institute for Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Melanie Reay
- Leeds Institute for Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Katherine Paradine
- Leeds Institute for Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - David J. A. Scott
- Leeds Institute for Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom
- Leeds Vascular Institute, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - 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 Teaching Hospitals NHS Trust, Leeds, United Kingdom
| |
Collapse
|
14
|
Busch A, Bleichert S, Ibrahim N, Wortmann M, Eckstein HH, Brostjan C, Wagenhäuser MU, Goergen CJ, Maegdefessel L. Translating mouse models of abdominal aortic aneurysm to the translational needs of vascular surgery. JVS Vasc Sci 2021; 2:219-234. [PMID: 34778850 PMCID: PMC8577080 DOI: 10.1016/j.jvssci.2021.01.002] [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: 07/13/2020] [Accepted: 01/04/2021] [Indexed: 01/03/2023] Open
Abstract
Introduction Abdominal aortic aneurysm (AAA) is a condition that has considerable socioeconomic impact and an eventual rupture is associated with high mortality and morbidity. Despite decades of research, surgical repair remains the treatment of choice and no medical therapy is currently available. Animal models and, in particular, murine models, of AAA are a vital tool for experimental in vivo research. However, each of the different models has individual limitations and provide only partial mimicry of human disease. This narrative review addresses the translational potential of the available mouse models, highlighting unanswered questions from a clinical perspective. It is based on a thorough presentation of the available literature and more than a decade of personal experience, with most of the available models in experimental and translational AAA research. Results From all the models published, only the four inducible models, namely the angiotensin II model (AngII), the porcine pancreatic elastase perfusion model (PPE), the external periadventitial elastase application (ePPE), and the CaCl2 model have been widely used by different independent research groups. Although the angiotensin II model provides features of dissection and aneurysm formation, the PPE model shows reliable features of human AAA, especially beyond day 7 after induction, but remains technically challenging. The translational value of ePPE as a model and the combination with β-aminopropionitrile to induce rupture and intraluminal thrombus formation is promising, but warrants further mechanistic insights. Finally, the external CaCl2 application is known to produce inflammatory vascular wall thickening. Unmet translational research questions include the origin of AAA development, monitoring aneurysm growth, gender issues, and novel surgical therapies as well as novel nonsurgical therapies. Conclusion New imaging techniques, experimental therapeutic alternatives, and endovascular treatment options provide a plethora of research topics to strengthen the individual features of currently available mouse models, creating the possibility of shedding new light on translational research questions.
Collapse
Affiliation(s)
- Albert Busch
- Department for Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany.,Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), Berlin, Germany
| | - Sonja Bleichert
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Nahla Ibrahim
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Markus Wortmann
- Department of Vascular and Endovascular Surgery, Universitaetsklinik Heidelberg, Heidelberg, Germany
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
| | - Christine Brostjan
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Markus U Wagenhäuser
- Department of Vascular and Endovascular Surgery, Heinrich-Heine-University Medical Center Düsseldorf, Düsseldorf, Germany
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Ind
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany.,Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), Berlin, Germany
| |
Collapse
|
15
|
Lavin B, Lacerda S, Andia ME, Lorrio S, Bakewell R, Smith A, Rashid I, Botnar RM, Phinikaridou A. Tropoelastin: an in vivo imaging marker of dysfunctional matrix turnover during abdominal aortic dilation. Cardiovasc Res 2020; 116:995-1005. [PMID: 31282949 PMCID: PMC7104357 DOI: 10.1093/cvr/cvz178] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/05/2019] [Indexed: 12/15/2022] Open
Abstract
Aims Dysfunctional matrix turnover is present at sites of abdominal aortic aneurysm (AAA) and leads to the accumulation of monomeric tropoelastin rather than cross-linked elastin. We used a gadolinium-based tropoelastin-specific magnetic resonance contrast agent (Gd-TESMA) to test whether quantifying regional tropoelastin turnover correlates with aortic expansion in a murine model. The binding of Gd-TESMA to excised human AAA was also assessed. Methods and results We utilized the angiotensin II (Ang II)-infused apolipoprotein E gene knockout (ApoE-/-) murine model of aortic dilation and performed in vivo imaging of tropoelastin by administering Gd-TESMA followed by late gadolinium enhancement (LGE) magnetic resonance imaging (MRI) and T1 mapping at 3 T, with subsequent ex vivo validation. In a cross-sectional study (n = 66; control = 11, infused = 55) we found that Gd-TESMA enhanced MRI was elevated and confined to dilated aortic segments (control: LGE=0.13 ± 0.04 mm2, control R1= 1.1 ± 0.05 s-1 vs. dilated LGE=1.0 ± 0.4 mm2, dilated R1 =2.4 ± 0.9 s-1) and was greater in segments with medium (8.0 ± 3.8 mm3) and large (10.4 ± 4.1 mm3) compared to small (3.6 ± 2.1 mm3) vessel volume. Furthermore, a proof-of-principle longitudinal study (n = 19) using Gd-TESMA enhanced MRI demonstrated a greater proportion of tropoelastin: elastin expression in dilating compared to non-dilating aortas, which correlated with the rate of aortic expansion. Treatment with pravastatin and aspirin (n = 10) did not reduce tropoelastin turnover (0.87 ± 0.3 mm2 vs. 1.0 ± 0.44 mm2) or aortic dilation (4.86 ± 2.44 mm3 vs. 4.0 ± 3.6 mm3). Importantly, Gd-TESMA-enhanced MRI identified accumulation of tropoelastin in excised human aneurysmal tissue (n = 4), which was confirmed histologically. Conclusion Tropoelastin MRI identifies dysfunctional matrix remodelling that is specifically expressed in regions of aortic aneurysm or dissection and correlates with the development and rate of aortic expansion. Thus, it may provide an additive imaging marker to the serial assessment of luminal diameter for surveillance of patients at risk of or with established aortopathy.
Collapse
Affiliation(s)
- Begoña Lavin
- School of Biomedical Engineering and Imaging Sciences, Department of Biomedical Engineering, King's College London, 3rd Floor, Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK.,Cardiovascular Division, BHF Centre of Excellence, King's College London, London, UK
| | - Sara Lacerda
- School of Biomedical Engineering and Imaging Sciences, Department of Biomedical Engineering, King's College London, 3rd Floor, Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK.,Cardiovascular Division, BHF Centre of Excellence, King's College London, London, UK.,Centre de Biophysique Moléculaire, CNRS, Orléans, France
| | - Marcelo E Andia
- School of Biomedical Engineering and Imaging Sciences, Department of Biomedical Engineering, King's College London, 3rd Floor, Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK.,Radiology Department, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Silvia Lorrio
- School of Biomedical Engineering and Imaging Sciences, Department of Biomedical Engineering, King's College London, 3rd Floor, Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK.,Cardiovascular Division, BHF Centre of Excellence, King's College London, London, UK
| | - Robert Bakewell
- School of Biomedical Engineering and Imaging Sciences, Department of Biomedical Engineering, King's College London, 3rd Floor, Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK
| | - Alberto Smith
- Cardiovascular Division, Academic Department of Vascular Surgery, King's College London, London, UK
| | - Imran Rashid
- School of Biomedical Engineering and Imaging Sciences, Department of Biomedical Engineering, King's College London, 3rd Floor, Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK
| | - René M Botnar
- School of Biomedical Engineering and Imaging Sciences, Department of Biomedical Engineering, King's College London, 3rd Floor, Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK.,Cardiovascular Division, BHF Centre of Excellence, King's College London, London, UK.,Wellcome Trust and EPSRC Medical Engineering Center, King's College London, London, UK.,Pontificia Universidad Católica de Chile, Escuela de Ingeniería, Santiago, Chile
| | - Alkystis Phinikaridou
- School of Biomedical Engineering and Imaging Sciences, Department of Biomedical Engineering, King's College London, 3rd Floor, Lambeth Wing, St Thomas' Hospital, London SE1 7EH, UK.,Cardiovascular Division, BHF Centre of Excellence, King's College London, London, UK
| |
Collapse
|
16
|
Risk Factors and Mouse Models of Abdominal Aortic Aneurysm Rupture. Int J Mol Sci 2020; 21:ijms21197250. [PMID: 33008131 PMCID: PMC7583758 DOI: 10.3390/ijms21197250] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/19/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) rupture is an important cause of death in older adults. In clinical practice, the most established predictor of AAA rupture is maximum AAA diameter. Aortic diameter is commonly used to assess AAA severity in mouse models studies. AAA rupture occurs when the stress (force per unit area) on the aneurysm wall exceeds wall strength. Previous research suggests that aortic wall structure and strength, biomechanical forces on the aorta and cellular and proteolytic composition of the AAA wall influence the risk of AAA rupture. Mouse models offer an opportunity to study the association of these factors with AAA rupture in a way not currently possible in patients. Such studies could provide data to support the use of novel surrogate markers of AAA rupture in patients. In this review, the currently available mouse models of AAA and their relevance to the study of AAA rupture are discussed. The review highlights the limitations of mouse models and suggests novel approaches that could be incorporated in future experimental AAA studies to generate clinically relevant results.
Collapse
|
17
|
Aslanidou L, Trachet B, Sasset L, Lovric G, Stergiopulos N, Di Lorenzo A. Early Morphofunctional Changes in AngII-Infused Mice Contribute to Regional Onset of Aortic Aneurysm and Dissection. J Vasc Res 2020; 57:367-375. [PMID: 32937637 DOI: 10.1159/000509545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/15/2020] [Indexed: 12/20/2022] Open
Abstract
Aortic aneurysms and dissections are silent and lethal conditions, whose pathogenesis remains incompletely understood. Although angiotensin II (AngII)-infused ApoE-/- mice have been widely used to study aortic aneurysm and dissection, early morphofunctional alterations preceding the onset of these conditions remain unknown. The goal of this study was to unveil early morphofunctional changes underlying the onset of aneurysm and dissection. At 3 days post-AngII infusion, suprarenal abdominal aorta presented significant volumetric dilatation and microstructural damage. Ex vivo assessment of vascular reactivity of the suprarenal dissection-prone aorta and its side branches, showed an endothelial and contractile dysfunctions that were severe in the suprarenal aorta, moderate distally, and absent in the side branches, mirroring the susceptibility to dissection of these different vascular segments. Early and specific morphofunctional changes of the suprarenal aorta may contribute to the regional onset of aortic aneurysm and dissection by exacerbating the biomechanical burden arising from its side branches.
