1
|
Ye L, Chang CC, Li Q, Tintut Y, Hsu JJ. Advanced Imaging Techniques for Atherosclerosis and Cardiovascular Calcification in Animal Models. J Cardiovasc Dev Dis 2024; 11:410. [PMID: 39728300 DOI: 10.3390/jcdd11120410] [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: 10/22/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
The detection and assessment of atherosclerosis and cardiovascular calcification can inform risk stratification and therapies to reduce cardiovascular morbidity and mortality. In this review, we provide an overview of current and emerging imaging techniques for assessing atherosclerosis and cardiovascular calcification in animal models. Traditional imaging modalities, such as computed tomography (CT) and magnetic resonance imaging (MRI), offer non-invasive approaches of visualizing atherosclerotic calcification in vivo; integration of these techniques with positron emission tomography (PET) imaging adds molecular imaging capabilities, such as detection of metabolically active microcalcifications with 18F-sodium fluoride. Photoacoustic imaging provides high contrast that enables in vivo evaluation of plaque composition, yet this method is limited by optical penetration depth. Light-sheet fluorescence microscopy provides high-resolution, three-dimensional imaging of cardiovascular structures and has been used for ex vivo assessment of atherosclerotic calcification, but its limited tissue penetration and requisite complex sample preparation preclude its use in vivo to evaluate cardiac tissue. Overall, with these evolving imaging tools, our understanding of cardiovascular calcification development in animal models is improving, and the combination of traditional imaging techniques with emerging molecular imaging modalities will enhance our ability to investigate therapeutic strategies for atherosclerotic calcification.
Collapse
Affiliation(s)
- Lifang Ye
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China
- Department of Medicine, University of California, 650 Charles E Young Dr. S, Center for Health Sciences, Room A2-237, Los Angeles, CA 90095, USA
| | - Chih-Chiang Chang
- Department of Medicine, University of California, 650 Charles E Young Dr. S, Center for Health Sciences, Room A2-237, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Qian Li
- Department of Medicine, University of California, 650 Charles E Young Dr. S, Center for Health Sciences, Room A2-237, Los Angeles, CA 90095, USA
| | - Yin Tintut
- Department of Medicine, University of California, 650 Charles E Young Dr. S, Center for Health Sciences, Room A2-237, Los Angeles, CA 90095, USA
- Department of Physiology, University of California, Los Angeles, CA 90095, USA
- Department of Orthopedic Surgery, University of California, Los Angeles, CA 90404, USA
| | - Jeffrey J Hsu
- Department of Medicine, University of California, 650 Charles E Young Dr. S, Center for Health Sciences, Room A2-237, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Department of Medicine, Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, CA 90073, USA
| |
Collapse
|
2
|
Ji C, Wang X, Xue B, Li S, Li J, Qiao B, Du J, Yin M, Wang Y. A fluorescent nano vector for early diagnosis and enhanced Interleukin-33 therapy of thoracic aortic dissection. Biomaterials 2023; 293:121958. [PMID: 36566550 DOI: 10.1016/j.biomaterials.2022.121958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
Thoracic aortic dissection (TAD) is the most devastating complication of vascular disease. The accuracy of the clinical diagnosis and treatment of TAD at the early stage is still limited. Herein, we report a nano-delivery strategy for early diagnosis and the first case of interleukin-33 (IL-33) based therapy for the effective intervention of TAD. A targeted fluorescent nano vector (FNV) is designed to co-assemble with IL-33, which protects IL-33 and prolongs its half-life. With specific targeting ability to the thoracic aorta, FNV can diagnose TAD at its early stage through fluorescent imaging. FNV@IL-33 nanocomplex presents better therapeutic effects on mice TAD progression compared with that of IL-33 alone by reducing smooth muscle apoptosis. Administration of FNV@IL-33 two weeks before onset, the development of TAD is greatly intervened. Our study provides a novel approach for early diagnosis and effective IL-33 therapy of TAD, which opens attractive opportunities for clinical prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Chendong Ji
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xue Wang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), And Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, 100029, Beijing, China
| | - Bingjie Xue
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), And Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, 100029, Beijing, China
| | - Shuolin Li
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Jianhao Li
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Bokang Qiao
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), And Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, 100029, Beijing, China
| | - Jie Du
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), And Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, 100029, Beijing, China.
| | - Meizhen Yin
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China.
| | - Yuan Wang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), And Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, 100029, Beijing, China.
| |
Collapse
|
3
|
Cilengitide Inhibits Neovascularization in a Rabbit Abdominal Aortic Plaque Model by Impairing the VEGF Signaling. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5954757. [PMID: 34888383 PMCID: PMC8651393 DOI: 10.1155/2021/5954757] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 11/28/2022]
Abstract
Background Cilengitide is a selective αvβ3 and αvβ5 integrin inhibitor. We sought to investigate the effect of cilengitide on the neovascularization of abdominal aortic plaques in rabbits and explore its underlying antiangiogenic mechanism on human umbilical vein endothelial cells (HUVECs). Materials and Methods For the in vivo experiment, the abdominal aortic plaque model of rabbits was established and injected with different doses of cilengitide or saline for 14 consecutive days. Conventional ultrasound (CUS) and contrast-enhanced ultrasound (CEUS) were applied to measure the vascular structure and blood flow parameters. CD31 immunofluorescence staining was performed for examining neovascularization. Relative expressions of vascular endothelial growth factor (VEGF) and integrin of the plaque were determined. For in vitro experiments, HUVECs were tested for proliferation, migration, apoptosis, and tube formation in the presence of different doses of cilengitide. Relative expressions of VEGF, integrin, and Ras/ERK/AKT signaling pathways were determined for the exploration of underlying mechanism. Results CEUS showed modestly increased size and eccentricity index (EI) of plaques in the control group. Different degrees of reduced size and EI of plaques were observed in two cilengitide treatment groups. The expressions of VEGF and integrin in the plaque were inhibited after 14 days of cilengitide treatment. The neovascularization and apoptosis of the abdominal aorta were also significantly alleviated by cilengitide treatment. For in vitro experiments, cilengitide treatment was found to inhibit the proliferation, migration, and tube formation of HUVECs. However, cilengitide did not induce the apoptosis of HUVECs. A higher dose of cilengitide inhibited the mRNA expression of VEGF-A, β3, and β5, but not αV. Lastly, cilengitide treatment significantly inhibited the Ras/ERK/AKT pathway in the HUVECs. Conclusions. This study showed that cilengitide effectively inhibited the growth of plaque size by inhibiting the angiogenesis of the abdominal aortic plaques and blocking the VEGF-mediated angiogenic effect on HUVECs.
Collapse
|
4
|
Yu K, Yu Y, Yao Y, Wu Z, Fu S, Cheng RH, Chen YW, Chen HY, Zhou J, Hwang DW, Ding S. A Polypeptide-Based, Membrane-Penetrating, Target-Specific Contrast Agent for Magnetic Resonance Molecular Imaging. ACS APPLIED BIO MATERIALS 2021; 4:1597-1604. [DOI: 10.1021/acsabm.0c01442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Kaichao Yu
- Department of Chemistry and Centre for Nanoscience and Nanotechnology, National Sun Yat-sen University, 70 Lien-Hai Road, Kaohsiung 80424, Taiwan
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yamian Yu
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yao Yao
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhen Wu
- Department of Chemistry and Centre for Nanoscience and Nanotechnology, National Sun Yat-sen University, 70 Lien-Hai Road, Kaohsiung 80424, Taiwan
| | - Shitao Fu
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ren-Hao Cheng
- Department of Chemistry and Centre for Nanoscience and Nanotechnology, National Sun Yat-sen University, 70 Lien-Hai Road, Kaohsiung 80424, Taiwan
| | - Yu-Wen Chen
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd. Nankang, Taipei 115, Taiwan
| | - Hsuan-Ying Chen
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung 80708, Taiwan
| | - Jinlan Zhou
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dennis W. Hwang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd. Nankang, Taipei 115, Taiwan
| | - Shangwu Ding
- Department of Chemistry and Centre for Nanoscience and Nanotechnology, National Sun Yat-sen University, 70 Lien-Hai Road, Kaohsiung 80424, Taiwan
| |
Collapse
|
5
|
Jiang L, Zhu H, Li Y, Wu X, Wang H, Cheng Z. Detecting Vulnerable Atherosclerotic Plaques by 68Ga-Labeled Divalent Cystine Knot Peptide. Mol Pharm 2019; 16:1350-1357. [PMID: 30742442 DOI: 10.1021/acs.molpharmaceut.8b01291] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Lei Jiang
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Hong Zhu
- Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Shanghai Key Laboratory of Stomatology, Ninth People’s Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yi Li
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Xiaodong Wu
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Huoqiang Wang
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California 94305-5484, United States
| |
Collapse
|
6
|
Marjanovic M, Nguyen FT, Ahmad A, Huang PC, Suslick KS, Boppart SA. Characterization of Magnetic Nanoparticle-Seeded Microspheres for Magnetomotive and Multimodal Imaging. IEEE JOURNAL OF SELECTED TOPICS IN QUANTUM ELECTRONICS : A PUBLICATION OF THE IEEE LASERS AND ELECTRO-OPTICS SOCIETY 2019; 25:7101314. [PMID: 30880897 PMCID: PMC6413528 DOI: 10.1109/jstqe.2018.2856582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Magnetic iron-oxide nanoparticles have been developed as contrast agents in magnetic resonance imaging (MRI) and as therapeutic agents in magnetic hyperthermia. They have also recently been demonstrated as contrast and elastography agents in magnetomotive optical coherence tomography and elastography (MM-OCT and MM-OCE, respectively). Protein-shell microspheres containing suspensions of these magnetic nanoparticles in lipid cores, and with functionalized outer shells for specific targeting, have also been demonstrated as efficient contrast agents for imaging modalities such as MM-OCT and MRI, and can be easily modified for other modalities such as ultrasound, fluorescence, and luminescence imaging. By leveraging the benefits of these various imaging modalities with the use of only a single agent, a magnetic microsphere, it becomes possible to use a widefield imaging method (such as MRI or small animal fluorescence imaging) to initially locate the agent, and then use MM-OCT to obtain dynamic contrast images with cellular level morphological resolution. In addition to multimodal contrast-enhanced imaging, these microspheres could serve as drug carriers for targeted delivery under image guidance. Although the preparation and surface modifications of protein microspheres containing iron oxide nanoparticles has been previously described and feasibility studies conducted, many questions regarding their production and properties remain. Since the use of multifunctional microspheres could have high clinical relevance, here we report a detailed characterization of their properties and behavior in different environments to highlight their versatility. The work presented here is an effort for the development and optimization of nanoparticle-based microspheres as multi-modal contrast agents that can bridge imaging modalities on different size scales, especially for their use in MM-OCT and MRI imaging.
