1
|
Wang H, Zhang R, Jia X, Gao S, Gao T, Fan K, Li Y, Wang S, Qiao M, Yan S, Hui H, Dong H. Highly sensitive magnetic particle imaging of abdominal aortic aneurysm NETosis with anti-Ly6G iron oxide nanoparticles. Cell Death Discov 2024; 10:395. [PMID: 39237520 PMCID: PMC11377588 DOI: 10.1038/s41420-024-02156-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024] Open
Abstract
Abdominal aortic aneurysms (AAA) are a significant health concern in developed countries due to their considerable mortality rate. The crucial factor of the progression of AAA is the release of neutrophils and neutrophil extracellular traps (NETs). Magnetic particle imaging (MPI) is a new imaging technique that offers the capability to detect superparamagnetic iron oxide nanoparticles (SPION) with exceptional sensitivity. We aimed to investigate the functional imaging of MPI for the detection and monitoring of neutrophil infiltration within AAA. A novel multimodal imaging agent targeting neutrophils, PEG-Fe3O4-Ly6G-Cy7 nanoparticles (Ly6G NPs), were designed by coupling Fe3O4 nanoparticles with Ly6G antibodies and Cy7. The targeting and sensitivity of Ly6G NPs were assessed using MPI and fluorescence imaging (FLI) in the AAA mouse model. After the inhibition of NETosis, the degree of neutrophil infiltration and AAA severity were assessed using MPI with Ly6G NPs. Ly6G NPs accurately localized and quantitatively analyzed AAA lesion sites in mice using MPI/FLI/CT. Compared to the control group, elevated MPI and FLI signal intensities were detected at the abdominal aortic lesion site, and neutrophil infiltration and NETs accumulation were detected by histological analysis in the AAA models. After the inhibition of NETs accumulation in vivo, pathological damage in the abdominal aorta was significantly reduced, along with a decrease in the accumulation of Ly6G NPs and MPI signals. This multimodal MPI strategy revealed that nanoparticles targeting Ly6G can be used to detect neutrophil infiltration within AAA and monitor AAA severity.
Collapse
Affiliation(s)
- Heng Wang
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Ruijing Zhang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaohua Jia
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- Department of Ultrasound, Shuozhou Grand Hospital of Shanxi Medical University, Shuozhou, 036000, China
| | - Siqi Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Tingting Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Keyi Fan
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Yaling Li
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Shule Wang
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Maolin Qiao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Sheng Yan
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Hui Hui
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- National Key Laboratory of Kidney Diseases, Beijing, 100853, China.
| | - Honglin Dong
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
2
|
Zhu J, Meganathan I, MacAruthur R, Kassiri Z. Inflammation in Abdominal Aortic Aneurysm: Cause or Comorbidity? Can J Cardiol 2024:S0828-282X(24)00926-7. [PMID: 39181326 DOI: 10.1016/j.cjca.2024.08.274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024] Open
Abstract
Aortic aneurysm is a potentially deadly disease. It is chronic degeneration of the aortic wall that involves an inflammatory response and the immune system, aberrant remodelling of the extracellular matrix, and maladaptive transformation of the aortic cells. This review article focuses on the role of the inflammatory cells in abdominal aortic aneurysm. Studies in human aneurysmal specimens and animal models have identified various inflammatory cell types that could contribute to formation or expansion of aneurysms. These include the commonly studied leukocytes (neutrophils and macrophages) as well as the less commonly explored natural killer cells, dendritic cells, T cells, and B cells. Despite the well-demonstrated contribution of inflammatory cells and the related signalling pathways to development and expansion of aneurysms, anti-inflammatory therapy approaches have demonstrated limitations and may require additional considerations such as a combinational approach in targeting multiple pathways for significant beneficial outcomes.
Collapse
Affiliation(s)
- Jiechun Zhu
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ilamaran Meganathan
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Roderick MacAruthur
- Department of Cardiac Surgery, Mazankowski Alberta Heart Institute, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
3
|
Liang M, Li F, Wang Y, Chen H, Tian J, Zhao Z, Schneider KH, Li G. Woven Vascular Stent-Grafts with Surface Modification of Silk Fibroin-Based Paclitaxel/Metformin Microspheres. Bioengineering (Basel) 2023; 10:bioengineering10040399. [PMID: 37106586 PMCID: PMC10136065 DOI: 10.3390/bioengineering10040399] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
In-stent restenosis caused by tumor ingrowth increases the risk of secondary surgery for patients with abdominal aortic aneurysms (AAA) because conventional vascular stent grafts suffer from mechanical fatigue, thrombosis, and endothelial hyperplasia. For that, we report a woven vascular stent-graft with robust mechanical properties, biocompatibility, and drug delivery functions to inhibit thrombosis and the growth of AAA. Paclitaxel (PTX)/metformin (MET)-loaded silk fibroin (SF) microspheres were self-assembly synthesized by emulsification-precipitation technology and layer-by-layer coated on the surface of a woven stent via electrostatic bonding. The woven vascular stent-graft before and after coating drug-loaded membranes were characterized and analyzed systematically. The results show that small-sized drug-loaded microspheres increased the specific surface area and promoted the dissolution/release of drugs. The stent-grafts with drug-loaded membranes exhibited a slow drug-release profile more for than 70 h and low water permeability at 158.33 ± 17.56 mL/cm2·min. The combination of PTX and MET inhibited the growth of human umbilical vein endothelial cells. Therefore, it was possible to generate dual-drug-loaded woven vascular stent-grafts to achieve the more effective treatment of AAA.
Collapse
Affiliation(s)
- Mengdi Liang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- Jiangsu Advanced Textile Engineering Technology Center, Nantong 226007, China
| | - Fang Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- Jiangsu Advanced Textile Engineering Technology Center, Nantong 226007, China
| | - Yongfeng Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- Jiangsu Advanced Textile Engineering Technology Center, Nantong 226007, China
| | - Hao Chen
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- Jiangsu Advanced Textile Engineering Technology Center, Nantong 226007, China
| | - Jingjing Tian
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- Jiangsu Advanced Textile Engineering Technology Center, Nantong 226007, China
| | - Zeyu Zhao
- Department of Applied Physics, The Hong Kong Polytechnic University, 11 Yukchoi Rd, Hung Hom, Kowloon, Hong Kong 999077, China
| | - Karl H Schneider
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Gang Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- Jiangsu Advanced Textile Engineering Technology Center, Nantong 226007, China
| |
Collapse
|
4
|
Exploring the Effect and Mechanism of Si-Miao-Yong-An Decoction on Abdominal Aortic Aneurysm Based on Mice Experiment and Bioinformatics Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4766987. [PMID: 35685724 PMCID: PMC9173986 DOI: 10.1155/2022/4766987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/26/2022] [Accepted: 05/19/2022] [Indexed: 12/02/2022]
Abstract
Background Abdominal aortic aneurysm (AAA) is a fatal disease characterized by high morbidity and mortality in old population. Globally, effective drugs for AAA are still limited. Si-Miao-Yong-An decoction (SMYAD), a traditional Chinese medicine (TCM) formula with a high medical value, was reported to be successfully used in an old AAA patient. Thus, we reason that SMYAD may serve as a potential anti-AAA regime. Objective The exact effects and detailed mechanisms of SMYAD on AAA were explored by using the experimental study and bioinformatics analysis. Methods Firstly, C57BL/6N mice induced by Bap and Ang II were utilized to reproduce the AAA model, and the effects of SMYAD were systematically assessed according to histology, immunohistochemistry, and enzyme-linked immunosorbent assay (ELISA). Then, network pharmacology was applied to identify the biological processes, pathways, and hub targets of SMYAD against AAA; moreover, molecular docking was utilized to identify the binding ability and action targets. Results In an animal experiment, SMYAD was found to effectively alleviate the degree of pathological expansion of abdominal aorta and reduce the incidence of Bap/Ang II-induced AAA, along with reducing the damage to elastic lamella, attenuating infiltration of macrophage, and lowering the circulating IL-6 level corresponding to the animal study, and network pharmacology revealed the detailed mechanisms of SMYAD on AAA that were related to pathways of inflammatory response, defense response, apoptotic, cell migration and adhesion, and reactive oxygen species metabolic process. Then, seven targets, IL-6, TNF, HSP90AA1, RELA, PTGS2, ESR1, and MMP9, were identified as hub targets of SMYAD against AAA. Furthermore, molecular docking verification revealed that the active compounds of SMYAD had good binding ability and clear binding site with core targets related to AAA formation. Conclusion SMYAD can suppress AAA development through multicompound, multitarget, and multipathway, which provides a research direction for further study.
Collapse
|
5
|
Different gene co-expression patterns of aortic intima-media and adventitia in thoracic aortic aneurysm. Gene 2022; 819:146233. [PMID: 35121027 DOI: 10.1016/j.gene.2022.146233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 01/04/2022] [Accepted: 01/18/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Due to permanent aortic dilation, thoracic aortic aneurysm (TAA) is a life-threatening disease. Once ruptured, TAA has a high lethality and disability rate. Although studies have focused on transcriptomic alterations in TAA, more detailed analysis is still lacking, especially the different aortic intima-media and adventitia roles. This study aimed to identify the different co-expression patterns between the aortic intima-media and the adventitia underlying the aortic dilation. METHODS We analyzed the gene expression profiles obtained from Gene Expression Omnibus (GEO, GSE26155) database. With a false discovery rate (FDR) < 0.05 and |log2FC| ≥ 1, 56 and 33 differential genes in the intima-media and adventitia, respectively, between the non-dilated and dilated status. Gene ontology (GO) and gene set enrichment analysis revealed that degranulation and activation of neutrophils play an essential role in the intima-media of dilated aortas. Through weighted gene co-expression network analysis (WGCNA), we identified essential co-expressed modules and hub genes to explore the biological functions of the dysregulated genes. RESULTS Functional pathway analysis suggested that lipid metabolism, C-C motif chemokine pathways were significantly enriched in the adventitia, whereas ribosome proteins and related mRNA translation pathways were closely related to intima and media. Furthermore, the ssGSEA analysis indicated that macrophages, helper T cells, and neutrophils were higher in the intima-media of the dilated thoracic aorta. Finally, we validated the critical findings of the study with the murine model of TAA. CONCLUSION This study identified and verified hub genes and pathways in aortic intima-media and adventitia prominently associated with aortic dilation, providing practical understanding in the perspective of searching for new molecular targets.