Collapse
Affiliation(s)
- Lydia Aslanidou
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Bram Trachet
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,BioMMeda, Ghent University, Ghent, Belgium
| | - Linda Sasset
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Goran Lovric
- Centre d'Imagerie BioMédicale, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
| | - Nikolaos Stergiopulos
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA,
| |
Collapse
|
18
|
Golledge J, Krishna SM, Wang Y. Mouse models for abdominal aortic aneurysm. Br J Pharmacol 2020; 179:792-810. [PMID: 32914434 DOI: 10.1111/bph.15260] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/25/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) rupture is estimated to cause 200,000 deaths each year. Currently, the only treatment for AAA is surgical repair; however, this is only indicated for large asymptomatic, symptomatic or ruptured aneurysms, is not always durable, and is associated with a risk of serious perioperative complications. As a result, patients with small asymptomatic aneurysms or who are otherwise unfit for surgery are treated conservatively, but up to 70% of small aneurysms continue to grow, increasing the risk of rupture. There is thus an urgent need to develop drug therapies effective at slowing AAA growth. This review describes the commonly used mouse models for AAA. Recent research in these models highlights key roles for pathways involved in inflammation and cell turnover in AAA pathogenesis. There is also evidence for long non-coding RNAs and thrombosis in aneurysm pathology. Further well-designed research in clinically relevant models is expected to be translated into effective AAA drugs.
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, The Townsville University Hospital, Townsville, Queensland, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - Smriti Murali Krishna
- 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, The Townsville University Hospital, Townsville, Queensland, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - Yutang Wang
- Discipline of Life Sciences, School of Health and Life Sciences, Federation University Australia, Ballarat, Victoria, Australia
| |
Collapse
|
19
|
X-ray Micro-Computed Tomography: An Emerging Technology to Analyze Vascular Calcification in Animal Models. Int J Mol Sci 2020; 21:ijms21124538. [PMID: 32630604 PMCID: PMC7352990 DOI: 10.3390/ijms21124538] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 12/15/2022] Open
Abstract
Vascular calcification describes the formation of mineralized tissue within the blood vessel wall, and it is highly associated with increased cardiovascular morbidity and mortality in patients with chronic kidney disease, diabetes, and atherosclerosis. In this article, we briefly review different rodent models used to study vascular calcification in vivo, and critically assess the strengths and weaknesses of the current techniques used to analyze and quantify calcification in these models, namely 2-D histology and the o-cresolphthalein assay. In light of this, we examine X-ray micro-computed tomography (µCT) as an emerging complementary tool for the analysis of vascular calcification in animal models. We demonstrate that this non-destructive technique allows us to simultaneously quantify and localize calcification in an intact vessel in 3-D, and we consider recent advances in µCT sample preparation techniques. This review also discusses the potential to combine 3-D µCT analyses with subsequent 2-D histological, immunohistochemical, and proteomic approaches in correlative microscopy workflows to obtain rich, multifaceted information on calcification volume, calcification load, and signaling mechanisms from within the same arterial segment. In conclusion we briefly discuss the potential use of µCT to visualize and measure vascular calcification in vivo in real-time.
Collapse
|
20
|
Bersi MR, Acosta Santamaría VA, Marback K, Di Achille P, Phillips EH, Goergen CJ, Humphrey JD, Avril S. Multimodality Imaging-Based Characterization of Regional Material Properties in a Murine Model of Aortic Dissection. Sci Rep 2020; 10:9244. [PMID: 32514185 PMCID: PMC7280301 DOI: 10.1038/s41598-020-65624-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/04/2020] [Indexed: 01/21/2023] Open
Abstract
Chronic infusion of angiotensin-II in atheroprone (ApoE-/-) mice provides a reproducible model of dissection in the suprarenal abdominal aorta, often with a false lumen and intramural thrombus that thickens the wall. Such lesions exhibit complex morphologies, with different regions characterized by localized changes in wall composition, microstructure, and properties. We sought to quantify the multiaxial mechanical properties of murine dissecting aneurysm samples by combining in vitro extension-distension data with full-field multimodality measurements of wall strain and thickness to inform an inverse material characterization using the virtual fields method. A key advance is the use of a digital volume correlation approach that allows for characterization of properties not only along and around the lesion, but also across its wall. Specifically, deformations are measured at the adventitial surface by tracking motions of a speckle pattern using a custom panoramic digital image correlation technique while deformations throughout the wall and thrombus are inferred from optical coherence tomography. These measurements are registered and combined in 3D to reconstruct the reference geometry and compute the 3D finite strain fields in response to pressurization. Results reveal dramatic regional variations in material stiffness and strain energy, which reflect local changes in constituent area fractions obtained from histology but emphasize the complexity of lesion morphology and damage within the dissected wall. This is the first point-wise biomechanical characterization of such complex, heterogeneous arterial segments. Because matrix remodeling is critical to the formation and growth of these lesions, we submit that quantification of regional material properties will increase the understanding of pathological mechanical mechanisms underlying aortic dissection.
Collapse
Affiliation(s)
- Matthew R Bersi
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | | | - Karl Marback
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Paolo Di Achille
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Evan H Phillips
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - 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
| | - Stéphane Avril
- Mines Saint-Etienne, University of Lyon, University Jean Monnet, INSERM, Saint-Etienne, France.
| |
Collapse
|
21
|
Zhang H, Liao M, Cao M, Qiu Z, Yan X, Zhou Y, Wu H, Wang Y, Zheng J, Ding J, Wang M, Liao Y, Chen X. ATRQβ-001 Vaccine Prevents Experimental Abdominal Aortic Aneurysms. J Am Heart Assoc 2019; 8:e012341. [PMID: 31512549 PMCID: PMC6817999 DOI: 10.1161/jaha.119.012341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background We have developed a peptide vaccine named ATRQβ‐001, which was proved to retard signal transduction initiated by angiotensin II (Ang II). Ang II was implicated in abdominal aortic aneurysm (AAA) progression, but whether the ATRQβ‐001 vaccine would prevent AAA is unknown. Methods and Results Ang II‐infused ApoE−/− mice and calcium phosphate‐induced AAA in C57BL/6 mice were used to verify the efficiency of ATRQβ‐001 vaccine in AAA. Results demonstrated that the vaccine effectively restrained the aneurysmal dilation and vascular wall destruction of aorta in both animal models, beyond anti‐hypertensive effects. In Ang II‐induced AAA vascular sections, Immunohistochemical staining showed that the vaccine notably constrained vascular inflammation and vascular smooth muscle cell (VSMC) phenotypic transition, concurrently reduced macrophages infiltration. In cultured VSMC, the anti‐ATR‐001 antibody inhibited osteopontin secretion induced by Ang II, thereby impeded macrophage migration while co‐culture. Furthermore, metalloproteinases and other matrix proteolytic enzymes were also found to be limited by the vaccine in vivo and in vitro. Conclusions ATRQβ‐001 vaccine prevented AAA initiation and progression in both Ang II and calcium phosphate‐induced AAA models. And the beneficial effects were played beyond decrease of blood pressure, which provided a novel and promising method to take precautions against AAA.
Collapse
Affiliation(s)
- Hongrong Zhang
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Mengyang Liao
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Mingsi Cao
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Zhihua Qiu
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xiaole Yan
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yanzhao Zhou
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Hailang Wu
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yingxuan Wang
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jiayu Zheng
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jiaxing Ding
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Min Wang
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yuhua Liao
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xiao Chen
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
22
|
Abstract
BACKGROUND The authors recently found that a CD31 agonist peptide reaches macrophages in injured aortas and exerts beneficial effects on apolipoprotein E-knockout (Apo E-/-) mice subjected to angiotensin (Ang) II infusion, a model of experimental acute aortic dissection and intramural hematoma (ADIM). OBJECTIVES The purpose of this study was to evaluate the therapeutic potential of a drug-suitable agonist peptide in experimental ADIM. METHODS P8RI, a retro-inverso sequence of the best candidate identified by functional in vitro screening of a peptide library, passed an absorption, distribution, metabolism, excretion and toxicology analysis. Apo E-/- mice (male, 28-week-old) implanted with Ang II-releasing pumps received P8RI (2.5 mg/kg/day) or vehicle from day 14 (n = 10/group). Leukocytes were analyzed by flow cytometry. Healing features of human and mouse dissected aortic segments were assessed by histology and immunofluorescence. The effect of CD31 on macrophages was evaluated using cells from CD31-/- mice and P8RI, in vitro. RESULTS Human and experimental ADIM were characterized by the infiltration of proinflammatory macrophages. The absence of CD31 enhanced the proinflammatory polarization of macrophages, whereas the CD31 agonist P8RI favored reparative macrophages both in vitro and in vivo. The administration of P8RI after the occurrence of ADIM prevented aneurysmal transformation by promoting the resolution of intramural hematoma and the production of collagen in dissected aortas in vivo, associated with enrichment of M2 macrophages at the site of injury. CONCLUSIONS CD31 signaling promotes the switching of proinflammatory macrophages to the reparative phenotype and favors the healing of experimental dissected aortas. Treatment with a drug-suitable CD31 agonist may facilitate the clinical management of ADIM.
Collapse
|
23
|
Adam M, Kooreman NG, Jagger A, Wagenhäuser MU, Mehrkens D, Wang Y, Kayama Y, Toyama K, Raaz U, Schellinger IN, Maegdefessel L, Spin JM, Hamming JF, Quax PHA, Baldus S, Wu JC, Tsao PS. Systemic Upregulation of IL-10 (Interleukin-10) Using a Nonimmunogenic Vector Reduces Growth and Rate of Dissecting Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2019; 38:1796-1805. [PMID: 29880489 DOI: 10.1161/atvbaha.117.310672] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective- Recruitment of immunologic competent cells to the vessel wall is a crucial step in formation of abdominal aortic aneurysms (AAA). Innate immunity effectors (eg, macrophages), as well as mediators of adaptive immunity (eg, T cells), orchestrate a local vascular inflammatory response. IL-10 (interleukin-10) is an immune-regulatory cytokine with a crucial role in suppression of inflammatory processes. We hypothesized that an increase in systemic IL-10-levels would mitigate AAA progression. Approach and Results- Using a single intravenous injection protocol, we transfected an IL-10 transcribing nonimmunogenic minicircle vector into the Ang II (angiotensin II)-ApoE-/- infusion mouse model of AAA. IL-10 minicircle transfection significantly reduced average aortic diameter measured via ultrasound at day 28 from 166.1±10.8% (control) to 131.0±5.8% (IL-10 transfected). Rates of dissecting AAA were reduced by IL-10 treatment, with an increase in freedom from dissecting AAA from 21.5% to 62.3%. Using flow cytometry of aortic tissue from minicircle IL-10-treated animals, we found a significantly higher percentage of CD4+/CD25+/Foxp3 (forkhead box P3)+ regulatory T cells, with fewer CD8+/GZMB+ (granzyme B) cytotoxic T cells. Furthermore, isolated aortic macrophages produced less TNF-α (tumor necrosis factor-α), more IL-10, and were more likely to be MRC1 (mannose receptor, C type 1)-positive alternatively activated macrophages. These results concurred with gene expression analysis of lipopolysaccharide-stimulated and Ang II-primed human peripheral blood mononuclear cells. Conclusions- Taken together, we provide an effective gene therapy approach to AAA in mice by enhancing antiinflammatory and dampening proinflammatory pathways through minicircle-induced augmentation of systemic IL-10 expression.