Collapse
Affiliation(s)
- Marina Marjanovic
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Freddy T Nguyen
- University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA. He is now with the Massachusetts Institute of Technology, Cambridge, MA, 02139 USA
| | - Adeel Ahmad
- University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA. He is now with Texas Instruments.
| | - Pin-Chieh Huang
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Kenneth S Suslick
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Stephen A Boppart
- Department of Electrical and Computer Engineering and Bioengineering, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA (phone: 217-244-7479; fax: 217-333-5833; )
| |
Collapse
|
7
|
Su T, Wang YB, Han D, Wang J, Qi S, Gao L, Shao YH, Qiao HY, Chen JW, Liang SH, Nie YZ, Li JY, Cao F. Multimodality Imaging of Angiogenesis in a Rabbit Atherosclerotic Model by GEBP11 Peptide Targeted Nanoparticles. Am J Cancer Res 2017; 7:4791-4804. [PMID: 29187904 PMCID: PMC5706100 DOI: 10.7150/thno.20767] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 09/16/2017] [Indexed: 12/12/2022] Open
Abstract
Background and Aims: Angiogenesis is an important pathological process during progression of plaque formation, which can result in plaque hemorrhage and vulnerability. This study aims to explore non-invasive imaging of angiogenesis in atherosclerotic plaque through magnetic resonance imaging (MRI) and positron emission tomography (PET) by using GEBP11 peptide targeted magnetic iron oxide nanoparticles in a rabbit model of atherosclerosis. Methods: The dual-modality imaging probe was constructed by coupling 2, 3-dimercaptosuccinnic acid-coated paramagnetic nanoparticles (DMSA-MNPs) and the PET 68Ga chelator 1,4,7-triazacyclononane-N, N', N''-triacetic acid (NOTA) to GEBP11 peptide. The atherosclerosis model was induced in New Zealand white rabbits by abdominal aorta balloon de-endothelialization and atherogenic diet for 12 weeks. The plaque areas in abdominal artery were detected by ultrasound imaging and Oil Red O staining. Immunofluorescence staining and Prussian blue staining were applied respectively to investigate the affinity of GEBP11 peptide. MTT and flow cytometric analysis were performed to detect the effects of NGD-MNPs on cell proliferation, cell cycle and apoptosis in Human umbilical vein endothelial cells (HUVECs). In vivo MRI and PET imaging of atherosclerotic plaque were carried out at different time points after intravenous injection of nanoparticles. Results: The NGD-MNPs with hydrodynamic diameter of 130.8 nm ± 7.1 nm exhibited good imaging properties, high stability, low immunogenicity and little cytotoxicity. In vivo PET/MR imaging revealed that 68Ga-NGD-MNPs were successfully applied to visualize atherosclerotic plaque angiogenesis in the rabbit abdominal aorta. Prussian blue and CD31 immunohistochemical staining confirmed that NGD-MNPs were well co-localized within the blood vessels' plaques. Conclusion:68Ga-NGD-MNPs might be a promising MR and PET dual imaging probe for visualizing the vulnerable plaques.
Collapse
|
8
|
Meloni MM, Barton S, Xu L, Kaski JC, Song W, He T. Contrast agents for cardiovascular magnetic resonance imaging: an overview. J Mater Chem B 2017; 5:5714-5725. [DOI: 10.1039/c7tb01241a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Contrast agents for Cardiovascular Magnetic Resonance (CMR) play a major role in research and clinical cardiology.
Collapse
Affiliation(s)
- Marco M. Meloni
- Molecular and Clinical Sciences Research Institute
- St George's, University of London
- London
- UK
- School of Pharmacy and Chemistry
| | - Stephen Barton
- School of Pharmacy and Chemistry
- Kingston University
- London
- UK
| | - Lei Xu
- Department of Radiology
- Beijing Anzhen Hospital
- Beijing
- China
| | - Juan C. Kaski
- Molecular and Clinical Sciences Research Institute
- St George's, University of London
- London
- UK
| | - Wenhui Song
- UCL Centre for Biomaterials
- Division of surgery & Interventional Science
- University College of London
- London
- UK
| | - Taigang He
- Molecular and Clinical Sciences Research Institute
- St George's, University of London
- London
- UK
- Royal Brompton Hospital
| |
Collapse
|
9
|
Expression Profiling of Genes Related to Endothelial Cells Biology in Patients with Type 2 Diabetes and Patients with Prediabetes. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1845638. [PMID: 27781209 PMCID: PMC5066000 DOI: 10.1155/2016/1845638] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/18/2016] [Accepted: 08/30/2016] [Indexed: 12/31/2022]
Abstract
Endothelial dysfunction appears to be an early sign indicating vascular damage and predicts the progression of atherosclerosis and cardiovascular disorders. Extensive clinical and experimental evidence suggests that endothelial dysfunction occurs in Type 2 Diabetes Mellitus (T2DM) and prediabetes patients. This study was carried out with an aim to appraise the expression levels in the peripheral blood of 84 genes related to endothelial cells biology in patients with diagnosed T2DM or prediabetes, trying to identify new genes whose expression might be changed under these pathological conditions. The study covered a total of 45 participants. The participants were divided into three groups: group 1, patients with T2DM; group 2, patients with prediabetes; group 3, control group. The gene expression analysis was performed using the Endothelial Cell Biology RT2 Profiler PCR Array. In the case of T2DM, 59 genes were found to be upregulated, and four genes were observed to be downregulated. In prediabetes patients, increased expression was observed for 49 genes, with two downregulated genes observed. Our results indicate that diabetic and prediabetic conditions change the expression levels of genes related to endothelial cells biology and, consequently, may increase the risk for occurrence of endothelial dysfunction.
Collapse
|
10
|
Molecular Imaging of Vulnerable Atherosclerotic Plaques in Animal Models. Int J Mol Sci 2016; 17:ijms17091511. [PMID: 27618031 PMCID: PMC5037788 DOI: 10.3390/ijms17091511] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/24/2016] [Accepted: 08/31/2016] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is characterized by intimal plaques of the arterial vessels that develop slowly and, in some cases, may undergo spontaneous rupture with subsequent heart attack or stroke. Currently, noninvasive diagnostic tools are inadequate to screen atherosclerotic lesions at high risk of acute complications. Therefore, the attention of the scientific community has been focused on the use of molecular imaging for identifying vulnerable plaques. Genetically engineered murine models such as ApoE−/− and ApoE−/−Fbn1C1039G+/− mice have been shown to be useful for testing new probes targeting biomarkers of relevant molecular processes for the characterization of vulnerable plaques, such as vascular endothelial growth factor receptor (VEGFR)-1, VEGFR-2, intercellular adhesion molecule (ICAM)-1, P-selectin, and integrins, and for the potential development of translational tools to identify high-risk patients who could benefit from early therapeutic interventions. This review summarizes the main animal models of vulnerable plaques, with an emphasis on genetically altered mice, and the state-of-the-art preclinical molecular imaging strategies.
Collapse
|
11
|
Preclinical models of atherosclerosis. The future of Hybrid PET/MR technology for the early detection of vulnerable plaque. Expert Rev Mol Med 2016; 18:e6. [PMID: 27056676 DOI: 10.1017/erm.2016.5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases are the leading cause of death in developed countries. The aetiology is currently multifactorial, thus making them very difficult to prevent. Preclinical models of atherothrombotic diseases, including vulnerable plaque-associated complications, are now providing significant insights into pathologies like atherosclerosis, and in combination with the most recent advances in new non-invasive imaging technologies, they have become essential tools to evaluate new therapeutic strategies, with which can forecast and prevent plaque rupture. Positron emission tomography (PET)/computed tomography imaging is currently used for plaque visualisation in clinical and pre-clinical cardiovascular research, albeit with significant limitations. However, the combination of PET and magnetic resonance imaging (MRI) technologies is still the best option available today, as combined PET/MRI scans provide simultaneous data acquisition together with high quality anatomical information, sensitivity and lower radiation exposure for the patient. The coming years may represent a new era for the implementation of PET/MRI in clinical practice, but first, clinically efficient attenuation correction algorithms and research towards multimodal reagents and safety issues should be validated at the preclinical level.
Collapse
|
12
|
Vancraeynest D, Roelants V, Bouzin C, Hanin FX, Walrand S, Bol V, Bol A, Pouleur AC, Pasquet A, Gerber B, Lesnik P, Huby T, Jamar F, Vanoverschelde JL. αVβ3 integrin-targeted microSPECT/CT imaging of inflamed atherosclerotic plaques in mice. EJNMMI Res 2016; 6:29. [PMID: 27009066 PMCID: PMC4805679 DOI: 10.1186/s13550-016-0184-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/15/2016] [Indexed: 12/11/2022] Open
Abstract
Background αVβ3-integrin is expressed by activated endothelial cells and macrophages in atherosclerotic plaques and may represent a valuable marker of high-risk plaques. We evaluated 99mTc-maraciclatide, an integrin-specific tracer, for imaging vascular inflammation in atherosclerotic lesions in mice. Methods Apolipoprotein E-negative (ApoE−/−) mice on a Western diet (n = 10) and normally fed adult C57BL/6 control mice (n = 4) were injected with 99mTc-maraciclatide (51.8 ± 3.7 MBq). A blocking peptide was infused in three ApoE−/− mice; this condition served as another control. After 90 min, the animals were imaged via single-photon emission computed tomography (SPECT). While maintained in the same position, the mice were transferred to computed tomography (CT) to obtain contrast-enhanced images of the aortic arch. Images from both modalities were fused, and signal was quantified in the aortic arch and in the vena cava for subtraction of blood-pool activity. The aorta was carefully dissected after imaging for gamma counting, autoradiography, and histology. Results Tracer uptake was significantly higher in ApoE−/− mice than in both groups of control mice (1.56 ± 0.33 vs. 0.82 ± 0.24 vs. 0.98 ± 0.11, respectively; P = 0.006). Furthermore, higher tracer activity was detected via gamma counting in the aorta of hypercholesterolemic mice than in both groups of control mice (1.52 ± 0.43 vs. 0.78 ± 0.19 vs. 0.47 ± 0.31 99mTc-maraciclatide %ID/g, respectively; P = 0.018). Autoradiography showed significantly higher tracer uptake in the atherosclerotic aorta than in the control aorta (P = 0.026). Finally, in the atherosclerotic aorta, immunostaining indicated that the integrin signal came predominantly from macrophages and was correlated with the macrophage CD68 immunomarker (r = 0.73). Conclusions 99mTc-maraciclatide allows in vivo detection of inflamed atherosclerotic plaques in mice and may represent a non-invasive approach for identifying high-risk plaques in patients.