Collapse
|
6
|
Xu T, Wang S, Li X, Li X, Qu K, Tong H, Zhang R, Bai S, Fan J. Lithium chloride represses abdominal aortic aneurysm via regulating GSK3β/SIRT1/NF-κB signaling pathway. Free Radic Biol Med 2021; 166:1-10. [PMID: 33588051 DOI: 10.1016/j.freeradbiomed.2021.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/14/2021] [Accepted: 02/04/2021] [Indexed: 10/22/2022]
Abstract
Lithium chloride (LiCl), a pharmacological compound, was effective in reducing inflammation, but whether it can protect against abdominal aortic aneurysm (AAA) is largely unknown. This study is designed to investigate therapeutic effects of LiCl on AAA and the potential mechanism. Rat AAA models were induced by periaortic application of CaCl2. AAA rats were treated by daily intraperitoneal injection of LiCl or vehicle alone to study the protection effects of LiCl in vivo. Rat primary vascular smooth muscle cells (VSMCs) stimulated with tumor necrosis factor (TNF)-α served as an in vitro model. LiCl treatment prevented the development of AAA through inhibiting the inflammatory cells infiltration and inflammatory cytokines overproduction, as well as attenuating superoxide production and elastin degradation in aorta of AAA rats. Additionally, the downregulation of p-GSK3β(Ser9) and SIRT1, upregulation of NF-κB(p-65), MMP-2 and MMP-9 in AAA were abolished by LiCl treatment. In vitro by upregulating p-GSK3β(Ser9), LiCl significantly induced SIRT1 expression, along with inhibition of the NF-κB activation and decreased elastin level elicited in VSMCs by TNF-α stimulation. SIRT1 activator SRT1720 achieved similar repressive effects as LiCl on TNF-α-induced NF-κB activation and decreased elastin in VSMCs. Moreover, administration of LiCl also caused regression of established rats AAA. This study provided the first evidence that LiCl prevented the development of AAA through inhibiting inflammation, MMPs, and superoxide production, and facilitating the biosynthesis of elastin. The beneficial effect of LiCl may be mediated by regulation GSK3β/SIRT1/NF-κB cascade.
Collapse
Affiliation(s)
- Tong Xu
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Shoushuai Wang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Xiang Li
- Department of Cell Biology, Key Laboratory of Cell Biology of National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology of Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Xiuquan Li
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Kaiyun Qu
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Hao Tong
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Ruijie Zhang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Shuling Bai
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Jun Fan
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China.
| |
Collapse
|
7
|
Bell M, Gandhi R, Shawer H, Tsoumpas C, Bailey MA. Imaging Biological Pathways in Abdominal Aortic Aneurysms Using Positron Emission Tomography. Arterioscler Thromb Vasc Biol 2021; 41:1596-1606. [PMID: 33761759 DOI: 10.1161/atvbaha.120.315812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Michael Bell
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Richa Gandhi
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Heba Shawer
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Charalampos Tsoumpas
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Marc A Bailey
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| |
Collapse
|
8
|
Brangsch J, Reimann C, Kaufmann JO, Adams LC, Onthank D, Thöne-Reineke C, Robinson S, Wilke M, Weller M, Buchholz R, Karst U, Botnar R, Hamm B, Makowski MR. Molecular MR-Imaging for Noninvasive Quantification of the Anti-Inflammatory Effect of Targeting Interleukin-1β in a Mouse Model of Aortic Aneurysm. Mol Imaging 2020; 19:1536012120961875. [PMID: 33216687 PMCID: PMC7682246 DOI: 10.1177/1536012120961875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Molecular-MRI is a promising imaging modality for the assessment of abdominal aortic aneurysms (AAAs). Interleukin-1β (IL-1β) represents a new therapeutic tool for AAA-treatment, since pro-inflammatory cytokines are key-mediators of inflammation. This study investigates the potential of molecular-MRI to evaluate therapeutic effects of an anti-IL-1β-therapy on AAA-formation in a mouse-model. Methods: Osmotic-minipumps were implanted in apolipoprotein-deficient-mice (N = 27). One group (Ang-II+01BSUR group, n = 9) was infused with angiotensin-II (Ang-II) for 4 weeks and received an anti-murine IL-1β-antibody (01BSUR) 3 times. One group (Ang-II-group, n = 9) was infused with Ang-II for 4 weeks but received no treatment. Control-group (n = 9) was infused with saline and received no treatment. MR-imaging was performed using an elastin-specific gadolinium-based-probe (0.2 mmol/kg). Results: Mice of the Ang-II+01BSUR-group showed a lower aortic-diameter compared to mice of the Ang-II-group and control mice (p < 0.05). Using the elastin-specific-probe, a significant decrease in elastin-destruction was observed in mice of the Ang-II+01BSUR-group. In vivo MR-measurements correlated well with histopathology (y = 0.34x-13.81, R2 = 0.84, p < 0.05), ICP-MS (y = 0.02x+2.39; R2 = 0.81, p < 0.05) and LA-ICP-MS. Immunofluorescence and western-blotting confirmed a reduced IL-1β-expression. Conclusions: Molecular-MRI enables the early visualization and quantification of the anti-inflammatory-effects of an IL-1β-inhibitor in a mouse-model of AAAs. Responders and non-responders could be identified early after the initiation of the therapy using molecular-MRI.
Collapse
Affiliation(s)
- Julia Brangsch
- Department of Radiology, 14903Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Berlin, Germany
| | - Carolin Reimann
- Department of Radiology, 14903Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Berlin, Germany
| | - Jan Ole Kaufmann
- Department of Radiology, 14903Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Division 1.5 Protein Analysis, Federal Institute for Materials Research and Testing (BAM), Berlin, Germany.,Department of Chemistry, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lisa Christine Adams
- Department of Radiology, 14903Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - David Onthank
- 128865Lantheus Medical Imaging, North Billerica, MA, USA
| | - Christa Thöne-Reineke
- Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Berlin, Germany
| | - Simon Robinson
- 128865Lantheus Medical Imaging, North Billerica, MA, USA
| | - Marco Wilke
- Division 1.5 Protein Analysis, Federal Institute for Materials Research and Testing (BAM), Berlin, Germany
| | - Michael Weller
- Division 1.5 Protein Analysis, Federal Institute for Materials Research and Testing (BAM), Berlin, Germany
| | - Rebecca Buchholz
- Institute of Inorganic and Analytical Chemistry, 9185Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, 9185Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Rene Botnar
- School of Biomedical Engineering and Imaging Sciences, 4616King's College London, St Thomas' Hospital, London, United Kingdom.,Wellcome Trust/EPSRC Centre for Medical Engineering, 4616King's College London, United Kingdom.,BHF Centre of Excellence, 4616King's College London, Denmark Hill Campus, London, United Kingdom.,Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bernd Hamm
- Department of Radiology, 14903Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Marcus Richard Makowski
- Department of Radiology, 14903Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,School of Biomedical Engineering and Imaging Sciences, 4616King's College London, St Thomas' Hospital, London, United Kingdom.,BHF Centre of Excellence, 4616King's College London, Denmark Hill Campus, London, United Kingdom.,Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| |
Collapse
|
9
|
Mangarova DB, Brangsch J, Mohtashamdolatshahi A, Kosch O, Paysen H, Wiekhorst F, Klopfleisch R, Buchholz R, Karst U, Taupitz M, Schnorr J, Hamm B, Makowski MR. Ex vivo magnetic particle imaging of vascular inflammation in abdominal aortic aneurysm in a murine model. Sci Rep 2020; 10:12410. [PMID: 32709967 PMCID: PMC7381631 DOI: 10.1038/s41598-020-69299-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/26/2020] [Indexed: 01/06/2023] Open
Abstract
Abdominal aortic aneurysms (AAAs) are currently one of the leading causes of death in developed countries. Inflammation is crucial in the disease progression, having a substantial impact on various determinants in AAAs development. Magnetic particle imaging (MPI) is an innovative imaging modality, enabling the highly sensitive detection of magnetic nanoparticles (MNPs), suitable as surrogate marker for molecular targeting of vascular inflammation. For this study, Apolipoprotein E-deficient-mice underwent surgical implantation of osmotic minipumps with constant Angiotensin II infusion. After 3 and 4 weeks respectively, in-vivo-magnetic resonance imaging (MRI), ex-vivo-MPI and ex-vivo-magnetic particle spectroscopy (MPS) were performed. The results were validated by histological analysis, immunohistology and laser ablation-inductively coupled plasma-mass spectrometry. MR-angiography enabled the visualization of aneurysmal development and dilatation in the experimental group. A close correlation (R = 0.87) with histological area assessment was measured. Ex-vivo-MPS revealed abundant iron deposits in AAA samples and ex-vivo histopathology measurements were in good agreement (R = 0.76). Ex-vivo-MPI and MPS results correlated greatly (R = 0.99). CD68-immunohistology stain and Perls’-Prussian-Blue-stain confirmed the colocalization of macrophages and MNPs. This study demonstrates the feasibility of ex-vivo-MPI for detecting inflammation in AAA. The quantitative ability for mapping MNPs establishes MPI as a promising tool for monitoring inflammatory progression in AAA in an experimental setting.