Collapse
Affiliation(s)
- Matti Adam
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.).,Department of Cardiovascular Medicine, Cologne Cardiovascular Research Center, University of Cologne, University Heart Center, Germany (M.A., D.M., S.B.).,VA Palo Alto Health Care System, CA (M.A., A.J., M.U.W., Y.K., K.T., U.R., I.N.S., J.M.S., P.S.T.)
| | - Nigel Geoffrey Kooreman
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.).,Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (N.G.K., J.F.H., P.H.A.Q.)
| | - Ann Jagger
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.).,VA Palo Alto Health Care System, CA (M.A., A.J., M.U.W., Y.K., K.T., U.R., I.N.S., J.M.S., P.S.T.)
| | - Markus U Wagenhäuser
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.).,VA Palo Alto Health Care System, CA (M.A., A.J., M.U.W., Y.K., K.T., U.R., I.N.S., J.M.S., P.S.T.)
| | - Dennis Mehrkens
- Department of Cardiovascular Medicine, Cologne Cardiovascular Research Center, University of Cologne, University Heart Center, Germany (M.A., D.M., S.B.)
| | - Yongming Wang
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.)
| | - Yosuke Kayama
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.).,VA Palo Alto Health Care System, CA (M.A., A.J., M.U.W., Y.K., K.T., U.R., I.N.S., J.M.S., P.S.T.)
| | - Kensuke Toyama
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.).,VA Palo Alto Health Care System, CA (M.A., A.J., M.U.W., Y.K., K.T., U.R., I.N.S., J.M.S., P.S.T.)
| | - Uwe Raaz
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.).,VA Palo Alto Health Care System, CA (M.A., A.J., M.U.W., Y.K., K.T., U.R., I.N.S., J.M.S., P.S.T.).,Heart Center, Georg-August-University Göttingen, Germany (U.R., I.N.S.)
| | - Isabel N Schellinger
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.).,VA Palo Alto Health Care System, CA (M.A., A.J., M.U.W., Y.K., K.T., U.R., I.N.S., J.M.S., P.S.T.).,Heart Center, Georg-August-University Göttingen, Germany (U.R., I.N.S.)
| | - Lars Maegdefessel
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.).,Department of Medicine, Karolinska Institutet, Stockholm, Sweden (L.M.)
| | - Joshua M Spin
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.).,VA Palo Alto Health Care System, CA (M.A., A.J., M.U.W., Y.K., K.T., U.R., I.N.S., J.M.S., P.S.T.)
| | - Jaap F Hamming
- Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (N.G.K., J.F.H., P.H.A.Q.)
| | - Paul H A Quax
- Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (N.G.K., J.F.H., P.H.A.Q.)
| | - Stephan Baldus
- Department of Cardiovascular Medicine, Cologne Cardiovascular Research Center, University of Cologne, University Heart Center, Germany (M.A., D.M., S.B.)
| | - Joseph C Wu
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.)
| | - Philip S Tsao
- From the Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, CA (M.A., N.G.K., A.J., M.U.W., Y.W., YK., K.T., U.R., I.N.S., L.M., J.M.S., J.C.W., P.S.T.).,VA Palo Alto Health Care System, CA (M.A., A.J., M.U.W., Y.K., K.T., U.R., I.N.S., J.M.S., P.S.T.)
| |
Collapse
|
24
|
Michel JB, Jondeau G, Milewicz DM. From genetics to response to injury: vascular smooth muscle cells in aneurysms and dissections of the ascending aorta. Cardiovasc Res 2019; 114:578-589. [PMID: 29360940 DOI: 10.1093/cvr/cvy006] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 01/16/2018] [Indexed: 12/20/2022] Open
Abstract
Vascular smooth muscle cells (vSMCs) play a crucial role in both the pathogenesis of Aneurysms and Dissections of the ascending thoracic aorta (TAAD) in humans and in the associated adaptive compensatory responses, since thrombosis and inflammatory processes are absent in the majority of cases. Aneurysms and dissections share numerous characteristics, including aetiologies and histopathological alterations: vSMC disappearance, medial areas of mucoid degeneration, and extracellular matrix (ECM) breakdown. Three aetiologies predominate in TAAD in humans: (i) genetic causes in heritable familial forms, (ii) an association with bicuspid aortic valves, and (iii) a sporadic degenerative form linked to the aortic aging process. Genetic forms include mutations in vSMC genes encoding for molecules of the ECM or the TGF-β pathways, or participating in vSMC tone. On the other hand, aneurysms and dissections, whatever their aetiologies, are characterized by an increase in wall permeability leading to transmural advection of plasma proteins which could interact with vSMCs and ECM components. In this context, blood-borne plasminogen appears to play an important role, because its outward convection through the wall is increased in TAAD, and it could be converted to active plasmin at the vSMC membrane. Active plasmin can induce vSMC disappearance, proteolysis of adhesive proteins, activation of MMPs and release of TGF-β from its ECM storage sites. Conversely, vSMCs could respond to aneurysmal biomechanical and proteolytic injury by an epigenetic phenotypic switch, including constitutional overexpression and nuclear translocation of Smad2 and an increase in antiprotease and ECM protein synthesis. In contrast, such an epigenetic phenomenon is not observed in dissections. In this context, dysfunction of proteins involved in vSMC tone are interesting to study, particularly in interaction with plasma protein transport through the wall and TGF-β activation, to establish the relationship between these dysfunctions and ECM proteolysis.
Collapse
Affiliation(s)
- Jean-Baptiste Michel
- UMR 1148, Laboratory for Translational Vascular Science, Inserm and Paris 7- Denis Diderot University, Xavier Bichat Hospital, 75018 Paris, France
| | - Guillaume Jondeau
- UMR 1148, Laboratory for Translational Vascular Science, Inserm and Paris 7- Denis Diderot University, Xavier Bichat Hospital, 75018 Paris, France.,Cardiology Department, National Reference Center for Marfan Syndrome and Related Diseases, APHP Hopital Bichat, 75018 Paris
| | - Dianna M Milewicz
- Division of Medical Genetics, Department of Internal Medicine, University of Texas Medical School at Houston, Houston, TX 77030, USA
| |
Collapse
|
25
|
Aslanidou L, Ferraro M, Lovric G, Bersi MR, Humphrey JD, Segers P, Trachet B, Stergiopulos N. Co-localization of microstructural damage and excessive mechanical strain at aortic branches in angiotensin-II-infused mice. Biomech Model Mechanobiol 2019; 19:81-97. [PMID: 31273562 DOI: 10.1007/s10237-019-01197-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 06/26/2019] [Indexed: 02/07/2023]
Abstract
Animal models of aortic aneurysm and dissection can enhance our limited understanding of the etiology of these lethal conditions particularly because early-stage longitudinal data are scant in humans. Yet, the pathogenesis of often-studied mouse models and the potential contribution of aortic biomechanics therein remain elusive. In this work, we combined micro-CT and synchrotron-based imaging with computational biomechanics to estimate in vivo aortic strains in the abdominal aorta of angiotensin-II-infused ApoE-deficient mice, which were compared with mouse-specific aortic microstructural damage inferred from histopathology. Targeted histology showed that the 3D distribution of micro-CT contrast agent that had been injected in vivo co-localized with precursor vascular damage in the aortic wall at 3 days of hypertension, with damage predominantly near the ostia of the celiac and superior mesenteric arteries. Computations similarly revealed higher mechanical strain in branching relative to non-branching regions, thus resulting in a positive correlation between high strain and vascular damage in branching segments that included the celiac, superior mesenteric, and right renal arteries. These results suggest a mechanically driven initiation of damage at these locations, which was supported by 3D synchrotron imaging of load-induced ex vivo delaminations of angiotensin-II-infused suprarenal abdominal aortas. That is, the major intramural delamination plane in the ex vivo tested aortas was also near side branches and specifically around the celiac artery. Our findings thus support the hypothesis of an early mechanically mediated formation of microstructural defects at aortic branching sites that subsequently propagate into a macroscopic medial tear, giving rise to aortic dissection in angiotensin-II-infused mice.
Collapse
Affiliation(s)
- Lydia Aslanidou
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Mauro Ferraro
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Goran Lovric
- Centre d'Imagerie BioMédicale, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
| | - Matthew R Bersi
- Department of Biomedical Engineering, Yale University, New Haven, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, USA
| | | | - Bram Trachet
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- bioMMeda, Ghent University, Ghent, Belgium
| | - Nikos Stergiopulos
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
26
|
Trachet B, Ferraro M, Lovric G, Aslanidou L, Logghe G, Segers P, Stergiopulos N. Synchrotron-based visualization and segmentation of elastic lamellae in the mouse carotid artery during quasi-static pressure inflation. J R Soc Interface 2019; 16:20190179. [PMID: 31238834 DOI: 10.1098/rsif.2019.0179] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In computational aortic biomechanics, aortic and arterial tissue are typically modelled as a homogeneous layer, making abstraction not only of the layered structure of intima, media and adventitia but also of the microstructure that exists within these layers. Here, we present a novel method to visualize the microstructure of the tunica media along the entire circumference of the vessel. To that end, we developed a pressure-inflation device that is compatible with synchrotron-based phase-contrast imaging. Using freshly excised left common carotid arteries from n = 12 mice, we visualized how the lamellae and interlamellar layers inflate as the luminal pressure is increased from 0 to 120 mm Hg in quasi-static steps. A graph-based segmentation algorithm subsequently allowed us to automatically segment each of the three lamellae, resulting in a three-dimensional geometry that represents lamellae, interlamellar layers and adventitia at nine different pressure levels. Our results demonstrate that the three elastic lamellae unfold and stretch simultaneously as luminal pressure is increased. In the long term, we believe that the results presented in this work can be a first step towards a better understanding of the mechanics of the arterial microstructure.
Collapse
Affiliation(s)
- Bram Trachet
- 1 Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne , Lausanne , Switzerland.,2 IBiTech-bioMMeda , Ghent University, Ghent , Belgium
| | - Mauro Ferraro
- 1 Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| | - Goran Lovric
- 3 Centre d'Imagerie BioMédicale, Ecole Polytechnique Fédérale de Lausanne , Lausanne , Switzerland.,4 Swiss Light Source, Paul Scherrer Institute , Villigen , Switzerland
| | - Lydia Aslanidou
- 1 Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| | | | | | - Nikolaos Stergiopulos
- 1 Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| |
Collapse
|
27
|
Adelsperger AR, Phillips EH, Ibriga HS, Craig BA, Green LA, Murphy MP, Goergen CJ. Development and growth trends in angiotensin II-induced murine dissecting abdominal aortic aneurysms. Physiol Rep 2019; 6:e13668. [PMID: 29696811 PMCID: PMC5917066 DOI: 10.14814/phy2.13668] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/05/2018] [Accepted: 03/07/2018] [Indexed: 11/24/2022] Open
Abstract
Abdominal aortic aneurysms are pathological dilations that can suddenly rupture, causing more than 15,000 deaths in the U.S. annually. Current treatment focuses on observation until an aneurysm's size warrants surgical intervention. Thus, there is a need for therapeutic intervention to inhibit growth of smaller aneurysms. An experimental aneurysm model that infuses angiotensin II into apolipoprotein E‐deficient mice is widely used to investigate underlying pathological mechanisms and potential therapeutics, but this model has two caveats: (1) aneurysms do not always form, and (2) aneurysm severity and growth is inconsistent among animals. Here we use high‐frequency ultrasound to collect data from angiotensin II‐induced aneurysms to develop prediction models of both aneurysm formation and growth. Baseline measurements of aortic diameter, volume/length, and strain were used with animal mass and age in a quadratic discriminant analysis and logistic regression to build two statistical models to predict disease status. Longitudinal ultrasound data were also acquired from mice with aneurysms to quantify aneurysm diameter, circumferential strain, blood flow velocity, aneurysm volume/length, and thrombus and open‐false lumen volumes over 28 days. Measurements taken at aneurysm diagnosis were used with branching artery information to produce a multiple linear regression model to predict final aneurysm volume/length. All three statistical models could be useful in future aneurysm therapeutic studies to better delineate the effects of preventative and suppressive treatments from normal variations in the angiotensin II aneurysm model.
