1
|
Shabbir R, Hayat Malik MN, Zaib M, Alamgeer, Jahan S, Khan MT. Amino Acid Conjugates of 2-Mercaptobenzimidazole Ameliorates High-Fat Diet-Induced Hyperlipidemia in Rats via Attenuation of HMGCR, APOB, and PCSK9. ACS OMEGA 2022; 7:40502-40511. [PMID: 36385864 PMCID: PMC9647896 DOI: 10.1021/acsomega.2c05735] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/19/2022] [Indexed: 07/28/2023]
Abstract
PURPOSE This study was designed to explore the antihyperlipidemic effects of amino acid derivatives of 2-mercaptobenzimidazole (4J and 4K) in high-fat diet (HFD)-fed rats. METHODS Male Sprague-Dawley rats were divided into nine groups which received either standard diet or HFD for 28 days. Blood samples were taken on 27th day from HFD-fed rats to ensure hyperlipidemia. HFD-induced hyperlipidemic rats later received daily dosing of either vehicle or simvastatin (SIM; 20 mg/kg) or 4J/4K compounds (10, 20, and 30 mg/kg) for 12 consecutive days. On 40th day, animals were sacrificed, and blood samples were collected for the determination of serum lipid profile and liver function parameters. Liver samples were harvested for histopathological, antioxidant, and qPCR analyses. Molecular docking of tested compounds with HMGCR was also performed to assess the binding affinities. RESULTS 4J and 4K dose dependently decreased serum total cholesterol, triglycerides, low-density lipoprotein, very low-density lipoproteins, alanine transaminase (ALT), and aspartate aminotransferase (AST) levels while significantly alleviated high-density lipoproteins. However, SIM failed to reduce AST and ALT levels. Moreover, tested compounds displayed antioxidant effects by inducing superoxide dismutase and glutathione levels. Histopathology data also displayed protective effects of 4J and 4K against HFD-induced fatty changes and hepatic damage. In addition, 4J and 4K downregulated transcript levels of HMGCR, APOB, PCSK9, and VCAM1, and molecular docking analysis also supported the experimental data. CONCLUSION It is conceivable from this study that 4J and 4K exert their antihyperlipidemic effects by modulating multiple targets regulating lipid levels.
Collapse
Affiliation(s)
- Ramla Shabbir
- Department
of Pharmacology, Faculty of Pharmacy, The
University of Lahore, Lahore 54590, Pakistan
| | | | - Maryam Zaib
- Department
of Pharmacology, Faculty of Pharmacy, The
University of Lahore, Lahore 54590, Pakistan
| | - Alamgeer
- University
College of Pharmacy, University of the Punjab, Lahore 54590, Pakistan
| | - Shah Jahan
- Department
of Immunology, University of Health Sciences, Lahore 54600, Pakistan
| | - Muhammad Tariq Khan
- Department
of Pharmacy, Capital University of Science
and Technology, Islamabad 44000, Pakistan
| |
Collapse
|
2
|
Qiu P, Xu Y. The construction of multifunctional nanoparticles system for dual-modal imaging and arteriosclerosis targeted therapy. Am J Transl Res 2021; 13:4026-4039. [PMID: 34149996 PMCID: PMC8205662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/14/2021] [Indexed: 06/12/2023]
Abstract
Atherosclerosis is a major risk factor for the development of cardiovascular disease. Unfortunately, due to relatively low sensitivities and poor resolution, the results of surgical resection are often largely unsatisfactory. Moreover, many chemotherapeutic agents, such as curcumin (Cur), are restricted by the low blood-brain barrier (BBB) permeability. Recently, nanotechnology proposes new opportunities to overcome these treatment barriers. In this study, superparamagnetic iron oxide nanoparticles (SPIO) was prepared by the high-temperature solid-state method, and then loaded into amphiphilic polymer DSPE-PEG to form SDP nanoparticles by hydrogen bonding in oil phase. The curcumin was encapsulated in SDP nanoparticles by self-assembly. Finally, vascular cell adhesion molecule-1 (VCAM-1) and Cy5.5 were conjugated on into SDP/Cur nanoparticles by amidation reaction. The average particle size of the prepared multifunctional SDP-VCAM-1/Cur/Cy5.5 nanoparticles is 124.4 nm, which can provide the sustained release of Cur. Moreover, the nanoparticles are proved to have superparamagnetic properties and fluorescence properties. In vitro cell experiments show that nanoparticles have excellent biocompatibility, blood compatibility and macrophage targeting. These results show that SDP-VCAM-1/Cur/Cy5.5 nanoparticles can be used not only as dual imaging probe for magnetic resonance (MR) and fluorescence imaging, but also as carriers to deliver chemotherapeutic drugs to inflammatory tissue, thus providing a promising opportunity for the treatment, molecular imaging and targeted therapy in atherosclerosis due to their established specificity and safety.
Collapse
Affiliation(s)
- Pengda Qiu
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University No. 63, Duobao Road, Liwan District, Guangzhou 510150, Guangdong, P. R. China
| | - Yunhong Xu
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University No. 63, Duobao Road, Liwan District, Guangzhou 510150, Guangdong, P. R. China
| |
Collapse
|
3
|
VCAM-1 Target in Non-Invasive Imaging for the Detection of Atherosclerotic Plaques. BIOLOGY 2020; 9:biology9110368. [PMID: 33138124 PMCID: PMC7692297 DOI: 10.3390/biology9110368] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023]
Abstract
Simple Summary Cardiovascular diseases are the first cause of morbimortality worldwide. They are mainly caused by atherosclerosis, with progressive plaque formation in the arterial wall. In this context, several imaging techniques have been developed to screen, detect and quantify atherosclerosis. Early screening improves primary prevention and promotes the prescription of adequate medication before adverse clinical events. In this review, we focus on the imaging of vascular cell adhesion molecule-1, an adhesion molecule involved in the first stages of the development of atherosclerosis. This molecule could therefore be a promising target to detect early atherosclerosis non-invasively. Potential clinical applications are critically discussed. Abstract Atherosclerosis is a progressive chronic arterial disease characterised by atheromatous plaque formation in the intima of the arterial wall. Several invasive and non-invasive imaging techniques have been developed to detect and characterise atherosclerosis in the vessel wall: anatomic/structural imaging, functional imaging and molecular imaging. In molecular imaging, vascular cell adhesion molecule-1 (VCAM-1) is a promising target for the non-invasive detection of atherosclerosis and for the assessment of novel antiatherogenic treatments. VCAM-1 is an adhesion molecule expressed on the activated endothelial surface that binds leucocyte ligands and therefore promotes leucocyte adhesion and transendothelial migration. Hence, for several years, there has been an increase in molecular imaging methods for detecting VCAM-1 in MRI, PET, SPECT, optical imaging and ultrasound. The use of microparticles of iron oxide (MPIO), ultrasmall superparamagnetic iron oxide (USPIO), microbubbles, echogenic immunoliposomes, peptides, nanobodies and other nanoparticles has been described. However, these approaches have been tested in animal models, and the remaining challenge is bench-to-bedside development and clinical applicability.
Collapse
|
4
|
Current Advances in the Diagnostic Imaging of Atherosclerosis: Insights into the Pathophysiology of Vulnerable Plaque. Int J Mol Sci 2020; 21:ijms21082992. [PMID: 32340284 PMCID: PMC7216001 DOI: 10.3390/ijms21082992] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/02/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is a lipoprotein-driven inflammatory disorder leading to a plaque formation at specific sites of the arterial tree. After decades of slow progression, atherosclerotic plaque rupture and formation of thrombi are the major factors responsible for the development of acute coronary syndromes (ACSs). In this regard, the detection of high-risk (vulnerable) plaques is an ultimate goal in the management of atherosclerosis and cardiovascular diseases (CVDs). Vulnerable plaques have specific morphological features that make their detection possible, hence allowing for identification of high-risk patients and the tailoring of therapy. Plaque ruptures predominantly occur amongst lesions characterized as thin-cap fibroatheromas (TCFA). Plaques without a rupture, such as plaque erosions, are also thrombi-forming lesions on the most frequent pathological intimal thickening or fibroatheromas. Many attempts to comprehensively identify vulnerable plaque constituents with different invasive and non-invasive imaging technologies have been made. In this review, advantages and limitations of invasive and non-invasive imaging modalities currently available for the identification of plaque components and morphologic features associated with plaque vulnerability, as well as their clinical diagnostic and prognostic value, were discussed.
Collapse
|
5
|
Molecular imaging of inflammation - Current and emerging technologies for diagnosis and treatment. Pharmacol Ther 2020; 211:107550. [PMID: 32325067 DOI: 10.1016/j.pharmthera.2020.107550] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022]
Abstract
Inflammation is a key factor in multiple diseases including primary immune-mediated inflammatory diseases e.g. rheumatoid arthritis but also, less obviously, in many other common conditions, e.g. cardiovascular disease and diabetes. Together, chronic inflammatory diseases contribute to the majority of global morbidity and mortality. However, our understanding of the underlying processes by which the immune response is activated and sustained is limited by a lack of cellular and molecular information obtained in situ. Molecular imaging is the visualization, detection and quantification of molecules in the body. The ability to reveal information on inflammatory biomarkers, pathways and cells can improve disease diagnosis, guide and monitor therapeutic intervention and identify new targets for research. The optimum molecular imaging modality will possess high sensitivity and high resolution and be capable of non-invasive quantitative imaging of multiple disease biomarkers while maintaining an acceptable safety profile. The mainstays of current clinical imaging are computed tomography (CT), magnetic resonance imaging (MRI), ultrasound (US) and nuclear imaging such as positron emission tomography (PET). However, none of these have yet progressed to routine clinical use in the molecular imaging of inflammation, therefore new approaches are required to meet this goal. This review sets out the respective merits and limitations of both established and emerging imaging modalities as clinically useful molecular imaging tools in addition to potential theranostic applications.