Collapse
Affiliation(s)
- David Vancraeynest
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium. .,Division of Cardiology, Cliniques Universitaires St-Luc, Avenue Hippocrate, 10-2881, B-1200, Brussels, Belgium.
| | - Véronique Roelants
- Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Caroline Bouzin
- IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - François-Xavier Hanin
- Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Stephan Walrand
- Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Vanesa Bol
- Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Anne Bol
- Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Anne-Catherine Pouleur
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Division of Cardiology, Cliniques Universitaires St-Luc, Avenue Hippocrate, 10-2881, B-1200, Brussels, Belgium
| | - Agnès Pasquet
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Division of Cardiology, Cliniques Universitaires St-Luc, Avenue Hippocrate, 10-2881, B-1200, Brussels, Belgium
| | - Bernhard Gerber
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Division of Cardiology, Cliniques Universitaires St-Luc, Avenue Hippocrate, 10-2881, B-1200, Brussels, Belgium
| | - Philippe Lesnik
- INSERM UMR_S 1166, Integrative Biology of Atherosclerosis Team, Université Pierre et Marie Curie-Paris6 and institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, 75013, Paris, France
| | - Thierry Huby
- INSERM UMR_S 1166, Integrative Biology of Atherosclerosis Team, Université Pierre et Marie Curie-Paris6 and institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, 75013, Paris, France
| | - François Jamar
- Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Jean-Louis Vanoverschelde
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Division of Cardiology, Cliniques Universitaires St-Luc, Avenue Hippocrate, 10-2881, B-1200, Brussels, Belgium
| |
Collapse
|
13
|
Ariyani W, Iwasaki T, Miyazaki W, Khongorzul E, Nakajima T, Kameo S, Koyama H, Tsushima Y, Koibuchi N. Effects of Gadolinium-Based Contrast Agents on Thyroid Hormone Receptor Action and Thyroid Hormone-Induced Cerebellar Purkinje Cell Morphogenesis. Front Endocrinol (Lausanne) 2016; 7:115. [PMID: 27617003 PMCID: PMC4999949 DOI: 10.3389/fendo.2016.00115] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 08/09/2016] [Indexed: 11/26/2022] Open
Abstract
Gadolinium (Gd)-based contrast agents (GBCAs) are used in diagnostic imaging to enhance the quality of magnetic resonance imaging or angiography. After intravenous injection, GBCAs can accumulate in the brain. Thyroid hormones (THs) are critical for the development and functional maintenance of the central nervous system. TH actions in brain are mainly exerted through nuclear TH receptors (TRs). We examined the effects of GBCAs on TR-mediated transcription in CV-1 cells using transient transfection-based reporter assay and TH-mediated cerebellar Purkinje cell morphogenesis in primary culture. We also measured the cellular accumulation and viability of Gd after representative GBCA treatments in cultured CV-1 cells. Both linear (Gd-diethylene triamine pentaacetic acid-bis methyl acid, Gd-DTPA-BMA) and macrocyclic (Gd-tetraazacyclododecane tetraacetic acid, Gd-DOTA) GBCAs were accumulated without inducing cell death in CV-1 cells. By contrast, Gd chloride (GdCl3) treatment induced approximately 100 times higher Gd accumulation and significantly reduced the number of cells. Low doses of Gd-DTPA-BMA (10(-8) to 10(-6)M) augmented TR-mediated transcription, but the transcription was suppressed at higher dose (10(-5) to 10(-4)M), with decreased β-galactosidase activity indicating cellular toxicity. TR-mediated transcription was not altered by Gd-DOTA or GdCl3, but the latter induced a significant reduction in β-galactosidase activity at high doses, indicating cellular toxicity. In cerebellar cultures, the dendrite arborization of Purkinje cells induced by 10(-9)M T4 was augmented by low-dose Gd-DTPA-BMA (10(-7)M) but was suppressed by higher dose (10(-5)M). Such augmentation by low-dose Gd-DTPA-BMA was not observed with 10(-9)M T3, probably because of the greater dendrite arborization by T3; however, the arborization by T3 was suppressed by a higher dose of Gd-DTPA-BMA (10(-5)M) as seen in T4 treatment. The effect of Gd-DOTA on dendrite arborization was much weaker than that of the other compounds. These results indicate that exposure to specific GBCAs may, at least in part, cause toxic effects in the brain by disrupting the action of THs on TRs. The toxic effects of GBCAs may depend on the chemical structure of GBCA and the dose. Thus, it is very important to choose appropriate GBCAs for imaging to prevent adverse side effects.
Collapse
Affiliation(s)
- Winda Ariyani
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Toshiharu Iwasaki
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
- Department of Liberal Arts and Human Development, Kanagawa University of Human Services, Kanagawa, Japan
| | - Wataru Miyazaki
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Erdene Khongorzul
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Takahito Nakajima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Satomi Kameo
- Department of Public Health, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hiroshi Koyama
- Department of Public Health, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
- *Correspondence: Noriyuki Koibuchi,
| |
Collapse
|
14
|
|
15
|
Jiang L, Tu Y, Kimura RH, Habte F, Chen H, Cheng K, Shi H, Gambhir SS, Cheng Z. 64Cu-Labeled Divalent Cystine Knot Peptide for Imaging Carotid Atherosclerotic Plaques. J Nucl Med 2015; 56:939-44. [PMID: 25908832 DOI: 10.2967/jnumed.115.155176] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/08/2015] [Indexed: 01/24/2023] Open
Abstract
UNLABELLED The rupture of vulnerable atherosclerotic plaques that lead to stroke and myocardial infarction may be induced by macrophage infiltration and augmented by the expression of integrin αvβ3. Indeed, atherosclerotic angiogenesis may be a promising marker of inflammation. In this study, an engineered integrin αvβ3-targeting PET probe, (64)Cu-NOTA-3-4A, derived from a divalent knottin miniprotein was evaluated in a mouse model for carotid atherosclerotic plaques. METHODS Atherosclerotic plaques in BALB/C mice, maintained on a high-fat diet, were induced with streptozotocin injection and carotid artery ligation and verified by MR imaging. Knottin 3-4A was synthesized by solid-phase peptide synthesis chemistry and coupled to 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) before radiolabeling with (64)Cu. PET probe stability in mouse serum was evaluated. Mice with carotid atherosclerotic plaques were injected via the tail vein with (64)Cu-NOTA-3-4A or (18)F-FDG, followed by small-animal PET/CT imaging at different time points. Receptor targeting specificity of the probe was verified by coinjection of c(RGDyK) administered in molar excess. Subsequently, carotid artery dissection and immunofluorescence staining were performed to evaluate target expression. RESULTS (64)Cu-NOTA-3-4A was synthesized in high radiochemical purity and yield and demonstrated molecular stability in both phosphate-buffered saline and mouse serum at 4 h. Small-animal PET/CT showed that (64)Cu-NOTA-3-4A accumulated at significantly higher levels in the neovasculature of carotid atherosclerotic plaques (7.41 ± 1.44 vs. 0.67 ± 0.23 percentage injected dose/gram, P < 0.05) than healthy or normal vessels at 1 h after injection. (18)F-FDG also accumulated in atherosclerotic lesions at 0.5 and 1 h after injection but at lower plaque-to-normal tissue ratios than (64)Cu-NOTA-3-4A. For example, plaque-to-normal carotid artery ratios for (18)F-FDG and (64)Cu-NOTA-3-4A at 1 h after injection were 3.75 and 14.71 (P < 0.05), respectively. Furthermore, uptake of (64)Cu-NOTA-3-4A in atherosclerotic plaques was effectively blocked (∼90% at 1 h after injection) by coinjection of c(RGDyK). Immunostaining confirmed integrin αvβ3 expression in both the infiltrating macrophages and the neovasculature of atherosclerotic plaques. CONCLUSION (64)Cu-NOTA-3-4A demonstrates specific accumulation in carotid atherosclerotic plaques in which macrophage infiltration and angiogenesis are responsible for elevated integrin αvβ3 levels. Therefore, (64)Cu-NOTA-3-4A may demonstrate clinical utility as a PET probe for atherosclerosis imaging or for the evaluation of therapies used to treat atherosclerosis.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; and Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California
| | - Yingfeng Tu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California
| | - Richard H Kimura
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California
| | - Frezghi Habte
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California
| | - Hao Chen
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California
| | - Kai Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; and
| | - Sanjiv Sam Gambhir
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California
| |
Collapse
|
16
|
Sharifi S, Seyednejad H, Laurent S, Atyabi F, Saei AA, Mahmoudi M. Superparamagnetic iron oxide nanoparticles for in vivo molecular and cellular imaging. CONTRAST MEDIA & MOLECULAR IMAGING 2015; 10:329-55. [PMID: 25882768 DOI: 10.1002/cmmi.1638] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 01/30/2015] [Accepted: 02/06/2015] [Indexed: 12/16/2022]
Abstract
In the last decade, the biomedical applications of nanoparticles (NPs) (e.g. cell tracking, biosensing, magnetic resonance imaging (MRI), targeted drug delivery, and tissue engineering) have been increasingly developed. Among the various NP types, superparamagnetic iron oxide NPs (SPIONs) have attracted considerable attention for early detection of diseases due to their specific physicochemical properties and their molecular imaging capabilities. A comprehensive review is presented on the recent advances in the development of in vitro and in vivo SPION applications for molecular imaging, along with opportunities and challenges.