Collapse
Affiliation(s)
- Amelia R Adelsperger
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Evan H Phillips
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Hilda S Ibriga
- Department of Statistics, Purdue University, West Lafayette, Indiana
| | - Bruce A Craig
- Department of Statistics, Purdue University, West Lafayette, Indiana
| | - Linden A Green
- IU Health Center for Aortic Disease/Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| |
Collapse
|
28
|
Sun D, Zhang M, Li Y, Mei S, Qin J, Yan J. c‑Jun/Ap‑1 is upregulated in an Ang II‑induced abdominal aortic aneurysm formation model and mediates Chop expression in mouse aortic smooth muscle cells. Mol Med Rep 2019; 19:3459-3468. [PMID: 30864718 PMCID: PMC6472129 DOI: 10.3892/mmr.2019.10017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 02/08/2019] [Indexed: 01/30/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is an asymptomatic, potentially lethal disease whose ruptures have a high mortality rate. An effective pharmacological approach to decrease expansion or prevent the rupture of AAAs in humans remains lacking. Previous studies have suggested that activator protein 1 (c-Jun/AP-1) and C/EBP homologous protein (Chop) are involved in the development of AAA. The purpose of the present study was to investigate whether c-Jun/AP-1 mediates Chop overexpression in AAA. c-Jun/AP-1 and Chop protein levels were determined in an angiotensin II (Ang II)-induced AAA model using apolipoprotein E-deficient mice. Additionally, mouse aortic smooth muscle cells (MOVAS cell line) were treated with Ang II. Apoptosis was evaluated via TUNEL assay, MOVAS cell migration ability was assessed by monolayer wound healing assay and the levels of c-Jun/AP-1 and Chop were determined by western blotting, immunofluorescence and immunocytochemical assays. Following c-Jun silencing using c-Jun-specific small interfering (si)RNA, Chop expression was evaluated. Furthermore, chromatin immunoprecipitation (ChIP) was used to investigate whether c-Jun/Ap-1 binds directly to the DNA damage-inducible transcript 3 protein (Ddit3) promoter. It was observed that c-Jun/AP-1 and Chop were synchronously overexpressed in Ang II-induced AAA and Ang II-treated cells, and that apoptosis and migration were induced by Ang II. In addition, Chop was suppressed when c-Jun was silenced by targeted siRNA. Notably, the ChIP assay demonstrated that the DNA fragments pulled down by primary antibodies against c-Jun/Ap-1 were able to be amplified by (Ddit3) promoter-specific primers. c-Jun/AP-1 may therefore mediate Chop expression in MOVAS cells via Ddit3. These results suggested that c-Jun/AP-1 may be a novel target for AAA therapy.
Collapse
Affiliation(s)
- Dating Sun
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Mingxi Zhang
- Division of Cardiology, Department of Internal Medicine, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430030, P.R. China
| | - Yuanyuan Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shuai Mei
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jin Qin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jiangtao Yan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
29
|
In Vivo Detection and Measurement of Aortic Aneurysm and Dissection in Mouse Models Using Microcomputed Tomography with Contrast Agent. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:5940301. [PMID: 30956627 PMCID: PMC6431409 DOI: 10.1155/2019/5940301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/11/2019] [Indexed: 02/03/2023]
Abstract
Objectives The aim of this study was to evaluate the potential of microcomputed tomography (micro-CT) using the intravascular contrast agent ExiTron nano 12000 for aorta imaging and monitoring the dynamic changing process of the aorta in mouse models with aortic aneurysm and dissection. Materials and Methods Experiments were performed on healthy mice and mice with aortic dissection. Mice that were developing aortic dissection and healthy mice underwent micro-CT imaging after injection of ExiTron nano 12000. Time-dependent signal enhancement (at 1, 2, 3, 6, and 12 hours after intravenous injection of the contrast agent, respectively) in the aorta of healthy mice was measured to confirm the optimal imaging time of aorta. Various contrast agent doses (70, 100, and 150 μl per 25 g mouse, respectively) were investigated to determine the optimal required dose for imaging of the aorta. The mice were scanned with micro-CT at 1, 14, and 28 days after onset of aneurysm and dissection to monitor the dynamic changing process of the aorta. Mouse aortas were stained with hematoxylin and eosin staining, and the diameter of the aorta was measured and compared with those obtained by micro-CT. Results Time-dependent signal enhancement in the aorta shows that the contrast agent has a long blood half-life of 6 hours, with a peak enhancement at 2 hours after injection. Injection of 100 μl ExiTron nano 12000 per 25 g mouse allows for effective visualization of the aorta. Micro-CT combined with contrast agent can monitor the changing process of the aorta in the mouse model of aortic aneurysm and dissection dynamically. The values of the diameter of the aortas obtained from the in vivo micro-CT imaging were compared with those obtained from histology and showed a significant correlation (R2 = 0.96). Conclusions These data demonstrate that in vivo micro-CT is an accurate and feasible technique to detect aortic aneurysm or dissection in a mouse model, and the micro-CT technique using the innovative contrast agent ExiTron nano 12000 allows for monitoring various processes dynamically such as aortic remodeling in longitudinal studies.
Collapse
|
30
|
López-Guimet J, Peña-Pérez L, Bradley RS, García-Canadilla P, Disney C, Geng H, Bodey AJ, Withers PJ, Bijnens B, Sherratt MJ, Egea G. MicroCT imaging reveals differential 3D micro-scale remodelling of the murine aorta in ageing and Marfan syndrome. Am J Cancer Res 2018; 8:6038-6052. [PMID: 30613281 PMCID: PMC6299435 DOI: 10.7150/thno.26598] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/12/2018] [Indexed: 12/13/2022] Open
Abstract
Aortic wall remodelling is a key feature of both ageing and genetic connective tissue diseases, which are associated with vasculopathies such as Marfan syndrome (MFS). Although the aorta is a 3D structure, little attention has been paid to volumetric assessment, primarily due to the limitations of conventional imaging techniques. Phase-contrast microCT is an emerging imaging technique, which is able to resolve the 3D micro-scale structure of large samples without the need for staining or sectioning. Methods: Here, we have used synchrotron-based phase-contrast microCT to image aortae of wild type (WT) and MFS Fbn1C1039G/+ mice aged 3, 6 and 9 months old (n=5). We have also developed a new computational approach to automatically measure key histological parameters. Results: This analysis revealed that WT mice undergo age-dependent aortic remodelling characterised by increases in ascending aorta diameter, tunica media thickness and cross-sectional area. The MFS aortic wall was subject to comparable remodelling, but the magnitudes of the changes were significantly exacerbated, particularly in 9 month-old MFS mice with ascending aorta wall dilations. Moreover, this morphological remodelling in MFS aorta included internal elastic lamina surface breaks that extended throughout the MFS ascending aorta and were already evident in animals who had not yet developed aneurysms. Conclusions: Our 3D microCT study of the sub-micron wall structure of whole, intact aorta reveals that histological remodelling of the tunica media in MFS could be viewed as an accelerated ageing process, and that phase-contrast microCT combined with computational image analysis allows the visualisation and quantification of 3D morphological remodelling in large volumes of unstained vascular tissues.
Collapse
|
31
|
Portier I, Martinod K, Desender L, Vandeputte N, Deckmyn H, Vanhoorelbeke K, De Meyer SF. von Willebrand factor deficiency does not influence angiotensin II-induced abdominal aortic aneurysm formation in mice. Sci Rep 2018; 8:16645. [PMID: 30413751 PMCID: PMC6226453 DOI: 10.1038/s41598-018-35029-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) refers to a localized dilation of the abdominal aorta that exceeds the normal diameter by 50%. AAA pathophysiology is characterized by progressive inflammation, vessel wall destabilization and thrombus formation. Our aim was to investigate the potential involvement of von Willebrand factor (VWF), a thrombo-inflammatory plasma protein, in AAA pathophysiology using a dissection-based and angiotensin II infusion-induced AAA mouse model. AAA formation was induced in both wild-type and VWF-deficient mice by subcutaneous implantation of an osmotic pump, continuously releasing 1000 ng/kg/min angiotensin II. Survival was monitored, but no significant difference was observed between both groups. After 28 days, the suprarenal aortic segment of the surviving mice was harvested. Both AAA incidence and severity were similar in wild-type and VWF-deficient mice, indicating that AAA formation was not significantly influenced by the absence of VWF. Although VWF plasma levels increased after the infusion period, these increases were not correlated with AAA progression. Also detailed histological analyses of important AAA hallmarks, including elastic degradation, intramural thrombus formation and leukocyte infiltration, did not reveal differences between both groups. These data suggest that, at least in the angiotensin II infusion-induced AAA mouse model, the role of VWF in AAA pathophysiology is limited.