Collapse
|
6
|
Peptide-based nanosystems for vascular cell adhesion molecule-1 targeting: a real opportunity for therapeutic and diagnostic agents in inflammation associated disorders. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
7
|
Li M, Sagastume EE, Lee D, McAlister D, DeGraffenreid AJ, Olewine KR, Graves S, Copping R, Mirzadeh S, Zimmerman BE, Larsen R, Johnson FL, Schultz MK. 203/212Pb Theranostic Radiopharmaceuticals for Image-guided Radionuclide Therapy for Cancer. Curr Med Chem 2020; 27:7003-7031. [PMID: 32720598 PMCID: PMC10613023 DOI: 10.2174/0929867327999200727190423] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/25/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
Receptor-targeted image-guided Radionuclide Therapy (TRT) is increasingly recognized as a promising approach to cancer treatment. In particular, the potential for clinical translation of receptor-targeted alpha-particle therapy is receiving considerable attention as an approach that can improve outcomes for cancer patients. Higher Linear-energy Transfer (LET) of alpha-particles (compared to beta particles) for this purpose results in an increased incidence of double-strand DNA breaks and improved-localized cancer-cell damage. Recent clinical studies provide compelling evidence that alpha-TRT has the potential to deliver a significantly more potent anti-cancer effect compared with beta-TRT. Generator-produced 212Pb (which decays to alpha emitters 212Bi and 212Po) is a particularly promising radionuclide for receptor-targeted alpha-particle therapy. A second attractive feature that distinguishes 212Pb alpha-TRT from other available radionuclides is the possibility to employ elementallymatched isotope 203Pb as an imaging surrogate in place of the therapeutic radionuclide. As direct non-invasive measurement of alpha-particle emissions cannot be conducted using current medical scanner technology, the imaging surrogate allows for a pharmacologically-inactive determination of the pharmacokinetics and biodistribution of TRT candidate ligands in advance of treatment. Thus, elementally-matched 203Pb labeled radiopharmaceuticals can be used to identify patients who may benefit from 212Pb alpha-TRT and apply appropriate dosimetry and treatment planning in advance of the therapy. In this review, we provide a brief history on the use of these isotopes for cancer therapy; describe the decay and chemical characteristics of 203/212Pb for their use in cancer theranostics and methodologies applied for production and purification of these isotopes for radiopharmaceutical production. In addition, a medical physics and dosimetry perspective is provided that highlights the potential of 212Pb for alpha-TRT and the expected safety for 203Pb surrogate imaging. Recent and current preclinical and clinical studies are presented. The sum of the findings herein and observations presented provide evidence that the 203Pb/212Pb theranostic pair has a promising future for use in radiopharmaceutical theranostic therapies for cancer.
Collapse
Affiliation(s)
- Mengshi Li
- Department of Radiology, The University of Iowa, Iowa City, IA USA
- Viewpoint Molecular Targeting, Inc., Coralville, IA USA
| | | | - Dongyoul Lee
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA
| | | | | | | | - Stephen Graves
- Department of Radiology, The University of Iowa, Iowa City, IA USA
| | - Roy Copping
- Oak Ridge National Laboratory, The US Department of Energy, Oak Ridge TN USA
| | - Saed Mirzadeh
- Oak Ridge National Laboratory, The US Department of Energy, Oak Ridge TN USA
| | - Brian E. Zimmerman
- The National Institute of Standards and Technology, Gaithersburg, MD, USA
| | | | - Frances L. Johnson
- Viewpoint Molecular Targeting, Inc., Coralville, IA USA
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa USA
| | - Michael K. Schultz
- Department of Radiology, The University of Iowa, Iowa City, IA USA
- Viewpoint Molecular Targeting, Inc., Coralville, IA USA
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA
- Department of Chemistry, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
8
|
Wei Q, Wang J, Shi W, Zhang B, Jiang H, Du M, Mei H, Hu Y. Improved in vivo detection of atherosclerotic plaques with a tissue factor-targeting magnetic nanoprobe. Acta Biomater 2019; 90:324-336. [PMID: 30954623 DOI: 10.1016/j.actbio.2019.04.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 03/22/2019] [Accepted: 04/03/2019] [Indexed: 11/19/2022]
Abstract
Rupture of atherosclerotic plaques causes acute cardiovascular and cerebrovascular pathology. Tissue factor (TF) is a key factor that affects the development of atherosclerotic plaques and the formation of thrombus and thus constitutes a potential target for the detection of atherosclerotic plaques. In this study, the conjugation of the fusion protein 'enhanced green fluorescent protein with the first epidermal growth factor domain' (EGFP-EGF1) and superparamagnetic iron oxide nanoparticles (EGFP-EGF1-SPIONs) was explored for molecular imaging of TF-positive atherosclerotic plaques. EGFP-EGF1-SPIONs showed improved accuracy, superior contrast effects, and better cytocompatibility compared with common contrast agents in the detection of atherosclerotic plaques of apolipoprotein E knockout (ApoE-/-) mice using magnetic resonance imaging. In conclusion, EGFP-EGF1-SPION is a promising TF-targeting nanoprobe to precisely and specifically detect atherosclerotic plaques, which may improve molecular imaging diagnosis of cardiovascular and cerebrovascular events for the comprehensive evaluation of atherosclerosis. STATEMENT OF SIGNIFICANCE: Traditional methods can only display the status of atherosclerosis, but not forecast the progress of lesions efficiently. It remains challenging to evaluate the plaques specifically and sensitively. In this study, we constructed a tissue factor-targeted magnetic nanoprobe to specifically detect plaques by magnetic resonance imaging in vivo, which will improve the diagnostic technology for atherosclerotic plaques and offer molecular level guidance to treat atherosclerosis. Furthermore, this strategy has critical clinical significance on prevention, diagnosis and therapeutic evaluation of cardio-cerebral vascular events.
Collapse
Affiliation(s)
- Qiuzhe Wei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China; Targeted Biotherapy Key Laboratory of Ministry of Education, Wuhan, Hubei, PR China
| | - Jing Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Wei Shi
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China; Targeted Biotherapy Key Laboratory of Ministry of Education, Wuhan, Hubei, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China; Targeted Biotherapy Key Laboratory of Ministry of Education, Wuhan, Hubei, PR China
| | - Huiwen Jiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China
| | - Mengyi Du
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China; Targeted Biotherapy Key Laboratory of Ministry of Education, Wuhan, Hubei, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China; Targeted Biotherapy Key Laboratory of Ministry of Education, Wuhan, Hubei, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China.
| |
Collapse
|
9
|
Zaman RT, Yousefi S, Chibana H, Ikeno F, Long SR, Gambhir SS, Chin FT, McConnell MV, Xing L, Yeung A. In Vivo Translation of the CIRPI System: Revealing Molecular Pathology of Rabbit Aortic Atherosclerotic Plaques. J Nucl Med 2019; 60:1308-1316. [PMID: 30737298 DOI: 10.2967/jnumed.118.222471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/13/2018] [Indexed: 01/13/2023] Open
Abstract
Thin-cap fibroatheroma (TCFA) are the unstable lesions in coronary artery disease that are prone to rupture, resulting in substantial morbidity and mortality worldwide. However, their small size and complex morphologic and biologic features make early detection and risk assessment difficult. We tested our newly developed catheter-based Circumferential-Intravascular-Radioluminescence-Photoacoustic-Imaging (CIRPI) system in vivo to enable detection and characterization of vulnerable plaque structure and biology in rabbit abdominal aorta. Methods: The CIRPI system includes a novel optical probe combining circumferential radioluminescence imaging and photoacoustic tomography (PAT). The probe's CaF2:Eu-based scintillating imaging window captures radioluminescence images (360° view) of plaques by detecting β-particles during 18F-FDG decay. A tunable laser-based PAT characterizes tissue constituents of plaque at 7 different wavelengths-540 and 560 nm (calcification), 920 nm (cholesteryl ester), 1040 nm (phospholipids), 1180 nm (elastin/collagen), 1210 nm (cholesterol), and 1235 nm (triglyceride). A single B-scan is concatenated from 330 A-lines captured during a 360° rotation. The abdominal aorta was imaged in vivo in both atherosclerotic rabbits (Watanabe Heritable Hyper Lipidemic [WHHL], 13-mo-old male, n = 5) and controls (New Zealand White, n = 2). Rabbits were fasted for 6 h before 5.55 × 107 Bq (1.5 mCi) of 18F-FDG were injected 1 h before the imaging procedure. Rabbits were anesthetized, and the right or left common carotid artery was surgically exposed. An 8 French catheter sheath was inserted into the common carotid artery, and a 0.035-cm (0.014-in) guidewire was advanced to the iliac artery, guided by x-ray fluoroscopy. A bare metal stent was implanted in the dorsal abdominal aorta as a landmark, followed by the 7 French imaging catheters that were advanced up to the proximal stent edge. Our CIRPI and clinical optical coherence tomography (OCT) were performed using pullback and nonocclusive flushing techniques. After imaging with the CIRPI system, the descending aorta was flushed with contrast agent, and OCT images were obtained with a pullback speed of 20 mm/s, providing images at 100 frames/s. Results were verified with histochemical analysis. Results: Our CIRPI system successfully detected the locations and characterized both stable and vulnerable aortic plaques in vivo among all WHHL rabbits. Calcification was detected from the stable plaque (540 and 560 nm), whereas TCFA exhibited phospholipids/cholesterol (1040 nm, 1210 nm). These findings were further verified with the clinical OCT system showing an area of low attenuation filled with lipids within TCFA. PAT images illustrated broken elastic fiber/collagen that could be verified with the histochemical analysis. All WHHL rabbits exhibited sparse to severe macrophages. Only 4 rabbits showed both moderate-to-severe level of calcifications and cholesterol clefts. However, all rabbits exhibited broken elastic fibers and collagen deposition. Control rabbits showed normal wall thickness with no presence of plaque tissue compositions. These findings were verified with OCT and histochemical analysis. Conclusion: Our novel multimodality hybrid system has been successfully translated to in vivo evaluation of atherosclerotic plaque structure and biology in a preclinical rabbit model. This system proposed a paradigm shift that unites molecular and pathologic imaging technologies. Therefore, the system may enhance the clinical evaluation of TCFA, as well as expand our understanding of coronary artery disease.