Collapse
Affiliation(s)
- Dilyana B Mangarova
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany. .,Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Str. 15, Building 12, 14163, Berlin, Germany.
| | - Julia Brangsch
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.,Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Königsweg 67, Building 21, 14163, Berlin, Germany
| | - Azadeh Mohtashamdolatshahi
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Olaf Kosch
- Department 8.2-Biosignals, Physikalisch-Technische Bundesanstalt Berlin, Abbestrasse 2-12, 10587, Berlin, Germany
| | - Hendrik Paysen
- Department 8.2-Biosignals, Physikalisch-Technische Bundesanstalt Berlin, Abbestrasse 2-12, 10587, Berlin, Germany
| | - Frank Wiekhorst
- Department 8.2-Biosignals, Physikalisch-Technische Bundesanstalt Berlin, Abbestrasse 2-12, 10587, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Str. 15, Building 12, 14163, Berlin, Germany
| | - Rebecca Buchholz
- Institute of Inorganic and Analytical Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstr. 30, 48149, Münster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstr. 30, 48149, Münster, Germany
| | - Matthias Taupitz
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Jörg Schnorr
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Bernd Hamm
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Marcus R Makowski
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.,Department of Diagnostic and Interventional Radiology, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| |
Collapse
|
10
|
Abstract
The objective of this research was to estimate whether a [99mTc]duramycin probe can be used for apoptosis imaging in patients with aortic aneurysm (AA). Vascular smooth muscle cell (SMC) apoptosis has an important influence on AA development. Thus, non-invasive imaging of SMC apoptosis may be able to evaluate AA progress and risk stratification. SMCs were treated with hydrogen peroxide (H2O2; 200 μΜ) or culture medium as a control. Apoptosis was measured using flow cytometry and [99mTc]duramycin to detect the binding efficiency to apoptotic SMCs. C57/BL6 mice were administered angiotensin-II and beta-aminopropionitrile (BAPN) subcutaneously to establish an AA model, or saline for controls. Aortic specimens underwent pathological evaluation and their aortic diameters were measured after 6 weeks. Micro-SPECT/CT scanning of [99mTc]duramycin and 18F-FDG PET detection were performed. SMCs treated with H2O2 showed more apoptosis compared with the control group (67.2 ± 3.8% vs. 16.1 ± 0.6%, P < 0.01). The experimental group showed a high rate of AA formation (70%) compared with no AA formation in the control group. The average aorta diameter was higher and [99mTc]duramycin uptake at the AA site was higher in the experimental group compared with the control group. Compared with the normal aorta in the control group, AA in experiment group had more severe medial degeneration, elastic fiber reduction and fracture, and collagen degeneration. TUNEL staining verified the higher apoptosis rate at the AA site in experiment group compared with the control group (63.9 ± 3.7% in ascending AA, 66.4 ± 4.0% in thoracic AA, vs. 3.5 ± 0.3% in normal aorta, P < 0.01). [99mTc]Duramycin may be an effective probe to evaluate apoptosis in AA.
Collapse
|
11
|
English SJ, Sastriques SE, Detering L, Sultan D, Luehmann H, Arif B, Heo GS, Zhang X, Laforest R, Zheng J, Lin CY, Gropler RJ, Liu Y. CCR2 Positron Emission Tomography for the Assessment of Abdominal Aortic Aneurysm Inflammation and Rupture Prediction. Circ Cardiovasc Imaging 2020; 13:e009889. [PMID: 32164451 PMCID: PMC7101060 DOI: 10.1161/circimaging.119.009889] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND The monocyte chemoattractant protein-1/CCR2 (chemokine receptor 2) axis plays an important role in abdominal aortic aneurysm (AAA) pathogenesis, with effects on disease progression and anatomic stability. We assessed the expression of CCR2 in a rodent model and human tissues, using a targeted positron emission tomography radiotracer (64Cu-DOTA-ECL1i). METHODS AAAs were generated in Sprague-Dawley rats by exposing the infrarenal, intraluminal aorta to PPE (porcine pancreatic elastase) under pressure to induce aneurysmal degeneration. Heat-inactivated PPE was used to generate a sham operative control. Rat AAA rupture was stimulated by the administration of β-aminopropionitrile, a lysyl oxidase inhibitor. Biodistribution was performed in wild-type rats at 1 hour post tail vein injection of 64Cu-DOTA-ECL1i. Dynamic positron emission tomography/computed tomography imaging was performed in rats to determine the in vivo distribution of radiotracer. RESULTS Biodistribution showed fast renal clearance. The localization of radiotracer uptake in AAA was verified with high-resolution computed tomography. At day 7 post-AAA induction, the radiotracer uptake (standardized uptake value [SUV]=0.91±0.25) was approximately twice that of sham-controls (SUV=0.47±0.10; P<0.01). At 14 days post-AAA induction, radiotracer uptake by either group did not significantly change (AAA SUV=0.86±0.17 and sham-control SUV=0.46±0.10), independent of variations in aortic diameter. Competitive CCR2 receptor blocking significantly decreased AAA uptake (SUV=0.42±0.09). Tracer uptake in AAAs that subsequently ruptured (SUV=1.31±0.14; P<0.005) demonstrated uptake nearly twice that of nonruptured AAAs (SUV=0.73±0.11). Histopathologic characterization of rat and human AAA tissues obtained from surgery revealed increased expression of CCR2 that was co-localized with CD68+ macrophages. Ex vivo autoradiography demonstrated specific binding of 64Cu-DOTA-ECL1i to CCR2 in both rat and human aortic tissues. CONCLUSIONS CCR2 positron emission tomography is a promising new biomarker for the noninvasive assessment of AAA inflammation that may aid in associated rupture prediction.
Collapse
MESH Headings
- Aneurysm, Ruptured/diagnosis
- Aneurysm, Ruptured/genetics
- Aneurysm, Ruptured/metabolism
- Animals
- Aorta, Abdominal/diagnostic imaging
- Aorta, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/diagnosis
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Biomarkers/metabolism
- Fluorodeoxyglucose F18/pharmacology
- Gene Expression Regulation
- Male
- Positron-Emission Tomography/methods
- Prognosis
- RNA/genetics
- Radiopharmaceuticals/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, CCR2/biosynthesis
- Receptors, CCR2/genetics
Collapse
Affiliation(s)
- Sean J. English
- Department of Surgery, Section of Vascular Surgery, Washington University, St. Louis, MO
| | - Sergio E. Sastriques
- Department of Surgery, Section of Vascular Surgery, Washington University, St. Louis, MO
| | - Lisa Detering
- Department of Radiology, Washington University, St. Louis, MO
| | - Deborah Sultan
- Department of Radiology, Washington University, St. Louis, MO
| | - Hannah Luehmann
- Department of Radiology, Washington University, St. Louis, MO
| | - Batool Arif
- Department of Surgery, Section of Vascular Surgery, Washington University, St. Louis, MO
| | - Gyu Seong Heo
- Department of Radiology, Washington University, St. Louis, MO
| | - Xiaohui Zhang
- Department of Radiology, Washington University, St. Louis, MO
| | | | - Jie Zheng
- Department of Radiology, Washington University, St. Louis, MO
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington University, St. Louis, MO
| | | | - Yongjian Liu
- Department of Radiology, Washington University, St. Louis, MO
| |
Collapse
|
12
|
Brangsch J, Reimann C, Kaufmann JO, Adams LC, Onthank DC, Thöne-Reineke C, Robinson SP, Buchholz R, Karst U, Botnar RM, Hamm B, Makowski MR. Concurrent Molecular Magnetic Resonance Imaging of Inflammatory Activity and Extracellular Matrix Degradation for the Prediction of Aneurysm Rupture. Circ Cardiovasc Imaging 2020; 12:e008707. [PMID: 30871334 DOI: 10.1161/circimaging.118.008707] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Molecular magnetic resonance imaging is a promising modality for the characterization of abdominal aortic aneurysms (AAAs). The combination of different molecular imaging biomarkers may improve the assessment of the risk of rupture. This study investigates the feasibility of imaging inflammatory activity and extracellular matrix degradation by concurrent dual-probe molecular magnetic resonance imaging in an AAA mouse model. METHODS Osmotic minipumps with a continuous infusion of Ang II (angiotensin II; 1000 ng/[kg·min]) to induce AAAs were implanted in apolipoprotein-deficient mice (N=58). Animals were assigned to 2 groups. In group 1 (longitudinal group, n=13), imaging was performed once after 1 week with a clinical dose of a macrophage-specific iron oxide-based probe (ferumoxytol, 4 mgFe/kg, surrogate marker for inflammatory activity) and an elastin-specific gadolinium-based probe (0.2 mmol/kg, surrogate marker for extracellular matrix degradation). Animals were then monitored with death as end point. In group 2 (week-by-week-group), imaging with both probes was performed after 1, 2, 3, and 4 weeks (n=9 per group). Both probes were evaluated in 1 magnetic resonance session. RESULTS The combined assessment of inflammatory activity and extracellular matrix degradation was the strongest predictor of AAA rupture (sensitivity 100%; specificity 89%; area under the curve, 0.99). Information from each single probe alone resulted in lower predictive accuracy. In vivo measurements for the elastin- and iron oxide-probe were in good agreement with ex vivo histopathology (Prussian blue-stain: R2=0.96, P<0.001; Elastica van Giesson stain: R2=0.79, P<0.001). Contrast-to-noise ratio measurements for the iron oxide and elastin-probe were in good agreement with inductively coupled mass spectroscopy ( R2=0.88, R2=0.75, P<0.001) and laser ablation coupled to inductively coupled plasma-mass spectrometry. CONCLUSIONS This study demonstrates the potential of the concurrent assessment of inflammatory activity and extracellular matrix degradation by dual-probe molecular magnetic resonance imaging in an AAA mouse model. Based on the combined information from both molecular probes, the rupture of AAAs could reliably be predicted.
Collapse
Affiliation(s)
- Julia Brangsch
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (J.B., C.R., J.O.K., L.C.A., B.H., M.R.M.).,Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Germany (J.B., C.R., C.T.-R.)
| | - Carolin Reimann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (J.B., C.R., J.O.K., L.C.A., B.H., M.R.M.).,Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Germany (J.B., C.R., C.T.-R.)
| | - Jan O Kaufmann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (J.B., C.R., J.O.K., L.C.A., B.H., M.R.M.).,Federal Institute for Materials Research and Testing (BAM), Division 1.5 Protein Analysis, Berlin, Germany (J.O.K.).,Department of Chemistry, Humboldt-Universität zu Berlin, Germany (J.O.K.)
| | - Lisa C Adams
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (J.B., C.R., J.O.K., L.C.A., B.H., M.R.M.)
| | - David C Onthank
- Lantheus Medical Imaging, North Billerica, MA (D.C.O., S.P.R.)
| | - Christa Thöne-Reineke
- Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Germany (J.B., C.R., C.T.-R.)
| | | | - Rebecca Buchholz
- Institute of Inorganic and Analytical Chemistry, Westfälische Wilhelms-Universität Münster, Germany (R.B., U.K.)