Collapse
Affiliation(s)
- Irina Portier
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Kimberly Martinod
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Linda Desender
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Nele Vandeputte
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Hans Deckmyn
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium.
| |
Collapse
|
32
|
Trachet B, Lovric G, Villanueva-Perez P, Aslanidou L, Ferraro M, Logghe G, Stergiopulos N, Segers P. Synchrotron-based phase contrast imaging of cardiovascular tissue in mice—grating interferometry or phase propagation? Biomed Phys Eng Express 2018. [DOI: 10.1088/2057-1976/aaeb65] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
33
|
Verbrugghe P, Verhoeven J, Clijsters M, Vervoort D, Coudyzer W, Verbeken E, Meuris B, Herijgers P. Creation of Abdominal Aortic Aneurysms in Sheep by Extrapolation of Rodent Models: Is It Feasible? Ann Vasc Surg 2018; 52:225-236. [DOI: 10.1016/j.avsg.2018.02.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 10/14/2022]
|
34
|
Bersi MR, Khosravi R, Wujciak AJ, Harrison DG, Humphrey JD. Differential cell-matrix mechanoadaptations and inflammation drive regional propensities to aortic fibrosis, aneurysm or dissection in hypertension. J R Soc Interface 2018; 14:rsif.2017.0327. [PMID: 29118111 DOI: 10.1098/rsif.2017.0327] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 10/17/2017] [Indexed: 01/01/2023] Open
Abstract
The embryonic lineage of intramural cells, microstructural organization of the extracellular matrix, local luminal and wall geometry, and haemodynamic loads vary along the length of the aorta. Yet, it remains unclear why certain diseases manifest differentially along the aorta. Toward this end, myriad animal models provide insight into diverse disease conditions-including fibrosis, aneurysm and dissection-but inherent differences across models impede general interpretations. We examined region-specific cellular, matrix, and biomechanical changes in a single experimental model of hypertension and atherosclerosis, which commonly coexist. Our findings suggest that (i) intramural cells within the ascending aorta are unable to maintain the intrinsic material stiffness of the wall, which ultimately drives aneurysmal dilatation, (ii) a mechanical stress-initiated, inflammation-driven remodelling within the descending aorta results in excessive fibrosis, and (iii) a transient loss of adventitial collagen within the suprarenal aorta contributes to dissection propensity. Smooth muscle contractility helps to control wall stress in the infrarenal aorta, which maintains mechanical properties near homeostatic levels despite elevated blood pressure. This early mechanoadaptation of the infrarenal aorta does not preclude subsequent acceleration of neointimal formation, however. Because region-specific conditions may be interdependent, as, for example, diffuse central arterial stiffening can increase cyclic haemodynamic loads on an aneurysm that is developing proximally, there is a clear need for more systematic assessments of aortic disease progression, not simply a singular focus on a particular region or condition.
Collapse
Affiliation(s)
- M R Bersi
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - R Khosravi
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - A J Wujciak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - D G Harrison
- Department of Medicine, Vanderbilt University, Nashville, TN, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN, 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
|
35
|
Propagation-based phase-contrast synchrotron imaging of aortic dissection in mice: from individual elastic lamella to 3D analysis. Sci Rep 2018; 8:2223. [PMID: 29396472 PMCID: PMC5797148 DOI: 10.1038/s41598-018-20673-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/23/2018] [Indexed: 11/09/2022] Open
Abstract
In order to show the advantage and potential of propagation-based phase-contrast synchrotron imaging in vascular pathology research, we analyzed aortic medial ruptures in BAPN/AngII-infused mice, a mouse model for aortic dissection. Ascending and thoraco-abdominal samples from n = 3 control animals and n = 10 BAPN/AngII-infused mice (after 3, 7 and 14 days of infusion, total of 24 samples) were scanned. A steep increase in the number of ruptures was already noted after 3 days of BAPN/AngII-infusion. The largest ruptures were found at the latest time points. 133 ruptures affected only the first lamella while 135 ruptures affected multiple layers. Medial ruptures through all lamellar layers, leading to false channel formation and intramural hematoma, occurred only in the thoraco-abdominal aorta and interlamellar hematoma formation in the ascending aorta could be directly related to ruptures of the innermost lamellae. The advantages of this technique are (i) ultra-high resolution that allows to visualize the individual elastic lamellae in the aorta; (ii) quantitative and qualitative analysis of medial ruptures; (iii) 3D analysis of the complete aorta; (iv) high contrast for qualitative information extraction, reducing the need for histology coupes; (v) earlier detection of (micro-) ruptures.
Collapse
|
36
|
Ohno-Urabe S, Aoki H, Nishihara M, Furusho A, Hirakata S, Nishida N, Ito S, Hayashi M, Yasukawa H, Imaizumi T, Akashi H, Tanaka H, Fukumoto Y. Role of Macrophage Socs3 in the Pathogenesis of Aortic Dissection. J Am Heart Assoc 2018; 7:JAHA.117.007389. [PMID: 29343476 PMCID: PMC5850160 DOI: 10.1161/jaha.117.007389] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Aortic dissection (AD) is a life-threatening medical emergency caused by the abrupt destruction of the intimomedial layer of the aortic walls. Given that previous studies have reported the involvement of proinflammatory cytokine interleukin-6 in AD pathogenesis, we investigated the role of signal transduction and activator of transcription 3 signaling, a downstream pathway of interleukin-6 in macrophages in pathogenesis of AD. METHODS AND RESULTS We characterized the pathological and molecular events triggered by aortic stress, which can lead to AD. Aortic stress on the suprarenal aorta because of infrarenal aorta stiffening and angiotensin II infusion for 1 week caused focal medial rupture at the branching point of the celiac trunk and superior mesenteric artery. This focal medial rupture healed in 6 weeks in wild-type (WT) mice, but progressed to AD in mice with macrophage-specific deletion of Socs3 gene (mSocs3-KO). mSocs3-KO mice showed premature activation of cell proliferation, an inflammatory response, and skewed differentiation of macrophages toward the tissue-destructive phenotype. Concomitantly, they showed aberrant phenotypic modulation of smooth muscle cells and transforming growth factor beta signaling, which are likely to participate in tissue repair. Human AD samples revealed signal transduction and activator of transcription 3 activation in adventitial macrophages adjacent to the site of tissue destruction. CONCLUSIONS These findings suggest that AD development is preceded by focal medial rupture, in which macrophage Socs3 maintains proper inflammatory response and differentiation of SMCs, thus promoting fibrotic healing to prevent tissue destruction and AD development. Understanding the sequence of the pathological and molecular events preceding AD development will help predict and prevent AD development and progression.
Collapse
Affiliation(s)
- Satoko Ohno-Urabe
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hiroki Aoki
- Cardiovascular Research Institute, Kurume University, Kurume, Japan
| | - Michihide Nishihara
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Aya Furusho
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Saki Hirakata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Norifumi Nishida
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Sohei Ito
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Makiko Hayashi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hideo Yasukawa
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | | | - Hidetoshi Akashi
- Division of Cardiovascular Surgery, Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Hiroyuki Tanaka
- Division of Cardiovascular Surgery, Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Yoshihiro Fukumoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
37
|
Tsai SH, Wang JC, Liao WI, Hsu YJ, Lin CY, Liao MT, Huang PH, Lin SJ. Fucoidan attenuates angiotensin II-induced abdominal aortic aneurysms through the inhibition of c-Jun N-terminal kinase and nuclear factor κB activation. J Vasc Surg 2017; 68:72S-81S.e1. [PMID: 29290496 DOI: 10.1016/j.jvs.2017.09.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/17/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Rupture of abdominal aortic aneurysm (AAA) is one of the leading causes of sudden death among the elderly. Most incidental AAAs are below the threshold for intervention at the time of detection; however, there is no evidence that commonly used cardiovascular drugs have clinical beneficial effects on AAA progression. Therefore, in addition to current cardiovascular risk-reducing treatments, an adjunctive medical therapy targeting the regulation of extracellular matrix metabolism is still required in the clinical setting. Fucoidan is an extract of brown seaweed and a sulfated polysaccharide. Emerging evidence suggests that fucoidan has potential cardiovascular applications. Numerous investigations of fucoidan in diseases of the cardiovascular system have mainly focused on its pleiotropic anti-inflammatory effects. Specifically, fucoidan has been shown to have matrix metalloproteinase (MMP)-reducing effects in several studies. We aimed to evaluate the beneficial effect of fucoidan on aneurysmal growth in a murine model of aortic aneurysm and further provide a rationale for using fucoidan as a medical adjunctive therapy. METHODS A murine model of angiotensin II (Ang II)-induced AAA was used to assess the therapeutic effects of fucoidan on AAA growth in vivo. The characteristics and quantification of AAAs were determined in situ. Human umbilical vein endothelial cells were used for studying the involved pathways in vitro. Western blotting was used to detect the involved signaling pathways both in vivo and in vitro. RESULTS Treatment with fucoidan significantly reduced the incidence of AAA formation. Administration of fucoidan significantly attenuated Ang II-induced aortic expansion from 1.56 ± 0.76 mm to 1.09 ± 0.30 mm. Administration of fucoidan significantly suppressed MMP-2 and MMP-9 activities and reduced the grade of elastin degradation in vivo. In vitro, we found that fucoidan could ameliorate the Ang II-induced phosphorylation of c-Jun N-terminal kinase and nuclear factor κB p65, and it further reduced MMP and reactive oxygen species production. CONCLUSIONS Fucoidan inhibits the progression of experimental AAA growth through the attenuation of proinflammatory nuclear factor κB and c-Jun N-terminal kinase activation. Fucoidan could be a potential medical adjunctive therapy for small AAAs.
Collapse
Affiliation(s)
- Shih-Hung Tsai
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Jen-Chun Wang
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Yuan Lin
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
38
|
A novel reproducible model of aortic aneurysm rupture. Surgery 2017; 163:397-403. [PMID: 29195736 DOI: 10.1016/j.surg.2017.10.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Given the unknown biologic antecedents before aortic aneurysm rupture, the purpose of this study was to establish a reproducible model of aortic aneurysm rupture. METHODS We fed 7-week-old apolipoprotein E deficient mice a high-fat diet for 4 weeks and osmotic infusion pumps containing Angiotensin II were implanted. Angiotensin II was delivered continuously for 4 weeks at either 1,000 ng/kg/min (n = 25) or 2,000 ng/kg/min (n = 29). A third group (n = 14) were given Angiotensin II at 2,000 ng/kg/min and 0.2% β-aminopropionitrile dissolved in drinking water. Surviving mice were killed 28 days after pump placement, aortic diameters were measured, and molecular analyses were performed. RESULTS Survival at 28 days was significantly different among groups with 80% survival in the 1,000 ng/kg/min group, 52% in the 2,000 ng/kg/min group, and only 14% in the Angiotensin II/β-aminopropionitrile group (P = .0001). Concordantly, rupture rates were statistically different among groups (8% versus 38% versus 79%, P < .0001). Rates of abdominal aortic aneurysm were 48%, 55%, and 93%, respectively, with statistically higher rates in the Angiotensin II/β-aminopropionitrile group compared with both the 1,000 ng and 2,000 ng Angiotensin II groups (P = .006 and P = .0165, respectively). Rates of thoracic aortic aneurysm formation were 12%, 52%, and 79% in the 3 groups with a statistically higher rate in the Angiotensin II/β-aminopropionitrile group compared with 1,000 ng group (P < .0001). CONCLUSIONS A reproducible model of aortic aneurysm rupture was developed with a high incidence of abdominal and thoracic aortic aneurysm. This model should enable further studies investigating the pathogenesis of aortic rupture, as well as allow for targeted strategies to prevent human aortic aneurysm rupture.