Collapse
Affiliation(s)
- Raiyan T Zaman
- Department of Radiology, Harvard Medical School, Boston, Massachusetts .,Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, Massachusetts.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Siavash Yousefi
- Division of Medical Physics, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Hidetoshi Chibana
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Fumiaki Ikeno
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Steven R Long
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Sanjiv S Gambhir
- Department of Radiology, Stanford University School of Medicine, Stanford, California.,Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, California.,Department of Bioengineering, Stanford University Schools of Medicine and of Engineering, Stanford, California; and
| | - Frederick T Chin
- Department of Radiology, Stanford University School of Medicine, Stanford, California.,Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, California
| | - Michael V McConnell
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California.,Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, California.,Verily Life Sciences, San Francisco, California
| | - Lei Xing
- Division of Medical Physics, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.,Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, California
| | - Alan Yeung
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
10
|
Mota R, Campen MJ, Cuellar ME, Garver WS, Hesterman J, Qutaish M, Daniels T, Nysus M, Wagner CR, Norenberg JP. 111In-DANBIRT In Vivo Molecular Imaging of Inflammatory Cells in Atherosclerosis. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:6508724. [PMID: 30538613 PMCID: PMC6257909 DOI: 10.1155/2018/6508724] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/24/2018] [Accepted: 10/23/2018] [Indexed: 11/25/2022]
Abstract
Atherosclerosis-related morbidity and mortality remain a global concern. Atherosclerotic disease follows a slow and silent progression, and the transition from early-stage lesions to vulnerable plaques remains difficult to diagnose. Inflammation is a key component of the development of atherosclerotic plaque and consequent life-threatening complications. This study assessed 111In-DANBIRT as an in vivo, noninvasive SPECT/CT imaging probe targeting an inflammatory marker, Lymphocyte Function Associated Antigen-1 (LFA-1), in atherosclerotic plaques. Methods. Selective binding of 111In-DANBIRT was assessed using Sprague-Dawley rats exposed to filtered air and ozone (1 ppm) by inhalation for 4 hours to induce a circulating leukocytosis and neutrophilia in peripheral blood. After 24 hours, whole blood was collected and incubated with radiolabeled DANBIRT (68Ga-DANBIRT and 111In-DANBIRT). Isolated cell component smeared slides using cytospin technique were stained with Wright-Giemsa stain. Apolipoprotein E-deficient (apoE-/-) mice were fed either a normal diet or a high-fat diet (HFD) for 8 weeks. Longitudinal SPECT/CT imaging was performed 3 hours after administration at baseline, 4, and 8 weeks of HFD diet, followed by tissue harvesting for biodistribution, serum lipid analysis, and histology. 3D autoradiography was performed in both groups 24 hours after administration of 111In-DANBIRT. Results. Increased specific uptake of radiolabeled DANBIRT by neutrophils in the ozone-exposed group was evidenced by the acute immune response due to 4-hour ozone exposure. Molecular imaging performed at 3 hours using SPECT/CT imaging evidenced an exponential longitudinal increase in 111In-DANBIRT uptake in atherosclerosis lesions in HFD-fed mice compared to normal-diet-fed mice. Such results were consistent with increased immune response to vascular injury in cardiovascular and also immune tissues, correlated by 24 hours after administration of 3D autoradiography. Histologic analysis confirmed atherosclerotic disease progression with an increased vascular lesion area in HFD-fed mice compared to normal-diet-fed mice. Conclusion. 111In-DANBIRT is a promising molecular imaging probe to assess inflammation in evolving atheroma and atherosclerotic plaque.
Collapse
Affiliation(s)
- Roberto Mota
- Radiopharmaceutical Sciences, University of New Mexico (UNM), Albuquerque, NM, USA
- Department of Surgery, Division of Vascular Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Matthew E. Cuellar
- Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - William S. Garver
- Department of Biochemistry & Molecular Biology, School of Medicine, UNM, Albuquerque, NM, USA
| | | | | | - Tamara Daniels
- Radiopharmaceutical Sciences, University of New Mexico (UNM), Albuquerque, NM, USA
| | - Monique Nysus
- Radiopharmaceutical Sciences, University of New Mexico (UNM), Albuquerque, NM, USA
| | - Carston R. Wagner
- Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey P. Norenberg
- Radiopharmaceutical Sciences, University of New Mexico (UNM), Albuquerque, NM, USA
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, UNM, Albuquerque, NM, USA
| |
Collapse
|
11
|
Zaman RT, Yousefi S, Long SR, Saito T, Mandella M, Qiu Z, Chen R, Contag CH, Gambhir SS, Chin FT, Khuri-Yakub BT, McConnell MV, Shung KK, Xing L. A Dual-Modality Hybrid Imaging System Harnesses Radioluminescence and Sound to Reveal Molecular Pathology of Atherosclerotic Plaques. Sci Rep 2018; 8:8992. [PMID: 29895966 PMCID: PMC5997702 DOI: 10.1038/s41598-018-26696-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 05/14/2018] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis is a progressive inflammatory condition caused by an unstable lesion, called thin-cap fibro atheromata (TCFA) that underlies coronary artery disease (CAD)-one of the leading causes of death worldwide. Therefore, early clinical diagnosis and effective risk stratification is important for CAD management as well as preventing progression to catastrophic events. However, early detection could be difficult due to their small size, motion, obscuring 18F-FDG uptake by adjacent myocardium, and complex morphological/biological features. To overcome these limitations, we developed a catheter-based Circumferential-Intravascular-Radioluminescence-Photoacoustic-Imaging (CIRPI) system that can detect vulnerable plaques in coronary arteries and characterizes them with respect to pathology and biology. Our CIRPI system combined two imaging modalities: Circumferential Radioluminescence Imaging (CRI) and PhotoAcoustic Tomography (PAT) within a novel optical probe. The probe's CaF2:Eu based scintillating imaging window provides a 360° view of human (n = 7) and murine carotid (n = 10) arterial plaques by converting β-particles into visible photons during 18F-FDG decay. A 60× and 63× higher radioluminescent signals were detected from the human and murine plaque inflammations, respectively, compared to the control. The system's photoacoustic imaging provided a comprehensive analysis of the plaque compositions and its morphologic information. These results were further verified with IVIS-200, immunohistochemical analysis, and autoradiography.
Collapse
Affiliation(s)
- Raiyan T Zaman
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, USA.
- Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, USA.
- Department of Radiology, Harvard medical School, Boston, MA, 02115, USA.