Collapse
Affiliation(s)
- Shahriar Sharifi
- Department of Biomaterials Science and Technology, University of Twente, The Netherlands
| | - Hajar Seyednejad
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA
| | - Sophie Laurent
- Department of General, Organic, and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, B-7000, Mons, Belgium.,CMMI - Center for Microscopy and Molecular Imaging, Rue Adrienne Bolland 8, B-6041, Gosselies, Belgium
| | - Fatemeh Atyabi
- Nanotechnology Research Center and Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ata Saei
- Nanotechnology Research Center and Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Morteza Mahmoudi
- Nanotechnology Research Center and Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
17
|
Guo S, Shen S, Wang J, Wang H, Li M, Liu Y, Hou F, Liao Y, Bin J. Detection of high-risk atherosclerotic plaques with ultrasound molecular imaging of glycoprotein IIb/IIIa receptor on activated platelets. Am J Cancer Res 2015; 5:418-30. [PMID: 25699100 PMCID: PMC4329504 DOI: 10.7150/thno.10020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 12/28/2014] [Indexed: 12/23/2022] Open
Abstract
Objective: Ultrasound molecular imaging (UMI) of glycoprotein (GP) IIb/IIIa receptor on activated platelets offers a unique means of identifying high-risk atherosclerosis. We hypothesized that contrast-enhanced ultrasound with microbubbles (MBs) targeted to GP IIb/IIIa could be used to detect and quantify activated platelets on the surface of advanced plaques. Methods and Results: A mouse model of advanced atherosclerosis was generated by maintaining apolipoprotein E-deficient (ApoE-/-) mice on a hypercholesterolemic diet (HCD). The three other experimental groups consisted of ApoE-/- and wild-type (C57BL/6) mice fed a normal chow diet and C57BL/6 mice on an HCD diet. Plaque formation was confirmed by histological and immunohistochemical methods using light, fluorescence, and electron microscopy. Mice were injected with a lipid MB-conjugated cyclic Arg-Gly-Asp peptide or nonspecific control peptide, and the abdominal aorta was examined by UMI. The accumulation of GP IIb/IIIa and activated platelets on the surface of atherosclerotic plaques was highest in the ApoE-/-+HCD group, followed by ApoE-/-+chow, C57BL/6+HCD, and C57BL/6+chow groups (P<0.05). Notably, GP IIb/IIIa expression was associated with the vulnerability index and necrotic center/fiber cap ratio (P<0.05), and contrast video intensity from adhered cyclic Arg-Gly-Asp-modified MBs (MB-cRGDs) was correlated with GP IIb/IIIa expression on the plaque surface (P<0.05). Conclusion: GP IIb/IIIa of activated platelets on the atherosclerotic endothelium is a biomarker for high-risk plaques that can be quantified by UMI using MB-cRGDs, providing a noninvasive means for detecting high-risk plaques and preventing acute cardiovascular events.
Collapse
|
18
|
Li J, Dobrucki LW, Marjanovic M, Chaney EJ, Suslick KS, Boppart SA. Enhancement and wavelength-shifted emission of Cerenkov luminescence using multifunctional microspheres. Phys Med Biol 2015; 60:727-39. [PMID: 25555157 DOI: 10.1088/0031-9155/60/2/727] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cerenkov luminescence (CL) imaging is a new molecular imaging modality that utilizes the photons emitted during radioactive decay when charged particles travel faster than the phase velocity of light in a dielectric medium. Here we present a novel agent to convert and increase CL emission at longer wavelengths using multimodal protein microspheres (MSs). The (64)Cu-labeled protein microspheres contain quantum dots (QDs) encapsulated within a high-refractive-index-oil core. Dark box imaging of the MSs was conducted to demonstrate the improvement in CL emission at longer wavelengths. To illustrate the versatile design of these MSs and the potential of CL in disease diagnosis, these MSs were utilized for in vitro cell targeting and ex vivo CL-excited QD fluorescence (CL-FL) imaging of atherosclerotic plaques in rats. It was shown that by utilizing both QDs and MSs with a high-refractive-index-oil core, the CL emission increases by four-fold at longer wavelengths. Furthermore, we demonstrate that these MSs generate both an in vivo and ex vivo contrast signal. The design concept of utilizing QDs and high-index core MSs may contribute to future developments of in vivo CL imaging.
Collapse
Affiliation(s)
- Joanne Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA. Department of Nuclear, Plasma, and Radiological Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA. Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | | | | | | | | | | |
Collapse
|
19
|
Abran M, Cloutier G, Cardinal MHR, Chayer B, Tardif JC, Lesage F. Development of a photoacoustic, ultrasound and fluorescence imaging catheter for the study of atherosclerotic plaque. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2014; 8:696-703. [PMID: 25350946 DOI: 10.1109/tbcas.2014.2360560] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Atherosclerotic cardiovascular diseases are a major cause of death in industrialized countries. Molecular imaging modalities are increasingly recognized to be a promising avenue towards improved diagnosis and for the evaluation of new drug therapies. In this work, we present an acquisition system and associated catheter enabling simultaneous photoacoustic, ultrasound and fluorescence imaging of arteries designed for in vivo imaging. The catheter performance is evaluated in tissue-mimicking phantoms. Simultaneous imaging with three modalities is demonstrated at frame rates of 30 images per second for ultrasound and fluorescence and 1 image per 13 seconds for photoacoustic. Acquired radio-frequency ultrasound data could be processed to obtain radial strain elastograms. With motorized pullback, 3D imaging of phantoms was performed using the three modalities.
Collapse
|
20
|
Iwaki S, Hokamura K, Ogawa M, Takehara Y, Muramatsu Y, Yamane T, Hirabayashi K, Morimoto Y, Hagisawa K, Nakahara K, Mineno T, Terai T, Komatsu T, Ueno T, Tamura K, Adachi Y, Hirata Y, Arita M, Arai H, Umemura K, Nagano T, Hanaoka K. A design strategy for small molecule-based targeted MRI contrast agents: their application for detection of atherosclerotic plaques. Org Biomol Chem 2014; 12:8611-8. [DOI: 10.1039/c4ob01270d] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
21
|
Teresa Albelda M, Garcia-España E, Frias JC. Visualizing the atherosclerotic plaque: a chemical perspective. Chem Soc Rev 2014; 43:2858-76. [PMID: 24526041 DOI: 10.1039/c3cs60410a] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Atherosclerosis is the major underlying pathologic cause of coronary artery disease. An early detection of the disease can prevent clinical sequellae such as angina, myocardial infarction, and stroke. The different imaging techniques employed to visualize the atherosclerotic plaque provide information of diagnostic and prognostic value. Furthermore, the use of contrast agents helps to improve signal-to-noise ratio providing better images. For nuclear imaging techniques and optical imaging these agents are absolutely necessary. We report on the different contrast agents that have been used, are used or may be used in future in animals, humans, or excised tissues for the distinct imaging modalities for atherosclerotic plaque imaging.
Collapse
Affiliation(s)
- Ma Teresa Albelda
- Universidad de Valencia, Instituto de Ciencia Molecular, Edificio de Institutos de Paterna, c/ Catedrático José Beltrán 2, 46071 Valencia, Spain
| | | | | |
Collapse
|
22
|
Wang X, Xing X, Zhang B, Liu F, Cheng Y, Shi D. Surface engineered antifouling optomagnetic SPIONs for bimodal targeted imaging of pancreatic cancer cells. Int J Nanomedicine 2014; 9:1601-1615. [PMID: 24741308 PMCID: PMC3970947 DOI: 10.2147/ijn.s58334] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Targeted imaging contrast agents for early pancreatic ductal adenocarcinoma diagnosis was developed using superparamagnetic iron oxide nanoparticles (SPIONs). For phase transfer of SPIONs, the hydrophobic SPIONs are first treated with tetrafluoroborate and then capped by bovine serum albumin (BSA) via ligand exchange. It was experimentally found that nitrosyl tetrafluoroborate pretreatment and proper structures of molecules are essential to the effective surface functionalization of SPIONs. Nonspecific binding was found to be significantly reduced by BSA surface functionalized hydrophobic SPIONs (BSA·SPIONs). The BSA·SPIONs were monodispersed with an average size of approximately 18.0 nm and stable in a wide pH range and various ionic strengths even after 7 days of storage. The longitudinal and transverse proton relaxation rate (r1, r2) values of the BSA·SPIONs were determined to be 11.6 and 154.2 s(-1) per mM of Fe(3+) respectively. The r2/r1 ratio of 13.3 ensured its application as the T2-weighted magnetic resonance imaging contrast agents. When conjugated with near-infrared fluorescent dye and monoclonal antibody, the (dye)BSA·SPION-monoclonal antibody bioconjugates showed excellent targeting capability with minimal nonspecific binding in the bimodal imaging of pancreatic cancer cells. The experimental approach is facile, environmentally benign, and straightforward, which presents great promise in early cancer diagnosis.
Collapse
Affiliation(s)
- Xiaohui Wang
- Radiology Department of the Tenth People’s Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaohong Xing
- Radiology Department of the Tenth People’s Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Bingbo Zhang
- Radiology Department of the Tenth People’s Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Fengjun Liu
- Radiology Department of the Tenth People’s Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Yingsheng Cheng
- Department of Radiology, Shanghai Sixth People’s Hospital, Shanghai Jiaotong University, Shanghai, People’s Republic of China
| | - Donglu Shi
- Radiology Department of the Tenth People’s Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Materials Science and Engineering Program, Department of Mechanical and Materials Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
23
|
Beer AJ, Pelisek J, Heider P, Saraste A, Reeps C, Metz S, Seidl S, Kessler H, Wester HJ, Eckstein HH, Schwaiger M. PET/CT imaging of integrin αvβ3 expression in human carotid atherosclerosis. JACC Cardiovasc Imaging 2014; 7:178-87. [PMID: 24412187 DOI: 10.1016/j.jcmg.2013.12.003] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 12/05/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The goal of this study was to evaluate the feasibility of [(18)F]Galacto-RGD positron emission tomography (PET)/computed tomography (CT) imaging of αvβ3 expression in human carotid plaques. BACKGROUND The integrin αvβ3 is expressed by macrophages and angiogenic endothelial cells in atherosclerotic lesions and thus is a marker of plaque inflammation and, potentially, of plaque vulnerability. [(18)F]Galacto-RGD is a PET tracer binding specifically to αvβ3. Therefore, [(18)F]Galacto-RGD PET/CT imaging of αvβ3 expression in human carotid plaques might provide a novel noninvasive biomarker of plaque vulnerability. METHODS [(18)F]Galacto-RGD PET/CT imaging was performed in 10 patients with high-grade carotid artery stenosis scheduled for carotid endarterectomy. Tracer uptake was measured in the stenotic areas of the carotid arteries, as well as on the contralateral side, and was corrected for blood pool activity, measured in the distal common carotid artery (target-to-background [TB] ratio). TB ratio was correlated with immunohistochemistry of αvβ3 expression (LM609), macrophage density (CD68), and microvessel density (CD31) of the surgical specimen. In addition, ex vivo autoradiography of the surgical specimen with [(18)F]Galacto-RGD and competition experiments with an unlabeled αvβ3-specific RGD peptide were performed. RESULTS [(18)F]Galacto-RGD PET/CT showed significantly higher TB ratios in stenotic areas compared with nonstenotic areas (p = 0.01). TB ratios correlated significantly with αvβ3 expression (R = 0.787, p = 0.026) and intensity of ex vivo autoradiography (R = 0.733, p = 0.038). Binding to atherosclerotic plaques was efficiently blocked in ex vivo competition experiments. A weak-to-moderate correlation was found with macrophage density (R = 0.367, p = 0.299) and microvessel density (R = 0.479, p = 0.176), which did not reach statistical significance. CONCLUSIONS [(18)F]Galacto-RGD PET/CT shows specific tracer accumulation in human atherosclerotic carotid plaques, which correlates with αvβ3 expression. Based on these initial data, larger prospective studies are now warranted to evaluate the potential of molecular imaging of αvβ3 expression for assessment of plaque inflammation in patients.
