1
|
Pickett JR, Wu Y, Ta HT. VCAM-1 as a common biomarker in inflammatory bowel disease and colorectal cancer: unveiling the dual anti-inflammatory and anti-cancer capacities of anti-VCAM-1 therapies. Cancer Metastasis Rev 2025; 44:40. [PMID: 40095109 PMCID: PMC11913972 DOI: 10.1007/s10555-025-10258-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Vascular cell adhesion molecule (VCAM)-1 has garnered significant research attention due to its potential as a disease biomarker and drug target across several inflammatory pathologies-including atherosclerosis, asthma, rheumatoid arthritis, and inflammatory bowel disease (IBD). The VCAM-1 protein has also been noted for its functional involvement in cancer metastasis and drug resistance to conventional chemotherapeutics. Although the anti-inflammatory and anti-cancer facets of VCAM-1 antagonisation have been examined separately, there is yet to be a review that explicitly addresses the functional interrelationship between these mechanisms. Furthermore, the pleiotropic mechanisms of anti-VCAM-1 therapies may present a useful paradigm for designing drug candidates with synergistic anti-inflammatory and anti-tumorigenic effects. The pathological overlap between inflammatory bowel disease (IBD) and colitis-associated colorectal cancer (CRC) serves as the quintessential disease model to observe this therapeutic duality. This review thereby details the adhesive mechanisms of VCAM-1 in colorectal disease-specifically, driving immune cell infiltration during IBD and tumour cell metastasis in CRC-and posits the potential of this receptor as a common drug target for both diseases. To explore this hypothesis, the current progress of novel VCAM-1-directed drug candidates in experimental models of IBD and CRC is also discussed.
Collapse
Affiliation(s)
- Jessica R Pickett
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia
| | - Yuao Wu
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia
| | - Hang Thu Ta
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia.
| |
Collapse
|
2
|
Castro R, Adair JH, Mastro AM, Neuberger T, Matters GL. VCAM-1-targeted nanoparticles to diagnose, monitor and treat atherosclerosis. Nanomedicine (Lond) 2024; 19:723-735. [PMID: 38420919 DOI: 10.2217/nnm-2023-0282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
Vascular cell adhesion molecule-1 (VCAM-1) was identified over 2 decades ago as an endothelial adhesion receptor involved in leukocyte recruitment and cell-based immune responses. In atherosclerosis, a chronic inflammatory disease of the blood vessels that is the leading cause of death in the USA, endothelial VCAM-1 is robustly expressed beginning in the early stages of the disease. The interactions of circulating immune cells with VCAM-1 on the activated endothelial cell surface promote the uptake of monocytes and the progression of atherosclerotic lesions in susceptible vessels. Herein, we review the role of VCAM-1 in atherosclerosis and the use of VCAM-1 binding peptides, antibodies and aptamers as targeting agents for nanoplatforms for early detection and treatment of atherosclerotic disease.
Collapse
Affiliation(s)
- Rita Castro
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Pharmaceutical Sciences & Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisboa, Portugal
| | - James H Adair
- Department of Materials Science, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Pharmacology, The Pennsylvania State University, University Park, PA 16802, USA
| | | | - Thomas Neuberger
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of The Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Gail L Matters
- Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
3
|
Tao Y, Lan X, Zhang Y, Fu C, Liu L, Cao F, Guo W. Biomimetic nanomedicines for precise atherosclerosis theranostics. Acta Pharm Sin B 2023; 13:4442-4460. [PMID: 37969739 PMCID: PMC10638499 DOI: 10.1016/j.apsb.2022.11.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/13/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis (AS) is a leading cause of the life-threatening cardiovascular disease (CVD), creating an urgent need for efficient, biocompatible therapeutics for diagnosis and treatment. Biomimetic nanomedicines (bNMs) are moving closer to fulfilling this need, pushing back the frontier of nano-based drug delivery systems design. This review seeks to outline how these nanomedicines (NMs) might work to diagnose and treat atherosclerosis, to trace the trajectory of their development to date and in the coming years, and to provide a foundation for further discussion about atherosclerotic theranostics.
Collapse
Affiliation(s)
- Ying Tao
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Biomedical Engineering & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Xinmiao Lan
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Yang Zhang
- Department of Cardiology, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Chenxing Fu
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Lu Liu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR 999077, China
| | - Feng Cao
- Department of Cardiology, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Biomedical Engineering & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| |
Collapse
|
4
|
Li J, Centurion F, Chen R, Gu Z. Intravascular Imaging of Atherosclerosis by Using Engineered Nanoparticles. BIOSENSORS 2023; 13:319. [PMID: 36979531 PMCID: PMC10046792 DOI: 10.3390/bios13030319] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 06/18/2023]
Abstract
Atherosclerosis is a leading cause of morbidity and mortality, and high-risk atherosclerotic plaques can result in myocardial infarction, stroke, and/or sudden death. Various imaging and sensing techniques (e.g., ultrasound, optical coherence tomography, fluorescence, photoacoustic) have been developed for scanning inside blood vessels to provide accurate detection of high-risk atherosclerotic plaques. Nanoparticles have been utilized in intravascular imaging to enable targeted detection of high-risk plaques, to enhance image contrast, and in some applications to also provide therapeutic functions of atherosclerosis. In this paper, we review the recent progress on developing nanoparticles for intravascular imaging of atherosclerosis. We discuss the basic nanoparticle design principles, imaging modalities and instrumentations, and common targets for atherosclerosis. The review is concluded and highlighted with discussions on challenges and opportunities for bringing nanoparticles into in vivo (pre)clinical intravascular applications.
Collapse
Affiliation(s)
- Jiawen Li
- School of Electrical and Mechanical Engineering, University of Adelaide, Adelaide, SA 5005, Australia
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, SA 5005, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Franco Centurion
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Rouyan Chen
- School of Electrical and Mechanical Engineering, University of Adelaide, Adelaide, SA 5005, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Zi Gu
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine (ACN), University of New South Wales, Sydney, NSW 2052, Australia
- UNSW RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
5
|
Tu Y, Ma X, Chen H, Fan Y, Jiang L, Zhang R, Cheng Z. Molecular Imaging of Matrix Metalloproteinase-2 in Atherosclerosis Using a Smart Multifunctional PET/MRI Nanoparticle. Int J Nanomedicine 2022; 17:6773-6789. [PMID: 36600879 PMCID: PMC9805955 DOI: 10.2147/ijn.s385679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/09/2022] [Indexed: 12/29/2022] Open
Abstract
Background Matrix metalloproteinases from macrophages are important intraplaque components that play pivotal roles in plaque progression and regression. This study sought to develop a novel multifunctional positron emission tomography (PET) and magnetic resonance imaging (MRI) contrast agents based on MMP-2 cleavable nanoparticles to noninvasive assessment of MMP-2 activity in mouse carotid atherosclerotic plaques. Results Macrophage-rich vascular lesions were induced by carotid ligation plus high-fat diet and streptozotocin-induced diabetes in CL57/BL6 mice. To render iron oxide nanoparticles (IONP) specific for the extracellular MMP-2, the magnetic nanoparticle base material has been derivatized with 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) for the nuclear tracer 64Cu labeling and the MMP-2-cleavable peptide modified with polyethylene glycol 2000, yielding a multi-modality reporter (64Cu-NOTA-IONP@MMP2c-PEG2K, MMP2cNPs) for PET/MR imaging. Small animal PET imaging and biodistribution data revealed that MMP2cNPs exhibited remarkable plaque uptake (3.06 ± 0.87% ID/g and 1.83 ± 0.28% ID/g at 4 and 12 h, respectively). And MMP2cNPs were rapidly cleared from the contralateral normal carotid artery, resulting in excellent plaque-to-normal carotid artery contrasts. Furthermore, in vivo MRI showed a preferential accumulation of MMP2cNPs in atherosclerotic lesions compared with the non-cleavable reference compound, MMP2ncNPs. In addition, histological analyses revealed iron accumulations in the carotid atherosclerotic plaque, in colocalization with MMP-2 expression and macrophages. Conclusion Using a combination of innovative imaging modalities, in this study, we demonstrate the feasibility of applying the novel smart MMP2cNPs as a PET/MR hybrid imaging contrast agent for detection of MMP-2 in atherosclerotic plaque in vivo.
Collapse
Affiliation(s)
- Yingfeng Tu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China,Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Stanford, CA, USA
| | - Xiaowei Ma
- Department of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Hao Chen
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Stanford, CA, USA,Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Yuhua Fan
- College of Pharmacy, Harbin Medical University, Daqing, Heilongjiang, People’s Republic of China
| | - Lei Jiang
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Stanford, CA, USA
| | - Ruiping Zhang
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Stanford, CA, USA,The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, People’s Republic of China,Ruiping Zhang, Department of Radiology, the Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, People’s Republic of China, Email
| | - Zhen Cheng
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Stanford, CA, USA,Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China,Correspondence: Zhen Cheng, Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Canary Center at Stanford for Cancer Early Detection, 1201 Welch Road, Lucas Expansion, P095, Stanford University, Stanford, CA, 94305, USA, Tel +01-650-723-7866, Email
| |
Collapse
|
6
|
Xu W, Yang X, Li Y, Jiang G, Jia S, Gong Z, Mao Y, Zhang S, Teng Y, Zhu J, He Q, Wan L, Liang D, Li Y, Hu Z, Zheng H, Liu X, Zhang N. Deep Learning-Based Automated Detection of Arterial Vessel Wall and Plaque on Magnetic Resonance Vessel Wall Images. Front Neurosci 2022; 16:888814. [PMID: 35720719 PMCID: PMC9198483 DOI: 10.3389/fnins.2022.888814] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/21/2022] [Indexed: 12/04/2022] Open
Abstract
Purpose To develop and evaluate an automatic segmentation method of arterial vessel walls and plaques, which is beneficial for facilitating the arterial morphological quantification in magnetic resonance vessel wall imaging (MRVWI). Methods MRVWI images acquired from 124 patients with atherosclerotic plaques were included. A convolutional neural network-based deep learning model, namely VWISegNet, was used to extract the features from MRVWI images and calculate the category of each pixel to facilitate the segmentation of vessel wall. Two-dimensional (2D) cross-sectional slices reconstructed from all plaques and 7 main arterial segments of 115 patients were used to build and optimize the deep learning model. The model performance was evaluated on the remaining nine-patient test set using the Dice similarity coefficient (DSC) and average surface distance (ASD). Results The proposed automatic segmentation method demonstrated satisfactory agreement with the manual method, with DSCs of 93.8% for lumen contours and 86.0% for outer wall contours, which were higher than those obtained from the traditional U-Net, Attention U-Net, and Inception U-Net on the same nine-subject test set. And all the ASD values were less than 0.198 mm. The Bland-Altman plots and scatter plots also showed that there was a good agreement between the methods. All intraclass correlation coefficient values between the automatic method and manual method were greater than 0.780, and greater than that between two manual reads. Conclusion The proposed deep learning-based automatic segmentation method achieved good consistency with the manual methods in the segmentation of arterial vessel wall and plaque and is even more accurate than manual results, hence improved the convenience of arterial morphological quantification.