- Massachusetts General Hospital 149 13th Street, Room 5406 Charlestown, Massachusetts, 02129, USA.
| | - Siavash Yousefi
- Division of Medical Physics, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, USA
| | - Steven R Long
- Department of Pathology, Stanford University School of Medicine, Stanford, USA
| | - Toshinobu Saito
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Michael Mandella
- Department of Pediatrics (Neonatology), Stanford University School of Medicine, Stanford, USA
| | - Zhen Qiu
- Department of Radiology, Stanford University School of Medicine, Stanford, USA
- Michigan State University, Michigan, USA
| | - Ruimin Chen
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Stanford, USA
| | - Christopher H Contag
- Department of Pediatrics (Neonatology), Stanford University School of Medicine, Stanford, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, USA
- Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, USA
- Department of Bioengineering, Stanford University Schools of Medicine and of Engineering, Stanford, USA
| | - Sanjiv S Gambhir
- Department of Radiology, Stanford University School of Medicine, Stanford, USA
- Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, USA
- Department of Bioengineering, Stanford University Schools of Medicine and of Engineering, Stanford, USA
| | - Frederick T Chin
- Department of Radiology, Stanford University School of Medicine, Stanford, USA
- Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, USA
| | | | - Michael V McConnell
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, USA
- Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, USA
| | - K Kirk Shung
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Stanford, USA
| | - Lei Xing
- Division of Medical Physics, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, USA
- Molecular Imaging Program at Stanford University (MIPS), Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
12
|
Moccetti F, Weinkauf CC, Davidson BP, Belcik JT, Marinelli ER, Unger E, Lindner JR. Ultrasound Molecular Imaging of Atherosclerosis Using Small-Peptide Targeting Ligands Against Endothelial Markers of Inflammation and Oxidative Stress. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:1155-1163. [PMID: 29548756 DOI: 10.1016/j.ultrasmedbio.2018.01.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 06/08/2023]
Abstract
The aim of this study was to evaluate a panel of endothelium-targeted microbubble (MB) ultrasound contrast agents bearing small peptide ligands as a human-ready approach for molecular imaging of markers of high-risk atherosclerotic plaque. Small peptide ligands with established affinity for human P-selectin, VCAM-1, LOX-1 and von Willebrand factor (VWF) were conjugated to the surface of lipid-stabilized MBs. Contrast-enhanced ultrasound (CEUS) molecular imaging of the thoracic aorta was performed in wild-type and gene-targeted mice with advanced atherosclerosis (DKO). Histology was performed on carotid endarterectomy samples from patients undergoing surgery for unstable atherosclerosis to assess target expression in humans. In DKO mice, CEUS signal for all four targeted MBs was significantly higher than that for control MBs, and was three to sevenfold higher than in wild-type mice, with the highest signal achieved for VCAM-1 and VWF. All molecular targets were present on the patient plaque surface but expression was greatest for VCAM-1 and VWF. We conclude that ultrasound contrast agents bearing small peptide ligands feasible for human use can be targeted against endothelial cell adhesion molecules for inflammatory cells and platelets for imaging advanced atherosclerotic disease.
Collapse
Affiliation(s)
- Federico Moccetti
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Craig C Weinkauf
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
| | - Brian P Davidson
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA; Portland Veterans Affairs Medical Center, Portland, Oregon, USA
| | - J Todd Belcik
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | | | | | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA; Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA.
| |
Collapse
|
13
|
Zhang X, Liu C, Hu F, Zhang Y, Wang J, Gao Y, Jiang Y, Zhang Y, Lan X. PET Imaging of VCAM-1 Expression and Monitoring Therapy Response in Tumor with a 68Ga-Labeled Single Chain Variable Fragment. Mol Pharm 2018; 15:609-618. [PMID: 29308904 DOI: 10.1021/acs.molpharmaceut.7b00961] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Vascular cell adhesion molecule-1 (VCAM-1) is a transmembrane glycoprotein closely related to tumorigenicity as well as tumor metastasis. It is also a well-known candidate for detecting tumors. LY2409881, an IKKβ inhibitor, could induce apoptosis of VCAM-1 positive cells. Our purpose is to prepare a novel tracer to evaluate its feasibility of detecting VCAM-1 expression and monitoring LY2409881 tumor curative effect. The tracer was prepared by conjugating the single chain variable fragment (scFv) of VCAM-1 and NOTA-NHS-ester and then labeled with 68Ga. 68Ga-NOTA-VCAM-1scFv was successfully prepared with high radiochemical yield. VCAM-1 overexpression and underexpression melanoma cell lines, B16F10 and A375m, were used in this study. The results of microPET/CT imaging in small animals indicated that the uptake of 68Ga-NOTA-VCAM-1scFv in B16F10 tumor was much higher than that of A375m, which was also confirmed by the biodistribution and autoradiography results. LY2409881 inhibits the growth of B16F10 melanoma in vivo by inducing dose- and time-dependent growth inhibition and apoptosis of the cells. The LY2409881 treated group and DMSO control group were established and imaged by microPET/CT. In the LY2409881 group, uptake of the tracer in tumor was decreased at the first week, and then gradually recovered to the initial level. In DMSO control, the uptake of the tracer remained at the same level during the whole time. The results suggested that LY2409881 inhibits the expression of VCAM-1 and suppresses tumor growth. 68Ga-NOTA-VCAM-1scFv, an easily synthesized probe, has a potential clinical application in the visual monitoring of IKKβ inhibitor intervention on VCAM-1 positive tumors.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Chunbao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Fan Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yingying Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University , Xi'an, 710032, China
| | - Yongheng Gao
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University , Xi'an, 710032, China
| | - Yaqun Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| |
Collapse
|
14
|
MacRitchie N, Grassia G, Noonan J, Garside P, Graham D, Maffia P. Molecular imaging of atherosclerosis: spotlight on Raman spectroscopy and surface-enhanced Raman scattering. Heart 2017; 104:460-467. [PMID: 29061690 PMCID: PMC5861389 DOI: 10.1136/heartjnl-2017-311447] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/25/2017] [Accepted: 09/25/2017] [Indexed: 01/03/2023] Open
Abstract
To accurately predict atherosclerotic plaque progression, a detailed phenotype of the lesion at the molecular level is required. Here, we assess the respective merits and limitations of molecular imaging tools. Clinical imaging includes contrast-enhanced ultrasound, an inexpensive and non-toxic technique but with poor sensitivity. CT benefits from high spatial resolution but poor sensitivity coupled with an increasing radiation burden that limits multiplexing. Despite high sensitivity, positron emission tomography and single-photon emission tomography have disadvantages when applied to multiplex molecular imaging due to poor spatial resolution, signal cross talk and increasing radiation dose. In contrast, MRI is non-toxic, displays good spatial resolution but poor sensitivity. Preclinical techniques include near-infrared fluorescence (NIRF), which provides good spatial resolution and sensitivity; however, multiplexing with NIRF is limited, due to photobleaching and spectral overlap. Fourier transform infrared spectroscopy and Raman spectroscopy are label-free techniques that detect molecules based on the vibrations of chemical bonds. Both techniques offer fast acquisition times with Raman showing superior spatial resolution. Raman signals are inherently weak; however, leading to the development of surface-enhanced Raman spectroscopy (SERS) that offers greatly increased sensitivity due to using metallic nanoparticles that can be functionalised with biomolecules targeted against plaque ligands while offering high multiplexing potential. This asset combined with high spatial resolution makes SERS an exciting prospect as a diagnostic tool. The ongoing refinements of SERS technologies such as deep tissue imaging and portable systems making SERS a realistic prospect for translation to the clinic.
Collapse
Affiliation(s)
- Neil MacRitchie
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,British Society for Cardiovascular Research, UK
| | - Gianluca Grassia
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jonathan Noonan
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,British Society for Cardiovascular Research, UK
| | - Paul Garside
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Duncan Graham
- Centre for Molecular Nanometrology, Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,British Society for Cardiovascular Research, UK.,Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
15
|
Anwaier G, Chen C, Cao Y, Qi R. A review of molecular imaging of atherosclerosis and the potential application of dendrimer in imaging of plaque. Int J Nanomedicine 2017; 12:7681-7693. [PMID: 29089763 PMCID: PMC5656339 DOI: 10.2147/ijn.s142385] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Despite the fact that technological advancements have been made in diagnosis and treatment, cardiovascular diseases (CVDs) remain the leading cause of mortality and morbidity worldwide. Early detection of atherosclerosis (AS), especially vulnerable plaques, plays a crucial role in the prevention of acute coronary syndrome (ACS). Targeting the critical cytokines and molecules that are upregulated during the biological process of AS by in vivo molecular imaging has been widely used in plaque imaging. With their three-dimensional architecture, composition, and abundant terminal functional groups, dendrimers provide a platform for multitargeting and multimodal imaging. Thus, modified dendrimers with the key molecules upregulated in AS plaques will be an innovative attempt to achieve targeted imaging of AS plaques specifically and efficiently. This review was aimed to address some recent works on imaging of AS plaques using various types of image technology and further discuss the applications of dendrimers, an innovative yet seldom used method in imaging of AS plaques due to some limitations and challenges, and we highlight the bright future of the modified dendrimers in characterizing AS plaques.