Collapse
Affiliation(s)
- Ambros J Beer
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany.
| | - Jaroslav Pelisek
- Department of Vascular Surgery, Technische Universität München, Munich, Germany
| | - Peter Heider
- Department of Vascular Surgery, Technische Universität München, Munich, Germany
| | - Antti Saraste
- Turku PET Centre and Department of Cardiology, Turku, Finland
| | - Christian Reeps
- Department of Vascular Surgery, Technische Universität München, Munich, Germany
| | - Stephan Metz
- Department of Radiology, Technische Universität München, Munich, Germany
| | - Stefan Seidl
- Department of Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Horst Kessler
- Institute for Advanced Study and Center of Integrated Protein Science, Technische Universität München, Department Chemie, Garching, Germany; Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hans-Jürgen Wester
- Chair of Pharmaceutical Radiochemistry, Technische Universität München, Garching, Germany
| | | | - Markus Schwaiger
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| |
Collapse
|
24
|
Cellular density effect on RGD ligand internalization in glioblastoma for MRI application. PLoS One 2013; 8:e82777. [PMID: 24386117 PMCID: PMC3873929 DOI: 10.1371/journal.pone.0082777] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/28/2013] [Indexed: 01/12/2023] Open
Abstract
Cellular density is a parameter measured for glioma grade and invasiveness diagnosis. The characterization of the cellular density can be performed, non invasively, by magnetic resonance imaging (MRI), since, this technique displays a good resolution. Nevertheless MRI sensitivity is critical. Development of smart contrast agents appears useful to increase MRI signal to noise ratio (SNR). Tumor invasiveness is correlated with high expression of integrins that can be targeted by RGD motif. In this study, MRI contrast agents or fluorescent probes linked to RGD-peptides were used, in a glioma model, to assess the relation between RGD uptake/signal improvement/cell density and consequently tumor invasiveness. Experiments were performed in vitro with U87-MG glioma cells. Flow cytometry and microscopy experiments with RGD and iRGD-alexa488 demonstrated that cell internalization was dependent on cell density. The internalization involved a clathrin-dependent endocytosis. Cytoskeleton and particularly the microtubules were concerned. Actin filaments played a minor role. The internalization was also dependent on the glycolysis and the oxidative phosphorylations. The cellular density modulated the importance of the endocytosis pathways and of the metabolism but not the cytoskeleton contribution. The internalization of the RGD-peptide associated to gadolinium chelate increased the SNR of U87 cells. Moreover, following the cell density augmentation, the SNR increased with a low amplitude but a trend was clearly determined. In conclusion, RGD-peptide internalization appeared, in vitro, as a marker of cellular density. In perspective, the combination of these peptides with contrast agents associated to more sensitive MRI techniques could improve the MRI signal allowing the characterization of cellular density for tumor diagnosis.
Collapse
|
25
|
Wildgruber M, Swirski FK, Zernecke A. Molecular imaging of inflammation in atherosclerosis. Am J Cancer Res 2013; 3:865-84. [PMID: 24312156 PMCID: PMC3841337 DOI: 10.7150/thno.5771] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 04/29/2013] [Indexed: 01/13/2023] Open
Abstract
Acute rupture of vulnerable plaques frequently leads to myocardial infarction and stroke. Within the last decades, several cellular and molecular players have been identified that promote atherosclerotic lesion formation, maturation and plaque rupture. It is now widely recognized that inflammation of the vessel wall and distinct leukocyte subsets are involved throughout all phases of atherosclerotic lesion development. The mechanisms that render a stable plaque unstable and prone to rupture, however, remain unknown and the identification of the vulnerable plaque remains a major challenge in cardiovascular medicine. Imaging technologies used in the clinic offer minimal information about the underlying biology and potential risk for rupture. New imaging technologies are therefore being developed, and in the preclinical setting have enabled new and dynamic insights into the vessel wall for a better understanding of this complex disease. Molecular imaging has the potential to track biological processes, such as the activity of cellular and molecular biomarkers in vivo and over time. Similarly, novel imaging technologies specifically detect effects of therapies that aim to stabilize vulnerable plaques and silence vascular inflammation. Here we will review the potential of established and new molecular imaging technologies in the setting of atherosclerosis, and discuss the cumbersome steps required for translating molecular imaging approaches into the clinic.
Collapse
|
26
|
Specific somatostatin receptor II expression in arterial plaque: 68Ga-DOTATATE autoradiographic, immunohistochemical and flow cytometric studies in apoE-deficient mice. Atherosclerosis 2013; 230:33-9. [DOI: 10.1016/j.atherosclerosis.2013.06.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 06/14/2013] [Accepted: 06/19/2013] [Indexed: 12/12/2022]
|
27
|
Feasibility and kinetic characteristics of 68Ga-NOTA-RGD PET for in vivo atherosclerosis imaging. Ann Nucl Med 2013; 27:847-54. [DOI: 10.1007/s12149-013-0757-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 07/09/2013] [Indexed: 12/13/2022]
|
28
|
den Adel B, Daemen MJ, Poelmann RE, van der Weerd L. Molecular Magnetic Resonance Imaging for the Detection of Vulnerable Plaques: Is It Possible?: Retracted. Arterioscler Thromb Vasc Biol 2013:ATVBAHA.112.300108. [PMID: 23413424 DOI: 10.1161/atvbaha.112.300108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 01/13/2013] [Indexed: 11/16/2022]
Abstract
Recent advances in molecular resonance imaging of atherosclerosis enable to visualize atherosclerotic plaques in vivo using molecular targeted contrast agents. This offers opportunities to study atherosclerosis development and plaque vulnerability noninvasively. In this review, we discuss MRI contrast agents targeted toward atherosclerotic plaques and illustrate how these new imaging platforms could assist in our understanding of atherogenesis and atheroprogression. In particular, we highlight the challenges and limitations of the different contrast agents and hurdles for clinical application. We describe the most promising existing compounds to detect atherosclerosis and plaque vulnerability. Of particular interest are the fibrin-targeted compounds that detect thrombi and, furthermore, the contrast agents targeted to integrins that allow to visualize plaque neovascularization. Moreover, vascular cell adhesion molecule 1-targeted iron oxides seem promising for early detection of atherosclerosis. These targeted MRI contrast agents, however promising and well characterized in (pre)clinical models, lack specificity for plaque vulnerability.
Collapse
Affiliation(s)
- Brigit den Adel
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (B.d.A., R.E.P., L.v.d.W.)
| | | | | | | |
Collapse
|
29
|
Dixit S, Das M, Alwarappan S, Goicochea NL, Howell M, Mohapatra S, Mohapatra S. Phospholipid micelle encapsulated gadolinium oxide nanoparticles for imaging and gene delivery. RSC Adv 2013; 3:2727-2735. [PMID: 24724012 PMCID: PMC3979490 DOI: 10.1039/c2ra22293k] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We encapsulated gadolinium oxide (Gd2O3) nanoparticles within phospholipid micelles as a novel low cytotoxic T1-weighted MRI imaging contrast agent (MGdNPs) that can also deliver small molecules such as DNA plasmids. MGdNPs show relatively good MRI relaxivity values, negligible cytotoxicity, excellent cellular uptake and expression of DNA plasmids in vivo. Biodistribution studies in mice show that intranasal and intraperitoneal administration of MGdNPs can effectively target specific organs.
Collapse
Affiliation(s)
- Suraj Dixit
- Nanomedicine Research Center, USF Morsani College of Medicine, Tampa, FL, USA. Fax: +1-813-974-8907; Tel: +1-813-974-8568
| | - Mahasweta Das
- Nanomedicine Research Center, USF Morsani College of Medicine, Tampa, FL, USA. Fax: +1-813-974-8907; Tel: +1-813-974-8568
- Department of Internal Medicine, Division of Translational Medicine, USF Morsani College of Medicine, Tampa, FL, USA
| | - Subbiah Alwarappan
- Nanomedicine Research Center, USF Morsani College of Medicine, Tampa, FL, USA. Fax: +1-813-974-8907; Tel: +1-813-974-8568
| | - Nancy L. Goicochea
- Nanomedicine Research Center, USF Morsani College of Medicine, Tampa, FL, USA. Fax: +1-813-974-8907; Tel: +1-813-974-8568
| | - Mark Howell
- Department of Molecular Medicine, USF Morsani College of Medicine, Tampa, FL, USA
| | - Subhra Mohapatra
- Nanomedicine Research Center, USF Morsani College of Medicine, Tampa, FL, USA. Fax: +1-813-974-8907; Tel: +1-813-974-8568
- Department of Molecular Medicine, USF Morsani College of Medicine, Tampa, FL, USA
- James A Haley Veteran’s Hospital and Medical Research Center, Tampa, FL, USA
| | - Shyam Mohapatra
- Nanomedicine Research Center, USF Morsani College of Medicine, Tampa, FL, USA. Fax: +1-813-974-8907; Tel: +1-813-974-8568
- Department of Internal Medicine, Division of Translational Medicine, USF Morsani College of Medicine, Tampa, FL, USA
- James A Haley Veteran’s Hospital and Medical Research Center, Tampa, FL, USA
| |
Collapse
|
30
|
Kasim S, Moran D, McFadden E. Vulnerable plaque: from bench to bedside; local pacification versus systemic therapy. Heart Views 2013; 13:139-45. [PMID: 23439781 PMCID: PMC3573359 DOI: 10.4103/1995-705x.105731] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Critical coronary stenoses accounts for a small proportion of acute coronary syndromes and sudden death. The majority are caused by coronary thromboses that arise from a nonangiographically obstructive atheroma. Recent developments in noninvasive imaging of so-called vulnerable plaques created opportunities to direct treatment to prevent morbidity and mortality associated with these high-risk lesions. This review covers therapy employed in the past, present, and potentially in the future as the natural history of plaque assessment unfolds.