Collapse
Affiliation(s)
- Wenjing Xu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Information Technology, Beijing University of Technology, Beijing, China
| | - Xiong Yang
- United Imaging Healthcare Co., Ltd., Shanghai, China
| | - Yikang Li
- Department of Computing, Imperial College London, London, United Kingdom
| | - Guihua Jiang
- Department of Radiology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Sen Jia
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhenhuan Gong
- United Imaging Healthcare Co., Ltd., Shanghai, China
| | - Yufei Mao
- United Imaging Healthcare Co., Ltd., Shanghai, China
| | - Shuheng Zhang
- United Imaging Healthcare Co., Ltd., Shanghai, China
| | - Yanqun Teng
- United Imaging Healthcare Co., Ltd., Shanghai, China
| | - Jiayu Zhu
- United Imaging Healthcare Co., Ltd., Shanghai, China
| | - Qiang He
- United Imaging Healthcare Co., Ltd., Shanghai, China
| | - Liwen Wan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dong Liang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ye Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhanli Hu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Na Zhang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
7
|
Zhang M, Xie Z, Long H, Ren K, Hou L, Wang Y, Xu X, Lei W, Yang Z, Ahmed S, Zhang H, Zhao G. Current advances in the imaging of atherosclerotic vulnerable plaque using nanoparticles. Mater Today Bio 2022; 14:100236. [PMID: 35341094 PMCID: PMC8943324 DOI: 10.1016/j.mtbio.2022.100236] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/13/2022] [Accepted: 03/05/2022] [Indexed: 01/29/2023]
Abstract
Vulnerable atherosclerotic plaques of the artery wall that pose a significant risk of cardio-cerebral vascular accidents remain the global leading cause of morbidity and mortality. Thus, early delineation of vulnerable atherosclerotic plaques is of clinical importance for prevention and treatment. The currently available imaging technologies mainly focus on the structural assessment of the vascular wall. Unfortunately, several disadvantages in these strategies limit the improvement in imaging effect. Nanoparticle technology is a novel diagnostic strategy for targeting and imaging pathological biomarkers. New functionalized nanoparticles that detect hallmarks of vulnerable plaques are promising for advance further control of this critical illness. The review aims to address the current opportunities and challenges for the use of nanoparticle technology in imagining vulnerable plaques.
Collapse
|
8
|
Tiwari A, Elgrably B, Saar G, Vandoorne K. Multi-Scale Imaging of Vascular Pathologies in Cardiovascular Disease. Front Med (Lausanne) 2022; 8:754369. [PMID: 35071257 PMCID: PMC8766766 DOI: 10.3389/fmed.2021.754369] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Cardiovascular disease entails systemic changes in the vasculature. The endothelial cells lining the blood vessels are crucial in the pathogenesis of cardiovascular disease. Healthy endothelial cells direct the blood flow to tissues as vasodilators and act as the systemic interface between the blood and tissues, supplying nutrients for vital organs, and regulating the smooth traffic of leukocytes into tissues. In cardiovascular diseases, when inflammation is sensed, endothelial cells adjust to the local or systemic inflammatory state. As the inflamed vasculature adjusts, changes in the endothelial cells lead to endothelial dysfunction, altered blood flow and permeability, expression of adhesion molecules, vessel wall inflammation, thrombosis, angiogenic processes, and extracellular matrix production at the endothelial cell level. Preclinical multi-scale imaging of these endothelial changes using optical, acoustic, nuclear, MRI, and multimodal techniques has progressed, due to technical advances and enhanced biological understanding on the interaction between immune and endothelial cells. While this review highlights biological processes that are related to changes in the cardiac vasculature during cardiovascular diseases, it also summarizes state-of-the-art vascular imaging techniques. The advantages and disadvantages of the different imaging techniques are highlighted, as well as their principles, methodologies, and preclinical and clinical applications with potential future directions. These multi-scale approaches of vascular imaging carry great potential to further expand our understanding of basic vascular biology, to enable early diagnosis of vascular changes and to provide sensitive diagnostic imaging techniques in the management of cardiovascular disease.
Collapse
Affiliation(s)
- Ashish Tiwari
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Betsalel Elgrably
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Galit Saar
- Biomedical Core Facility, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Katrien Vandoorne
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
9
|
Zia A, Wu Y, Nguyen T, Wang X, Peter K, Ta HT. The choice of targets and ligands for site-specific delivery of nanomedicine to atherosclerosis. Cardiovasc Res 2021; 116:2055-2068. [PMID: 32077918 DOI: 10.1093/cvr/cvaa047] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/23/2019] [Accepted: 02/17/2020] [Indexed: 12/22/2022] Open
Abstract
As nanotechnologies advance into clinical medicine, novel methods for applying nanomedicine to cardiovascular diseases are emerging. Extensive research has been undertaken to unlock the complex pathogenesis of atherosclerosis. However, this complexity presents challenges to develop effective imaging and therapeutic modalities for early diagnosis and acute intervention. The choice of ligand-receptor system vastly influences the effectiveness of nanomedicine. This review collates current ligand-receptor systems used in targeting functionalized nanoparticles for diagnosis and treatment of atherosclerosis. Our focus is on the binding affinity and selectivity of ligand-receptor systems, as well as the relative abundance of targets throughout the development and progression of atherosclerosis. Antibody-based targeting systems are currently the most commonly researched due to their high binding affinities when compared with other ligands, such as antibody fragments, peptides, and other small molecules. However, antibodies tend to be immunogenic due to their size. Engineering antibody fragments can address this issue but will compromise their binding affinity. Peptides are promising ligands due to their synthetic flexibility and low production costs. Alongside the aforementioned binding affinity of ligands, the choice of target and its abundance throughout distinct stages of atherosclerosis and thrombosis is relevant to the intended purpose of the nanomedicine. Further studies to investigate the components of atherosclerotic plaques are required as their cellular and molecular profile shifts over time.
Collapse
Affiliation(s)
- Adil Zia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yuao Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Tuan Nguyen
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xiaowei Wang
- Baker Heart and Diabetes Institute, Melbourne, VIC 3000, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, VIC 3000, Australia
| | - Hang T Ta
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
10
|
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
|
11
|
Development of an LDL Receptor-Targeted Peptide Susceptible to Facilitate the Brain Access of Diagnostic or Therapeutic Agents. BIOLOGY 2020; 9:biology9070161. [PMID: 32664518 PMCID: PMC7407834 DOI: 10.3390/biology9070161] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/30/2020] [Accepted: 07/08/2020] [Indexed: 12/02/2022]
Abstract
Blood-brain barrier (BBB) crossing and brain penetration are really challenging for the delivery of therapeutic agents and imaging probes. The development of new crossing strategies is needed, and a wide range of approaches (invasive or not) have been proposed so far. The receptor-mediated transcytosis is an attractive mechanism, allowing the non-invasive penetration of the BBB. Among available targets, the low-density lipoprotein (LDL) receptor (LDLR) shows favorable characteristics mainly because of the lysosome-bypassed pathway of LDL delivery to the brain, allowing an intact discharge of the carried ligand to the brain targets. The phage display technology was employed to identify a dodecapeptide targeted to the extracellular domain of LDLR (ED-LDLR). This peptide was able to bind the ED-LDLR in the presence of natural ligands and dissociated at acidic pH and in the absence of calcium, in a similar manner as the LDL. In vitro, our peptide was endocytosed by endothelial cells through the caveolae-dependent pathway, proper to the LDLR route in BBB, suggesting the prevention of its lysosomal degradation. The in vivo studies performed by magnetic resonance imaging and fluorescent lifetime imaging suggested the brain penetration of this ED-LDLR-targeted peptide.
Collapse
|
12
|
Fanfone D, Stanicki D, Nonclercq D, Port M, Vander Elst L, Laurent S, Muller RN, Saussez S, Burtea C. Molecular Imaging of Galectin-1 Expression as a Biomarker of Papillary Thyroid Cancer by Using Peptide-Functionalized Imaging Probes. BIOLOGY 2020; 9:biology9030053. [PMID: 32183292 PMCID: PMC7150867 DOI: 10.3390/biology9030053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 01/11/2023]
Abstract
Thyroid cancers are the most frequent endocrine cancers and their incidence is increasing worldwide. Thyroid nodules occur in over 19–68% of the population, but only 7–15% of them are diagnosed as malignant. Diagnosis relies on a fine needle aspiration biopsy, which is often inconclusive and about 90% of thyroidectomies are performed for benign lesions. Galectin-1 has been proposed as a confident biomarker for the discrimination of malignant from benign nodules. We previously identified by phage display two peptides (P1 and P7) targeting galectin-1, with the goal of developing imaging probes for non-invasive diagnosis of thyroid cancer. The peptides were coupled to ultra-small superparamagnetic particles of iron oxide (USPIO) or to a near-infrared dye (CF770) for non-invasive detection of galectin-1 expression in a mouse model of papillary thyroid cancer (PTC, as the most frequent one) by magnetic resonance imaging and fluorescence lifetime imaging. The imaging probes functionalized with the two peptides presented comparable image enhancement characteristics. However, those coupled to P7 were more favorable, and showed decreased retention by the liver and spleen (known for their galectin-1 expression) and high sensitivity (75%) and specificity (100%) of PTC detection, which confirm the aptitude of this peptide to discriminate human malignant from benign nodules (80% sensitivity, 100% specificity) previously observed by immunohistochemistry.