Collapse
|
39
|
Horimatsu T, Kim HW, Weintraub NL. The Role of Perivascular Adipose Tissue in Non-atherosclerotic Vascular Disease. Front Physiol 2017; 8:969. [PMID: 29234289 PMCID: PMC5712360 DOI: 10.3389/fphys.2017.00969] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/14/2017] [Indexed: 12/30/2022] Open
Abstract
Perivascular adipose tissue (PVAT) surrounds most large blood vessels and plays an important role in vascular homeostasis. PVAT releases various chemokines and adipocytokines, functioning in an endocrine and paracrine manner to regulate vascular signaling and inflammation. Mounting evidence suggests that PVAT plays an important role in atherosclerosis and hypertension; however, the role of PVAT in non-atherosclerotic vascular diseases, including neointimal formation, aortic aneurysm, arterial stiffness and vasculitis, has received far less attention. Increasing evidence suggests that PVAT responds to mechanical endovascular injury and regulates the subsequent formation of neointima via factors that promote smooth muscle cell growth, adventitial inflammation and neovascularization. Circumstantial evidence also links PVAT to the pathogenesis of aortic aneurysms and vasculitic syndromes, such as Takayasu's arteritis, where infiltration and migration of inflammatory cells from PVAT into the vascular wall may play a contributory role. Moreover, in obesity, PVAT has been implicated to promote stiffness of elastic arteries via the production of reactive oxygen species. This review will discuss the growing body of data and mechanisms linking PVAT to the pathogenesis of non-atherosclerotic vascular diseases in experimental animal models and in humans.
Collapse
Affiliation(s)
- Tetsuo Horimatsu
- Division of Cardiology, Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Ha Won Kim
- Division of Cardiology, Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Neal L Weintraub
- Division of Cardiology, Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
40
|
Ferraro M, Trachet B, Aslanidou L, Fehervary H, Segers P, Stergiopulos N. Should We Ignore What We Cannot Measure? How Non-Uniform Stretch, Non-Uniform Wall Thickness and Minor Side Branches Affect Computational Aortic Biomechanics in Mice. Ann Biomed Eng 2017; 46:159-170. [PMID: 29071528 DOI: 10.1007/s10439-017-1945-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/14/2017] [Indexed: 12/18/2022]
Abstract
In order to advance the state-of-the-art in computational aortic biomechanics, we investigated the influence of (i) a non-uniform wall thickness, (ii) minor aortic side branches and (iii) a non-uniform axial stretch distribution on the location of predicted hotspots of principal strain in a mouse model for dissecting aneurysms. After 3 days of angiotensin II infusion, a murine abdominal aorta was scanned in vivo with contrast-enhanced micro-CT. The animal was subsequently sacrificed and its aorta was scanned ex vivo with phase-contrast X-ray tomographic microscopy (PCXTM). An automatic morphing framework was developed to map the non-pressurized, non-stretched PCXTM geometry onto the pressurized, stretched micro-CT geometry. The output of the morphing model was a structural FEM simulation where the output strain distribution represents an estimation of the wall deformation, not only due to the pressurization, but also due to the local axial stretch field. The morphing model also included minor branches and a mouse-specific wall thickness. A sensitivity study was then performed to assess the influence of each of these novel features on the outcome of the simulations. The results were supported by comparing the computed hotspots of principal strain to hotspots of early vascular damage as detected on PCXTM. Non-uniform axial stretch, non-uniform wall thickness and minor subcostal arteries significantly alter the locations of calculated hotspots of maximal principal strain. Even if experimental data on these features are often not available in clinical practice, one should be aware of the important implications that simplifications in the model might have on the final simulated result.
Collapse
Affiliation(s)
- Mauro Ferraro
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, LHTC STI IBI EPFL, MED 32924 Station 9, 1015, Lausanne, Switzerland.
| | - Bram Trachet
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, LHTC STI IBI EPFL, MED 32924 Station 9, 1015, Lausanne, Switzerland
- IBiTech - bioMMeda, Ghent University, Ghent, Belgium
| | - Lydia Aslanidou
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, LHTC STI IBI EPFL, MED 32924 Station 9, 1015, Lausanne, Switzerland
| | | | | | | |
Collapse
|
41
|
Lareyre F, Clément M, Raffort J, Pohlod S, Patel M, Esposito B, Master L, Finigan A, Vandestienne M, Stergiopulos N, Taleb S, Trachet B, Mallat Z. TGFβ (Transforming Growth Factor-β) Blockade Induces a Human-Like Disease in a Nondissecting Mouse Model of Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2017; 37:2171-2181. [PMID: 28912363 DOI: 10.1161/atvbaha.117.309999] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/21/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Current experimental models of abdominal aortic aneurysm (AAA) do not accurately reproduce the major features of human AAA. We hypothesized that blockade of TGFβ (transforming growth factor-β) activity-a guardian of vascular integrity and immune homeostasis-would impair vascular healing in models of nondissecting AAA and would lead to sustained aneurysmal growth until rupture. APPROACH AND RESULTS Here, we test this hypothesis in the elastase-induced AAA model in mice. We analyze AAA development and progression using ultrasound in vivo, synchrotron-based ultrahigh resolution imaging ex vivo, and a combination of biological, histological, and flow cytometry-based cellular and molecular approaches in vitro. Systemic blockade of TGFβ using a monoclonal antibody induces a transition from a self-contained aortic dilatation to a model of sustained aneurysmal growth, associated with the formation of an intraluminal thrombus. AAA growth is associated with wall disruption but no medial dissection and culminates in fatal transmural aortic wall rupture. TGFβ blockade enhances leukocyte infiltration both in the aortic wall and the intraluminal thrombus and aggravates extracellular matrix degradation. Early blockade of IL-1β or monocyte-dependent responses substantially limits AAA severity. However, blockade of IL-1β after disease initiation has no effect on AAA progression to rupture. CONCLUSIONS Endogenous TGFβ activity is required for the healing of AAA. TGFβ blockade may be harnessed to generate new models of AAA with better relevance to the human disease. We expect that the new models will improve our understanding of the pathophysiology of AAA and will be useful in the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Fabien Lareyre
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Marc Clément
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Juliette Raffort
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Stefanie Pohlod
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Meghana Patel
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Bruno Esposito
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Leanne Master
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Alison Finigan
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Marie Vandestienne
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Nikolaos Stergiopulos
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Soraya Taleb
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Bram Trachet
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine, University of Cambridge, UK (F.L., M.C., J.R., M.P., L.M., A.F., Z.M.); Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institute for Research on Cancer and Aging in Nice, France (F.L., J.R.); University Hospital of Nice, France (F.L, J.R.); Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (B.E., M.V., S.T., Z.M.); Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Switzerland (S.P., N.S., B.T.); and IBiTech-bioMMeda (Institute Biomedical Technology-Biofluid, Tissue and Solid Mechanics for Medical Applications), Ghent University, Belgium (N.S., B.T.).
| |
Collapse
|
42
|
Doderer SA, Gäbel G, Kokje VBC, Northoff BH, Holdt LM, Hamming JF, Lindeman JHN. Adventitial adipogenic degeneration is an unidentified contributor to aortic wall weakening in the abdominal aortic aneurysm. J Vasc Surg 2017; 67:1891-1900.e4. [PMID: 28912007 DOI: 10.1016/j.jvs.2017.05.088] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 05/01/2017] [Indexed: 02/09/2023]
Abstract
OBJECTIVE The processes driving human abdominal aortic aneurysm (AAA) progression are not fully understood. Although antiinflammatory and proteolytic strategies effectively quench aneurysm progression in preclinical models, so far all clinical interventions failed. These observations hint at an incomplete understanding of the processes involved in AAA progression and rupture. Interestingly, strong clinical and molecular associations exist between popliteal artery aneurysms (PAAs) and AAAs; however, PAAs have an extremely low propensity to rupture. We thus reasoned that differences between these aneurysms may provide clues toward (auxiliary) processes involved in AAA-related wall debilitation. A better understanding of the pathophysiologic processes driving AAA growth can contribute to pharmaceutical treatments in the future. METHODS Aneurysmal wall samples were collected during open elective and emergency repair. Control perirenal aorta was obtained during kidney transplantation, and reference popliteal tissue obtained from the anatomy department. This study incorporates various techniques including (immuno)histochemistry, Western Blot, quantitative polymerase chain reaction, microarray, and cell culture. RESULTS Histologic evaluation of AAAs, PAAs, and control aorta shows extensive medial (PAA) and transmural fibrosis (AAA), and reveals abundant adventitial adipocytes aggregates as an exclusive phenomenon of AAAs (P < .001). Quantitative polymerase chain reaction, immunohistochemistry, Western blotting, and microarray analysis showed enrichment of adipogenic mediators (C/EBP family P = .027; KLF5 P < .000; and peroxisome proliferator activated receptor-γ, P = .032) in AAA tissue. In vitro differentiation tests indicated a sharply increased adipogenic potential of AAA adventitial mesenchymal cells (P < .0001). Observed enrichment of adipocyte-related genes and pathways in ruptured AAA (P < .0003) supports an association between the extent of fatty degeneration and rupture. CONCLUSIONS This translational study identifies extensive adventitial fatty degeneration as an ignored and distinctive feature of AAA disease. Enrichment of adipocyte genesis and adipocyte-related genes in ruptured AAA point to an association between the extent of fatty degeneration and rupture. This observation may (partly) explain the failure of medical therapy and could provide a lead for pharmaceutical alleviation of AAA progression.
Collapse
Affiliation(s)
- Stefan A Doderer
- Department of Vascular Surgery, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Gabor Gäbel
- Department of Vascular and Endovascular Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Vivianne B C Kokje
- Department of Vascular Surgery, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Bernd H Northoff
- Institute of Laboratory Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lesca M Holdt
- Institute of Laboratory Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jaap F Hamming
- Department of Vascular Surgery, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Jan H N Lindeman
- Department of Vascular Surgery, Leiden University Medical Center (LUMC), Leiden, The Netherlands.
| |
Collapse
|
43
|
Trachet B, Aslanidou L, Piersigilli A, Fraga-Silva RA, Sordet-Dessimoz J, Villanueva-Perez P, Stampanoni MF, Stergiopulos N, Segers P. Angiotensin II infusion into ApoE-/- mice: a model for aortic dissection rather than abdominal aortic aneurysm? Cardiovasc Res 2017; 113:1230-1242. [DOI: 10.1093/cvr/cvx128] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/26/2017] [Indexed: 01/13/2023] Open
Affiliation(s)
- Bram Trachet
- IBiTech–bioMMeda, Ghent University-iMinds Medical IT, De Pintelaan 185 Blok B, 9000 Ghent, Belgium
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Lydia Aslanidou
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Rodrigo A. Fraga-Silva
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | | | - Marco F.M. Stampanoni
- Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland
- Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland
| | - Nikolaos Stergiopulos
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Patrick Segers
- IBiTech–bioMMeda, Ghent University-iMinds Medical IT, De Pintelaan 185 Blok B, 9000 Ghent, Belgium
| |
Collapse
|
44
|
Bellini C, Kristofik NJ, Bersi MR, Kyriakides TR, Humphrey JD. A hidden structural vulnerability in the thrombospondin-2 deficient aorta increases the propensity to intramural delamination. J Mech Behav Biomed Mater 2017; 71:397-406. [PMID: 28412645 DOI: 10.1016/j.jmbbm.2017.01.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/23/2017] [Accepted: 01/27/2017] [Indexed: 12/22/2022]
Abstract
Mice lacking thrombospondin-2 (TSP2) represent an animal model of impaired collagen fibrillogenesis. Collagen constitutes ~1/3 of the wall of the normal murine descending thoracic aorta (DTA) and is thought to confer mechanical strength at high pressures. Microstructural analysis of the DTA from TSP2-null mice revealed irregular and disorganized collagen fibrils in the adventitia and at the interface between the media and adventitia. Yet, biaxial mechanical tests performed under physiologic loading conditions showed that most mechanical metrics, including stress and stiffness, were not different between mutant and control DTAs at 20- and 40-weeks of age, thus suggesting that the absence of TSP2 is well compensated under normal conditions. A detailed bilayered analysis of the wall mechanics predicted, however, that the adventitia of TSP2-null DTAs fails to engage at high pressures, which could render the media vulnerable to mechanical damage. Failure tests confirmed that the pressure at which the DTA ruptures is significantly lower in 20-week-old TSP2-null mice compared to age-matched controls (640±37 vs. 1120±45mmHg). Moreover, half of the 20-week-old and all 40-week-old mutant DTAs failed by delamination, not rupture. This delamination occurred at the interface between the media and the adventitia, with separation planes often observed at ~45 degrees with respect to the circumferential/axial directions. Combined with the observed microstructural anomalies, our theoretical-experimental biomechanical results suggest that TSP2-null DTAs are more susceptible to material failure when exposed to high pressures and this vulnerability may result from a reduced resistance to shear loading at the medial/adventitial border.