Collapse
Affiliation(s)
- Sazzli Kasim
- Cardiology Unit, Medical Faculty, UiTM Sg Buloh, Selangor, Malaysia ; Division of Cardiology, Cork University Hospital, Cork, Ireland
| | | | | |
Collapse
|
31
|
Molecular imaging to identify the vulnerable plaque--from basic research to clinical practice. Mol Imaging Biol 2013; 14:523-33. [PMID: 22983911 DOI: 10.1007/s11307-012-0586-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cardiovascular disease (CVD) is still the leading cause of death in the Western World. Adverse outcomes of CVD include stroke, myocardial infarction, and heart failure. Atherosclerosis is considered to be the major cause of CVD and is estimated to cause half of all deaths in developed countries. Atherosclerotic lesions of the vessel wall may obstruct blood flow mechanically through stenosis, but rupture of atherosclerotic plaques causing formation of occlusive thrombi is far more prevalent. Unfortunately, conventional diagnostic tools fail to assess whether a plaque is vulnerable to rupture. Research over the past decade identified the biological processes that are implicated in the course towards plaque rupture, like cell death and inflammation. Knowledge about plaque biology propelled the development of imaging techniques that target biologic processes in order to predict the vulnerable plaque. This paper discusses novel and existing molecular imaging targets and addresses advantages and disadvantages of these targets and respective imaging techniques in respect of clinical application and socio-economic impact.
Collapse
|
32
|
Interventional radionuclide therapy of hepatocellular carcinoma: Assessment of intratumoral retention of HPMA copolymers. Chem Res Chin Univ 2013. [DOI: 10.1007/s40242-013-2089-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
33
|
Zhao XQ, Kerwin WS. Utilizing imaging tools in lipidology: examining the potential of MRI for monitoring cholesterol therapy. ACTA ACUST UNITED AC 2012. [PMID: 23197995 DOI: 10.2217/clp.12.33] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Lipid abnormalities play important roles in the development of atherosclerosis. Lipid therapies result in alterations in atherosclerotic plaques including halting of progression of the plaque, lipid transport out of the plaque and reducing inflammatory activity, which lead to plaque morphologies that are less prone to disruption, the main cause of clinical events. In order to investigate and monitor plaque morphological changes during lipid therapy in vivo we need an imaging method that can provide accurate assessment of plaque tissue components and activity. MRI of atherosclerosis has been validated as a reliable assessment of the size of the vessel lumen, but also the size of the plaque, its tissue composition and plaque activity, including inflammation. The purpose of this review is to summarize the state of evidence for the direct assessment of atherosclerotic plaque and its change by MRI, and to establish the proven role of MRI of atherosclerosis in pharmaceutical trials with lipid therapy.
Collapse
Affiliation(s)
- Xue-Qiao Zhao
- University of Washington School of Medicine, Seattle, WA 98105, USA
| | | |
Collapse
|
34
|
Quantitative Longitudinal Imaging of Vascular Inflammation and Treatment by Ezetimibe in apoE Mice by FMT Using New Optical Imaging Biomarkers of Cathepsin Activity and α(v)β(3) Integrin. INTERNATIONAL JOURNAL OF MOLECULAR IMAGING 2012; 2012:189254. [PMID: 23119157 PMCID: PMC3483711 DOI: 10.1155/2012/189254] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 09/06/2012] [Indexed: 12/02/2022]
Abstract
Inflammation as a core pathological event of atherosclerotic lesions is associated with the secretion of cathepsin proteases and the expression of αvβ3 integrin. We employed fluorescence molecular tomographic (FMT) noninvasive imaging of these molecular activities using cathepsin sensing (ProSense, CatB FAST) and αvβ3 integrin (IntegriSense) near-infrared fluorescence (NIRF) agents. A statistically significant increase in the ProSense and IntegriSense signal was observed within the chest region of apoE−/− mice (P < 0.05) versus C57BL/6 mice starting 25 and 22 weeks on high cholesterol diet, respectively. In a treatment study using ezetimibe (7 mg/kg), there was a statistically significant reduction in the ProSense and CatB FAST chest signal of treated (P < 0.05) versus untreated apoE−/− mice at 31 and 21 weeks on high cholesterol diet, respectively. The signal of ProSense and CatB FAST correlated with macrophage counts and was found associated with inflammatory cells by fluorescence microscopy and flow cytometry of cells dissociated from aortas. This report demonstrates that cathepsin and αvβ3 integrin NIRF agents can be used as molecular imaging biomarkers for longitudinal detection of atherosclerosis, and cathepsin agents can monitor anti-inflammatory effects of ezetimibe with applications in preclinical testing of therapeutics and potentially for early diagnosis of atherosclerosis in patients.
Collapse
|
35
|
Gadolinium-Based Contrast Agents for Vessel Wall Magnetic Resonance Imaging (MRI) of Atherosclerosis. CURRENT CARDIOVASCULAR IMAGING REPORTS 2012; 6:11-24. [PMID: 23539505 DOI: 10.1007/s12410-012-9177-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease due to atherosclerosis is the number one killer in the Western world, and threatens to become the major cause of morbidity and mortality worldwide. It is therefore paramount to develop non-invasive methods for the detection of high-risk, asymptomatic individuals before the onset of clinical symptoms or events. In the recent past, great strides have been made in the understanding of the pathological mechanisms involved in the atherosclerotic cascade down to the molecular details. This has allowed the development of contrast agents that can aid in the in vivo characterization of these processes. Gadolinium chelates are among the contrast media most commonly used in MR imaging. Originally used for MR angiography for the detection and quantification of vascular stenosis, more recently they have been applied to improve characterization of atherosclerotic plaques. In this manuscript, we will briefly review gadolinium-chelates (Gd) based contrast agents for non-invasive MR imaging of atherosclerosis. We will first describe Gd-based non-targeted FDA approved agents, used routinely in clinical practice for the evaluation of neovascularization in other diseases. Secondly, we will describe non-specific and specific targeted contrast agents, which have great potential for dissecting specific biological processes in the atherosclerotic cascade. Lastly, we will briefly compare Gd-based agents to others commonly used in MRI and to other imaging modalities.
Collapse
|
36
|
Phinikaridou A, Andia ME, Shah AM, Botnar RM. Advances in molecular imaging of atherosclerosis and myocardial infarction: shedding new light on in vivo cardiovascular biology. Am J Physiol Heart Circ Physiol 2012; 303:H1397-410. [PMID: 23064836 DOI: 10.1152/ajpheart.00583.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Molecular imaging of the cardiovascular system heavily relies on the development of new imaging probes and technologies to facilitate visualization of biological processes underlying or preceding disease. Molecular imaging is a highly active research discipline that has seen tremendous growth over the past decade. It has broadened our understanding of oncologic, neurologic, and cardiovascular diseases by providing new insights into the in vivo biology of disease progression and therapeutic interventions. As it allows for the longitudinal evaluation of biological processes, it is ideally suited for monitoring treatment response. In this review, we will concentrate on the major accomplishments and advances in the field of molecular imaging of atherosclerosis and myocardial infarction with a special focus on magnetic resonance imaging.
Collapse
Affiliation(s)
- Alkystis Phinikaridou
- Division of Imaging Science and Biomedical Engineering, King's College London, United Kingdom.
| | | | | | | |
Collapse
|
37
|
Abstract
Despite recent progress, cardiovascular and allied metabolic disorders remain a worldwide health challenge. We must identify new targets for therapy, develop new agents for clinical use, and deploy them in a clinically effective and cost-effective manner. Molecular imaging of atherosclerotic lesions has become a major experimental tool in the last decade, notably by providing a direct gateway to the processes involved in atherogenesis and its complications. This review summarizes the current status of molecular imaging approaches that target the key processes implicated in plaque formation, development, and disruption and highlights how the refinement and application of such tools might aid the development and evaluation of novel therapeutics.
Collapse
Affiliation(s)
- Thibaut Quillard
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
38
|
Wu X, Burden-Gulley SM, Yu GP, Tan M, Lindner D, Brady-Kalnay SM, Lu ZR. Synthesis and evaluation of a peptide targeted small molecular Gd-DOTA monoamide conjugate for MR molecular imaging of prostate cancer. Bioconjug Chem 2012; 23:1548-56. [PMID: 22812444 DOI: 10.1021/bc300009t] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tumor extracellular matrix has an abundance of cancer related proteins that can be used as biomarkers for cancer molecular imaging. Innovative design and development of safe and effective targeted contrast agents to these biomarkers would allow effective MR cancer molecular imaging with high spatial resolution. In this study, we synthesized a low molecular weight CLT1 peptide targeted Gd(III) chelate CLT1-dL-(Gd-DOTA)(4) specific to clotted plasma proteins in tumor stroma for cancer MR molecular imaging. CLT1-dL-(Gd-DOTA)(4) was synthesized by conjugating four Gd-DOTA monoamide chelates to a CLT1 peptide via generation 1 lysine dendrimer. The T(1) relaxivity of CLT1-dL-(Gd-DOTA)(4) was 40.4 mM(-1) s(-1) per molecule (10.1 mM(-1) s(-1) per Gd) at 37 °C and 1.5 T. Fluorescence imaging showed high binding specificity of CLT1 to orthotopic PC3 prostate tumor in mice. The contrast agent resulted in improved tumor contrast enhancement in male athymic nude mice bearing orthotopic PC3 prostate tumor xenograft at a dose of 0.03 mmol Gd/kg. The peptide targeted MRI contrast agent is promising for high-resolution MR molecular imaging of prostate tumor.
Collapse
Affiliation(s)
- Xueming Wu
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Saraste A, Laitinen I, Weidl E, Wildgruber M, Weber AW, Nekolla SG, Hölzlwimmer G, Esposito I, Walch A, Leppänen P, Lisinen I, Luppa PB, Ylä-Herttuala S, Wester HJ, Knuuti J, Schwaiger M. Diet intervention reduces uptake of αvβ3 integrin-targeted PET tracer 18F-galacto-RGD in mouse atherosclerotic plaques. J Nucl Cardiol 2012; 19:775-84. [PMID: 22527796 DOI: 10.1007/s12350-012-9554-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 03/26/2012] [Indexed: 01/16/2023]
Abstract
BACKGROUND Expression of α(v)β(3) integrin has been proposed as a marker for atherosclerotic lesion inflammation. We studied whether diet intervention reduces uptake of α(v)β(3) integrin-targeted positron emission tomography tracer (18)F-galacto-RGD in mouse atherosclerotic plaques. METHODS AND RESULTS Hypercholesterolemic LDLR(-/-) ApoB(100/100) mice on high-fat diet for 4 months were randomized to further 3 months on high-fat diet (high-fat group, n = 8) or regular mouse chow (intervention group, n = 7). Intima-media ratio describing plaque burden was comparable between intervention and high-fat groups (2.0 ± 0.5 vs 2.3 ± 0.8, P = .5). Uptake of (18)F-galacto-RGD in the aorta was lower in the intervention than high-fat group (%ID/g 0.16 vs 0.23, P < .01). Autoradiography showed 35% lower uptake of (18)F-galacto-RGD in the atherosclerotic plaques in the intervention than high-fat group (P = .007). Uptake of (18)F-galacto-RGD in plaques correlated with uptake of (3)H-deoxyglucose and nuclear density, which was lower in the intervention than high-fat group (P = .01). Flow cytometry demonstrated macrophages expressing α(v) and β(3) integrins in the aorta. CONCLUSIONS Uptake of (18)F-galacto-RGD in mouse atherosclerotic lesions was reduced by lipid-lowering diet intervention. Expression of α(v)β(3) integrin is a potential target for evaluation of therapy response in atherosclerosis.