Collapse
Affiliation(s)
- Gulinigaer Anwaier
- Peking University Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of education, Peking University Health Science Center.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing.,School of Basic Medical Science, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Cong Chen
- Peking University Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of education, Peking University Health Science Center.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing
| | - Yini Cao
- Peking University Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of education, Peking University Health Science Center.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing
| | - Rong Qi
- Peking University Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of education, Peking University Health Science Center.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing.,School of Basic Medical Science, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| |
Collapse
|
16
|
Cellular Uptake of Plain and SPION-Modified Microbubbles for Potential Use in Molecular Imaging. Cell Mol Bioeng 2017; 10:537-548. [PMID: 29151981 PMCID: PMC5662700 DOI: 10.1007/s12195-017-0504-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 08/01/2017] [Indexed: 12/31/2022] Open
Abstract
Introduction Both diagnostic ultrasound (US) and magnetic resonance imaging (MRI) accuracy can be improved by using contrast enhancement. For US gas-filled microbubbles (MBs) or silica nanoparticles (SiNPs), and for MRI superparamagnetic or paramagnetic agents, contribute to this. However, interactions of MBs with the vascular wall and cells are not fully known for all contrast media. Methods We studied the in vitro interactions between three types of non-targeted air-filled MBs with a polyvinyl-alcohol shell and murine macrophages or endothelial cells. The three MB types were plain MBs and two types that were labelled (internally and externally) with superparamagnetic iron oxide nanoparticles (SPIONs) for US/MRI bimodality. Cells were incubated with MBs and imaged by microscopy to evaluate uptake and adhesion. Interactions were quantified and the MB internalization was confirmed by fluorescence quenching of non-internalized MBs. Results Macrophages internalized each MB type within different time frames: plain MBs 6 h, externally labelled MBs 25 min and internally labelled MBs 2 h. An average of 0.14 externally labelled MBs per cell were internalized after 30 min and 1.34 after 2 h; which was 113% more MBs than the number of internalized internally labelled MBs. The macrophages engulfed these three differently modified new MBs at various rate, whereas endothelial cells did not engulf MBs. Conclusions Polyvinyl-alcohol MBs are not taken up by endothelial cells. The MB uptake by macrophages is promoted by SPION labelling, in particular external such, which may be important for macrophage targeting.
Collapse
|
17
|
Meletta R, Slavik R, Mu L, Rancic Z, Borel N, Schibli R, Ametamey SM, Krämer SD, Müller Herde A. Cannabinoid receptor type 2 (CB2) as one of the candidate genes in human carotid plaque imaging: Evaluation of the novel radiotracer [ 11 C]RS-016 targeting CB2 in atherosclerosis. Nucl Med Biol 2017; 47:31-43. [DOI: 10.1016/j.nucmedbio.2017.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 12/15/2016] [Accepted: 01/05/2017] [Indexed: 01/15/2023]
|
18
|
Liang G, Vo D, Nguyen PK. Fundamentals of Cardiovascular Molecular Imaging: a Review of Concepts and Strategies. CURRENT CARDIOVASCULAR IMAGING REPORTS 2017. [DOI: 10.1007/s12410-017-9403-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
19
|
Leukocyte trafficking-associated vascular adhesion protein 1 is expressed and functionally active in atherosclerotic plaques. Sci Rep 2016; 6:35089. [PMID: 27731409 PMCID: PMC5059718 DOI: 10.1038/srep35089] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/23/2016] [Indexed: 12/15/2022] Open
Abstract
Given the important role of inflammation and the potential association of the leukocyte trafficking-associated adhesion molecule vascular adhesion protein 1 (VAP-1) with atherosclerosis, this study examined whether functional VAP-1 is expressed in atherosclerotic lesions and, if so, whether it could be targeted by positron emission tomography (PET). First, immunohistochemistry revealed that VAP-1 localized to endothelial cells of intra-plaque neovessels in human carotid endarterectomy samples from patients with recent ischemic symptoms. In low-density lipoprotein receptor-deficient mice expressing only apolipoprotein B100 (LDLR-/-ApoB100/100), VAP-1 was expressed on endothelial cells lining inflamed atherosclerotic lesions; normal vessel walls in aortas of C57BL/6N control mice were VAP-1-negative. Second, we discovered that the focal uptake of VAP-1 targeting sialic acid-binding immunoglobulin-like lectin 9 based PET tracer [68Ga]DOTA-Siglec-9 in atherosclerotic plaques was associated with the density of activated macrophages (r = 0.58, P = 0.022). As a final point, we found that the inhibition of VAP-1 activity with small molecule LJP1586 decreased the density of macrophages in inflamed atherosclerotic plaques in mice. Our results suggest for the first time VAP-1 as a potential imaging target for inflamed atherosclerotic plaques, and corroborate VAP-1 inhibition as a therapeutic approach in the treatment of atherosclerosis.
Collapse
|
20
|
Pedrigi RM, Mehta VV, Bovens SM, Mohri Z, Poulsen CB, Gsell W, Tremoleda JL, Towhidi L, de Silva R, Petretto E, Krams R. Influence of shear stress magnitude and direction on atherosclerotic plaque composition. ROYAL SOCIETY OPEN SCIENCE 2016; 3:160588. [PMID: 27853578 PMCID: PMC5099003 DOI: 10.1098/rsos.160588] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/19/2016] [Indexed: 05/19/2023]
Abstract
The precise flow characteristics that promote different atherosclerotic plaque types remain unclear. We previously developed a blood flow-modifying cuff for ApoE-/- mice that induces the development of advanced plaques with vulnerable and stable features upstream and downstream of the cuff, respectively. Herein, we sought to test the hypothesis that changes in flow magnitude promote formation of the upstream (vulnerable) plaque, whereas altered flow direction is important for development of the downstream (stable) plaque. We instrumented ApoE-/- mice (n = 7) with a cuff around the left carotid artery and imaged them with micro-CT (39.6 µm resolution) eight to nine weeks after cuff placement. Computational fluid dynamics was then performed to compute six metrics that describe different aspects of atherogenic flow in terms of wall shear stress magnitude and/or direction. In a subset of four imaged animals, we performed histology to confirm the presence of advanced plaques and measure plaque length in each segment. Relative to the control artery, the region upstream of the cuff exhibited changes in shear stress magnitude only (p < 0.05), whereas the region downstream of the cuff exhibited changes in shear stress magnitude and direction (p < 0.05). These data suggest that shear stress magnitude contributes to the formation of advanced plaques with a vulnerable phenotype, whereas variations in both magnitude and direction promote the formation of plaques with stable features.
Collapse
Affiliation(s)
- Ryan M. Pedrigi
- Department of Bioengineering, Imperial College London, London, UK
| | - Vikram V. Mehta
- Department of Bioengineering, Imperial College London, London, UK
| | - Sandra M. Bovens
- Department of Bioengineering, Imperial College London, London, UK
| | - Zahra Mohri
- Department of Bioengineering, Imperial College London, London, UK
| | | | - Willy Gsell
- MRC-Clinical Sciences Centre, Imperial College London, London, UK
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jordi L. Tremoleda
- MRC-Clinical Sciences Centre, Imperial College London, London, UK
- Centre for Trauma Sciences, Queen Mary University of London, London, UK
| | - Leila Towhidi
- Department of Bioengineering, Imperial College London, London, UK
| | - Ranil de Silva
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Enrico Petretto
- MRC-Clinical Sciences Centre, Imperial College London, London, UK
- Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Rob Krams
- Department of Bioengineering, Imperial College London, London, UK
- Author for correspondence: Rob Krams e-mail:
| |
Collapse
|
21
|
Liu C, Zhang X, Song Y, Wang Y, Zhang F, Zhang Y, Zhang Y, Lan X. SPECT and fluorescence imaging of vulnerable atherosclerotic plaque with a vascular cell adhesion molecule 1 single-chain antibody fragment. Atherosclerosis 2016; 254:263-270. [PMID: 27680307 DOI: 10.1016/j.atherosclerosis.2016.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 08/02/2016] [Accepted: 09/06/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND AIMS Early detection and evaluation of vulnerable atherosclerotic plaque are important for risk stratification and timely intervention, and vascular cell adhesion molecule 1 (VCAM1) assists in adhesion and recruitment of inflammatory cells to vulnerable lesions. We labeled a single-chain variable fragment (scFv) of VCAM1 with 99mtechnetium (99mTc) and fluorescent markers to investigate its potential utility in detecting vulnerable plaques in animal models of atherosclerosis. METHODS We labeled VCAM1 scFv with 99mTc and cyanine5 (CY5) and evaluated the probes on apolipoprotein E gene-deficient mice and New Zealand White rabbits with induced atherosclerosis. Histopathology and Western blot examinations confirmed atherosclerotic plaque and VCAM1 expression in the aortas. In vivo biodistribution of 99mTc-scFv-VCAM1 was studied. Abdominal organs of mice were removed after CY5-scFv-VCAM1 administration for aortic fluorescence imaging. Rabbits SPECT imaging of 99mTc-scFv-VCAM1 was performed and autoradiography (ARG) of the aortas was checked to confirm the tracer uptake. RESULTS The radiochemical purity of 99mTc-scFv-VCAM1 was 98.72± 1.04% (n = 5) and its specific activity was 7.8 MBq/μg. Biodistribution study indicated predominant probe clearance by kidneys. In fluorescence imaging, stronger signal from CY5-scFv-VCAM1 in the aorta was observed in atherosclerotic mice than that in controls. SPECT imaging with 99mTc-scFv-VCAM1 showed tracer uptake in the abdominal aorta and the aortic arch of atherosclerotic animals. ARG confirmed tracer uptake in the aortas of atherosclerotic rabbits, with higher uptake ratios of aortic arch/descending aorta in experimental animals (4.45 ± 0.63, n = 5) than controls (1.12 ± 0.15, n = 5; p < 0.05). CONCLUSIONS SPECT and fluorescence imaging results showed the feasibility and effectiveness of detecting vulnerable plaque with scFv of VCAM1, indicating its potential for early diagnosis and evaluation of atherosclerosis.