Collapse
Affiliation(s)
- Deborah Fanfone
- Department of General, Organic and Biomedical Chemistry, UMONS, Avenue Victor Maistriau 19, 7000 Mons, Belgium; (D.F.); (L.V.E.); (S.L.); (R.N.M.)
| | - Dimitri Stanicki
- Center for Microscopy and Molecular Imaging, Rue Adrienne Bolland, 8, 6041 Charleroi, Belgium;
| | - Denis Nonclercq
- Laboratory of Histology, Faculty of Medicine and Pharmacy, University of Mons–UMONS, Avenue du Champ de Mars 6, 7000 Mons, Belgium;
| | - Marc Port
- Laboratoire de Génomique, Bioinformatique et Chimie Moléculaire (EA 7528), Equipe Chimie Moléculaire, Conservatoire National des Arts et Métiers (CNAM), HESAM Université, 75003 Paris, France;
| | - Luce Vander Elst
- Department of General, Organic and Biomedical Chemistry, UMONS, Avenue Victor Maistriau 19, 7000 Mons, Belgium; (D.F.); (L.V.E.); (S.L.); (R.N.M.)
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, UMONS, Avenue Victor Maistriau 19, 7000 Mons, Belgium; (D.F.); (L.V.E.); (S.L.); (R.N.M.)
- Center for Microscopy and Molecular Imaging, Rue Adrienne Bolland, 8, 6041 Charleroi, Belgium;
| | - Robert N. Muller
- Department of General, Organic and Biomedical Chemistry, UMONS, Avenue Victor Maistriau 19, 7000 Mons, Belgium; (D.F.); (L.V.E.); (S.L.); (R.N.M.)
- Center for Microscopy and Molecular Imaging, Rue Adrienne Bolland, 8, 6041 Charleroi, Belgium;
| | - Sven Saussez
- Laboratory of Human Anatomy and Experimental Oncology, UMONS, Avenue du Champ de Mars, 6, 7000 Mons, Belgium;
| | - Carmen Burtea
- Department of General, Organic and Biomedical Chemistry, UMONS, Avenue Victor Maistriau 19, 7000 Mons, Belgium; (D.F.); (L.V.E.); (S.L.); (R.N.M.)
- Correspondence: ; Tel.: +32-6537-3814
| |
Collapse
|
13
|
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]
|
14
|
Li J, Cha R, Luo H, Hao W, Zhang Y, Jiang X. Nanomaterials for the theranostics of obesity. Biomaterials 2019; 223:119474. [PMID: 31536920 DOI: 10.1016/j.biomaterials.2019.119474] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/01/2019] [Accepted: 09/03/2019] [Indexed: 02/06/2023]
Abstract
As a chronic and lifelong disease, obesity not only significant impairs health but also dramatically shortens life span (at least 10 years). Obesity requires a life-long effort for the successful treatment because a number of abnormalities would appear in the development of obesity. Nanomaterials possess large specific surface area, strong absorptivity, and high bioavailability, especially the good targeting properties and adjustable release rate, which would benefit the diagnosis and treatment of obesity and obesity-related metabolic diseases. Herein, we discussed the therapy and diagnosis of obesity and obesity-related metabolic diseases by using nanomaterials. Therapies of obesity with nanomaterials include improving intestinal health and reducing energy intake, targeting and treating functional cell abnormalities, regulating redox homeostasis, and removing free lipoprotein in blood. Diagnosis of obesity-related metabolic diseases would benefit the therapy of these diseases. The development of nanomaterials will promote the diagnosis and therapy of obesity and obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Juanjuan Li
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, PR China
| | - Ruitao Cha
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, PR China.
| | - Huize Luo
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, PR China
| | - Wenshuai Hao
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, PR China
| | - Yan Zhang
- Department of Cardiac Surgery, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100032, PR China.
| | - Xingyu Jiang
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, PR China; Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong, 518055, PR China; University of Chinese Academy of Sciences, 19 A Yuquan Road, Shijingshan District, Beijing, 100049, PR China.
| |
Collapse
|
15
|
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
|
16
|
MRI visualization of neuroinflammation using VCAM-1 targeted paramagnetic micelles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2341-2350. [DOI: 10.1016/j.nano.2017.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/05/2017] [Accepted: 10/13/2017] [Indexed: 01/29/2023]
|
17
|
Li X, Sui Z, Li X, Xu W, Guo Q, Sun J, Jing F. Perfluorooctylbromide nanoparticles for ultrasound imaging and drug delivery. Int J Nanomedicine 2018; 13:3053-3067. [PMID: 29872293 PMCID: PMC5975599 DOI: 10.2147/ijn.s164905] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Perfluorooctylbromide nanoparticles (PFOB NPs) are a type of multifunctional nanotechnology that has been studied for various medical applications. Commercial ultrasound contrast agents (UCAs) suffer from the following limitations: short half-lives in vivo, high background signal and restricted distribution in the vascular circulation due to their micrometer dimensions. PFOB NPs are new potential UCAs that persist for long periods in the circulatory system, possess a relatively stable echogenic response without increasing the background signal and exhibit lower acoustic attenuation than commercial UCAs. Furthermore, PFOB NPs may also serve as drug delivery vehicles in which drugs are dissolved in the outer lipid or polymer layer for subsequent delivery to target sites in site-targeted therapy. The use of PFOB NPs as carriers has the potential advantage of selectively delivering payloads to the target site while improving visualization of the site using ultrasound (US) imaging. Unfortunately, the application of PFOB NPs to the field of ultrasonography has been limited because of the low intensity of US reflection. Numerous researchers have realized the potential use of PFOB NPs as UCAs and thus have developed alternative approaches to apply PFOB NPs in ultrasonography. In this article, we review the latest approaches for using PFOB NPs to enhance US imaging in vivo. In addition, this article emphasizes the application of PFOB NPs as promising drug delivery carriers for cancer and atherosclerosis treatments, as PFOB NPs can transport different drug payloads for various applications with good efficacy. We also note the challenges and future study directions for the application of PFOB NPs as both a delivery system for therapeutic agents and a diagnostic agent for ultrasonography.
Collapse
Affiliation(s)
- Xiao Li
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Zhongguo Sui
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Xin Li
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Wen Xu
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Qie Guo
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Jialin Sun
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Fanbo Jing
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| |
Collapse
|
18
|
The Multifaceted Uses and Therapeutic Advantages of Nanoparticles for Atherosclerosis Research. MATERIALS 2018; 11:ma11050754. [PMID: 29738480 PMCID: PMC5978131 DOI: 10.3390/ma11050754] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 12/27/2022]
Abstract
Nanoparticles are uniquely suited for the study and development of potential therapies against atherosclerosis by virtue of their size, fine-tunable properties, and ability to incorporate therapies and/or imaging modalities. Furthermore, nanoparticles can be specifically targeted to the atherosclerotic plaque, evading off-target effects and/or associated cytotoxicity. There has been a wealth of knowledge available concerning the use of nanotechnologies in cardiovascular disease and atherosclerosis, in particular in animal models, but with a major focus on imaging agents. In fact, roughly 60% of articles from an initial search for this review included examples of imaging applications of nanoparticles. Thus, this review focuses on experimental therapy interventions applied to and observed in animal models. Particular emphasis is placed on how nanoparticle materials and properties allow researchers to learn a great deal about atherosclerosis. The objective of this review was to provide an update for nanoparticle use in imaging and drug delivery studies and to illustrate how nanoparticles can be used for sensing and modelling, for studying fundamental biological mechanisms, and for the delivery of biotherapeutics such as proteins, peptides, nucleic acids, and even cells all with the goal of attenuating atherosclerosis. Furthermore, the various atherosclerosis processes targeted mainly for imaging studies have been summarized in the hopes of inspiring new and exciting targeted therapeutic and/or imaging strategies.
Collapse
|
19
|
Jin K, Luo Z, Zhang B, Pang Z. Biomimetic nanoparticles for inflammation targeting. Acta Pharm Sin B 2018; 8:23-33. [PMID: 29872620 PMCID: PMC5985691 DOI: 10.1016/j.apsb.2017.12.002] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
There have been many recent exciting developments in biomimetic nanoparticles for biomedical applications. Inflammation, a protective response involving immune cells, blood vessels, and molecular mediators directed against harmful stimuli, is closely associated with many human diseases. As a result, biomimetic nanoparticles mimicking immune cells can help achieve molecular imaging and precise drug delivery to these inflammatory sites. This review is focused on inflammation-targeting biomimetic nanoparticles and will provide an in-depth look at the design of these nanoparticles to maximize their benefits for disease diagnosis and treatment.
Collapse
Key Words
- Biomimetic nanoparticles
- CAM, cellular adhesion molecule
- CCL5, chemokine (C-C motif) ligand 5
- CD40L, cluster of differentiation 40 ligand
- CTC, circulating tumor cell
- CTL, cytotoxic T cell or CD8+ T cell
- CXCL4, chemokine (C-X-C motif) ligand 4
- CXCR1, chemokine (C-X-C motif) receptor 1
- Cell membrane
- Cell membrane proteins
- Cy7, cyanine 7
- DC, dendritic cell
- DSPE-PEG, distearoyl Phosphoethanolamine-poly(ethylene glycol)
- GPIV, glycoprotein IV
- GPIX, glycoprotein IX
- GPIbα, glycoprotein Ibα
- GPV, glycoprotein V
- GPVI, glycoprotein VI
- HUVEC, umbilical cord vascular endothelial cell
- IBD, inflammatory bowel disease
- ICAM-1, intercellular cellular adhesion molecule-1
- IL, interleukin
- IgG, immunoglobulin G
- Immune cells
- Inflammation targeting
- LFA-1, lymphocyte function associated antigen-1
- LLV, leukocyte-like vector
- LPS, lipopolysaccharide
- MHC, major histocompatibility complex
- MRI, magnetic resonance imaging
- Mac-1, macrophage adhesion molecule-1
- Molecular imaging
- NM-NP, neutrophil membrane-coated nanoparticle
- PECAM-1, platelet-endothelial cellular adhesion molecule-1
- PLA-PEG, poly(lactic acid)-poly(ethylene glycol)
- PLGA, poly(lactic-co-glycolic acid)
- PNP, platelet membrane-cloaked nanoparticle
- PSGL-1, P-selectin glycoprotein ligand-1
- RA, rheumatoid arthritis
- RBC, red blood cell
- SLeX, sialyl lewis X
- SPIO, super paramagnetic iron oxide
- TGF-β, transforming growth factor β
- TNF-α, tumor necrosis factor-α
- Targeting ligands
- Th cell, T-helper cell or CD4+ T cell
- VCAM-1, vascular cellular adhesion molecule-1
- VLA-4, very late antigen-4
- VWF, Von Willebrand factor
- apoE–/– mice, Apolipoprotein e knockout mice
Collapse
Affiliation(s)
- Kai Jin
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Zimiao Luo
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
- Biomedical Engineering and Technology Institute, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Bo Zhang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| |
Collapse
|
20
|
André S, Ansciaux E, Saidi E, Larbanoix L, Stanicki D, Nonclercq D, Vander Elst L, Laurent S, Muller RN, Burtea C. Validation by Magnetic Resonance Imaging of the Diagnostic Potential of a Heptapeptide-Functionalized Imaging Probe Targeted to Amyloid-β and Able to Cross the Blood-Brain Barrier. J Alzheimers Dis 2017; 60:1547-1565. [DOI: 10.3233/jad-170563] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Séverine André
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Emilie Ansciaux
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Elamine Saidi
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | | | - Dimitri Stanicki
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | | | - Luce Vander Elst
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Robert N. Muller
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Carmen Burtea
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| |
Collapse
|
21
|
Savla R, Minko T. Nanoparticle design considerations for molecular imaging of apoptosis: Diagnostic, prognostic, and therapeutic value. Adv Drug Deliv Rev 2017; 113:122-140. [PMID: 27374457 DOI: 10.1016/j.addr.2016.06.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 06/21/2016] [Indexed: 12/13/2022]
Abstract
The present review analyzes various approaches for the design and synthesis of different nanoparticles for imaging and therapy. Nanoparticles for computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET) and optical imaging are discussed. The influence of nanoparticle size, shape, surface charge, composition, surface functionalization, active targeting and other factors on imaging and therapeutic efficacy is analyzed. Cyto- and genotoxicity of nanoparticles are also discussed. Special attention in the review is paid to the imaging of apoptotic tissues and cells in different diseases.