Collapse
Affiliation(s)
- Antti Saraste
- Nuklearmedizinische Klinik und Poliklinik, Nuklearmedizinische Klinik der TU München, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str 22, 81675 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Godby RC, Van Den Berg YW, Srinivasan R, Sturm R, Hui DY, Konieczny SF, Aronow BJ, Ozhegov E, Ruf W, Versteeg HH, Bogdanov VY. Nonproteolytic properties of murine alternatively spliced tissue factor: implications for integrin-mediated signaling in murine models. Mol Med 2012; 18:771-9. [PMID: 22481268 DOI: 10.2119/molmed.2011.00416] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 03/29/2012] [Indexed: 11/06/2022] Open
Abstract
This study was performed to determine whether murine alternatively spliced tissue factor (masTF) acts analogously to human alternatively spliced tissue factor (hasTF) in promoting neovascularization via integrin ligation. Immunohistochemical evaluation of a spontaneous murine pancreatic ductal adenocarcinoma model revealed increased levels of masTF and murine full-length tissue factor (mflTF) in tumor lesions compared with benign pancreas; furthermore, masTF colocalized with mflTF in spontaneous aortic plaques of Ldlr(-/-) mice, indicating that masTF is likely involved in atherogenesis and tumorigenesis. Recombinant masTF was used to perform in vitro and ex vivo studies examining its integrin-mediated biologic activity. Murine endothelial cells (ECs) rapidly adhered to masTF in a β3-dependent fashion. Using adult and embryonic murine ECs, masTF potentiated cell migration in transwell assays. Scratch assays were performed using murine and primary human ECs; the effects of masTF and hasTF were comparable in murine ECs, but in human ECs, the effects of hasTF were more pronounced. In aortic sprouting assays, the potency of masTF-triggered vessel growth was undistinguishable from that observed with hasTF. The proangiogenic effects of masTF were found to be Ccl2-mediated, yet independent of vascular endothelial growth factor. In murine ECs, masTF and hasTF upregulated genes involved in inflammatory responses; murine and human ECs stimulated with masTF and hasTF exhibited increased interaction with murine monocytic cells under orbital shear. We propose that masTF is a functional homolog of hasTF, exerting some of its key effects via β3 integrins. Our findings have implications for the development of murine models to examine the interplay between blood coagulation, atherosclerosis and cancer.
Collapse
Affiliation(s)
- Richard C Godby
- Department of Internal Medicine, Division of Hematology/Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Burtea C, Ballet S, Laurent S, Rousseaux O, Dencausse A, Gonzalez W, Port M, Corot C, Elst LV, Muller RN. Development of a Magnetic Resonance Imaging Protocol for the Characterization of Atherosclerotic Plaque by Using Vascular Cell Adhesion Molecule-1 and Apoptosis-Targeted Ultrasmall Superparamagnetic Iron Oxide Derivatives. Arterioscler Thromb Vasc Biol 2012; 32:e36-48. [DOI: 10.1161/atvbaha.112.245415] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objective—
Acute ischemic events are often caused by the disruption of lipid-rich plaques, which are frequently not angiographically visible. Vascular cell adhesion molecule-1 and apoptotic cell-targeted peptides studied during our previous work were conjugated to ultrasmall superparamagnetic iron oxide (USPIO) (USPIO-R832 for vascular cell adhesion molecule-1 targeting; USPIO-R826 for apoptosis targeting) and assessed by magnetic resonance imaging.
Methods and Results—
Apolipoprotein E knockout mice were injected with 0.1 mmol Fe/kg body weight and were imaged on a 4.7-T Bruker magnetic resonance imaging until 24 hours after contrast agent administration. Aortic samples were then harvested and examined by histochemistry, and the magnetic resonance images and histological micrographs were analyzed with ImageJ software. The plaques enhanced by USPIO-R832 contained macrophages concentrated in the cap and a large necrotic core, whereas USPIO-R826 produced a negative enhancement of plaques rich in macrophages and neutral fats concentrated inside the plaque. Both USPIO derivatives colocalized with their target on histological sections and were able to detect plaques with a vulnerable morphology, but each one is detecting a specific environment.
Conclusion—
Our vascular cell adhesion molecule-1 and apoptotic cell targeted USPIO derivatives seem to be highly promising tools for atherosclerosis imaging contributing to the detection of vulnerable plaques. They are able to attain their target in low doses and as fast as 30 minutes after administration.
Collapse
Affiliation(s)
- Carmen Burtea
- From the Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium (C.B., S.L., L.V.E., R.N.M.); and Biological Research (S.B., A.D., W.G.), Chemical Discovery (O.R., M.P.), Research Director (C.C.), Guerbet, Research Center, Aulnay-sous-Bois, France
| | - Sébastien Ballet
- From the Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium (C.B., S.L., L.V.E., R.N.M.); and Biological Research (S.B., A.D., W.G.), Chemical Discovery (O.R., M.P.), Research Director (C.C.), Guerbet, Research Center, Aulnay-sous-Bois, France
| | - Sophie Laurent
- From the Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium (C.B., S.L., L.V.E., R.N.M.); and Biological Research (S.B., A.D., W.G.), Chemical Discovery (O.R., M.P.), Research Director (C.C.), Guerbet, Research Center, Aulnay-sous-Bois, France
| | - Olivier Rousseaux
- From the Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium (C.B., S.L., L.V.E., R.N.M.); and Biological Research (S.B., A.D., W.G.), Chemical Discovery (O.R., M.P.), Research Director (C.C.), Guerbet, Research Center, Aulnay-sous-Bois, France
| | - Anne Dencausse
- From the Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium (C.B., S.L., L.V.E., R.N.M.); and Biological Research (S.B., A.D., W.G.), Chemical Discovery (O.R., M.P.), Research Director (C.C.), Guerbet, Research Center, Aulnay-sous-Bois, France
| | - Walter Gonzalez
- From the Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium (C.B., S.L., L.V.E., R.N.M.); and Biological Research (S.B., A.D., W.G.), Chemical Discovery (O.R., M.P.), Research Director (C.C.), Guerbet, Research Center, Aulnay-sous-Bois, France
| | - Marc Port
- From the Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium (C.B., S.L., L.V.E., R.N.M.); and Biological Research (S.B., A.D., W.G.), Chemical Discovery (O.R., M.P.), Research Director (C.C.), Guerbet, Research Center, Aulnay-sous-Bois, France
| | - Claire Corot
- From the Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium (C.B., S.L., L.V.E., R.N.M.); and Biological Research (S.B., A.D., W.G.), Chemical Discovery (O.R., M.P.), Research Director (C.C.), Guerbet, Research Center, Aulnay-sous-Bois, France
| | - Luce Vander Elst
- From the Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium (C.B., S.L., L.V.E., R.N.M.); and Biological Research (S.B., A.D., W.G.), Chemical Discovery (O.R., M.P.), Research Director (C.C.), Guerbet, Research Center, Aulnay-sous-Bois, France
| | - Robert N. Muller
- From the Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium (C.B., S.L., L.V.E., R.N.M.); and Biological Research (S.B., A.D., W.G.), Chemical Discovery (O.R., M.P.), Research Director (C.C.), Guerbet, Research Center, Aulnay-sous-Bois, France
| |
Collapse
|
42
|
Verwilst P, Eliseeva SV, Vander Elst L, Burtea C, Laurent S, Petoud S, Muller RN, Parac-Vogt TN, De Borggraeve WM. A Tripodal Ruthenium–Gadolinium Metallostar as a Potential αvβ3 Integrin Specific Bimodal Imaging Contrast Agent. Inorg Chem 2012; 51:6405-11. [DOI: 10.1021/ic300717m] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Peter Verwilst
- University of Leuven, Department
of Chemistry, Celestijnenlaan 200F−P.O. Box 2404, B-3001 Heverlee,
Belgium
| | - Svetlana V. Eliseeva
- University of Leuven, Department
of Chemistry, Celestijnenlaan 200F−P.O. Box 2404, B-3001 Heverlee,
Belgium
- Centre de Biophysique
Moléculaire,
UPR 4301 CNRS, Rue Charles Sadron, 45071 Orléans Cedex 2, France
- Le STUDIUM, Institute for Advanced Studies, Orléans & Tours, France
| | - Luce Vander Elst
- NMR and Molecular Imaging Laboratory,
Department of General, Organic and Biomedical Chemistry, University
of Mons-Hainaut, B-7000 Mons, Belgium
| | - Carmen Burtea
- NMR and Molecular Imaging Laboratory,
Department of General, Organic and Biomedical Chemistry, University
of Mons-Hainaut, B-7000 Mons, Belgium
| | - Sophie Laurent
- NMR and Molecular Imaging Laboratory,
Department of General, Organic and Biomedical Chemistry, University
of Mons-Hainaut, B-7000 Mons, Belgium
| | - Stéphane Petoud
- Centre de Biophysique
Moléculaire,
UPR 4301 CNRS, Rue Charles Sadron, 45071 Orléans Cedex 2, France
| | - Robert N. Muller
- NMR and Molecular Imaging Laboratory,
Department of General, Organic and Biomedical Chemistry, University
of Mons-Hainaut, B-7000 Mons, Belgium
- Center for Microscopy and Molecular
Imaging, Rue Adrienne Bolland 8, B-6041 Charleroi, Belgium
| | - Tatjana N. Parac-Vogt
- University of Leuven, Department
of Chemistry, Celestijnenlaan 200F−P.O. Box 2404, B-3001 Heverlee,
Belgium
| | - Wim M. De Borggraeve
- University of Leuven, Department
of Chemistry, Celestijnenlaan 200F−P.O. Box 2404, B-3001 Heverlee,
Belgium
| |
Collapse
|
43
|
Longenecker CT, Hoit BD. Imaging atherosclerosis in HIV: carotid intima-media thickness and beyond. Transl Res 2012; 159:127-39. [PMID: 22340762 DOI: 10.1016/j.trsl.2011.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/17/2011] [Accepted: 10/19/2011] [Indexed: 10/15/2022]
Abstract
Chronic immune activation and inflammation are associated with an increased risk of atherosclerosis in HIV-infected patients. In this review, we discuss the role of established and novel imaging modalities to define more accurately the structure and function of inflammation-mediated atherosclerosis in the context of HIV. Historically, carotid ultrasound studies were the first to show higher rates of subclinical atherosclerosis in HIV-infected subjects versus uninfected controls. However, computed tomography is the noninvasive gold standard for imaging the coronary arteries, and studies in HIV suggest a higher prevalence of noncalcified plaque. Endothelial dysfunction can be quantified by measuring flow-mediated brachial artery dilation by ultrasound and has been used extensively in antiretroviral switching trials and small pilot trials of therapeutics to assess cardiovascular risk in this population. In the future, novel imaging modalities such as intracoronary optical coherence tomography, positron emission tomography imaging of (18)F-fluorodeoxyglucose uptake, and molecular-targeted magnetic resonance imaging will characterize the burden of vulnerable plaque and other unique features of inflammatory atherosclerosis in HIV.