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, Westfälische Wilhelms-Universität Münster, Germany (R.B., U.K.)
| | - Rene M Botnar
- School of Biomedical Engineering and Imaging Sciences (R.M.B., M.R.M.), King's College London, United Kingdom.,BHF Centre of Excellence (R.M.B., M.R.M.), King's College London, United Kingdom.,Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago (R.M.B.)
| | - Bernd Hamm
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (J.B., C.R., J.O.K., L.C.A., B.H., M.R.M.)
| | - Marcus R Makowski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (J.B., C.R., J.O.K., L.C.A., B.H., M.R.M.).,School of Biomedical Engineering and Imaging Sciences (R.M.B., M.R.M.), King's College London, United Kingdom.,BHF Centre of Excellence (R.M.B., M.R.M.), King's College London, United Kingdom
| |
Collapse
|
13
|
Sharma N, Dev R, Belenchia AM, Aroor AR, Whaley-Connell A, Pulakat L, Hans CP. Deficiency of IL12p40 (Interleukin 12 p40) Promotes Ang II (Angiotensin II)-Induced Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2019; 39:212-223. [PMID: 30580570 DOI: 10.1161/atvbaha.118.311969] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Objective- Abdominal aortic aneurysm is caused by the accumulation of inflammatory cells in the aortic wall. Our recent studies demonstrated that inhibition of Notch signaling attenuates abdominal aortic aneurysm formation by shifting the macrophage balance towards anti-inflammatory (M2) phenotype. Using IL12p40-/- (interleukin 12 p40) mice, we investigated the effects of M2-predominant macrophages on the development of abdominal aortic aneurysm. Approach and Results- Male (8-10 week-old) wild-type and IL12p40-/- mice (n=15) on C57BL/6 background were infused with Ang II (angiotensin II, 1000 ng/kg per minute) by implanting osmotic pumps subcutaneously for 28 days. In the IL12p40-/- mice, Ang II significantly increased the maximal intraluminal diameter (9/15) as determined by transabdominal ultrasound imaging. In addition, IL12p40-deletion significantly increased aortic stiffness in response to Ang II as measured by pulse wave velocity and atomic force microscopy. Histologically, IL12p40-/- mice exhibited increased maximal external diameter of aorta and aortic lesions associated with collagen deposition and increased elastin fragmentation compared with wild-type mice infused with Ang II. Mechanistically, IL12p40 deficiency by siRNA (small interfering RNA) augmented the Tgfβ2-mediated Mmp2 expression in wild-type bone marrow-derived macrophages without affecting the expression of Mmp9. No such effects of IL12p40 deficiency on MMP2/MMP9 was observed in human aortic smooth muscle cells or fibroblasts. Depletion of macrophages in IL12p40-/- mice by clodronate liposomes significantly decreased the maximal external diameter of aorta and aortic stiffness in response to Ang II as determined by imaging and atomic force microscopy. Conclusions- IL12p40 depletion promotes the development of abdominal aortic aneurysm, in part, by facilitating recruitment of M2-like macrophages and potentiating aortic stiffness and fibrosis mediated by Tgfβ2.
Collapse
Affiliation(s)
- Neekun Sharma
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Rishabh Dev
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Anthony M Belenchia
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Annayya R Aroor
- Department of Medical Pharmacology and Physiology (A.R.A., C.P.H.), University of Missouri, Columbia
| | - Adam Whaley-Connell
- Harry S. Truman Memorial Veterans' Hospital (A.W.-C.), University of Missouri, Columbia
| | - Lakshmi Pulakat
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Chetan P Hans
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Department of Medical Pharmacology and Physiology (A.R.A., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| |
Collapse
|
14
|
Wang X, Lane BA, Eberth JF, Lessner SM, Vyavahare NR. Gold nanoparticles that target degraded elastin improve imaging and rupture prediction in an AngII mediated mouse model of abdominal aortic aneurysm. Theranostics 2019; 9:4156-4167. [PMID: 31281538 PMCID: PMC6592177 DOI: 10.7150/thno.34441] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/28/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Abdominal aortic aneurysms (AAA) are characterized by a progressive disruption and weakening of the extracellular matrix (ECM) leading to dilation of the aorta which can be fatal if not treated. Current diagnostic imaging modalities provides little insight on the varying degree of ECM degeneration that precedes rupture in AAAs. Targeted delivery of contrast agents such as gold nanoparticles (GNPs) that bind to degraded matrix could prove useful when combined with computed tomography (CT) to provide a non-invasive surrogate marker of AAA rupture potential. Methods: AAAs were induced by chronic infusion of angiotensin II (AngII) into low density-lipoprotein receptor-deficient (LDLr -/-) mice in combination with a high-fat diet. Abdominal ultrasound was used to monitor disease progression and to assess the circumferential strain throughout the cardiac cycle. At six weeks, GNPs conjugated with an elastin antibody (EL-GNP) were injected retro-orbitally. Mice were euthanized 24 hours after EL-GNP injection, and aortas were explanted and scanned ex-vivo with a micro-CT system. Histological assessment and 3D models of the aneurysms with micro-CT were used to determine the EL-GNPs distribution. Isolated vessel burst pressure testing was performed on each aneurysmal aorta to quantify rupture strength and to assess rupture location. Results: Aneurysms were found along the suprarenal aorta in AngII infused mice. Darkfield microscopy indicated EL-GNPs accumulation around the site of degraded elastin while avoiding the healthy and intact elastin fibers. Using nonlinear regression, the micro-CT signal intensity of EL-GNPs along the suprarenal aortas correlated strongly with burst pressures (R2=0.9415) but not the dilation as assessed by ultrasound measurements. Conclusions: Using an established mouse model of AAA, we successfully demonstrated in vivo targeting of EL-GNPs to damaged aortic elastin and correlated micro-CT-based signal intensities with burst pressures. Thus, we show that this novel targeting technique can be used as a diagnostic tool to predict the degree of elastin damage and therefore rupture potential in AAAs better than the extent of dilation.
Collapse
|
15
|
Zhang TH, Chi DC, Jiang WL, Qiang S. Marfan syndrome combined with huge abdominal aortic aneurysm size of 20 × 11 cm: A case report of surgical approach. Medicine (Baltimore) 2018; 97:e09398. [PMID: 30212924 PMCID: PMC6156054 DOI: 10.1097/md.0000000000009398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
RATIONALE Abdominal aortic aneurysm is one of the most common aneurisms. Patients presenting with secondary back pain should be given prompt medical attention. Herein, a rare case of a giant abdominal aortic aneurysm that was successfully treated with surgery is described. PATIENT CONCERNS A 33-year-old Chinese male suffered from Marfan syndrome combined with giant abdominal aortic aneurysm, and presented with back pain, fever, nausea, vomiting, abdominal distention, and constipation. After undergoing numerous tests, the patient underwent an abdominal aortic aneurysm resection coupled with artificial graft bypass. The patient's recovery was smooth, and he was discharged 14 days after the operation in stable condition. DIAGNOSIS Abdominal aortic aneurysm. INTERVENTIONS The patient underwent a surgery involving mass resection and artificial graft bypass. OUTCOME The patient was discharged 14 days after surgery in stable condition. LESSONS Giant abdominal aortic aneurysms are rarely seen, and aneurysmectomy associated with prosthetic vascular graft repair is an effective and standard treatment for such aneurysms.
Collapse
Affiliation(s)
| | | | | | - Shuai Qiang
- Department of Plastic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
16
|
Hu C, Zhu K, Li J, Wang C, Lai L. Molecular targets in aortic aneurysm for establishing novel management paradigms. J Thorac Dis 2017; 9:4708-4722. [PMID: 29268541 DOI: 10.21037/jtd.2017.10.63] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Aortic aneurysm (AA) is a lethal disease and presents a large challenge for surgeons in the clinic. Although surgical management remains the major choice of AA, operative mortality remains high. With advances in understanding of the mechanisms of AAs, molecular targets, such as matrix metalloproteinases (MMPs), D-dimer, and inflammation markers, including C-reactive protein, interleukins and phagocytes, are important in the pathology of development of AA. These markers may become important for improving the diagnostic quality and provide more therapeutic choices for treatment of AA. Although these new markers require long-term trials before they can be translated into the clinic, they can still be helpful in determining new directions. The main aim of this review is to discuss the current findings of molecular targets in progression of AA and discuss the potential application of these new targets for managing this disease.
Collapse
Affiliation(s)
- Chengkai Hu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Shanghai Institute of Cardiovascular Disease, Shanghai 200032, China
| | - Kai Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Shanghai Institute of Cardiovascular Disease, Shanghai 200032, China
| | - Jun Li
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Shanghai Institute of Cardiovascular Disease, Shanghai 200032, China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Shanghai Institute of Cardiovascular Disease, Shanghai 200032, China
| | - Lao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Shanghai Institute of Cardiovascular Disease, Shanghai 200032, China
| |
Collapse
|
17
|
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening disease associated with high morbidity, and high mortality in the event of aortic rupture. Major advances in open surgical and endovascular repair of AAA have been achieved during the past 2 decades. However, drug-based therapies are still lacking, highlighting a real need for better understanding of the molecular and cellular mechanisms involved in AAA formation and progression. The main pathological features of AAA include extracellular matrix remodelling associated with degeneration and loss of vascular smooth muscle cells and accumulation and activation of inflammatory cells. The inflammatory process has a crucial role in AAA and substantially influences many determinants of aortic wall remodelling. In this Review, we focus specifically on the involvement of monocytes and macrophages, summarizing current knowledge on the roles, origin, and functions of these cells in AAA development and its complications. Furthermore, we show and propose that distinct monocyte and macrophage subsets have critical and differential roles in initiation, progression, and healing of the aneurysmal process. On the basis of experimental and clinical studies, we review potential translational applications to detect, assess, and image macrophage subsets in AAA, and discuss the relevance of these applications for clinical practice.