Collapse
Affiliation(s)
- Ronak Savla
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, United States; Environmental and Occupational Health Sciences Institute, Piscataway, NJ 08854, United States.
| |
Collapse
|
22
|
Meloni MM, Barton S, Xu L, Kaski JC, Song W, He T. Contrast agents for cardiovascular magnetic resonance imaging: an overview. J Mater Chem B 2017; 5:5714-5725. [DOI: 10.1039/c7tb01241a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Contrast agents for Cardiovascular Magnetic Resonance (CMR) play a major role in research and clinical cardiology.
Collapse
Affiliation(s)
- Marco M. Meloni
- Molecular and Clinical Sciences Research Institute
- St George's, University of London
- London
- UK
- School of Pharmacy and Chemistry
| | - Stephen Barton
- School of Pharmacy and Chemistry
- Kingston University
- London
- UK
| | - Lei Xu
- Department of Radiology
- Beijing Anzhen Hospital
- Beijing
- China
| | - Juan C. Kaski
- Molecular and Clinical Sciences Research Institute
- St George's, University of London
- London
- UK
| | - Wenhui Song
- UCL Centre for Biomaterials
- Division of surgery & Interventional Science
- University College of London
- London
- UK
| | - Taigang He
- Molecular and Clinical Sciences Research Institute
- St George's, University of London
- London
- UK
- Royal Brompton Hospital
| |
Collapse
|
23
|
Khodabandehlou K, Masehi-Lano JJ, Poon C, Wang J, Chung EJ. Targeting cell adhesion molecules with nanoparticles using in vivo and flow-based in vitro models of atherosclerosis. Exp Biol Med (Maywood) 2017; 242:799-812. [PMID: 28195515 DOI: 10.1177/1535370217693116] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Atherosclerosis is a leading cause of death worldwide; in addition to lipid dysfunction, chronic arterial wall inflammation is a key component of atherosclerosis. Techniques that target cell adhesion molecules, which are overexpressed during inflammation, are effective methods to detect and treat atherosclerosis. Specifically, research groups have identified vascular cell adhesion molecule-1, intercellular adhesion molecule-1, platelet endothelial cell adhesion molecule, and selectins (E-selectin and P-selectin) as correlated to atherogenesis. In this review, we discuss recent strategies both in vivo and in vitro that target cell adhesion molecules. First, we discuss peptide-based and antibody (Ab)-based nanoparticles utilized in vivo for diagnostic, therapeutic, and theranostic applications. Second, we discuss flow-based in vitro models that serve to reduce the traditional disadvantages of in vivo studies such as variability, time to develop the disease, and ethical burden, but preserve physiological relevance. The knowledge gained from these targeting studies can be translated into clinical solutions for improved detection, prevention, and treatment of atherosclerosis. Impact statement As atherosclerosis remains the leading cause of death, there is an urgent need to develop better tools for treatment of the disease. The ability to improve current treatments relies on enhancing the accuracy of in vitro and in vivo atherosclerotic models. While in vivo models provide all the relevant testing parameters, variability between animals and among models used is a barrier to reproducible results and comparability of NP efficacy. In vitro cultures isolate cells into microenvironments that fail to take into account flow separation and shear stress, which are characteristics of atherosclerotic lesions. Flow-based in vitro models provide more physiologically relevant platforms, bridging the gap between in vivo and 2D in vitro models. This is the first review that presents recent advances regarding endothelial cell-targeting using adhesion molecules in light of in vivo and flow-based in vitro models, providing insights for future development of optimal strategies against atherosclerosis.
Collapse
Affiliation(s)
- Khosrow Khodabandehlou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Jacqueline J Masehi-Lano
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Christopher Poon
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
24
|
Screening for peptides targeted to IL-7Rα for molecular imaging of rheumatoid arthritis synovium. Arthritis Res Ther 2016; 18:230. [PMID: 27729062 PMCID: PMC5059943 DOI: 10.1186/s13075-016-1133-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/20/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Interleukin-7 receptor alpha (IL-7Rα) represents a biomarker with potential applications in rheumatoid arthritis (RA) diagnosis and therapy. We have therefore searched by phage display potential IL-7Rα specific peptides with the primary goal being to develop in vivo molecular imaging tools. METHODS IL-7Rα-targeted peptides were searched within a disulfide-constrained combinatorial phage displayed library of random linear heptapeptides. The apparent dissociation constant (Kd) and half maximal inhibition constant (IC50) were estimated for phage clones and synthesized peptides by ELISA. We used 5-Aza-2'-deoxycytidine (ADC)-stimulated Jurkat cells and human synovial tissue from patients with RA for in vitro characterization of peptides. For molecular imaging studies performed by magnetic resonance imaging (MRI), experimental arthritis was induced in DBA/1 male mice by immunization with an emulsion of complete Freund's adjuvant and type II collagen from chicken sternal cartilage. RESULTS After several steps of phage display and peptide screening, two IL-7Rα-specific heptapeptides (P258 and P725) were selected from the initial library, based on their affinity for the target (extracellular domain of IL-7Rα, which contains a fibronectin type III repeat-like sequence). P258 (a linear peptide obtained by removing the Cys-constraint) had the lowest affinity for fibronectin itself and was therefore proposed for molecular imaging. After grafting to ultra-small superparamagnetic particles of iron oxide (USPIO), P258 produced a strong negative contrast on MRI in mice with collagen-induced arthritis (CIA), even at 2 hours post injection. The co-localization of USPIO-P258 with IL-7Rα-expressing cells in the synovial tissue from CIA mice and its ability to discriminate the level of IL-7R expression and the disease severity confirmed its efficacy as an in vivo IL-7Rα imaging agent. Interestingly, the cyclic peptide (P725), which was less adequate for molecular imaging because of higher affinity for fibronectin, had a strong ability to compete with IL-7 for the IL-7Rα binding sites, making it a potential candidate for blocking applications. Accordingly, P725 prevented the signal transducer and activator of transcription 5 (STAT5) activation induced by IL-7 in ADC-stimulated Jurkat cells. CONCLUSIONS The two peptides identified in this work demonstrate that IL-7Rα targeting in RA presents potential applications for in vivo molecular imaging and putative blocking purposes.