Collapse
Affiliation(s)
- C Bellini
- Department of Biomedical Engineering Yale University, New Haven, CT, United States.
| | - N J Kristofik
- Department of Biomedical Engineering Yale University, New Haven, CT, United States
| | - M R Bersi
- Department of Biomedical Engineering Yale University, New Haven, CT, United States
| | - T R Kyriakides
- Department of Biomedical Engineering Yale University, New Haven, CT, United States; Department of Pathology, Yale School of Medicine, New Haven, CT, United States; Vascular Biology and Therapeutics Program Yale School of Medicine, New Haven, CT, United States
| | - J D Humphrey
- Department of Biomedical Engineering Yale University, New Haven, CT, United States; Vascular Biology and Therapeutics Program Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
45
|
Sénémaud J, Caligiuri G, Etienne H, Delbosc S, Michel JB, Coscas R. Translational Relevance and Recent Advances of Animal Models of Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2017; 37:401-410. [DOI: 10.1161/atvbaha.116.308534] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 12/21/2016] [Indexed: 01/11/2023]
Abstract
Human abdominal aortic aneurysm (AAA) pathophysiology is not yet completely understood. In conductance arteries, the insoluble extracellular matrix, synthesized by vascular smooth muscle cells, assumes the function of withstanding the intraluminal arterial blood pressure. Progressive loss of this function through extracellular matrix proteolysis is a main feature of AAAs. As most patients are now treated via endovascular approaches, surgical AAA specimens have become rare. Animal models provide valuable complementary insights into AAA pathophysiology. Current experimental AAA models involve induction of intraluminal dilation (nondissecting AAAs) or a contained intramural rupture (dissecting models). Although the ideal model should reproduce the histological characteristics and natural history of the human disease, none of the currently available animal models perfectly do so. Experimental models try to represent the main pathophysiological determinants of AAAs: genetic or acquired defects in extracellular matrix, loss of vascular smooth muscle cells, and innate or adaptive immune response. Nevertheless, most models are characterized by aneurysmal stabilization and healing after a few weeks because of cessation of the initial stimulus. Recent studies have focused on ways to optimize existing models to allow continuous aneurysmal growth. This review aims to discuss the relevance and recent advances of current animal AAA models.
Visual Overview—
An online visual overview is available for this article.
Collapse
Affiliation(s)
- Jean Sénémaud
- From the UMR 1148, Inserm-Paris7 - Denis Diderot University, Xavier Bichat Hospital, Paris, France (J.S., G.C., H.E., S.D., J.-B.M., R.C.); UMR 1173, Inserm-Paris11 - Faculty of Health Sciences Simone Veil, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Montigny-le-Bretonneux, France (R.C.); Department of Vascular Surgery, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt, France (R.C.); and UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines
| | - Giuseppina Caligiuri
- From the UMR 1148, Inserm-Paris7 - Denis Diderot University, Xavier Bichat Hospital, Paris, France (J.S., G.C., H.E., S.D., J.-B.M., R.C.); UMR 1173, Inserm-Paris11 - Faculty of Health Sciences Simone Veil, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Montigny-le-Bretonneux, France (R.C.); Department of Vascular Surgery, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt, France (R.C.); and UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines
| | - Harry Etienne
- From the UMR 1148, Inserm-Paris7 - Denis Diderot University, Xavier Bichat Hospital, Paris, France (J.S., G.C., H.E., S.D., J.-B.M., R.C.); UMR 1173, Inserm-Paris11 - Faculty of Health Sciences Simone Veil, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Montigny-le-Bretonneux, France (R.C.); Department of Vascular Surgery, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt, France (R.C.); and UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines
| | - Sandrine Delbosc
- From the UMR 1148, Inserm-Paris7 - Denis Diderot University, Xavier Bichat Hospital, Paris, France (J.S., G.C., H.E., S.D., J.-B.M., R.C.); UMR 1173, Inserm-Paris11 - Faculty of Health Sciences Simone Veil, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Montigny-le-Bretonneux, France (R.C.); Department of Vascular Surgery, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt, France (R.C.); and UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines
| | - Jean-Baptiste Michel
- From the UMR 1148, Inserm-Paris7 - Denis Diderot University, Xavier Bichat Hospital, Paris, France (J.S., G.C., H.E., S.D., J.-B.M., R.C.); UMR 1173, Inserm-Paris11 - Faculty of Health Sciences Simone Veil, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Montigny-le-Bretonneux, France (R.C.); Department of Vascular Surgery, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt, France (R.C.); and UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines
| | - Raphaël Coscas
- From the UMR 1148, Inserm-Paris7 - Denis Diderot University, Xavier Bichat Hospital, Paris, France (J.S., G.C., H.E., S.D., J.-B.M., R.C.); UMR 1173, Inserm-Paris11 - Faculty of Health Sciences Simone Veil, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Montigny-le-Bretonneux, France (R.C.); Department of Vascular Surgery, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt, France (R.C.); and UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines
| |
Collapse
|
46
|
Qin Y, Wang Y, Liu O, Jia L, Fang W, Du J, Wei Y. Tauroursodeoxycholic Acid Attenuates Angiotensin II Induced Abdominal Aortic Aneurysm Formation in Apolipoprotein E-deficient Mice by Inhibiting Endoplasmic Reticulum Stress. Eur J Vasc Endovasc Surg 2016; 53:337-345. [PMID: 27889204 DOI: 10.1016/j.ejvs.2016.10.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 10/27/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE/BACKGROUND Abdominal aortic aneurysm (AAA) is characterised by the infiltration of smooth muscle cell (SMC) apoptosis, inflammatory cells, neovascularisation, and degradation of the extracellular matrix. Previous work has shown that endoplasmic reticulum (ER) stress and SMC apoptosis were increased both in a mouse model and human thoracic aortic aneurysm. However, whether the ER stress is activated in AAA formation and whether suppressing ER stress attenuates AAA is unknown. METHODS Human AAA and control aorta samples were collected. Expression of ER stress chaperones glucose-regulated protein (GRP)-78 and GRP-94 was detected by immunohistochemical staining. The effect of ER stress inhibitor tauroursodeoxycholic acid (TUDCA) on AAA formation in angiotensin (Ang) II induced apolipoprotein E-/- mice was explored. Elastin staining was used to observe the rupture of elastic fragmentation. Immunohistochemistry and Western blot analysis were performed, to detect the protein expression of ER stress chaperones and apoptosis molecules. RESULTS There was significant upregulation of GRP-78 and GRP-94 in aneurysmal areas of human AAA and Ang II induced ApoE-/- mice (p < .05). TUDCA significantly attenuated the maximum diameters of abdominal aortas in Ang II induced ApoE-/- mice (p < .05). TUDCA significantly reduced expression of ER stress chaperones and the apoptotic cell numbers (p < .05). Furthermore, TUDCA significantly reduced expression of apoptosis molecules, such as caspase-3, caspase-12, C/EBP homologous protein, c-Jun N-terminal kinase activating transcription factor 4, X-box binding protein, and eukaryotic initiation factor 2α in Ang II induced ApoE-/- mice (p < .05). CONCLUSION The results suggest that ER stress is involved in human and Ang II induced AAA formation in ApoE-/- mice. TUDCA attenuates Ang II induced AAA formation in ApoE-/- mice by inhibiting ER stress mediated apoptosis.
Collapse
Affiliation(s)
- Y Qin
- The Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China; The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China.
| | - Y Wang
- The Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China; The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - O Liu
- The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - L Jia
- The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - W Fang
- The Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - J Du
- The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Y Wei
- The Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China.
| |
Collapse
|
47
|
Martorell S, Hueso L, Gonzalez-Navarro H, Collado A, Sanz MJ, Piqueras L. Vitamin D Receptor Activation Reduces Angiotensin-II-Induced Dissecting Abdominal Aortic Aneurysm in Apolipoprotein E-Knockout Mice. Arterioscler Thromb Vasc Biol 2016; 36:1587-97. [PMID: 27283745 DOI: 10.1161/atvbaha.116.307530] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 05/27/2016] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is a vascular disorder characterized by chronic inflammation of the aortic wall. Low concentrations of vitamin D3 are associated with AAA development; however, the potential direct effect of vitamin D3 on AAA remains unknown. This study evaluates the effect of oral treatment with the vitamin D3 receptor (VDR) ligand, calcitriol, on dissecting AAA induced by angiotensin-II (Ang-II) infusion in apoE(-/-) mice. APPROACH AND RESULTS Oral treatment with calcitriol reduced Ang-II-induced dissecting AAA formation in apoE(-/-) mice, which was unrelated to systolic blood pressure or plasma cholesterol concentrations. Immunohistochemistry and reverse-transcription polymerase chain reaction analysis demonstrated a significant increase in macrophage infiltration, neovessel formation, matrix metalloproteinase-2 and matrix metalloproteinase-9, chemokine (CCL2 [(C-C motif) ligand 2], CCL5 [(C-C motif) ligand 5], and CXCL1 [(C-X-C motif) ligand 1]) and vascular endothelial growth factor expression in suprarenal aortic walls of apoE(-/-) mice infused with Ang-II, and all were significantly reduced by cotreatment with calcitriol. Phosphorylation of extracellular signal-regulated kinases 1/2, p38 mitogen-activated protein kinase, and nuclear factor-κB was also decreased in the suprarenal aortas of apoE(-/-) mice cotreated with calcitriol. These effects were accompanied by a marked increase in VDR-retinoid X receptor (RXR) interaction in the aortas of calcitriol-treated mice. In vitro, VDR activation by calcitriol in human endothelial cells inhibited Ang-II-induced leukocyte-endothelial cell interactions, morphogenesis, and production of endothelial proinflammatory and angiogenic chemokines through VDR-RXR interactions, and knockdown of VDR or RXR abolished the inhibitory effects of calcitriol. CONCLUSIONS VDR activation reduces dissecting AAA formation induced by Ang-II in apoE(-/-) mice and may constitute a novel therapeutic strategy to prevent AAA progression.