Collapse
|
18
|
Emeto TI, Alele FO, Smith AM, Smith FM, Dougan T, Golledge J. Use of Nanoparticles As Contrast Agents for the Functional and Molecular Imaging of Abdominal Aortic Aneurysm. Front Cardiovasc Med 2017; 4:16. [PMID: 28386544 PMCID: PMC5362602 DOI: 10.3389/fcvm.2017.00016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/09/2017] [Indexed: 01/19/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a degenerative disease of the aorta common in adults older than 65 years of age. AAA is usually imaged using ultrasound or computed tomography. Molecular imaging technologies employing nanoparticles (NPs) have been proposed as novel ways to quantify pathological processes, such as inflammation, within AAAs as a means to identify the risk of rapid progression or rupture. This article reviews the current evidence supporting the role of NP-based imaging in the management of AAA. Currently, ultrasmall superparamagnetic NPs enhanced magnetic resonance imaging appears to hold the greatest potential for imaging macrophage-mediated inflammation in human AAA.
Collapse
Affiliation(s)
- Theophilus I Emeto
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia; Queensland Research Centre for Peripheral Vascular Diseases, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia
| | - Faith O Alele
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University , Townsville, QLD , Australia
| | - Amy M Smith
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University , Townsville, QLD , Australia
| | - Felicity M Smith
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University , Townsville, QLD , Australia
| | - Tammy Dougan
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Addenbrookes Hospital , Cambridge , UK
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Diseases, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia; Department of Vascular and Endovascular Surgery, The Townsville Hospital, Townsville, QLD, Australia
| |
Collapse
|
19
|
Brangsch J, Reimann C, Collettini F, Buchert R, Botnar RM, Makowski MR. Molecular Imaging of Abdominal Aortic Aneurysms. Trends Mol Med 2017; 23:150-164. [PMID: 28110838 DOI: 10.1016/j.molmed.2016.12.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/06/2016] [Accepted: 12/11/2016] [Indexed: 12/21/2022]
Abstract
Abdominal aortic aneurysms (AAAs) represent a vascular disease with severe complications. AAAs are currently the overall 10th leading cause of death in western countries and their incidence is rising. Although different diagnostic techniques are currently available in clinical practice, including ultrasound (US), magnetic resonance imaging (MRI), and computed tomography (CT), imaging-based prediction of life-threatening complications such as aneurysm-rupture remains challenging. Molecular imaging provides a novel diagnostic approach for in vivo visualization of biological processes and pathological alterations at a cellular and molecular level. Its overall aim is to improve our understanding of disease pathogenesis and to facilitate novel diagnostic pathways. This review outlines recent preclinical and clinical developments in molecular MRI, positron emission tomography (PET), and single-photon emission computed tomography (SPECT) for imaging of AAAs.
Collapse
Affiliation(s)
- Julia Brangsch
- Department of Radiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Carolin Reimann
- Department of Radiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Federico Collettini
- Department of Radiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Ralf Buchert
- Department of Radiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - René M Botnar
- Department of Radiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Division of Imaging Sciences and Biomedical Engineering, King's College London, London WC2R 2LS, UK; Wellcome Trust and Engineering and Physical Sciences Research Council (EPSRC) Medical Engineering Centre, King's College London, London SE1 7EH, UK; British Heart Foundation (BHF) Centre of Excellence, King's College London, London SE5 9NU, UK; National Institute for Health Research (NIHR) Biomedical Research Centre, King's College London, London SE1 9RT, UK
| | - Marcus R Makowski
- Department of Radiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; Division of Imaging Sciences and Biomedical Engineering, King's College London, London WC2R 2LS, UK.
| |
Collapse
|
20
|
Jalalzadeh H, Indrakusuma R, Planken RN, Legemate DA, Koelemay MJW, Balm R. Inflammation as a Predictor of Abdominal Aortic Aneurysm Growth and Rupture: A Systematic Review of Imaging Biomarkers. Eur J Vasc Endovasc Surg 2016; 52:333-42. [PMID: 27283346 DOI: 10.1016/j.ejvs.2016.05.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/02/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Methods are required to identify abdominal aortic aneurysms (AAAs) at increased risk of rupture. Inflammatory characteristics of AAA can be visualised using advanced imaging techniques and have been proposed as potential predictors of aneurysm progression. The objective of this review was to determine which inflammatory imaging biomarkers are associated with AAA growth and rupture. METHODS A systematic review was carried out in accordance with the PRISMA guidelines. The electronic databases of Medline (PubMed), Embase, and the Cochrane Library were searched up to January 1, 2016 for studies to determine the potential association between inflammatory imaging biomarkers and AAA growth or rupture. RESULTS Seven studies were included, comprising 202 AAA patients. (18)F-fluoro-deoxy-glucose positron emission tomography ((18)F-FDG PET-CT) was evaluated in six studies. Magnetic resonance imaging with ultrasmall superparamagnetic particles of iron oxide (USPIO-MRI) was evaluated in one study. Two of six (18)F-FDG PET-CT studies reported a significant negative correlation (r=.383, p = .015) or a significant negative association (p = .04). Four of six (18)F-FDG PET-CT studies reported no significant association between (18)F-FDG uptake and AAA growth. The single study investigating USPIO-MRI demonstrated that AAA growth was three times higher in patients with focal USPIO uptake in the AAA wall compared to patients with diffuse or no USPIO uptake in the wall (0.66 vs. 0.24 vs. 0.22 cm/y, p = .020). In the single study relating (18)F-FDG uptake results to AAA rupture, the association was not significant. CONCLUSIONS Current evidence shows contradictory associations between (18)F-FDG uptake and AAA growth. Data on the association with rupture are insufficient. Based on the currently available evidence, neither (18)F-FDG PET-CT nor USPIO-MRI can be implemented as growth or rupture prediction tools in daily practice. The heterogeneous results reflect the complex and partially unclear relationship between inflammatory processes and AAA progression.
Collapse
Affiliation(s)
- H Jalalzadeh
- Department of Vascular Surgery, Academic Medical Center, Amsterdam, The Netherlands
| | - R Indrakusuma
- Department of Vascular Surgery, Academic Medical Center, Amsterdam, The Netherlands
| | - R N Planken
- Department of Radiology, Academic Medical Center, Amsterdam, The Netherlands
| | - D A Legemate
- Department of Vascular Surgery, Academic Medical Center, Amsterdam, The Netherlands
| | - M J W Koelemay
- Department of Vascular Surgery, Academic Medical Center, Amsterdam, The Netherlands
| | - R Balm
- Department of Vascular Surgery, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Angiotensin II-induced TLR4 mediated abdominal aortic aneurysm in apolipoprotein E knockout mice is dependent on STAT3. J Mol Cell Cardiol 2015; 87:160-70. [PMID: 26299839 DOI: 10.1016/j.yjmcc.2015.08.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 08/12/2015] [Accepted: 08/14/2015] [Indexed: 11/22/2022]
Abstract
Abdominal Aortic Aneurysm (AAA) is a major cause of mortality and morbidity in men over 65 years of age. Male apolipoprotein E knockout (ApoE(-/-)) mice infused with angiotensin II (AngII) develop AAA. Although AngII stimulates both JAK/STAT and Toll-like receptor 4 (TLR4) signaling pathways, their involvement in AngII mediated AAA formation is unclear. Here we used the small molecule STAT3 inhibitor, S3I-201, the TLR4 inhibitor Eritoran and ApoE(-/-)TLR4(-/-) mice to evaluate the interaction between STAT3 and TLR4 signaling in AngII-induced AAA formation. ApoE(-/-) mice infused for 28 days with AngII developed AAAs and increased STAT3 activation and TLR4 expression. Moreover, AngII increased macrophage infiltration and the ratio of M1 (pro-inflammatory)/M2 (healing) macrophages in aneurysmal tissue as early as 7-10 days after AngII infusion. STAT3 inhibition with S3I-201 decreased the incidence and severity of AngII-induced AAA formation and decreased MMP activity and the ratio of M1/M2 macrophages. Furthermore, AngII-mediated AAA formation, MMP secretion, STAT3 phosphorylation and the ratio of M1/M2 macrophages were markedly decreased in ApoE(-/-)TLR4(-/-) mice, and in Eritoran-treated ApoE(-/-) mice. TLR4 and pSTAT3 levels were also increased in human aneurysmal tissue. These data support a role of pSTAT3 in TLR4 dependent AAA formation and possible therapeutic roles for TLR4 and/or STAT3 inhibition in AAA.