Collapse
|
25
|
Perreault A, Richter S, Bergman C, Wuest M, Wuest F. Targeting Phosphatidylserine with a 64Cu-Labeled Peptide for Molecular Imaging of Apoptosis. Mol Pharm 2016; 13:3564-3577. [DOI: 10.1021/acs.molpharmaceut.6b00666] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Amanda Perreault
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Susan Richter
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Cody Bergman
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Melinda Wuest
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Frank Wuest
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| |
Collapse
|
26
|
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
|
27
|
Pagoto A, Stefania R, Garello F, Arena F, Digilio G, Aime S, Terreno E. Paramagnetic Phospholipid-Based Micelles Targeting VCAM-1 Receptors for MRI Visualization of Inflammation. Bioconjug Chem 2016; 27:1921-30. [DOI: 10.1021/acs.bioconjchem.6b00308] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Amerigo Pagoto
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126-Torino, Italy
| | - Rachele Stefania
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126-Torino, Italy
| | - Francesca Garello
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126-Torino, Italy
| | - Francesca Arena
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126-Torino, Italy
| | - Giuseppe Digilio
- DISIT,
Università
del Piemonte Orientale “A. Avogadro”, Via T. Michel 11, 15121 Alessandria, Italy
| | - Silvio Aime
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126-Torino, Italy
| | - Enzo Terreno
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126-Torino, Italy
| |
Collapse
|
28
|
Van Koninckxloo A, Henoumont C, Laurent S, Muller RN, Vander Elst L. (1) H-NMR relaxometric studies of interaction between apoptosis specific MRI paramagnetic contrast agents and micellar models of apoptotic cells. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2016; 54:568-574. [PMID: 26647764 DOI: 10.1002/mrc.4397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 10/02/2015] [Accepted: 11/11/2015] [Indexed: 06/05/2023]
Abstract
(1) H-NMR was previously used to analyze the interaction between peptides (E3 and R826) selected by phage display to target apoptotic cells and phospholipidic models of these cells. In order to avoid the use of apoptotic cells and to obtain a fast evaluation of the efficiency of the potential MRI contrast agents obtained by grafting these peptides and their scramble analogs on a paramagnetic gadolinium complex, their proton relaxometric behavior was investigated in the presence of micelles mimicking healthy and apoptotic cells. Their preferential interaction with 1,2-dipalmitoyl-sn-glycero-3-phospho-l-serine micelles mimicking apoptotic cells as compared with 1,2-dipalmitoyl-sn-glycero-3-phosphocholine micelles modeling healthy cells was shown by nuclear magnetic relaxation dispersion profiles and the enhancement of the transverse proton relaxation rates at 60 MHz. The association constant values confirm the stronger interaction of the selected conjugated peptides (Ka Gd-PMN-E3(gadolinium 2,2',2'',2'''-[((4-carboxy)pyridine-2,6-diyl)bis(methylenenitrilo)]-tetrakis acetate) grafted with E3 peptide): 2.43 10(4) m(-1) ; Ka Gd-DTPA-R826(gadolinium ((1-p-isothiocyanatobenzyl)-diethylenetriaminepentaacetate) grafted with R826 peptide): 2.91 10(4) m(-1) ) as compared with their conjugated scrambles (Ka Gd-PMN-E3sc(gadolinium 2,2',2'',2'''-[((4-carboxy)pyridine-2,6-diyl)bis(methylenenitrilo)]-tetrakis acetate) grafted with E3 scramble peptide): 0.18 10(4) m(-1) ; Ka Gd-DTPA-R826sc(gadolinium ((1-p-isothiocyanatobenzyl)-diethylenetriaminepentaacetate) grafted with R826 scramble peptide): 0.32 10(4) m(-1) ) even if the conjugation of E3 and R826 seems to decrease their interaction. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Aurore Van Koninckxloo
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, 7000, Mons, Belgium
| | - Céline Henoumont
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, 7000, Mons, Belgium
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, 7000, Mons, Belgium
- CMMI - Center for Microscopy and Molecular Imaging, 6041, Gosselies, Belgium
| | - Robert N Muller
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, 7000, Mons, Belgium
- CMMI - Center for Microscopy and Molecular Imaging, 6041, Gosselies, Belgium
| | - Luce Vander Elst
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, 7000, Mons, Belgium
- CMMI - Center for Microscopy and Molecular Imaging, 6041, Gosselies, Belgium
| |
Collapse
|
29
|
Zhang J, Zu Y, Dhanasekara CS, Li J, Wu D, Fan Z, Wang S. Detection and treatment of atherosclerosis using nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27241794 DOI: 10.1002/wnan.1412] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/25/2016] [Accepted: 04/12/2016] [Indexed: 01/10/2023]
Abstract
Atherosclerosis is the key pathogenesis of cardiovascular disease, which is a silent killer and a leading cause of death in the United States. Atherosclerosis starts with the adhesion of inflammatory monocytes on the activated endothelial cells in response to inflammatory stimuli. These monocytes can further migrate into the intimal layer of the blood vessel where they differentiate into macrophages, which take up oxidized low-density lipoproteins and release inflammatory factors to amplify the local inflammatory response. After accumulation of cholesterol, the lipid-laden macrophages are transformed into foam cells, the hallmark of the early stage of atherosclerosis. Foam cells can die from apoptosis or necrosis, and the intracellular lipid is deposed in the artery wall forming lesions. The angiogenesis for nurturing cells is enhanced during lesion development. Proteases released from macrophages, foam cells, and other cells degrade the fibrous cap of the lesion, resulting in rupture of the lesion and subsequent thrombus formation. Thrombi can block blood circulation, which represents a major cause of acute heart events and stroke. There are generally no symptoms in the early stages of atherosclerosis. Current detection techniques cannot easily, safely, and effectively detect the lesions in the early stages, nor can they characterize the lesion features such as the vulnerability. While the available therapeutic modalities cannot target specific molecules, cells, and processes in the lesions, nanoparticles appear to have a promising potential in improving atherosclerosis detection and treatment via targeting the intimal macrophages, foam cells, endothelial cells, angiogenesis, proteolysis, apoptosis, and thrombosis. Indeed, many nanoparticles have been developed in improving blood lipid profile and decreasing inflammatory response for enhancing therapeutic efficacy of drugs and decreasing their side effects. WIREs Nanomed Nanobiotechnol 2017, 9:e1412. doi: 10.1002/wnan.1412 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Jia Zhang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | | | - Jun Li
- Laboratory Animal Center, Peking University, Beijing, PR China
| | - Dayong Wu
- Nutritional Immunology Laboratory, Jean Mayer Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Zhaoyang Fan
- Department of Electrical and Computer Engineering and Nano Tech Center, Texas Tech University, Lubbock, TX, USA
| | - Shu Wang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
30
|
Liu Z, Larsen BT, Lerman LO, Gray BD, Barber C, Hedayat AF, Zhao M, Furenlid LR, Pak KY, Woolfenden JM. Detection of atherosclerotic plaques in ApoE-deficient mice using (99m)Tc-duramycin. Nucl Med Biol 2016; 43:496-505. [PMID: 27236285 DOI: 10.1016/j.nucmedbio.2016.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/07/2016] [Accepted: 05/11/2016] [Indexed: 12/25/2022]
Abstract
UNLABELLED Apoptosis of macrophages and smooth muscle cells is linked to atherosclerotic plaque destabilization. The apoptotic cascade leads to exposure of phosphatidylethanolamine (PE) on the outer leaflet of the cell membrane, thereby making apoptosis detectable using probes targeting PE. The objective of this study was to exploit capabilities of a PE-specific imaging probe, (99m)Tc-duramycin, in localizing atherosclerotic plaque and assessing plaque evolution in apolipoprotein-E knockout (ApoE(-/-)) mice. METHODS Atherosclerosis was induced in ApoE(-/-) mice by feeding an atherogenic diet. (99m)Tc-duramycin images were acquired using a small-animal SPECT imager. Six ApoE(-/-) mice at 20weeks of age (Group I) were imaged and then sacrificed for ex vivo analyses. Six additional ApoE(-/-) mice (Group II) were imaged at 20 and 40weeks of age before sacrifice. Six ApoE wild-type (ApoE(+/+)) mice (Group III) were imaged at 40weeks as controls. Five additional ApoE(-/-) mice (40weeks of age) (Group IV) were imaged with a (99m)Tc-labeled inactive peptide, (99m)Tc-LinDUR, to assess (99m)Tc-duramycin targeting specificity. RESULTS Focal (99m)Tc-duramycin uptake in the ascending aorta and aortic arch was detected at 20 and 40weeks in the ApoE(-/-) mice but not in ApoE(+/+) mice. (99m)Tc-duramycin uptake in the aortic lesions increased 2.2-fold on quantitative imaging in the ApoE(-/-) mice between 20 and 40weeks. Autoradiographic and histological data indicated significantly increased (99m)Tc-duramycin uptake in the ascending aorta and aortic arch associated with advanced plaques. Quantitative autoradiography showed that the ratio of activity in the aortic arch to descending thoracic aorta, which had no plaques or radioactive uptake, was 2.1 times higher at 40weeks than at 20weeks (6.62±0.89 vs. 3.18±0.29, P<0.01). There was barely detectable focal uptake of (99m)Tc-duramycin in the aortic arch of ApoE(+/+) mice. No detectable (99m)Tc-LinDUR uptake was observed in the aortas of ApoE(-/-) mice. CONCLUSIONS PE-targeting properties of (99m)Tc-duramycin in the atherosclerotic mouse aortas were noninvasively characterized. (99m)Tc-duramycin is promising in localizing advanced atherosclerotic plaques.
Collapse
Affiliation(s)
- Zhonglin Liu
- Department of Medical Imaging, University of Arizona, Tucson, AZ, USA.
| | | | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Brian D Gray
- Molecular Targeting Technologies, Inc, West Chester, PA, USA
| | - Christy Barber
- Department of Medical Imaging, University of Arizona, Tucson, AZ, USA
| | - Ahmad F Hedayat
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Ming Zhao
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lars R Furenlid
- Department of Medical Imaging, University of Arizona, Tucson, AZ, USA
| | - Koon Y Pak
- Molecular Targeting Technologies, Inc, West Chester, PA, USA
| | | |
Collapse
|
31
|
Abstract
Molecular imaging offers great potential for noninvasive visualization and quantitation of the cellular and molecular components involved in atherosclerotic plaque stability. In this chapter, we review emerging molecular imaging modalities and approaches for quantitative, noninvasive detection of early biological processes in atherogenesis, including vascular endothelial permeability, endothelial adhesion molecule up-regulation, and macrophage accumulation, with special emphasis on mouse models. We also highlight a number of targeted imaging nanomaterials for assessment of advanced atherosclerotic plaques, including extracellular matrix degradation, proteolytic enzyme activity, and activated platelets using mouse models of atherosclerosis. The potential for clinical translation of molecular imaging nanomaterials for assessment of atherosclerotic plaque biology, together with multimodal approaches is also discussed.
Collapse
|
32
|
Urao N, Mirza RE, Heydemann A, Garcia J, Koh TJ. Thrombospondin-1 levels correlate with macrophage activity and disease progression in dysferlin deficient mice. Neuromuscul Disord 2016; 26:240-51. [PMID: 26927626 DOI: 10.1016/j.nmd.2016.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/24/2015] [Accepted: 01/14/2016] [Indexed: 10/22/2022]
Abstract
Dysferlinopathy is associated with accumulation of thrombospondin (TSP)-1 and macrophages, both of which may contribute to the pathogenesis of the disease. The purpose of this study was to determine whether TSP-1 levels can predict macrophage activity and disease progression in dysferlin deficient BlaJ mice, focusing on the early disease process. In 3 month-old BlaJ mice, muscle TSP-1 levels exhibited strong positive correlations with both accumulation of F4/80hi macrophages and with their in vivo phagocytic activity in psoas muscles as measured by magnetic resonance imaging and flow cytometry. Muscle TSP-1 levels also exhibited a strong negative correlation with muscle mass and strong positive correlations with histological measurements of muscle fiber infiltration and regeneration. Over the course of disease progression from 3 to 12 months of age, muscle TSP-1 levels showed more complicated relationships with macrophage activity and an inverse relationship with muscle mass. Importantly, blood TSP-1 levels showed strong correlations with macrophage activity and muscle degeneration, particularly early in disease progression in BlaJ mice. These data indicate that TSP-1 may contribute to a destructive macrophage response in dysferlinopathy and pose the intriguing possibility that TSP-1 levels may serve as a biomarker for disease progression.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA; Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rita E Mirza
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ahlke Heydemann
- Department of Physiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jesus Garcia
- Department of Physiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Timothy J Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA; Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
33
|
Lee N, Yoo D, Ling D, Cho MH, Hyeon T, Cheon J. Iron Oxide Based Nanoparticles for Multimodal Imaging and Magnetoresponsive Therapy. Chem Rev 2015; 115:10637-89. [PMID: 26250431 DOI: 10.1021/acs.chemrev.5b00112] [Citation(s) in RCA: 618] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Nohyun Lee
- School of Advanced Materials Engineering, Kookmin University , Seoul 136-702, Korea
| | - Dongwon Yoo
- Department of Chemistry, Yonsei University , Seoul 120-749, Korea
| | - Daishun Ling
- Center for Nanoparticle Research, Institute for Basic Science (IBS) , Seoul 151-742, Korea.,School of Chemical and Biological Engineering, Seoul National University , Seoul 151-742, Korea.,Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, PR China
| | - Mi Hyeon Cho
- Department of Chemistry, Yonsei University , Seoul 120-749, Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS) , Seoul 151-742, Korea.,School of Chemical and Biological Engineering, Seoul National University , Seoul 151-742, Korea
| | - Jinwoo Cheon
- Department of Chemistry, Yonsei University , Seoul 120-749, Korea
| |
Collapse
|
34
|
Bakermans AJ, Abdurrachim D, Moonen RPM, Motaal AG, Prompers JJ, Strijkers GJ, Vandoorne K, Nicolay K. Small animal cardiovascular MR imaging and spectroscopy. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2015; 88-89:1-47. [PMID: 26282195 DOI: 10.1016/j.pnmrs.2015.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/09/2015] [Accepted: 03/09/2015] [Indexed: 06/04/2023]
Abstract
The use of MR imaging and spectroscopy for studying cardiovascular disease processes in small animals has increased tremendously over the past decade. This is the result of the remarkable advances in MR technologies and the increased availability of genetically modified mice. MR techniques provide a window on the entire timeline of cardiovascular disease development, ranging from subtle early changes in myocardial metabolism that often mark disease onset to severe myocardial dysfunction associated with end-stage heart failure. MR imaging and spectroscopy techniques play an important role in basic cardiovascular research and in cardiovascular disease diagnosis and therapy follow-up. This is due to the broad range of functional, structural and metabolic parameters that can be quantified by MR under in vivo conditions non-invasively. This review describes the spectrum of MR techniques that are employed in small animal cardiovascular disease research and how the technological challenges resulting from the small dimensions of heart and blood vessels as well as high heart and respiratory rates, particularly in mice, are tackled.