Collapse
Affiliation(s)
- Sara Martorell
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.)
| | - Luisa Hueso
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.)
| | - Herminia Gonzalez-Navarro
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.)
| | - Aida Collado
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.)
| | - Maria-Jesus Sanz
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.).
| | - Laura Piqueras
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.).
| |
Collapse
|
48
|
Tai HC, Tsai PJ, Chen JY, Lai CH, Wang KC, Teng SH, Lin SC, Chang AYW, Jiang MJ, Li YH, Wu HL, Maeda N, Tsai YS. Peroxisome Proliferator-Activated Receptor γ Level Contributes to Structural Integrity and Component Production of Elastic Fibers in the Aorta. Hypertension 2016; 67:1298-308. [PMID: 27045031 DOI: 10.1161/hypertensionaha.116.07367] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 03/09/2016] [Indexed: 01/24/2023]
Abstract
Loss of integrity and massive disruption of elastic fibers are key features of abdominal aortic aneurysm (AAA). Peroxisome proliferator-activated receptor γ (PPARγ) has been shown to attenuate AAA through inhibition of inflammation and proteolytic degradation. However, its involvement in elastogenesis during AAA remains unclear. PPARγ was highly expressed in human AAA within all vascular cells, including inflammatory cells and fibroblasts. In the aortas of transgenic mice expressing PPARγ at 25% normal levels (Pparg(C) (/-) mice), we observed the fragmentation of elastic fibers and reduced expression of vital elastic fiber components of elastin and fibulin-5. These were not observed in mice with 50% normal PPARγ expression (Pparg(+/-) mice). Infusion of a moderate dose of angiotensin II (500 ng/kg per minute) did not induce AAA but Pparg(+/-) aorta developed flattened elastic lamellae, whereas Pparg(C/-) aorta showed severe destruction of elastic fibers. After infusion of angiotensin II at 1000 ng/kg per minute, 73% of Pparg(C/-) mice developed atypical suprarenal aortic aneurysms: superior mesenteric arteries were dilated with extensive collagen deposition in adventitia and infiltrations of inflammatory cells. Although matrix metalloproteinase inhibition by doxycycline somewhat attenuated the dilation of aneurysm, it did not reduce the incidence nor elastic lamella deterioration in angiotensin II-infused Pparg(C/-) mice. Furthermore, PPARγ antagonism downregulated elastin and fibulin-5 in fibroblasts, but not in vascular smooth muscle cells. Chromatin immunoprecipitation assay demonstrated PPARγ binding in the genomic sequence of fibulin-5 in fibroblasts. Our results underscore the importance of PPARγ in AAA development though orchestrating proper elastogenesis and preserving elastic fiber integrity.
Collapse
Affiliation(s)
- Haw-Chih Tai
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Pei-Jane Tsai
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Ju-Yi Chen
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Chao-Han Lai
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Kuan-Chieh Wang
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Shih-Hua Teng
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Shih-Chieh Lin
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Alice Y W Chang
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Meei-Jyh Jiang
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Yi-Heng Li
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Hua-Lin Wu
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Nobuyo Maeda
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Yau-Sheng Tsai
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.).
| |
Collapse
|
49
|
Raffort J, Lareyre F, Clement M, Mallat Z. Micro-RNAs in abdominal aortic aneurysms: insights from animal models and relevance to human disease. Cardiovasc Res 2016; 110:165-77. [PMID: 26965051 DOI: 10.1093/cvr/cvw046] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/28/2016] [Indexed: 01/09/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a major health concern and may be associated with high rates of mortality linked to acute complications. Diagnosis and treatment are, respectively, based on imaging and surgical techniques. Drug-based therapies are still mostly ineffective, which highlight a real unmet need. Major pathophysiological mechanisms leading to aneurysm formation involve inflammatory processes, degradation of the extracellular matrix, and loss of smooth muscle cells. However, the precise cellular and molecular pathways are still poorly understood. Recently, microRNAs have emerged as major intracellular players in a wide range of biological processes, and their stability in extracellular medium within microvesicles has led to propose them as mediators of intercellular crosstalk and as potential biomarkers and therapeutic targets in a variety of disease settings. To date, several studies have been performed to address the involvement of micro-RNAs (miRs) in aneurysm formation and complications. Here, we discuss the roles and implications of miRs in animal models and their relevance to human AAA.
Collapse
Affiliation(s)
- Juliette Raffort
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge CB20 SZ, UK University of Nice-Sophia Antipolis, Medical School, Nice 06107, France INSERM U1081, CNRS UMR7284, IRCAN, Nice, France Clinical Chemistry Laboratory, University Hospital of Nice, Nice, France
| | - Fabien Lareyre
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge CB20 SZ, UK University of Nice-Sophia Antipolis, Medical School, Nice 06107, France INSERM U1081, CNRS UMR7284, IRCAN, Nice, France Department of Vascular Surgery, University Hospital of Nice, Nice, France
| | - Marc Clement
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge CB20 SZ, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge CB20 SZ, UK Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Research Center, Paris 75015, France
| |
Collapse
|
50
|
Trachet B, Piersigilli A, Fraga-Silva RA, Aslanidou L, Sordet-Dessimoz J, Astolfo A, Stampanoni MFM, Segers P, Stergiopulos N. Ascending Aortic Aneurysm in Angiotensin II-Infused Mice: Formation, Progression, and the Role of Focal Dissections. Arterioscler Thromb Vasc Biol 2016; 36:673-81. [PMID: 26891740 DOI: 10.1161/atvbaha.116.307211] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 02/05/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To understand the anatomy and physiology of ascending aortic aneurysms in angiotensin II-infused ApoE(-/-) mice. APPROACH AND RESULTS We combined an extensive in vivo imaging protocol (high-frequency ultrasound and contrast-enhanced microcomputed tomography at baseline and after 3, 10, 18, and 28 days of angiotensin II infusion) with synchrotron-based ultrahigh resolution ex vivo imaging (phase contrast X-ray tomographic microscopy) in n=47 angiotensin II-infused mice and 6 controls. Aortic regurgitation increased significantly over time, as did the luminal volume of the ascending aorta. In the samples that were scanned ex vivo, we observed one or several focal dissections, with the largest located in the outer convex aspect of the ascending aorta. The volume of the dissections moderately correlated to the volume of the aneurysm as measured in vivo (r(2)=0.46). After 3 days of angiotensin II infusion, we found an interlaminar hematoma in 7/12 animals, which could be linked to an intimal tear. There was also a significant increase in single laminar ruptures, which may have facilitated a progressive enlargement of the focal dissections over time. At later time points, the hematoma was resorbed and the medial and adventitial thickness increased. Fatal transmural dissection occurred in 8/47 mice at an early stage of the disease, before adventita remodeling. CONCLUSIONS We visualized and quantified the dissections that lead to ascending aortic aneurysms in angiotensin II-infused mice and provided unique insight into the temporal evolution of these lesions.
Collapse
Affiliation(s)
- Bram Trachet
- From the Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (B.T., R.A.F.-S., L.A., N.S.); IBiTech-bioMMeda, Ghent University-iMinds Medical IT, Ghent, Belgium (B.T., P.S.); School of Life Sciences, PTEC GE, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (A.P.); Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland (A.P.); Histology Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (J.S.-D.); Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland (A.A., M.F.M.S.); and Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland (M.F.M.S.).
| | - Alessandra Piersigilli
- From the Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (B.T., R.A.F.-S., L.A., N.S.); IBiTech-bioMMeda, Ghent University-iMinds Medical IT, Ghent, Belgium (B.T., P.S.); School of Life Sciences, PTEC GE, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (A.P.); Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland (A.P.); Histology Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (J.S.-D.); Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland (A.A., M.F.M.S.); and Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland (M.F.M.S.)
| | - Rodrigo A Fraga-Silva
- From the Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (B.T., R.A.F.-S., L.A., N.S.); IBiTech-bioMMeda, Ghent University-iMinds Medical IT, Ghent, Belgium (B.T., P.S.); School of Life Sciences, PTEC GE, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (A.P.); Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland (A.P.); Histology Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (J.S.-D.); Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland (A.A., M.F.M.S.); and Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland (M.F.M.S.)
| | - Lydia Aslanidou
- From the Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (B.T., R.A.F.-S., L.A., N.S.); IBiTech-bioMMeda, Ghent University-iMinds Medical IT, Ghent, Belgium (B.T., P.S.); School of Life Sciences, PTEC GE, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (A.P.); Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland (A.P.); Histology Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (J.S.-D.); Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland (A.A., M.F.M.S.); and Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland (M.F.M.S.)
| | - Jessica Sordet-Dessimoz
- From the Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (B.T., R.A.F.-S., L.A., N.S.); IBiTech-bioMMeda, Ghent University-iMinds Medical IT, Ghent, Belgium (B.T., P.S.); School of Life Sciences, PTEC GE, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (A.P.); Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland (A.P.); Histology Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (J.S.-D.); Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland (A.A., M.F.M.S.); and Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland (M.F.M.S.)
| | - Alberto Astolfo
- From the Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (B.T., R.A.F.-S., L.A., N.S.); IBiTech-bioMMeda, Ghent University-iMinds Medical IT, Ghent, Belgium (B.T., P.S.); School of Life Sciences, PTEC GE, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (A.P.); Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland (A.P.); Histology Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (J.S.-D.); Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland (A.A., M.F.M.S.); and Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland (M.F.M.S.)
| | - Marco F M Stampanoni
- From the Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (B.T., R.A.F.-S., L.A., N.S.); IBiTech-bioMMeda, Ghent University-iMinds Medical IT, Ghent, Belgium (B.T., P.S.); School of Life Sciences, PTEC GE, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (A.P.); Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland (A.P.); Histology Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (J.S.-D.); Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland (A.A., M.F.M.S.); and Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland (M.F.M.S.)
| | - Patrick Segers
- From the Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (B.T., R.A.F.-S., L.A., N.S.); IBiTech-bioMMeda, Ghent University-iMinds Medical IT, Ghent, Belgium (B.T., P.S.); School of Life Sciences, PTEC GE, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (A.P.); Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland (A.P.); Histology Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (J.S.-D.); Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland (A.A., M.F.M.S.); and Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland (M.F.M.S.)
| | - Nikolaos Stergiopulos
- From the Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (B.T., R.A.F.-S., L.A., N.S.); IBiTech-bioMMeda, Ghent University-iMinds Medical IT, Ghent, Belgium (B.T., P.S.); School of Life Sciences, PTEC GE, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (A.P.); Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland (A.P.); Histology Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland (J.S.-D.); Swiss Light Source, Paul Scherrer Institut, Villigen, Switzerland (A.A., M.F.M.S.); and Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland (M.F.M.S.)
| |
Collapse
|