Collapse
|
22
|
Zhou Y, Wu W, Lindholt JS, Sukhova GK, Libby P, Yu X, Shi GP. Regulatory T cells in human and angiotensin II-induced mouse abdominal aortic aneurysms. Cardiovasc Res 2015; 107:98-107. [PMID: 25824145 PMCID: PMC4560044 DOI: 10.1093/cvr/cvv119] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 03/07/2015] [Accepted: 03/19/2015] [Indexed: 01/02/2023] Open
Abstract
AIMS Regulatory T cells (Tregs) protect mice from angiotensin II (Ang-II)-induced abdominal aortic aneurysms (AAA). This study tested whether AAA patients are Treg-insufficient and the Treg molecular mechanisms that control AAA pathogenesis. METHODS AND RESULTS ELISA determined the Foxp3 concentration in blood cell lysates from 485 AAA patients and 204 age- and sex-matched controls. AAA patients exhibited lower blood cell Foxp3 expression than controls (P < 0.0001). Pearson's correlation test demonstrated a significant but negative correlation between Foxp3 and AAA annual expansion rate before (r = -0.147, P = 0.007) and after (r = -0.153, P = 0.006) adjustment for AAA risk factors. AAA in apolipoprotein E-deficient (Apoe(-/-)) mice that received different doses of Ang-II exhibited a negative correlation of lesion Foxp3(+) Treg numbers with AAA size (r = -0.883, P < 0.0001). Adoptive transfer of Tregs from wild-type (WT) and IL10-deficient (Il10(-/-)) mice increased AAA lesion Treg content, but only WT mice Tregs reduced AAA size, AAA incidence, blood pressure, lesion macrophage and CD4(+) and CD8(+) T-cell accumulation, and angiogenesis with concurrent increase of lesion collagen content. Both AAA lesion immunostaining and plasma ELISA demonstrated that adoptive transfer of WT Tregs, but not Il10(-/-) Tregs, reduced the expression of MCP-1. In vitro cell culture and aortic ring assay demonstrated that only Tregs from WT mice, but not those from Il10(-/-) mice, reduced macrophage MCP-1 secretion, macrophage and vascular cell protease expression and activity, and aortic ring microvessel formation. CONCLUSION This study supports a protective role of Tregs in human and experimental AAA by releasing IL10 to suppress inflammatory cell chemotaxis, arterial wall remodelling, and angiogenesis.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA Department of Nephrology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wenxue Wu
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA College of Veterinary Medicine, China Agriculture University, Beijing 100193, China
| | - Jes S Lindholt
- Elitary Research Centre of Individualized Medicine in Arterial Diseases, Department of Cardiothoracic and Vascular Surgery, University Hospital of Odense, Odense DK-5000, Denmark
| | - Galina K Sukhova
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Peter Libby
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Xueqing Yu
- Department of Nephrology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| |
Collapse
|
23
|
McBride OMB, Berry C, Burns P, Chalmers RTA, Doyle B, Forsythe R, Garden OJ, Goodman K, Graham C, Hoskins P, Holdsworth R, MacGillivray TJ, McKillop G, Murray G, Oatey K, Robson JMJ, Roditi G, Semple S, Stuart W, van Beek EJR, Vesey A, Newby DE. MRI using ultrasmall superparamagnetic particles of iron oxide in patients under surveillance for abdominal aortic aneurysms to predict rupture or surgical repair: MRI for abdominal aortic aneurysms to predict rupture or surgery-the MA(3)RS study. Open Heart 2015; 2:e000190. [PMID: 25932334 PMCID: PMC4410138 DOI: 10.1136/openhrt-2014-000190] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/18/2015] [Indexed: 11/16/2022] Open
Abstract
Introduction Population screening for abdominal aortic aneurysms (AAA) halves the associated mortality and has led to the establishment of national screening programmes. Prediction of aneurysm growth and rupture is challenging and currently relies on serial diameter measurements with ultrasound. Recently, a novel MRI-based technique using ultrasmall superparamagnetic particles of iron oxide (USPIO) has demonstrated considerable promise as a method of identifying aneurysm inflammation and expansion. Methods and analysis The MA3RS study is a prospective observational multicentre cohort study of 350 patients with AAA in three centres across Scotland. All participants will undergo MRI with USPIO and aneurysm expansion will be measured over 2 years with CT in addition to standard clinical ultrasound surveillance. The relationship between mural USPIO uptake and subsequent clinical outcomes, including expansion, rupture and repair, will be evaluated and used to determine whether the technique augments standard risk prediction markers. To ensure adequate sensitivity to answer the primary question, we need to observe 130 events (composite of rupture or repair) with an estimated event rate of 41% over 2 years of follow-up. The MA3RS study is currently recruiting and expects to report in 2017. Discussion This is the first study to evaluate the use of USPIO-enhanced MRI to provide additional information to aid risk prediction models in patients with AAA. If successful, this study will lay the foundation for a large randomised controlled trial targeted at applying this technique to determine clinical management. Trial registration number Current Controlled Trials: ISRCTN76413758.
Collapse
Affiliation(s)
- Olivia M B McBride
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Colin Berry
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow , Glasgow , UK
| | - Paul Burns
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Roderick T A Chalmers
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Barry Doyle
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Rachael Forsythe
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - O James Garden
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Kirsteen Goodman
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Catriona Graham
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Peter Hoskins
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | | | - Thomas J MacGillivray
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Graham McKillop
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Gordon Murray
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Katherine Oatey
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Jennifer M J Robson
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow , Glasgow , UK
| | - Giles Roditi
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow , Glasgow , UK
| | - Scott Semple
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Wesley Stuart
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow , Glasgow , UK
| | - Edwin J R van Beek
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - Alex Vesey
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| | - David E Newby
- British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science , Edinburgh , UK
| |
Collapse
|
24
|
Trachet B, Fraga-Silva RA, Piersigilli A, Tedgui A, Sordet-Dessimoz J, Astolfo A, Van der Donckt C, Modregger P, Stampanoni MFM, Segers P, Stergiopulos N. Dissecting abdominal aortic aneurysm in Ang II-infused mice: suprarenal branch ruptures and apparent luminal dilatation. Cardiovasc Res 2014; 105:213-22. [DOI: 10.1093/cvr/cvu257] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
25
|
Esfahani DR, Viswanathan V, Alaraj A. Nanoparticles and stem cells - has targeted therapy for aneurysms finally arrived? Neurol Res 2014; 37:269-77. [PMID: 25082670 DOI: 10.1179/1743132814y.0000000435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Until recently, endovascular management of intracranial aneurysms has focused on mechanical and hemodynamic aspects: characterizing aneurysm morphology by angiogram, mechanical obstruction by detachable coils, and flow diversion with endovascular stents. Although now common practice, these interventions only ward off aneurysm rupture. The source of the problem, disease of the vessel wall itself, remains. New imaging technology and treatment modalities, however, are offering great promise to the field. In this review, we outline several new developments in the recent literature and pose potential adaptations toward cerebral aneurysms using them. The incidence, presentation, and contemporary endovascular treatment for aneurysms are first reviewed to lay the groundwork for new adaptations. Nanoparticles, including ultrasmall supraparagmenetic iron oxide particles (USPIOs), are next explored as a novel mechanism of predicting aneurysm wall instability and as an agent themselves for aneurysm occlusion. Cellular transplant grafts, bone marrow-derived stem cells (BM-MSCs), and endothelial progenitor cells (EPCs) are then investigated, with the role of cellular differentiation, chemokine secretion, and integration into the injured vascular wall receiving particular emphasis. Several promising translational papers are next discussed, with review of multiple studies that show benefit in aneurysm treatment and endovascular stenting using these agents as adjuncts. We next adapt these research findings into several potential applications we feel may be promising directions for the aspiring researcher. These new treatments may one day strengthen the arsenal of the endovascular neurosurgeon.
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Functional and molecular aortic imaging has shown great promise for evaluation of aortic disease, and may soon augment conventional assessment of aortic dimensions for the clinical management of patients. RECENT FINDINGS A range of imaging techniques is available for evaluation of patients with aortic disease. Magnetic resonance blood flow imaging can identify atherosclerosis prone aortic regions and may be useful for predicting aneurysm growth. Computational modeling can demonstrate significant differences in wall stress between abdominal aortic aneurysms of similar size and may better predict rupture than diameter alone. Metabolic imaging with fluorodeoxyglucose-PET [(FDG)-PET] can identify focal aortic wall inflammation that may portend rapid progression of disease. Molecular imaging with probes that target collagen and elastin can directly exhibit changes in the vessel wall associated with disease. SUMMARY The complexity of aortic disease is more fully revealed with new functional imaging techniques than with conventional anatomic analysis alone. This may better inform surveillance imaging regimens, medical management and decisions regarding early intervention for aortic disease.
Collapse
|
27
|
Golestani R, Sadeghi MM. Emergence of molecular imaging of aortic aneurysm: implications for risk stratification and management. J Nucl Cardiol 2014; 21:251-67; quiz 268-70. [PMID: 24381115 PMCID: PMC3991015 DOI: 10.1007/s12350-013-9845-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 11/19/2013] [Indexed: 12/21/2022]
Abstract
Imaging cellular and molecular processes associated with aneurysm expansion, dissection, and rupture can potentially transform the management of patients with thoracic and abdominal aortic aneurysm. Here, we review recent advances in molecular imaging of aortic aneurysm, focusing on imaging modalities with the greatest potential for clinical translation and application, PET, SPECT, and MRI. Inflammation (e.g., with (18)F-FDG, nanoparticles) and matrix remodeling (e.g., with matrix metalloproteinase-targeted tracers) are highlighted as promising targets for molecular imaging of aneurysm. Potential alternative or complementary approaches to molecular imaging for aneurysm risk stratification are briefly discussed.
Collapse
Affiliation(s)
- Reza Golestani
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
28
|
Siu KL, Miao XN, Cai H. Recoupling of eNOS with folic acid prevents abdominal aortic aneurysm formation in angiotensin II-infused apolipoprotein E null mice. PLoS One 2014; 9:e88899. [PMID: 24558445 PMCID: PMC3928303 DOI: 10.1371/journal.pone.0088899] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 01/13/2014] [Indexed: 01/31/2023] Open
Abstract
We have previously shown that eNOS uncoupling mediates abdominal aortic aneurysm (AAA) formation in hph-1 mice. In the present study we examined whether recoupling of eNOS prevents AAA formation in a well-established model of Angiotensin II-infused apolipoprotein E (apoE) null mice by targeting some common pathologies of AAA. Infusion of Ang II resulted in a 92% incidence rate of AAA in the apoE null animals. In a separate group, animals were treated orally with folic acid (FA), which is known to recouple eNOS through augmentation of dihydrofolate reductase (DHFR) function. This resulted in a reduction of AAA rate to 19.5%. Imaging with ultrasound showed that FA markedly inhibited expansion of abdominal aorta. FA also abolished elastin breakdown and macrophage infiltration in the AAA animals. The eNOS uncoupling activity, assessed by L-NAME-sensitive superoxide production, was minimal at baseline but greatly exaggerated with Ang II infusion, which was completely attenuated by FA. This was accompanied by markedly improved tetrahydrobiopterin and nitric oxide bioavailability. Furthermore, the expression and activity of DHFR was decreased in Ang II-infused apoE null mice specifically in the endothelial cells, while FA administration resulted in its recovery. Taken together, these data further establish a significant role of uncoupled eNOS in mediating AAA formation, and a universal efficacy of FA in preventing AAA formation via restoration of DHFR to restore eNOS function.