Collapse
Affiliation(s)
- Adrianus J Bakermans
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Desiree Abdurrachim
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Rik P M Moonen
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Abdallah G Motaal
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeanine J Prompers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Gustav J Strijkers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Katrien Vandoorne
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Klaas Nicolay
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
35
|
Juenet M, Varna M, Aid-Launais R, Chauvierre C, Letourneur D. Nanomedicine for the molecular diagnosis of cardiovascular pathologies. Biochem Biophys Res Commun 2015; 468:476-84. [PMID: 26129770 DOI: 10.1016/j.bbrc.2015.06.138] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 06/20/2015] [Indexed: 11/15/2022]
Abstract
Predicting acute clinical events caused by atherosclerotic plaque rupture remains a clinical challenge. Anatomic mapping of the vascular tree provided by standard imaging technologies is not always sufficient for a robust diagnosis. Yet biological mechanisms leading to unstable plaques have been identified and corresponding biomarkers have been described. Nanosystems charged with contrast agents and targeted towards these specific biomarkers have been developed for several types of imaging modalities. The first systems that have reached the clinic are ultrasmall superparamagnetic iron oxides for Magnetic Resonance Imaging. Their potential relies on their passive accumulation by predominant physiological mechanisms in rupture-prone plaques. Active targeting strategies are under development to improve their specificity and set up other types of nanoplatforms. Preclinical results show a huge potential of nanomedicine for cardiovascular diagnosis, as long as the safety of these nanosystems in the body is studied in depth.
Collapse
Affiliation(s)
- Maya Juenet
- Inserm, U1148, Cardiovascular Bio-Engineering, X. Bichat Hospital, 75018, Paris, France; Université Paris 13, Institut Galilée, Sorbonne Paris Cité, 75018, Paris, France
| | - Mariana Varna
- Inserm, U1148, Cardiovascular Bio-Engineering, X. Bichat Hospital, 75018, Paris, France; Université Paris 13, Institut Galilée, Sorbonne Paris Cité, 75018, Paris, France
| | - Rachida Aid-Launais
- Inserm, U1148, Cardiovascular Bio-Engineering, X. Bichat Hospital, 75018, Paris, France; Université Paris 13, Institut Galilée, Sorbonne Paris Cité, 75018, Paris, France
| | - Cédric Chauvierre
- Inserm, U1148, Cardiovascular Bio-Engineering, X. Bichat Hospital, 75018, Paris, France; Université Paris 13, Institut Galilée, Sorbonne Paris Cité, 75018, Paris, France.
| | - Didier Letourneur
- Inserm, U1148, Cardiovascular Bio-Engineering, X. Bichat Hospital, 75018, Paris, France; Université Paris 13, Institut Galilée, Sorbonne Paris Cité, 75018, Paris, France
| |
Collapse
|
36
|
De Wilde D, Trachet B, Van der Donckt C, Vandeghinste B, Descamps B, Vanhove C, De Meyer GRY, Segers P. Vulnerable Plaque Detection and Quantification with Gold Particle–Enhanced Computed Tomography in Atherosclerotic Mouse Models. Mol Imaging 2015; 14. [DOI: 10.2310/7290.2015.00009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- David De Wilde
- From the Biofluid, Tissue and Solid Mechanics for Medical Applications (bioMMeda), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-iMinds-IBiTech, Gent, Belgium; Institute for Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Division of Physiopharmacology, University of Antwerp, Wilrijk, Belgium; Medical Image and Signal Processing (MEDISIP), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-
| | - Bram Trachet
- From the Biofluid, Tissue and Solid Mechanics for Medical Applications (bioMMeda), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-iMinds-IBiTech, Gent, Belgium; Institute for Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Division of Physiopharmacology, University of Antwerp, Wilrijk, Belgium; Medical Image and Signal Processing (MEDISIP), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-
| | - Carole Van der Donckt
- From the Biofluid, Tissue and Solid Mechanics for Medical Applications (bioMMeda), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-iMinds-IBiTech, Gent, Belgium; Institute for Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Division of Physiopharmacology, University of Antwerp, Wilrijk, Belgium; Medical Image and Signal Processing (MEDISIP), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-
| | - Bert Vandeghinste
- From the Biofluid, Tissue and Solid Mechanics for Medical Applications (bioMMeda), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-iMinds-IBiTech, Gent, Belgium; Institute for Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Division of Physiopharmacology, University of Antwerp, Wilrijk, Belgium; Medical Image and Signal Processing (MEDISIP), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-
| | - Benedicte Descamps
- From the Biofluid, Tissue and Solid Mechanics for Medical Applications (bioMMeda), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-iMinds-IBiTech, Gent, Belgium; Institute for Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Division of Physiopharmacology, University of Antwerp, Wilrijk, Belgium; Medical Image and Signal Processing (MEDISIP), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-
| | - Christian Vanhove
- From the Biofluid, Tissue and Solid Mechanics for Medical Applications (bioMMeda), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-iMinds-IBiTech, Gent, Belgium; Institute for Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Division of Physiopharmacology, University of Antwerp, Wilrijk, Belgium; Medical Image and Signal Processing (MEDISIP), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-
| | - Guido R. Y. De Meyer
- From the Biofluid, Tissue and Solid Mechanics for Medical Applications (bioMMeda), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-iMinds-IBiTech, Gent, Belgium; Institute for Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Division of Physiopharmacology, University of Antwerp, Wilrijk, Belgium; Medical Image and Signal Processing (MEDISIP), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-
| | - Patrick Segers
- From the Biofluid, Tissue and Solid Mechanics for Medical Applications (bioMMeda), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-iMinds-IBiTech, Gent, Belgium; Institute for Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Division of Physiopharmacology, University of Antwerp, Wilrijk, Belgium; Medical Image and Signal Processing (MEDISIP), iMinds Medical IT, Department of Electronics and Information Systems, Ghent University-
| |
Collapse
|
37
|
Lin JB, Phillips EH, Riggins TE, Sangha GS, Chakraborty S, Lee JY, Lycke RJ, Hernandez CL, Soepriatna AH, Thorne BRH, Yrineo AA, Goergen CJ. Imaging of small animal peripheral artery disease models: recent advancements and translational potential. Int J Mol Sci 2015; 16:11131-77. [PMID: 25993289 PMCID: PMC4463694 DOI: 10.3390/ijms160511131] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 03/10/2015] [Indexed: 12/11/2022] Open
Abstract
Peripheral artery disease (PAD) is a broad disorder encompassing multiple forms of arterial disease outside of the heart. As such, PAD development is a multifactorial process with a variety of manifestations. For example, aneurysms are pathological expansions of an artery that can lead to rupture, while ischemic atherosclerosis reduces blood flow, increasing the risk of claudication, poor wound healing, limb amputation, and stroke. Current PAD treatment is often ineffective or associated with serious risks, largely because these disorders are commonly undiagnosed or misdiagnosed. Active areas of research are focused on detecting and characterizing deleterious arterial changes at early stages using non-invasive imaging strategies, such as ultrasound, as well as emerging technologies like photoacoustic imaging. Earlier disease detection and characterization could improve interventional strategies, leading to better prognosis in PAD patients. While rodents are being used to investigate PAD pathophysiology, imaging of these animal models has been underutilized. This review focuses on structural and molecular information and disease progression revealed by recent imaging efforts of aortic, cerebral, and peripheral vascular disease models in mice, rats, and rabbits. Effective translation to humans involves better understanding of underlying PAD pathophysiology to develop novel therapeutics and apply non-invasive imaging techniques in the clinic.
Collapse
Affiliation(s)
- Jenny B Lin
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Evan H Phillips
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Ti'Air E Riggins
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Gurneet S Sangha
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Sreyashi Chakraborty
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Janice Y Lee
- Psychological Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | - Roy J Lycke
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Clarissa L Hernandez
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Arvin H Soepriatna
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Bradford R H Thorne
- School of Sciences, Neuroscience, Purdue University, West Lafayette, IN 47907, USA.
| | - Alexa A Yrineo
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| |
Collapse
|
38
|
Burtea C, Laurent S, Crombez D, Delcambre S, Sermeus C, Millard I, Rorive S, Flamez D, Beckers MC, Salmon I, Vander Elst L, Eizirik DL, Muller RN. Development of a peptide-functionalized imaging nanoprobe for the targeting of (FXYD2)γa as a highly specific biomarker of pancreatic beta cells. CONTRAST MEDIA & MOLECULAR IMAGING 2015; 10:398-412. [PMID: 25930968 DOI: 10.1002/cmmi.1641] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 02/06/2015] [Accepted: 02/17/2015] [Indexed: 01/15/2023]
Abstract
Diabetes is characterized by a progressive decline of the pancreatic beta cell mass (BCM), which is responsible for insufficient insulin secretion and hyperglycaemia. There are currently no reliable methods to measure non-invasively the BCM in diabetic patients. Our work describes a phage display-derived peptide (P88) that is highly specific to (FXYD2)γa expressed by human beta cells and is proposed as a molecular vector for the development of functionalized imaging probes. P88 does not bind to the exocrine pancreas and is able to detect down to ~156 human pancreatic islets/mm(3) in vitro after conjugation to ultra-small particles of iron oxide (USPIO), as proven by the R2 measured on MR images. For in vivo evaluation, MRI studies were carried out on nude mice bearing Capan-2 tumours that also express (FXYD2)γa. A strong negative contrast was obtained subsequent to the injection of USPIO-P88, but not in negative controls. On human histological sections, USPIO-P88 seems to be specific to pancreatic beta cells, but not to duodenum, stomach or kidney tissues. USPIO-P88 thus represents a novel and promising tool for monitoring pancreatic BCM in diabetic patients. The quantitative correlation between BCM and R2 remains to be demonstrated in vivo, but the T2 mapping and the black pixel estimation after USPIO-P88 injection could provide important information for the future pancreatic BCM evaluation by MRI.