Collapse
Affiliation(s)
- Chris T Longenecker
- Harrington-McLaughlin Heart and Vascular Institute, University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | | |
Collapse
|
44
|
Yao Y, Jiang Y, Sheng Z, Zhang Y, An Y, Yan F, Ma G, Liu N, Teng G, Cheng Z. Analysis of in situ and ex vivo αVβ3 integrin expression during experimental carotid atherogenesis. Int J Nanomedicine 2012; 7:641-9. [PMID: 22334786 PMCID: PMC3278228 DOI: 10.2147/ijn.s28065] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Mural inflammation has been shown to contribute to the development of plaque, with the α(V)β(3) integrin highly expressed in atherosclerotic plaques. We herein examined α(V)β(3) integrin expression as a function of carotid atherosclerosis formation in the apolipoprotein E-deficient (apoE(-/-)) mouse. METHODS AND RESULTS Constrictive collars were placed around the left common carotid arteries of apo E(-/-) mice maintained on a high-fat diet (n = 14). Before and 21 days following collar placement, in vivo serial magnetic resonance imaging (MRI) measurements of the carotid aortic diameter were performed using a 7T magnetic resonance (MR) scanner. Near- infrared fluorescence (NIRF) imaging was performed (n = 6) using an in vivo imaging system 0-24 hours following administration of 1.0 nmol c(RGDyK)-Cy5.5 via the tail vein. A competition experiment was performed by the co-injection of a saturating dose of bicyclic RGD peptide H-Glu[cyclo(Arg-Gly-Asp-D-Tyr-Lys)]2 (n = 3). Following image acquisition and sacrifice at 24 hours after injection, carotid arteries were harvested for histological analyses. Neointima formation and arterial remodeling in the carotid arteries of apoE(-/-) mice were induced by the placement of a constrictive collar. Significantly greater fluorescent signals were obtained from constrictive collar left common carotid arteries as compared to uninvolved aortic segments in constrictive collar mice. Binding to stenotic lesions was efficiently blocked in competition experiments. Immunostaining confirmed the presence of mural α(V)β(3) integrin expression in macrophages in the neointima. Signal intensity increased in a macrophage density-dependent fashion in the stenotic segments. CONCLUSION Mural α(V)β(3) integrin expression, as determined using RGD-Cy5.5 near-infrared optical imaging, was increased in carotid arteries with constrictive collars in experimental mice. This expression can estimate the macrophage-bound inflammatory activity of atherosclerotic lesions.
Collapse
Affiliation(s)
- Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Translation initiation factors and active sites of protein synthesis co-localize at the leading edge of migrating fibroblasts. Biochem J 2011; 438:217-27. [PMID: 21539520 DOI: 10.1042/bj20110435] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cell migration is a highly controlled essential cellular process, often dysregulated in tumour cells, dynamically controlled by the architecture of the cell. Studies involving cellular fractionation and microarray profiling have previously identified functionally distinct mRNA populations specific to cellular organelles and architectural compartments. However, the interaction between the translational machinery itself and cellular structures is relatively unexplored. To help understand the role for the compartmentalization and localized protein synthesis in cell migration, we have used scanning confocal microscopy, immunofluorescence and a novel ribopuromycylation method to visualize translating ribosomes. In the present study we show that eIFs (eukaryotic initiation factors) localize to the leading edge of migrating MRC5 fibroblasts in a process dependent on TGN (trans-Golgi network) to plasma membrane vesicle transport. We show that eIF4E and eIF4GI are associated with the Golgi apparatus and membrane microdomains, and that a proportion of these proteins co-localize to sites of active translation at the leading edge of migrating cells.
Collapse
|
46
|
Mouse phenotyping with MRI. Methods Mol Biol 2011. [PMID: 21874500 DOI: 10.1007/978-1-61779-219-9_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The field of mouse phenotyping with magnetic resonance imaging (MRI) is rapidly growing, motivated by the need for improved tools for characterizing and evaluating mouse models of human disease. Image results can provide important comparisons of human conditions with mouse disease models, evaluations of treatment, development or disease progression, as well as direction for histological or other investigations. Effective mouse MRI studies require attention to many aspects of experiment design. In this chapter, we provide details and discussion of important practical considerations: hardware requirements, mouse handling for in vivo imaging, specimen preparation for ex vivo imaging, sequence and contrast agent selection, study size, and quantitative image analysis. We focus particularly on anatomical phenotyping, an important and accessible application that has shown a high potential for impact in many mouse models at our imaging center.
Collapse
|
47
|
Small GR, Ruddy TD. PET imaging of aortic atherosclerosis: Is combined imaging of plaque anatomy and function an amaranthine quest or conceivable reality? J Nucl Cardiol 2011; 18:717-28. [PMID: 21553158 DOI: 10.1007/s12350-011-9385-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Traditionally, blood vessels have been studied using contrast luminography to determine the site, extent and severity of luminal compromise by atherosclerotic deposits. Similar anatomical data can now be acquired non-invasively using ultrasound, computed tomography or magnetic resonance imaging. Plaque stability is an important determinant of subsequent vascular events and currently functional data on the stability of plaque is less well provided by these methods. The search for non-invasive techniques to image combined plaque anatomy and function has been pursued with visionary anticipation. This expectation may soon be realised as imaging with radionuclide-labelled atheroma-targeted contrast agents has demonstrated that plaque functional characteristics can now be shown. Increasingly positron emission tomography/computed tomography (PET/CT) imaging with (18)F fluorodexoyglucose (FDG) and other radionuclides is being used to determine culprit plaques in complex clinically scenarios. Clinically, this information may prove extremely valuable in the assessment of stable and unstable patients and its use in prime time medical practice is eagerly awaited. We will discuss the current clinical applications of functional atheroma imaging in the aorta and highlight the promising preclinical data on novel image biomarkers of plaque instability. If clinical science is able to successfully translate these advances in vascular imaging from the bench to the bedside, a new paradigm will be achieved in cardiovascular diagnostics.
Collapse
Affiliation(s)
- Gary R Small
- Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON, K1Y 4W7, Canada
| | | |
Collapse
|
48
|
Vancraeynest D, Pasquet A, Roelants V, Gerber BL, Vanoverschelde JLJ. Imaging the vulnerable plaque. J Am Coll Cardiol 2011; 57:1961-79. [PMID: 21565634 DOI: 10.1016/j.jacc.2011.02.018] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 02/09/2011] [Accepted: 02/15/2011] [Indexed: 12/27/2022]
Abstract
Cardiovascular diseases are still the primary causes of mortality in the United States and in Western Europe. Arterial thrombosis is triggered by a ruptured atherosclerotic plaque and precipitates an acute vascular event, which is responsible for the high mortality rate. These rupture-prone plaques are called "vulnerable plaques." During the past decades, much effort has been put toward accurately detecting the presence of vulnerable plaques with different imaging techniques. In this review, we provide an overview of the currently available invasive and noninvasive imaging modalities used to detect vulnerable plaques. We will discuss the upcoming challenges in translating these techniques into clinical practice and in assigning them their exact place in the decision-making process.
Collapse
Affiliation(s)
- David Vancraeynest
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Cliniques, Université Catholique de Louvain, Brussels, Belgium
| | | | | | | | | |
Collapse
|
49
|
Verwilst P, Eliseeva SV, Carron S, Vander Elst L, Burtea C, Dehaen G, Laurent S, Binnemans K, Muller RN, Parac-Vogt TN, De Borggraeve WM. A Modular Approach towards the Synthesis of Target-Specific MRI Contrast Agents. Eur J Inorg Chem 2011. [DOI: 10.1002/ejic.201100575] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
50
|
68Ga-DOTA-RGD peptide: biodistribution and binding into atherosclerotic plaques in mice. Eur J Nucl Med Mol Imaging 2011; 36:2058-67. [PMID: 19629477 DOI: 10.1007/s00259-009-1220-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Accepted: 07/01/2009] [Indexed: 10/20/2022]
Abstract
PURPOSE Increased expression of αvβ3/αvβ5 integrin is involved in angiogenesis and the inflammatory process in atherosclerotic plaques. The novel 68Ga-DOTA-RGD peptide binds with high affinity to αvβ3/αvβ5 integrin. The aim of this study was to investigate the uptake of the 68Ga-DOTA-RGD peptide in atherosclerotic plaques. METHODS Uptake of intravenously administered 68Ga-DOTA-RGD peptide was studied ex vivo in excised tissue samples and aortic sections of LDLR-/-ApoB100/100 atherosclerotic mice. The uptake of the tracer in aortic cryosections was examined by using digital autoradiography. Subsequently, the autoradiographs were combined with histological and immunohistological analysis of the sections. RESULTS DOTA-RGD peptide was successfully labelled with the generator-produced 68Ga. The tracer had reasonably good specific radioactivity (8.7 ± 1.1 GBq/μmol) and was quite stable in vivo. According to ex vivo biodistribution results, 68Ga-DOTA-RGD was cleared rapidly from the blood circulation and excreted through the kidneys to the urine with high radioactivity in the intestine, lungs, spleen and liver. Autoradiography results showed significantly higher uptake of 68Ga-DOTA-RGD peptide in the atherosclerotic plaques compared to healthy vessel wall (mean ratio ± SD 1.4 ± 0.1, p = 0.0004). Conclusion We observed that 68Ga-DOTA-RGD is accumulated into the plaques of atherosclerotic mice. However, this data only shows the feasibility of the approach, while the clinical significance still remains to be proven. Further studies are warranted to assess the uptake of this tracer into human atherosclerotic plaques.
Collapse
|