Collapse
MESH Headings
- Administration, Oral
- Angiotensin II/pharmacology
- Animals
- Aorta, Abdominal/diagnostic imaging
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/diagnostic imaging
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Biopterins/analogs & derivatives
- Biopterins/metabolism
- Folic Acid/administration & dosage
- Folic Acid/pharmacology
- Male
- Mice
- Mice, Knockout
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Radiography
- Superoxides/metabolism
- Tetrahydrofolate Dehydrogenase/metabolism
- Ultrasonography
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Kin Lung Siu
- Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Xiao Niu Miao
- Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Hua Cai
- Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
29
|
Ji K, Zhang Y, Jiang F, Qian L, Guo H, Hu J, Liao L, Tang J. Exploration of the mechanisms by which 3,4-benzopyrene promotes angiotensin II-induced abdominal aortic aneurysm formation in mice. J Vasc Surg 2013; 59:492-9. [PMID: 23676189 DOI: 10.1016/j.jvs.2013.03.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 02/26/2013] [Accepted: 03/04/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE This study examined the influence of 3,4-benzopyrene (BaP), a compound found in cigarette smoke, on the formation of angiotensin II (Ang II)-induced abdominal aortic aneurysm (AAA) formation in mice and the underlying mechanisms. METHODS C57/B6n mice were divided into four groups. The control group received a weekly intraperitoneal injection of medium-chain triglycerides. The Ang II group received a daily Ang II infusion (0.72 mg/kg) and a weekly intraperitoneal injection of medium-chain triglycerides. The Ang II/BaP group received a daily Ang II infusion (0.72 mg/kg) and a weekly intraperitoneal BaP injection (10 mg/kg, dissolved in medium-chain triglycerides). The BaP group received a weekly intraperitoneal BaP injection (10 mg/kg). After 5 weeks, abdominal aortic diameter was determined. Aortic tissues underwent hematoxylin and eosin, Masson, and immunochemistry staining for evaluation of vascular wall structure, collagen, macrophage infiltration, matrix metalloproteinases (MMPs), and apoptosis. RESULTS The Ang II infusion and BaP injection induced AAAs in 41.67% of mice vs 25% in the Ang II group (P < .05). The average aortic diameter increased in the Ang II/BaP group compared with the Ang II group (1.40 ± 0.25 vs 1.2 ± 0.23 mm; P < .05). Average aortic muscular cell apoptosis was higher in the Ang II/BaP group (31% ± 12%) than in the Ang II (19% ± 5%; P < .05) or BaP groups (23% ± 4%; P < .05). Aortic macrophage infiltration and expression of MMP-2, MMP-9, MMP-12, and nuclear factor-κB increased (0.56 ± 0.12, 0.47 ± 0.13, 0.49 ± 0.14, 0.49 ± 0.11, and 0.42 ± 0.12, respectively) in the Ang II/BaP group compared with the Ang II group (0.27 ± 0.08, 0.25 ± 0.06, 0.24 ± 0.09, 0.24 ± 0.09, and 0.23 ± 0.06, respectively; P < .05 for all). CONCLUSIONS BaP promotes Ang II-induced AAA formation in mice via elevating infiltration of macrophages, activating nuclear factor-κB, upregulating the expression of MMP-2, MMP-9, and MMP-12, and increasing the apoptosis of vascular muscle cells in its synergistic effect with Ang II in aortic wall.
Collapse
Affiliation(s)
- Kangting Ji
- Department of Cardiology, The Second Hospital Affiliated to Wenzhou Medical College, Wenzhou, China
| | - Yong Zhang
- Department of Cardiology, The Second Hospital Affiliated to Wenzhou Medical College, Wenzhou, China
| | - Fengchun Jiang
- Department of Cardiology, The Second Hospital Affiliated to Wenzhou Medical College, Wenzhou, China
| | - Lu Qian
- Department of Cardiology, The Second Hospital Affiliated to Wenzhou Medical College, Wenzhou, China
| | - Huihui Guo
- Department of Cardiology, The Second Hospital Affiliated to Wenzhou Medical College, Wenzhou, China
| | - Jianjian Hu
- Department of Cardiology, The Second Hospital Affiliated to Wenzhou Medical College, Wenzhou, China
| | - Lianming Liao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.
| | - Jifei Tang
- Department of Cardiology, The Second Hospital Affiliated to Wenzhou Medical College, Wenzhou, China.
| |
Collapse
|
30
|
Molecular imaging of experimental abdominal aortic aneurysms. ScientificWorldJournal 2013; 2013:973150. [PMID: 23737735 PMCID: PMC3655677 DOI: 10.1155/2013/973150] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 03/19/2013] [Indexed: 11/18/2022] Open
Abstract
Current laboratory research in the field of abdominal aortic aneurysm (AAA) disease often utilizes small animal experimental models induced by genetic manipulation or chemical application. This has led to the use and development of multiple high-resolution molecular imaging modalities capable of tracking disease progression, quantifying the role of inflammation, and evaluating the effects of potential therapeutics. In vivo imaging reduces the number of research animals used, provides molecular and cellular information, and allows for longitudinal studies, a necessity when tracking vessel expansion in a single animal. This review outlines developments of both established and emerging molecular imaging techniques used to study AAA disease. Beyond the typical modalities used for anatomical imaging, which include ultrasound (US) and computed tomography (CT), previous molecular imaging efforts have used magnetic resonance (MR), near-infrared fluorescence (NIRF), bioluminescence, single-photon emission computed tomography (SPECT), and positron emission tomography (PET). Mouse and rat AAA models will hopefully provide insight into potential disease mechanisms, and the development of advanced molecular imaging techniques, if clinically useful, may have translational potential. These efforts could help improve the management of aneurysms and better evaluate the therapeutic potential of new treatments for human AAA disease.
Collapse
|
31
|
Eagleton MJ. Inflammation in abdominal aortic aneurysms: cellular infiltrate and cytokine profiles. Vascular 2012; 20:278-83. [DOI: 10.1258/vasc.2011.201207] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Abdominal aortic aneurysm (AAA) pathogenesis occurs as a result of the altered homeostasis of the aortic vessel wall structural proteins. This results in weakening, and subsequent expansion, of the aorta leading to aneurysm formation. Multiple mechanisms are involved in this process, including genetic abnormalities, biomechanical wall stress, apoptosis, and proteolytic degradation of the aortic wall. One key hallmark of this pathology, which orchestrates the interaction of the various pathologic processes, is inflammation. The inflammatory process is characterized by the infiltration of a variety of cells, which leads to the upregulation of multiple cytokines. The balance of the cellular type and resultant cytokine milieu determines the ultimate fate of the aortic wall – healing, atherosclerosis or aneurysm formation. This review highlights some of the known cellular and cytokine inflammatory events that are involved in aortic aneurysm formation.
Collapse
Affiliation(s)
- Matthew J Eagleton
- Department of Vascular Surgery, Cleveland Clinic, Lerner College of Medicine-CWRU, Cleveland, OH 44195, USA
| |
Collapse
|
32
|
Bartoli MA, Kober F, Cozzone P, Thompson RW, Alessi MC, Bernard M. In vivo assessment of murine elastase-induced abdominal aortic aneurysm with high resolution magnetic resonance imaging. Eur J Vasc Endovasc Surg 2012; 44:475-81. [PMID: 22939881 DOI: 10.1016/j.ejvs.2012.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 08/01/2012] [Indexed: 11/30/2022]
Abstract
OBJECTIVES There are, to date, no published non-invasive or longitudinal studies performed in mice to measure aortic diameter and wall thickness in an elastase-induced abdominal aortic aneurysm. This MRI study at 11.75 T aimed at evaluating the reliability of longitudinal in vivo aortic diameter and wall thickness measurements in this particular model. METHODS Adult male C57BL/6 mice underwent transient elastase or heat-inactivated elastase perfusion (controls). Aortic dilatation was measured before, during and immediately after elastase perfusion, and again 14 days after, with a calibrated ocular grid. MRI was performed just before initial surgery and at day 14 before harvest using an 11.75 T MR microscopy imager. RESULTS Aortic diameter was significantly greater in elastase-perfused mice compared to controls as measured by optic grid (1.150 ± 0.153 mm vs 0.939 ± 0.07 mm, P = 0.038) and according to MRI measurement of the outer diameter on spin echo images (1.203 ± 0.105 mm vs 1070 ± 0.048 mm, P = 0.0067). Aortic wall thickness was found to be significantly increased in elastase-perfused mice at day 14. CONCLUSIONS This study demonstrates in the mouse elastase-induced aneurysm model that characterization of aneurysm development by its inner and outer vessel diameter and vessel wall thickness can be carried out longitudinally using high resolution MRI without significant mortality.
Collapse
Affiliation(s)
- M A Bartoli
- Aix-Marseille université, CNRS, CRMBM UMR, Marseille, France.