Collapse
Affiliation(s)
- Carmen Burtea
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Deborah Crombez
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Sébastien Delcambre
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Corine Sermeus
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Isabelle Millard
- Center for Diabetes Research, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
| | - Sandrine Rorive
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium.,DIAPath, Center for Microscopy and Molecular Imaging, 8 rue Adrienne Bolland, 6041, Gosselies, Belgium
| | - Daisy Flamez
- Center for Diabetes Research, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
| | - Marie-Claire Beckers
- Eurogentec S.A., Liège Science Park, Rue du Bois Saint-Jean 5, B-4102, Seraing, Belgium
| | - Isabelle Salmon
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium.,DIAPath, Center for Microscopy and Molecular Imaging, 8 rue Adrienne Bolland, 6041, Gosselies, Belgium
| | - Luce Vander Elst
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Decio L Eizirik
- Center for Diabetes Research, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
| | - Robert N Muller
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium.,Center for Microscopy and Molecular Imaging, 8 rue Adrienne Bolland, 6041, Gosselies, Belgium
| |
Collapse
|
39
|
Iron-based superparamagnetic nanoparticle contrast agents for MRI of infection and inflammation. AJR Am J Roentgenol 2015; 204:W302-13. [PMID: 25714316 DOI: 10.2214/ajr.14.12733] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE. In this article, we summarize the progress to date on the use of superparamagnetic iron oxide nanoparticles (SPIONs) as contrast agents for MRI of inflammatory processes. CONCLUSION. Phagocytosis by macrophages of injected SPIONs results in a prolonged shortening of both T2 and T2* leading to hypointensity of macrophage-infiltrated tissues in contrast-enhanced MR images. SPIONs as contrast agents are therefore useful for the in vivo MRI detection of macrophage infiltration, and there is substantial research and clinical interest in the use of SPION-based contrast agents for MRI of infection and inflammation. This technique has been used to identify active infection in patients with septic arthritis and osteomyelitis; importantly, the MRI signal intensity of the tissue has been found to return to its unenhanced value on successful treatment of the infection. In SPION contrast-enhanced MRI of vascular inflammation, animal studies have shown decreased macrophage uptake in atherosclerotic plaques after treatment with statin drugs. Human studies have shown that both coronary and carotid plaques that take up SPIONs are more prone to rupture and that abdominal aneurysms with increased SPION uptake are more likely to grow. Studies of patients with multiple sclerosis suggest that MRI using SPIONs may have increased sensitivity over gadolinium for plaque detection. Finally, SPIONs have enabled the tracking and imaging of transplanted stem cells in a recipient host.
Collapse
|
40
|
Affiliation(s)
- Ziad Mallat
- From the Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom; and Institut National de la Santé et de la Recherche Médicale, U970, Paris, France.
| |
Collapse
|
41
|
Sharifi S, Seyednejad H, Laurent S, Atyabi F, Saei AA, Mahmoudi M. Superparamagnetic iron oxide nanoparticles for in vivo molecular and cellular imaging. CONTRAST MEDIA & MOLECULAR IMAGING 2015; 10:329-55. [PMID: 25882768 DOI: 10.1002/cmmi.1638] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 01/30/2015] [Accepted: 02/06/2015] [Indexed: 12/16/2022]
Abstract
In the last decade, the biomedical applications of nanoparticles (NPs) (e.g. cell tracking, biosensing, magnetic resonance imaging (MRI), targeted drug delivery, and tissue engineering) have been increasingly developed. Among the various NP types, superparamagnetic iron oxide NPs (SPIONs) have attracted considerable attention for early detection of diseases due to their specific physicochemical properties and their molecular imaging capabilities. A comprehensive review is presented on the recent advances in the development of in vitro and in vivo SPION applications for molecular imaging, along with opportunities and challenges.
Collapse
Affiliation(s)
- Shahriar Sharifi
- Department of Biomaterials Science and Technology, University of Twente, The Netherlands
| | - Hajar Seyednejad
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA
| | - Sophie Laurent
- Department of General, Organic, and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, B-7000, Mons, Belgium.,CMMI - Center for Microscopy and Molecular Imaging, Rue Adrienne Bolland 8, B-6041, Gosselies, Belgium
| | - Fatemeh Atyabi
- Nanotechnology Research Center and Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ata Saei
- Nanotechnology Research Center and Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Morteza Mahmoudi
- Nanotechnology Research Center and Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
42
|
Zeng W, Wang X, Xu P, Liu G, Eden HS, Chen X. Molecular imaging of apoptosis: from micro to macro. Theranostics 2015; 5:559-82. [PMID: 25825597 PMCID: PMC4377726 DOI: 10.7150/thno.11548] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 02/18/2015] [Indexed: 12/21/2022] Open
Abstract
Apoptosis, or programmed cell death, is involved in numerous human conditions including neurodegenerative diseases, ischemic damage, autoimmune disorders and many types of cancer, and is often confused with other types of cell death. Therefore strategies that enable visualized detection of apoptosis would be of enormous benefit in the clinic for diagnosis, patient management, and development of new therapies. In recent years, improved understanding of the apoptotic machinery and progress in imaging modalities have provided opportunities for researchers to formulate microscopic and macroscopic imaging strategies based on well-defined molecular markers and/or physiological features. Correspondingly, a large collection of apoptosis imaging probes and approaches have been documented in preclinical and clinical studies. In this review, we mainly discuss microscopic imaging assays and macroscopic imaging probes, ranging in complexity from simple attachments of reporter moieties to proteins that interact with apoptotic biomarkers, to rationally designed probes that target biochemical changes. Their clinical translation will also be our focus.
Collapse
|
43
|
Abstract
Nanoparticles are frequently suggested as diagnostic agents. However, except for iron oxide nanoparticles, diagnostic nanoparticles have been barely incorporated into clinical use so far. This is predominantly due to difficulties in achieving acceptable pharmacokinetic properties and reproducible particle uniformity as well as to concerns about toxicity, biodegradation, and elimination. Reasonable indications for the clinical utilization of nanoparticles should consider their biologic behavior. For example, many nanoparticles are taken up by macrophages and accumulate in macrophage-rich tissues. Thus, they can be used to provide contrast in liver, spleen, lymph nodes, and inflammatory lesions (eg, atherosclerotic plaques). Furthermore, cells can be efficiently labeled with nanoparticles, enabling the localization of implanted (stem) cells and tissue-engineered grafts as well as in vivo migration studies of cells. The potential of using nanoparticles for molecular imaging is compromised because their pharmacokinetic properties are difficult to control. Ideal targets for nanoparticles are localized on the endothelial luminal surface, whereas targeted nanoparticle delivery to extravascular structures is often limited and difficult to separate from an underlying enhanced permeability and retention (EPR) effect. The majority of clinically used nanoparticle-based drug delivery systems are based on the EPR effect, and, for their more personalized use, imaging markers can be incorporated to monitor biodistribution, target site accumulation, drug release, and treatment efficacy. In conclusion, although nanoparticles are not always the right choice for molecular imaging (because smaller or larger molecules might provide more specific information), there are other diagnostic and theranostic applications for which nanoparticles hold substantial clinical potential.
Collapse
Affiliation(s)
- Fabian Kiessling
- From the Department of Experimental Molecular Imaging, RWTH-Aachen University, Aachen, Germany (F.K., M.E.M., T.L.); and Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY (J.G.)
| | | | | | | |
Collapse
|
44
|
Gauberti M, Montagne A, Quenault A, Vivien D. Molecular magnetic resonance imaging of brain-immune interactions. Front Cell Neurosci 2014; 8:389. [PMID: 25505871 PMCID: PMC4245913 DOI: 10.3389/fncel.2014.00389] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 10/31/2014] [Indexed: 01/09/2023] Open
Abstract
Although the blood-brain barrier (BBB) was thought to protect the brain from the effects of the immune system, immune cells can nevertheless migrate from the blood to the brain, either as a cause or as a consequence of central nervous system (CNS) diseases, thus contributing to their evolution and outcome. Accordingly, as the interface between the CNS and the peripheral immune system, the BBB is critical during neuroinflammatory processes. In particular, endothelial cells are involved in the brain response to systemic or local inflammatory stimuli by regulating the cellular movement between the circulation and the brain parenchyma. While neuropathological conditions differ in etiology and in the way in which the inflammatory response is mounted and resolved, cellular mechanisms of neuroinflammation are probably similar. Accordingly, neuroinflammation is a hallmark and a decisive player of many CNS diseases. Thus, molecular magnetic resonance imaging (MRI) of inflammatory processes is a central theme of research in several neurological disorders focusing on a set of molecules expressed by endothelial cells, such as adhesion molecules (VCAM-1, ICAM-1, P-selectin, E-selectin, …), which emerge as therapeutic targets and biomarkers for neurological diseases. In this review, we will present the most recent advances in the field of preclinical molecular MRI. Moreover, we will discuss the possible translation of molecular MRI to the clinical setting with a particular emphasis on myeloperoxidase imaging, autologous cell tracking, and targeted iron oxide particles (USPIO, MPIO).