Collapse
Affiliation(s)
- Chunbao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yiling Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yichun Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fengzhen Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yingying Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
22
|
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
|
23
|
Kazuma SM, Sultan D, Zhao Y, Detering L, You M, Luehmann HP, Abdalla DSP, Liu Y. Recent Advances of Radionuclide-Based Molecular Imaging of Atherosclerosis. Curr Pharm Des 2016; 21:5267-76. [PMID: 26369676 DOI: 10.2174/1381612821666150915104529] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is a systemic disease characterized by the development of multifocal plaque lesions within vessel walls and extending into the vascular lumen. The disease takes decades to develop symptomatic lesions, affording opportunities for accurate detection of plaque progression, analysis of risk factors responsible for clinical events, and planning personalized treatment. Of the available molecular imaging modalities, radionuclidebased imaging strategies have been favored due to their sensitivity, quantitative detection and pathways for translational research. This review summarizes recent advances of radiolabeled small molecules, peptides, antibodies and nanoparticles for atherosclerotic plaque imaging during disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, 63110, United States.
| |
Collapse
|
24
|
Bala G, Blykers A, Xavier C, Descamps B, Broisat A, Ghezzi C, Fagret D, Van Camp G, Caveliers V, Vanhove C, Lahoutte T, Droogmans S, Cosyns B, Devoogdt N, Hernot S. Targeting of vascular cell adhesion molecule-1 by 18F-labelled nanobodies for PET/CT imaging of inflamed atherosclerotic plaques. Eur Heart J Cardiovasc Imaging 2016; 17:1001-8. [PMID: 26800768 DOI: 10.1093/ehjci/jev346] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/12/2015] [Indexed: 12/30/2022] Open
Abstract
AIMS Positron emission tomography-computed tomography (PET-CT) is a highly sensitive clinical molecular imaging modality to study atherosclerotic plaque biology. Therefore, we sought to develop a new PET tracer, targeting vascular cell adhesion molecule (VCAM)-1 and validate it in a murine atherosclerotic model as a potential agent to detect atherosclerotic plaque inflammation. METHODS AND RESULTS The anti-VCAM-1 nanobody (Nb) (cAbVCAM-1-5) was radiolabelled with Fluorine-18 ((18)F), with a radiochemical purity of >98%. In vitro cell-binding studies showed specific binding of the tracer to VCAM-1 expressing cells. In vivo PET/CT imaging of ApoE(-/-) mice fed a Western diet or control mice was performed at 2h30 post-injection of [(18)F]-FB-cAbVCAM-1-5 or (18)F-control Nb. Additionally, plaque uptake in different aorta segments was evaluated ex vivo based on extent of atherosclerosis. Atherosclerotic lesions in the aortic arch of ApoE(-/-) mice, injected with [(18)F]-FB-anti-VCAM-1 Nb, were successfully identified using PET/CT imaging, while background signal was observed in the control groups. These results were confirmed by ex vivo analyses where uptake of [(18)F]-FB-cAbVCAM-1-5 in atherosclerotic lesions was significantly higher compared with control groups. Moreover, uptake increased with the increasing extent of atherosclerosis (Score 0: 0.68 ± 0.10, Score 1: 1.18 ± 0.36, Score 2: 1.49 ± 0.37, Score 3: 1.48 ± 0.38%ID/g, Spearman's r(2) = 0.675, P < 0.0001). High lesion-to-heart, lesion-to-blood, and lesion-to-control vessel ratios were obtained (12.4 ± 0.4, 3.3 ± 0.4, and 3.1 ± 0.6, respectively). CONCLUSION The [(18)F]-FB-anti-VCAM-1 Nb, cross-reactive for both mouse and human VCAM-1, allows non-invasive PET/CT imaging of VCAM-1 expression in atherosclerotic plaques in a murine model and may represent an attractive tool for imaging vulnerable atherosclerotic plaques in patients.
Collapse
Affiliation(s)
- Gezim Bala
- Centrum voor Hart-en Vaatziekten (CHVZ), UZ Brussel, Brussels, Belgium In Vivo Cellular and Molecular Imaging (ICMI), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels B-1090, Belgium
| | - Anneleen Blykers
- In Vivo Cellular and Molecular Imaging (ICMI), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels B-1090, Belgium
| | - Catarina Xavier
- In Vivo Cellular and Molecular Imaging (ICMI), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels B-1090, Belgium
| | - Benedicte Descamps
- iMinds-IBiTech-MEDISIP, Department of Electronics and Information Systems, Universiteit Gent, Ghent, Belgium
| | - Alexis Broisat
- Radiopharmaceutiques Biocliniques, INSERM, 1039-Université de Grenoble, La Tronche, France
| | - Catherine Ghezzi
- Radiopharmaceutiques Biocliniques, INSERM, 1039-Université de Grenoble, La Tronche, France
| | - Daniel Fagret
- Radiopharmaceutiques Biocliniques, INSERM, 1039-Université de Grenoble, La Tronche, France
| | - Guy Van Camp
- In Vivo Cellular and Molecular Imaging (ICMI), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels B-1090, Belgium
| | - Vicky Caveliers
- In Vivo Cellular and Molecular Imaging (ICMI), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels B-1090, Belgium Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Christian Vanhove
- iMinds-IBiTech-MEDISIP, Department of Electronics and Information Systems, Universiteit Gent, Ghent, Belgium
| | - Tony Lahoutte
- In Vivo Cellular and Molecular Imaging (ICMI), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels B-1090, Belgium Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Steven Droogmans
- Centrum voor Hart-en Vaatziekten (CHVZ), UZ Brussel, Brussels, Belgium In Vivo Cellular and Molecular Imaging (ICMI), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels B-1090, Belgium
| | - Bernard Cosyns
- Centrum voor Hart-en Vaatziekten (CHVZ), UZ Brussel, Brussels, Belgium In Vivo Cellular and Molecular Imaging (ICMI), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels B-1090, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging (ICMI), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels B-1090, Belgium
| | - Sophie Hernot
- In Vivo Cellular and Molecular Imaging (ICMI), Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels B-1090, Belgium
| |
Collapse
|
25
|
Wang Y, Chen J, Yang B, Qiao H, Gao L, Su T, Ma S, Zhang X, Li X, Liu G, Cao J, Chen X, Chen Y, Cao F. In vivo MR and Fluorescence Dual-modality Imaging of Atherosclerosis Characteristics in Mice Using Profilin-1 Targeted Magnetic Nanoparticles. Theranostics 2016; 6:272-86. [PMID: 26877785 PMCID: PMC4729775 DOI: 10.7150/thno.13350] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/29/2015] [Indexed: 01/27/2023] Open
Abstract
Aims: This study aims to explore non-invasive imaging of atherosclerotic plaque through magnetic resonance imaging (MRI) and near-infrared fluorescence (NIRF) by using profilin-1 targeted magnetic iron oxide nanoparticles (PF1-Cy5.5-DMSA-Fe3O4-NPs, denoted as PC-NPs) as multimodality molecular imaging probe in murine model of atherosclerosis. Methods and Results: PC-NPs were constructed by conjugating polyclonal profilin-1 antibody and NHS-Cy5.5 fluorescent dye to the surface of DMSA-Fe3O4-nanoparticles via condensation reaction. Murine atherosclerosis model was induced in apoE-/- mice by high fat and cholesterol diet (HFD) for 16 weeks. The plaque areas in aortic artery were detected with Oil Red O staining. Immunofluorescent staining and Western blot analysis were applied respectively to investigate profilin-1 expression. CCK-8 assay and transwell migration experiment were performed to detect vascular smooth muscle cells (VSMCs) proliferation. In vivo MRI and NIRF imaging of atherosclerotic plaque were carried out before and 36 h after intravenous injection of PC-NPs. Oil Red O staining showed that the plaque area was significantly increased in HFD group (p<0.05). Immunofluorescence staining revealed that profilin-1 protein was highly abundant within plaque in HFD group and co-localized with α-smooth muscle actin. Profilin-1 siRNA intervention could inhibit VSMCs proliferation and migration elicited by ox-LDL (p<0.05). In vivo MRI and NIRF imaging revealed that PC-NPs accumulated in atherosclerotic plaque of carotid artery. There was a good correlation between the signals of MRI and ex vivo fluorescence intensities of NIRF imaging in animals with PC-NPs injection. Conclusion: PC-NPs is a promising dual modality imaging probe, which may improve molecular diagnosis of plaque characteristics and evaluation of pharmaceutical interventions for atherosclerosis.
Collapse
|
26
|
Winkel LC, Hoogendoorn A, Xing R, Wentzel JJ, Van der Heiden K. Animal models of surgically manipulated flow velocities to study shear stress-induced atherosclerosis. Atherosclerosis 2015; 241:100-10. [PMID: 25969893 DOI: 10.1016/j.atherosclerosis.2015.04.796] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/12/2015] [Accepted: 04/22/2015] [Indexed: 10/23/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial tree that develops at predisposed sites, coinciding with locations that are exposed to low or oscillating shear stress. Manipulating flow velocity, and concomitantly shear stress, has proven adequate to promote endothelial activation and subsequent plaque formation in animals. In this article, we will give an overview of the animal models that have been designed to study the causal relationship between shear stress and atherosclerosis by surgically manipulating blood flow velocity profiles. These surgically manipulated models include arteriovenous fistulas, vascular grafts, arterial ligation, and perivascular devices. We review these models of manipulated blood flow velocity from an engineering and biological perspective, focusing on the shear stress profiles they induce and the vascular pathology that is observed.