| | | | | | | | | | | |
Collapse
|
33
|
Reeps C, Bundschuh RA, Pellisek J, Herz M, van Marwick S, Schwaiger M, Eckstein HH, Nekolla SG, Essler M. Quantitative assessment of glucose metabolism in the vessel wall of abdominal aortic aneurysms: correlation with histology and role of partial volume correction. Int J Cardiovasc Imaging 2012; 29:505-12. [PMID: 22772434 DOI: 10.1007/s10554-012-0090-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 06/27/2012] [Indexed: 11/26/2022]
Abstract
Inflammatory-proteolytic processes in the vessel wall are essential in the pathophysiology of abdominal aortic aneurysm (AAA). It has been demonstrated that, (18)F-FDG-PET/CT may be useful for detection of pathological wall metabolism and therefore risk stratification. Quantification of the FDG-uptake in AAA wall is hampered by partial-volume (PV)-effects. For correction and accurate quantitative (18)F-FDG-uptake analysis we designed and validated a novel IDL-based software in correlation to phantom studies, histopathology and clinical presentation of AAA patients. For in vivo studies 23 patients with symptomatic and asymptomatic AAA underwent (18)F-FDG-PET/CT before surgery. In areas with (18)F-FDG-uptake the maximum and mean standardized uptake values in the vessel wall with (PVC-SUV(max), PVC-SUV(mean)) and without (SUV(max), SUV(mean)) PV-correction were determined. Results were correlated with clinical presentation, corresponding macrophage-infiltration and MMP-2- and -9-expression in surgical specimens. In patients, SUV(max), SUV(mean) as well as PVC-SUV(max) or PVC-SUV(mean) enabled a highly significant (p < 0.005) discrimination of symptomatic and asymptomatic AAA. Uncorrected and corrected SUVs showed comparable correlations with macrophage-infiltration and MMP-9 expression. No correlation of (18)F-FDG-uptake and MMP-2 was found. In vivo correlations of detected FDG-uptake with clinical and histological results showed comparable results for corrected and uncorrected SUVs. PV-correction is not mandatory for qualitative clinical assessment of glucose metabolism in the vessel wall of AAA-patients but may be necessary to establish quantitative cut off values to stratify patients for aneurysm repair.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Aorta, Abdominal/diagnostic imaging
- Aorta, Abdominal/enzymology
- Aorta, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/diagnostic imaging
- Aortic Aneurysm, Abdominal/enzymology
- Aortic Aneurysm, Abdominal/metabolism
- Female
- Fluorodeoxyglucose F18/metabolism
- Humans
- Image Interpretation, Computer-Assisted
- Immunohistochemistry
- Linear Models
- Macrophages/metabolism
- Male
- Matrix Metalloproteinase 2/analysis
- Matrix Metalloproteinase 9/analysis
- Middle Aged
- Multimodal Imaging/instrumentation
- Phantoms, Imaging
- Positron-Emission Tomography
- Predictive Value of Tests
- Radiopharmaceuticals/metabolism
- Reproducibility of Results
- Software Validation
- Tomography, X-Ray Computed
Collapse
Affiliation(s)
- Christian Reeps
- Klinik für Gefäßchirurgie, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Yao Y, Wang Y, Zhang Y, Li Y, Sheng Z, Wen S, Ma G, Liu N, Fang F, Teng GJ. In vivo imaging of macrophages during the early-stages of abdominal aortic aneurysm using high resolution MRI in ApoE mice. PLoS One 2012; 7:e33523. [PMID: 22448249 PMCID: PMC3308989 DOI: 10.1371/journal.pone.0033523] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 02/15/2012] [Indexed: 01/15/2023] Open
Abstract
Background Angiotensin II (ANG II) promotes vascular inflammation and induces abdominal aortic aneurysm (AAA) in hyperlipidemic apolipoprotein E knock-out (apoE−/−) mice. The aim of the present study was to detect macrophage activities in an ANG II-induced early-stage AAA model using superparamagnetic iron oxide (SPIO) as a marker. Methodology/Principal Findings Twenty-six male apoE−/− mice received saline or ANG II (1000 or 500 ng/kg/min) infusion for 14 days. All animals underwent MRI scanning following administration of SPIO with the exception of three mice in the 1000 ng ANG II group, which were scanned without SPIO administration. MR imaging was performed using black-blood T2 to proton density -weighted multi-spin multi-echo sequence. In vivo MRI measurement of SPIO uptake and abdominal aortic diameter were obtained. Prussian blue, CD68,α-SMC and MAC3 immunohistological stains were used for the detection of SPIO, macrophages and smooth muscle cells. ANG II infusion with 1000 ng/kg/min induced AAA in all of the apoE−/− mice. ANG II infusion exhibited significantly higher degrees of SPIO uptake, which was detected using MRI as a distinct loss of signal intensity. The contrast-to-noise ratio value decreased in proportion to an increase in the number of iron-laden macrophages in the aneurysm. The aneurysmal vessel wall in both groups of ANG II treated mice contained more iron-positive macrophages than saline-treated mice. However, the presence of cells capable of phagocytosing haemosiderin in mural thrombi also induced low-signal-intensities via MRI imaging. Conclusions/Significance SPIO is taken up by macrophages in the shoulder and the outer layer of AAA. This alters the MRI signaling properties and can be used in imaging inflammation associated with AAA. It is important to compare images of the aorta before and after SPIO injection.
Collapse
Affiliation(s)
- Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Yuanyuan Wang
- Jiangsu Key Lab of Molecular and Function Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Yi Zhang
- Jiangsu Key Lab of Molecular and Function Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Yefei Li
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Zulong Sheng
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Song Wen
- Jiangsu Key Lab of Molecular and Function Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Naifeng Liu
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Fang Fang
- Jiangsu Key Lab of Molecular and Function Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Gao-Jun Teng
- Jiangsu Key Lab of Molecular and Function Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
- * E-mail:
| |
Collapse
|
35
|
Miyama N, Dua MM, Schultz GM, Kosuge H, Terashima M, Pisani LJ, Dalman RL, McConnell MV. Bioluminescence and Magnetic Resonance Imaging of Macrophage Homing to Experimental Abdominal Aortic Aneurysms. Mol Imaging 2012. [DOI: 10.2310/7290.2011.00033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Macrophage infiltration is a prominent feature of abdominal aortic aneurysm (AAA) progression. We used a combined imaging approach with bioluminescence (BLI) and magnetic resonance imaging (MRI) to study macrophage homing and accumulation in experimental AAA disease. Murine AAAs were created via intra-aortic infusion of porcine pancreatic elastase. Mice were imaged over 14 days after injection of prepared peritoneal macrophages. For BLI, macrophages were from transgenic mice expressing luciferase. For MRI, macrophages were labeled with iron oxide particles. Macrophage accumulation during aneurysm progression was observed by in situ BLI and by in vivo 7T MRI. Mice were sacrificed after imaging for histologic analysis. In situ BLI ( n = 32) demonstrated high signal in the AAA by days 7 and 14, which correlated significantly with macrophage number and aortic diameter. In vivo 7T MRI ( n = 13) at day 14 demonstrated T2* signal loss in the AAA and not in sham mice. Immunohistochemistry and Prussian blue staining confirmed the presence of injected macrophages in the AAA. BLI and MRI provide complementary approaches to track macrophage homing and accumulation in experimental AAAs. Similar dual imaging strategies may aid the study of AAA biology and the evaluation of novel therapies.
Collapse
Affiliation(s)
- Noriyuki Miyama
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Monica M. Dua
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Geoffrey M. Schultz
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Hisanori Kosuge
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Masahiro Terashima
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Laura J. Pisani
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Ronald L. Dalman
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Michael V. McConnell
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
36
|
Xu JM, Shi GP. Emerging role of mast cells and macrophages in cardiovascular and metabolic diseases. Endocr Rev 2012; 33:71-108. [PMID: 22240242 PMCID: PMC3365842 DOI: 10.1210/er.2011-0013] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 09/12/2011] [Indexed: 12/11/2022]
Abstract
Mast cells are essential in allergic immune responses. Recent discoveries have revealed their direct participation in cardiovascular diseases and metabolic disorders. Although more sophisticated mechanisms are still unknown, data from animal studies suggest that mast cells act similarly to macrophages and other inflammatory cells and contribute to human diseases through cell-cell interactions and the release of proinflammatory cytokines, chemokines, and proteases to induce inflammatory cell recruitment, cell apoptosis, angiogenesis, and matrix protein remodeling. Reduced cardiovascular complications and improved metabolic symptoms in animals receiving over-the-counter antiallergy medications that stabilize mast cells open another era of mast cell biology and bring new hope to human patients suffering from these conditions.
Collapse
Affiliation(s)
- Jia-Ming Xu
- Department of Medicine, Nanfang Hospital and Southern Medical University, Guangzhou 510515, China
| | | |
Collapse
|
37
|
Vérollet C, Charrière GM, Labrousse A, Cougoule C, Le Cabec V, Maridonneau-Parini I. Extracellular proteolysis in macrophage migration: Losing grip for a breakthrough. Eur J Immunol 2011; 41:2805-13. [DOI: 10.1002/eji.201141538] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
38
|
Choudhury RP. Macrophage detection in aortic aneurysm: the heat is on. Arterioscler Thromb Vasc Biol 2011; 31:723-4. [PMID: 21411752 DOI: 10.1161/atvbaha.111.223297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
39
|
Richards JMJ, Semple SI, MacGillivray TJ, Gray C, Langrish JP, Williams M, Dweck M, Wallace W, McKillop G, Chalmers RTA, Garden OJ, Newby DE. Abdominal aortic aneurysm growth predicted by uptake of ultrasmall superparamagnetic particles of iron oxide: a pilot study. Circ Cardiovasc Imaging 2011; 4:274-81. [PMID: 21304070 DOI: 10.1161/circimaging.110.959866] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Abdominal aortic aneurysms are a major cause of death. Prediction of aneurysm expansion and rupture is challenging and currently relies on the simple measure of aneurysm diameter. Using MRI, we aimed to assess whether areas of cellular inflammation correlated with the rate of abdominal aortic aneurysm expansion. METHODS AND RESULTS Stable patients (n=29; 27 male; age, 70±5 years) with asymptomatic abdominal aortic aneurysms (4.0 to 6.6 cm) were recruited from a surveillance program and imaged using a 3-T MRI scanner before and 24 to 36 hours after administration of ultrasmall superparamagnetic particles of iron oxide (USPIO). The change in T2* value on T2*-weighted imaging was used to detect accumulation of USPIO within the abdominal aortic aneurysm. Histological examination of aneurysm tissue confirmed colocalization and uptake of USPIO in areas with macrophage infiltration. Patients with distinct mural uptake of USPIO had a 3-fold higher growth rate (n=11, 0.66 cm/y; P=0.020) than those with no (n=6, 0.22 cm/y) or nonspecific USPIO uptake (n=8, 0.24 cm/y) despite having similar aneurysm diameters (5.4±0.6, 5.1±0.5, and 5.0±0.5 cm, respectively; P>0.05). In 1 patient with an inflammatory aneurysm, there was a strong and widespread uptake of USPIO extending beyond the aortic wall. CONCLUSIONS Uptake of USPIO in abdominal aortic aneurysms identifies cellular inflammation and appears to distinguish those patients with more rapidly progressive abdominal aortic aneurysm expansion. This technique holds major promise as a new method of risk-stratifying patients with abdominal aortic aneurysms that extends beyond the simple anatomic measure of aneurysm diameter. Clinical Trial Registration- URL: http://www.clinicaltrials.gov. Unique identifier: NCT00794092.
Collapse
Affiliation(s)
- Jennifer M J Richards
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|