Collapse
Affiliation(s)
- Maxime Gauberti
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université de Caen Basse-Normandie - GIP Cyceron Caen, France
| | - Axel Montagne
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université de Caen Basse-Normandie - GIP Cyceron Caen, France
| | - Aurélien Quenault
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université de Caen Basse-Normandie - GIP Cyceron Caen, France
| | - Denis Vivien
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université de Caen Basse-Normandie - GIP Cyceron Caen, France
| |
Collapse
|
45
|
Sadat U, Jaffer FA, van Zandvoort MAMJ, Nicholls SJ, Ribatti D, Gillard JH. Inflammation and neovascularization intertwined in atherosclerosis: imaging of structural and molecular imaging targets. Circulation 2014; 130:786-94. [PMID: 25156914 DOI: 10.1161/circulationaha.114.010369] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Umar Sadat
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.).
| | - Farouc A Jaffer
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Marc A M J van Zandvoort
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Stephen J Nicholls
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Domenico Ribatti
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Jonathan H Gillard
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| |
Collapse
|
46
|
Ansciaux E, Burtea C, Laurent S, Crombez D, Nonclercq D, Vander Elst L, Muller RN. In vitro and in vivo characterization of several functionalized ultrasmall particles of iron oxide, vectorized against amyloid plaques and potentially able to cross the blood-brain barrier: toward earlier diagnosis of Alzheimer's disease by molecular imag. CONTRAST MEDIA & MOLECULAR IMAGING 2014; 10:211-24. [DOI: 10.1002/cmmi.1626] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 07/13/2014] [Accepted: 08/25/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Emilie Ansciaux
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory; University of Mons; Avenue Maistriau 19, Mendeleev Building B-7000 Mons Belgium
| | - Carmen Burtea
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory; University of Mons; Avenue Maistriau 19, Mendeleev Building B-7000 Mons Belgium
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory; University of Mons; Avenue Maistriau 19, Mendeleev Building B-7000 Mons Belgium
| | - Deborah Crombez
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory; University of Mons; Avenue Maistriau 19, Mendeleev Building B-7000 Mons Belgium
| | - Denis Nonclercq
- Laboratory of Histology; University of Mons; Pentagon - 1B, 6 Avenue du Champ de Mars B-7000 Mons Belgium
| | - Luce Vander Elst
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory; University of Mons; Avenue Maistriau 19, Mendeleev Building B-7000 Mons Belgium
| | - Robert N. Muller
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory; University of Mons; Avenue Maistriau 19, Mendeleev Building B-7000 Mons Belgium
- Center for Microscopy and Molecular Imaging; 8, rue Adrienne Bolland 6041 Gosselies Belgium
| |
Collapse
|
47
|
Plaunt AJ, Harmatys KM, Wolter WR, Suckow MA, Smith BD. Library synthesis, screening, and discovery of modified Zinc(II)-Bis(dipicolylamine) probe for enhanced molecular imaging of cell death. Bioconjug Chem 2014; 25:724-37. [PMID: 24575875 PMCID: PMC3993938 DOI: 10.1021/bc500003x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
![]()
Zinc(II)-bis(dipicolylamine)
(Zn-BDPA) coordination complexes selectively
target the surfaces of dead and dying mammalian cells, and they have
promise as molecular probes for imaging cell death. A necessary step
toward eventual clinical imaging applications is the development of
next-generation Zn-BDPA complexes with enhanced affinity for the cell
death membrane biomarker, phosphatidylserine (PS). This study employed
an iterative cycle of library synthesis and screening, using a novel
rapid equilibrium dialysis assay, to discover a modified Zn-BDPA structure
with high and selective affinity for vesicles containing PS. The lead
structure was converted into a deep-red fluorescent probe and its
targeting and imaging performance was compared with an unmodified
control Zn-BDPA probe. The evaluation process included a series of
FRET-based vesicle titration studies, cell microscopy experiments,
and rat tumor biodistribution measurements. In all cases, the modified
probe exhibited comparatively higher affinity and selectivity for
the target membranes of dead and dying cells. The results show that
this next-generation deep-red fluorescent Zn-BDPA probe is well suited
for preclinical molecular imaging of cell death in cell cultures and
animal models. Furthermore, it should be possible to substitute the
deep-red fluorophore with alternative reporter groups that enable
clinically useful, deep-tissue imaging modalities, such as MRI and
nuclear imaging.
Collapse
Affiliation(s)
- Adam J Plaunt
- Department of Chemistry and Biochemistry, 236 Nieuwland Science Hall and ‡Department of Biological Science, Galvin Life Sciences, University of Notre Dame , Notre Dame, 46556 Indiana, United States
| | | | | | | | | |
Collapse
|
48
|
Bruckman M, Jiang K, Simpson EJ, Randolph LN, Luyt LG, Yu X, Steinmetz NF. Dual-modal magnetic resonance and fluorescence imaging of atherosclerotic plaques in vivo using VCAM-1 targeted tobacco mosaic virus. NANO LETTERS 2014; 14:1551-8. [PMID: 24499194 PMCID: PMC4169141 DOI: 10.1021/nl404816m] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 02/02/2014] [Indexed: 05/04/2023]
Abstract
The underlying cause of major cardiovascular events, such as myocardial infarctions and strokes, is atherosclerosis. For accurate diagnosis of this inflammatory disease, molecular imaging is required. Toward this goal, we sought to develop a nanoparticle-based, high aspect ratio, molecularly targeted magnetic resonance (MR) imaging contrast agent. Specifically, we engineered the plant viral nanoparticle platform tobacco mosaic virus (TMV) to target vascular cell adhesion molecule (VCAM)-1, which is highly expressed on activated endothelial cells at atherosclerotic plaques. To achieve dual optical and MR imaging in an atherosclerotic ApoE(-/-) mouse model, TMV was modified to carry near-infrared dyes and chelated Gd ions. Our results indicate molecular targeting of atherosclerotic plaques. On the basis of the multivalency and multifunctionality, the targeted TMV-based MR probe increased the detection limit significantly; the injected dose of Gd ions could be further reduced 400x compared to the suggested clinical use, demonstrating the utility of targeted nanoparticle cargo delivery.
Collapse
Affiliation(s)
- Michael
A. Bruckman
- Department of Biomedical Engineering, Department of Radiology, Department of Materials
Science and Engineering, and Department of Macromolecular Engineering, Case Western Reserve University Schools of Medicine
and Engineering, 10900
Euclid Avenue, Cleveland, Ohio 44106, United
States
| | - Kai Jiang
- Department of Biomedical Engineering, Department of Radiology, Department of Materials
Science and Engineering, and Department of Macromolecular Engineering, Case Western Reserve University Schools of Medicine
and Engineering, 10900
Euclid Avenue, Cleveland, Ohio 44106, United
States
| | - Emily J. Simpson
- Departments
of Chemistry, Oncology, Medical Imaging, The University of Western Ontario, London, Ontario N6A 4L6, Canada
| | - Lauren N. Randolph
- Department of Biomedical Engineering, Department of Radiology, Department of Materials
Science and Engineering, and Department of Macromolecular Engineering, Case Western Reserve University Schools of Medicine
and Engineering, 10900
Euclid Avenue, Cleveland, Ohio 44106, United
States
| | - Leonard G. Luyt
- Departments
of Chemistry, Oncology, Medical Imaging, The University of Western Ontario, London, Ontario N6A 4L6, Canada
| | - Xin Yu
- Department of Biomedical Engineering, Department of Radiology, Department of Materials
Science and Engineering, and Department of Macromolecular Engineering, Case Western Reserve University Schools of Medicine
and Engineering, 10900
Euclid Avenue, Cleveland, Ohio 44106, United
States
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering, Department of Radiology, Department of Materials
Science and Engineering, and Department of Macromolecular Engineering, Case Western Reserve University Schools of Medicine
and Engineering, 10900
Euclid Avenue, Cleveland, Ohio 44106, United
States
| |
Collapse
|
49
|
Lin K, Lloyd-Jones DM, Li D, Carr JC. Quantitative imaging biomarkers for the evaluation of cardiovascular complications in type 2 diabetes mellitus. J Diabetes Complications 2014; 28:234-42. [PMID: 24309215 DOI: 10.1016/j.jdiacomp.2013.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 09/19/2013] [Accepted: 09/19/2013] [Indexed: 01/24/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a prevalent condition in aged populations. Cardiovascular diseases are leading causes of death and disability in patients with T2DM. Traditional strategies for controlling the cardiovascular complications of diabetes primarily target a cluster of well-defined risk factors, such as hyperglycemia, lipid disorders and hypertension. However, there is controversy over some recent clinical trials aimed at evaluating efficacy of intensive treatments for T2DM. As a powerful tool for quantitative cardiovascular risk estimation, multi-disciplinary cardiovascular imaging have been applied to detect and quantify morphological and functional abnormalities in the cardiovascular system. Quantitative imaging biomarkers acquired with advanced imaging procedures are expected to provide new insights to stratify absolute cardiovascular risks and reduce the overall costs of health care for people with T2DM by facilitating the selection of optimal therapies. This review discusses principles of state-of-the-art cardiovascular imaging techniques and compares applications of those techniques in various clinical circumstances. Individuals measurements of cardiovascular disease burdens from multiple aspects, which are closely related to existing biomarkers and clinical outcomes, are recommended as promising candidates for quantitative imaging biomarkers to assess the responses of the cardiovascular system during diabetic regimens.
Collapse
Affiliation(s)
- Kai Lin
- Department of Radiology, Northwestern University Feinberg School of Medicine, 737N Michigan Avenue, Suite 1600, Chicago, IL 60611, USA
| | - Donald M Lloyd-Jones
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680N Lake shore drive, Suite 1400, Chicago, IL 60611, USA
| | - Debiao Li
- Department of Radiology, Northwestern University Feinberg School of Medicine, 737N Michigan Avenue, Suite 1600, Chicago, IL 60611, USA
| | - James C Carr
- Department of Radiology, Northwestern University Feinberg School of Medicine, 737N Michigan Avenue, Suite 1600, Chicago, IL 60611, USA.
| |
Collapse
|
50
|
Abstract
Understanding the pathophysiology of atherogenesis and the progression of atherosclerosis have been major goals of cardiovascular research during the previous decades. However, the complex molecular and cellular mechanisms underlying plaque destabilization remain largely obscure. Here, we review how lesional cells undergo cell death and how failed clearance exacerbates necrotic core formation. Advanced atherosclerotic lesions are further weakened by the pronounced local activity of matrix-degrading proteases as well as immature neovessels sprouting into the lesion. To stimulate translation of the current knowledge of molecular mechanisms of plaque destabilization into clinical studies, we further summarize available animal models of plaque destabilization. Based on the molecular mechanisms leading to plaque instability, we outline the current status of clinical and preclinical trials to induce plaque stability with a focus on induction of dead cell clearance, inhibition of protease activity, and dampening of inflammatory cell recruitment.
Collapse
|