Collapse
Affiliation(s)
- Leah C Winkel
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Ayla Hoogendoorn
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Ruoyu Xing
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Jolanda J Wentzel
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Kim Van der Heiden
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands.
| |
Collapse
|
27
|
Zaman RT, Kosuge H, Carpenter C, Sun C, McConnell MV, Xing L. Scintillating balloon-enabled fiber-optic system for radionuclide imaging of atherosclerotic plaques. J Nucl Med 2015; 56:771-7. [PMID: 25858046 DOI: 10.2967/jnumed.114.153239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/18/2015] [Indexed: 01/28/2023] Open
Abstract
UNLABELLED Atherosclerosis underlies coronary artery disease, the leading cause of death in the United States and worldwide. Detection of coronary plaque inflammation remains challenging. In this study, we developed a scintillating balloon-enabled fiber-optic radionuclide imaging (SBRI) system to improve the sensitivity and resolution of plaque imaging using (18)F-FDG, a marker of vascular inflammation, and tested it in a murine model. METHODS The fiber-optic system uses a Complementary Metal-Oxide Silicon (CMOS) camera with a distal ferrule terminated with a wide-angle lens. The novelty of this system is a scintillating balloon in the front of the wide-angle lens to image light from the decay of (18)F-FDG emission signal. To identify the optimal scintillating materials with respect to resolution, we calculated the modulation transfer function of yttrium-aluminum-garnet doped with cerium, anthracene, and calcium fluoride doped with europium (CaF2:Eu) phosphors using an edge pattern and a thin-line optical phantom. The scintillating balloon was then fabricated from 10 mL of silicone RTV catalyst mixed with 1 mL of base and 50 mg of CaF2:Eu per mL. The addition of a lutetium oxyorthosilicate scintillating crystal (500 μm thick) to the balloon was also investigated. The SBRI system was tested in a murine atherosclerosis model: carotid-ligated mice (n = 5) were injected with (18)F-FDG, followed by ex vivo imaging of the macrophage-rich carotid plaques and nonligated controls. Confirmatory imaging of carotid plaques and controls was also performed by an external optical imaging system and autoradiography. RESULTS Analyses of the different phosphors showed that CaF2:Eu enabled the best resolution of 1.2 μm. The SBRI system detected almost a 4-fold-higher radioluminescence signal from the ligated left carotid artery than the nonligated right carotid: 1.63 × 10(2) ± 4.01 × 10(1) vs. 4.21 × 10(1) ± 2.09 × 10(0) (photon counts), P = 0.006. We found no significant benefit to adding a scintillating crystal to the balloon: 1.65 × 10(2) ± 4.07 × 10(1) vs. 4.44 × 10(1) ± 2.17 × 10(0) (photon counts), P = 0.005. Both external optical imaging and autoradiography confirmed the high signal from the (18)F-FDG in carotid plaques versus controls. CONCLUSION This SBRI system provides high-resolution and sensitive detection of (18)F-FDG uptake by murine atherosclerotic plaques.
Collapse
Affiliation(s)
- Raiyan T Zaman
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California Division of Radiation Physics, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California; and
| | - Hisanori Kosuge
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Conroy Sun
- Division of Radiation Physics, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California; and
| | - Michael V McConnell
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Lei Xing
- Division of Radiation Physics, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California; and
| |
Collapse
|
28
|
Meletta R, Müller Herde A, Chiotellis A, Isa M, Rancic Z, Borel N, Ametamey SM, Krämer SD, Schibli R. Evaluation of the radiolabeled boronic acid-based FAP inhibitor MIP-1232 for atherosclerotic plaque imaging. Molecules 2015; 20:2081-99. [PMID: 25633335 PMCID: PMC6272135 DOI: 10.3390/molecules20022081] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 12/29/2014] [Accepted: 01/20/2015] [Indexed: 12/21/2022] Open
Abstract
Research towards the non-invasive imaging of atherosclerotic plaques is of high clinical priority as early recognition of vulnerable plaques may reduce the incidence of cardiovascular events. The fibroblast activation protein alpha (FAP) was recently proposed as inflammation-induced protease involved in the process of plaque vulnerability. In this study, FAP mRNA and protein levels were investigated by quantitative polymerase chain reaction and immunohistochemistry, respectively, in human endarterectomized carotid plaques. A published boronic-acid based FAP inhibitor, MIP-1232, was synthetized and radiolabeled with iodine-125. The potential of this radiotracer to image plaques was evaluated by in vitro autoradiography with human carotid plaques. Specificity was assessed with a xenograft with high and one with low FAP level, grown in mice. Target expression analyses revealed a moderately higher protein level in atherosclerotic plaques than normal arteries correlating with plaque vulnerability. No difference in expression was determined on mRNA level. The radiotracer was successfully produced and accumulated strongly in the FAP-positive SK-Mel-187 melanoma xenograft in vitro while accumulation was negligible in an NCI-H69 xenograft with low FAP levels. Binding of the tracer to endarterectomized tissue was similar in plaques and normal arteries, hampering its use for atherosclerosis imaging.
Collapse
Affiliation(s)
- Romana Meletta
- Department of Chemistry and Applied Bioscience of ETH Zurich, Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland.
| | - Adrienne Müller Herde
- Department of Chemistry and Applied Bioscience of ETH Zurich, Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland.
| | - Aristeidis Chiotellis
- Department of Chemistry and Applied Bioscience of ETH Zurich, Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland.
| | - Malsor Isa
- Department of Chemistry and Applied Bioscience of ETH Zurich, Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland.
| | - Zoran Rancic
- Division of Cardiovascular Surgery, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland.
| | - Nicole Borel
- Institute for Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, 8057 Zurich, Switzerland.
| | - Simon M Ametamey
- Department of Chemistry and Applied Bioscience of ETH Zurich, Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland.
| | - Stefanie D Krämer
- Department of Chemistry and Applied Bioscience of ETH Zurich, Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland.
| | - Roger Schibli
- Department of Chemistry and Applied Bioscience of ETH Zurich, Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland.
| |
Collapse
|
29
|
PARK SJ, KIM JY, TEOH CL, KANG NY, CHANG YT. New Targets of Molecular Imaging in Atherosclerosis: Prehension of Current Status. ANAL SCI 2015; 31:245-55. [DOI: 10.2116/analsci.31.245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Sung-Jin PARK
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Jun-Young KIM
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Chai Lean TEOH
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Nam-Young KANG
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Young-Tae CHANG
- Department of Chemistry & NUS MedChem Program of Life Sciences Institute, National University of Singapore
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| |
Collapse
|
30
|
Hong H, Chen F, Zhang Y, Cai W. New radiotracers for imaging of vascular targets in angiogenesis-related diseases. Adv Drug Deliv Rev 2014; 76:2-20. [PMID: 25086372 DOI: 10.1016/j.addr.2014.07.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 07/14/2014] [Accepted: 07/22/2014] [Indexed: 01/03/2023]
Abstract
Tremendous advances over the last several decades in positron emission tomography (PET) and single photon emission computed tomography (SPECT) allow for targeted imaging of molecular and cellular events in the living systems. Angiogenesis, a multistep process regulated by the network of different angiogenic factors, has attracted world-wide interests, due to its pivotal role in the formation and progression of different diseases including cancer, cardiovascular diseases (CVD), and inflammation. In this review article, we will summarize the recent progress in PET or SPECT imaging of a wide variety of vascular targets in three major angiogenesis-related diseases: cancer, cardiovascular diseases, and inflammation. Faster drug development and patient stratification for a specific therapy will become possible with the facilitation of PET or SPECT imaging and it will be critical for the maximum benefit of patients.
Collapse
|
31
|
Zaman RT, Kosuge H, Pratx G, Carpenter C, Xing L, McConnell MV. Fiber-optic system for dual-modality imaging of glucose probes 18F-FDG and 6-NBDG in atherosclerotic plaques. PLoS One 2014; 9:e108108. [PMID: 25233472 PMCID: PMC4169475 DOI: 10.1371/journal.pone.0108108] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 08/19/2014] [Indexed: 12/02/2022] Open
Abstract
Background Atherosclerosis is a progressive inflammatory condition that underlies coronary artery disease (CAD)–the leading cause of death in the United States. Thus, the ultimate goal of this research is to advance our understanding of human CAD by improving the characterization of metabolically active vulnerable plaques within the coronary arteries using a novel catheter-based imaging system. The aims of this study include (1) developing a novel fiber-optic imaging system with a scintillator to detect both 18F and fluorescent glucose probes, and (2) validating the system on ex vivo murine plaques. Methods A novel design implements a flexible fiber-optic catheter consisting of both a radio-luminescence and a fluorescence imaging system to detect radionuclide 18F-fluorodeoxyglucose (18F-FDG) and the fluorescent analog 6-(N-(7-Nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-6-Deoxyglucose (6-NBDG), respectively. Murine macrophage-rich atherosclerotic carotid plaques were imaged ex vivo after intravenous delivery of 18F-FDG or 6-NBDG. Confirmatory optical imaging by IVIS-200 and autoradiography were also performed. Results Our fiber-optic imaging system successfully visualized both 18F-FDG and 6-NBDG probes in atherosclerotic plaques. For 18F-FDG, the ligated left carotid arteries (LCs) exhibited 4.9-fold higher radioluminescence signal intensity compared to the non-ligated right carotid arteries (RCs) (2.6×104±1.4×103 vs. 5.4×103±1.3×103 A.U., P = 0.008). Similarly, for 6-NBDG, the ligated LCs emitted 4.3-fold brighter fluorescent signals than the control RCs (1.6×102±2.7×101 vs. 3.8×101±5.9 A.U., P = 0.002). The higher uptake of both 18F-FDG and 6-NBDG in ligated LCs were confirmed with the IVIS-200 system. Autoradiography further verified the higher uptake of 18F-FDG by the LCs. Conclusions This novel fiber-optic imaging system was sensitive to both radionuclide and fluorescent glucose probes taken up by murine atherosclerotic plaques. In addition, 6-NBDG is a promising novel fluorescent probe for detecting macrophage-rich atherosclerotic plaques.
Collapse
Affiliation(s)
- Raiyan T. Zaman
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Radiation Physics, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| | - Hisanori Kosuge
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Guillem Pratx
- Division of Radiation Physics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Colin Carpenter
- Division of Radiation Physics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Lei Xing
- Division of Radiation Physics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Michael V. McConnell
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
32
|
Tekabe Y, Kollaros M, Zerihoun A, Zhang G, Backer MV, Backer JM, Johnson LL. Imaging VEGF receptor expression to identify accelerated atherosclerosis. EJNMMI Res 2014; 4:41. [PMID: 26055940 PMCID: PMC4884015 DOI: 10.1186/s13550-014-0041-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/08/2014] [Indexed: 01/23/2023] Open
Abstract
Background The biology of the vulnerable plaque includes increased inflammation and rapid growth of vasa vasorum, processes that are associated with enhanced vascular endothelial growth factor (VEGF)/ imaging receptors for VEGF (VEGFR) signaling and are accelerated in diabetes. This study was designed to test the hypothesis that VEGFRs in atherosclerotic plaques with a SPECT tracer scVEGF-PEG-DOTA/99mTc (scV/Tc) can identify accelerated atherosclerosis in diabetes. Methods Male apolipoprotein E null (ApoE−/−) mice (6 weeks of age) were made diabetic (n = 10) or left as non-diabetic (n = 13). At 26 to 28 weeks of age, 5 non-diabetic mice were injected with functionally inactivated scV/Tc (in-scV/Tc) that does not bind to VEGF receptors, while 8 non-diabetic and 10 diabetic mice were injected with scV/Tc. After blood pool clearance, at 3 to 4 h post-injection, mice were injected with CT contrast agent and underwent SPECT/CT imaging. From the scans, regions of interest (ROI) were drawn on serial transverse sections comprising the proximal aorta and the percentage of injected dose (%ID) in ROIs was calculated. At the completion of imaging, mice were euthanized, proximal aorta explanted for gamma well counting to determine the percentage of injected dose per gram (%ID/g) uptake and immunohistochemical characterization. Results The uptake of scV/Tc in the proximal aorta, calculated from SPECT/CT co-registered scans as %ID, was significantly higher in the diabetic mice (0.036 ± 0.017%ID) compared to non-diabetic mice (0.017 ± 0.005%ID; P < 0.01), as was uptake measured as %ID/g in harvested aorta, 1.81 ± 0.50%ID/g in the diabetic group vs. 0.98 ± 0.25%ID/g in the non-diabetic group (P < 0.01). The nonspecific uptake of in-scV/Tc in proximal aorta was significantly lower than the uptake of functionally active scV/Tc. Immunostaining of the atherosclerotic lesions showed higher expression of VEGFR-1 and VEGFR-2 in the diabetic mice. Conclusion These initial results suggest that imaging VEGFR with scV/Tc shows promise as a non-invasive approach to identify accelerated atherosclerosis. Electronic supplementary material The online version of this article (doi:10.1186/s13550-014-0041-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yared Tekabe
- Department of Medicine, Columbia University Medical Center, 622 West 168th St, PH 10 center rm 203, New York, NY, 10032, USA,
| | | | | | | | | | | | | |
Collapse
|
33
|
Active targeting of early and mid-stage atherosclerotic plaques using self-assembled peptide amphiphile micelles. Biomaterials 2014; 35:8678-86. [PMID: 25043572 DOI: 10.1016/j.biomaterials.2014.06.054] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 06/26/2014] [Indexed: 11/21/2022]
Abstract
Inflammatory cell adhesion molecules expressed by endothelial cells on the luminal surface of atherosclerotic plaques, such as vascular cell adhesion molecule-1 (VCAM-1), provide a rational target for diagnostic and therapeutic delivery vehicles. Therefore, the potential of using spherical, self-assembled micelles synthesized from VCAM-1 targeted peptide amphiphile molecules was examined for the ability to specifically bind to both early and mid-stage atherosclerotic plaques. In vitro, cells incubated with VCAM-1 targeted and dye-labeled micelles show enhanced fluorescence signal as compared to cells incubated with a PEG micelle control. In vivo, VCAM-1 targeted and Cy7-labeled peptide amphiphile micelles were shown to specifically accumulate at atherosclerotic plaques in both early and mid-stage ApoE -/- mice through co-localization of Cy7 signal with anti-VCAM-1 antibody staining in fixed tissue. No specific accumulation was observed with a PEG micelle control. Histological analysis of excised tissue provided evidence for the in vivo biocompatibility of these micelle formulations as no tissue damage was observed. These results demonstrate that VCAM-1 targeted micelles have potential as a platform for targeted drug delivery to multiple stages of atherosclerotic plaque formation due to their established specificity and safety.
Collapse
|
34
|
Luehmann HP, Pressly ED, Detering L, Wang C, Pierce R, Woodard PK, Gropler RJ, Hawker CJ, Liu Y. PET/CT imaging of chemokine receptor CCR5 in vascular injury model using targeted nanoparticle. J Nucl Med 2014; 55:629-34. [PMID: 24591489 PMCID: PMC4255944 DOI: 10.2967/jnumed.113.132001] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED Inflammation plays important roles at all stages of atherosclerosis. Chemokine systems have major effects on the initiation and progression of atherosclerosis by controlling the trafficking of inflammatory cells in vivo through interaction with their receptors. Chemokine receptor 5 (CCR5) has been reported to be an active participant in the late stage of atherosclerosis and has the potential as a prognostic biomarker for plaque stability. However, its diagnostic potential has not yet been explored. The purpose of this study was to develop a targeted nanoparticle for sensitive and specific PET/CT imaging of the CCR5 receptor in an apolipoprotein E knock-out (ApoE(-/-)) mouse vascular injury model. METHODS The D-Ala1-peptide T-amide (DAPTA) peptide was selected as a targeting ligand for the CCR5 receptor. Through controlled conjugation and polymerization, a biocompatible poly(methyl methacrylate)-core/polyethylene glycol-shell amphiphilic comblike nanoparticle was prepared and labeled with (64)Cu for CCR5 imaging in the ApoE(-/-) wire-injury model. Immunohistochemistry, histology, and real-time reverse transcription polymerase chain reaction (RT-PCR) were performed to assess the disease progression and upregulation of CCR5 receptor. RESULTS The (64)Cu-DOTA-DAPTA tracer showed specific PET imaging of CCR5 in the ApoE(-/-) mice. The targeted (64)Cu-DOTA-DAPTA-comb nanoparticles showed extended blood signal and optimized biodistribution. The tracer uptake analysis showed significantly higher accumulations at the injury lesions than those acquired from the sham-operated sites. The competitive PET receptor blocking studies confirmed the CCR5 receptor-specific uptake. The assessment of (64)Cu-DOTA-DAPTA-comb in C57BL/6 mice and (64)Cu-DOTA-comb in ApoE(-/-) mice verified low nonspecific nanoparticle uptake. Histology, immunohistochemistry, and RT-PCR analyses verified the upregulation of CCR5 in the progressive atherosclerosis model. CONCLUSION This work provides a nanoplatform for sensitive and specific detection of CCR5's physiologic functions in an animal atherosclerosis model.
Collapse
Affiliation(s)
| | - Eric D. Pressly
- Department of Materials, Chemistry and Biochemistry, University of California Santa Barbara, California
| | - Lisa Detering
- Department of Radiology, Washington University, St. Louis, Missouri
| | - Cynthia Wang
- Department of Materials, Chemistry and Biochemistry, University of California Santa Barbara, California
| | - Richard Pierce
- Department of Medicine, Washington University, St. Louis, Missouri
| | | | | | - Craig J. Hawker
- Department of Materials, Chemistry and Biochemistry, University of California Santa Barbara, California
| | - Yongjian Liu
- Department of Radiology, Washington University, St. Louis, Missouri
| |
Collapse
|