1
|
Sekita A, Unterweger H, Berg S, Ohlmeyer S, Bäuerle T, Zheng KH, Coolen BF, Nederveen AJ, Cabella C, Rossi S, Stroes ESG, Alexiou C, Lyer S, Cicha I. Accumulation of Iron Oxide-Based Contrast Agents in Rabbit Atherosclerotic Plaques in Relation to Plaque Age and Vulnerability Features. Int J Nanomedicine 2024; 19:1645-1666. [PMID: 38406599 PMCID: PMC10893894 DOI: 10.2147/ijn.s430693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/14/2023] [Indexed: 02/27/2024] Open
Abstract
Purpose In this study, a detailed characterization of a rabbit model of atherosclerosis was performed to assess the optimal time frame for evaluating plaque vulnerability using superparamagnetic iron oxide nanoparticle (SPION)-enhanced magnetic resonance imaging (MRI). Methods The progression of atherosclerosis induced by ballooning and a high-cholesterol diet was monitored using angiography, and the resulting plaques were characterized using immunohistochemistry and histology. Morphometric analyses were performed to evaluate plaque size and vulnerability features. The accumulation of SPIONs (novel dextran-coated SPIONDex and ferumoxytol) in atherosclerotic plaques was investigated by histology and MRI and correlated with plaque age and vulnerability. Toxicity of SPIONDex was evaluated in rats. Results Weak positive correlations were detected between plaque age and intima thickness, and total macrophage load. A strong negative correlation was observed between the minimum fibrous cap thickness and plaque age as well as the mean macrophage load. The accumulation of SPION in the atherosclerotic plaques was detected by MRI 24 h after administration and was subsequently confirmed by Prussian blue staining of histological specimens. Positive correlations between Prussian blue signal in atherosclerotic plaques, plaque age, and macrophage load were detected. Very little iron was observed in the histological sections of the heart and kidney, whereas strong staining of SPIONDex and ferumoxytol was detected in the spleen and liver. In contrast to ferumoxytol, SPIONDex administration in rabbits was well tolerated without inducing hypersensitivity. The maximum tolerated dose in rat model was higher than 100 mg Fe/kg. Conclusion Older atherosclerotic plaques with vulnerable features in rabbits are a useful tool for investigating iron oxide-based contrast agents for MRI. Based on the experimental data, SPIONDex particles constitute a promising candidate for further clinical translation as a safe formulation that offers the possibility of repeated administration free from the risks associated with other types of magnetic contrast agents.
Collapse
Affiliation(s)
- Alexander Sekita
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Harald Unterweger
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sonja Berg
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sabine Ohlmeyer
- Institute of Radiology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tobias Bäuerle
- Preclinical Imaging Platform Erlangen (PIPE), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Kang H Zheng
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Bram F Coolen
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Aart J Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Claudia Cabella
- Bracco Imaging SpA, Centro Ricerche Bracco, Colleretto Giacosa, Turin, Italy
| | - Silvia Rossi
- Bracco Imaging SpA, Centro Ricerche Bracco, Colleretto Giacosa, Turin, Italy
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Christoph Alexiou
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Lyer
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Iwona Cicha
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
2
|
Zhang R, Lu K, Xiao L, Hu X, Cai W, Liu L, Liu Y, Li W, Zhou H, Qian Z, Wang S, Chen C, Zeng J, Gao M. Exploring atherosclerosis imaging with contrast-enhanced MRI using PEGylated ultrasmall iron oxide nanoparticles. Front Bioeng Biotechnol 2023; 11:1279446. [PMID: 37811376 PMCID: PMC10557075 DOI: 10.3389/fbioe.2023.1279446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Plaque rupture is a critical concern due to its potential for severe outcomes such as cerebral infarction and myocardial infarction, underscoring the urgency of noninvasive early diagnosis. Magnetic resonance imaging (MRI) has gained prominence in plaque imaging, leveraging its noninvasiveness, high spatial resolution, and lack of ionizing radiation. Ultrasmall iron oxides, when modified with polyethylene glycol, exhibit prolonged blood circulation and passive targeting toward plaque sites, rendering them conducive for MRI. In this study, we synthesized ultrasmall iron oxide nanoparticles of approximately 3 nm via high-temperature thermal decomposition. Subsequent surface modification facilitated the creation of a dual-modality magnetic resonance/fluorescence probe. Upon intravenous administration of the probes, MRI assessment of atherosclerotic plaques and diagnostic evaluation were conducted. The application of Flash-3D sequence imaging revealed vascular constriction at lesion sites, accompanied by a gradual signal amplification postprobe injection. T1-weighted imaging of the carotid artery unveiled a progressive signal ratio increase between plaques and controls within 72 h post-administration. Fluorescence imaging of isolated carotid arteries exhibited incremental lesion-to-control signal ratios. Additionally, T1 imaging of the aorta demonstrated an evolving signal enhancement over 48 h. Therefore, the ultrasmall iron oxide nanoparticles hold immense promise for early and noninvasive diagnosis of plaques, providing an avenue for dynamic evaluation over an extended time frame.
Collapse
Affiliation(s)
- Ruru Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Kuan Lu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Xiao
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xuelan Hu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Wu Cai
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Linjiang Liu
- Medical Imaging Department, Shenzhen Second People’s Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yan Liu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Weihua Li
- Medical Imaging Department, Shenzhen Second People’s Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Hui Zhou
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Zhiyuan Qian
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Sixia Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Can Chen
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Mingyuan Gao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
3
|
Cazelles A, Collard MK, Lalatonne Y, Doblas S, Zappa M, Labiad C, Cazals-Hatem D, Maggiori L, Treton X, Panis Y, Jarry U, Desvallées T, Eliat PA, Pineau R, Motte L, Letourneur D, Simon-Yarza T, Ogier-Denis E. A Preclinical Validation of Iron Oxide Nanoparticles for Treatment of Perianal Fistulizing Crohn's Disease. Int J Mol Sci 2022; 23:8324. [PMID: 35955465 PMCID: PMC9368411 DOI: 10.3390/ijms23158324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 07/25/2022] [Indexed: 11/26/2022] Open
Abstract
Fistulizing anoperineal lesions are severe complications of Crohn's disease (CD) that affect quality of life with a long-term risk of anal sphincter destruction, incontinence, permanent stoma, and anal cancer. Despite several surgical procedures, they relapse in about two-thirds of patients, mandating innovative treatments. Ultrasmall particles of iron oxide (USPIO) have been described to achieve in vivo rapid healing of deep wounds in the skin and liver of rats thanks to their nanobridging capability that could be adapted to fistula treatment. Our main purpose was to highlight preclinical data with USPIO for the treatment of perianal fistulizing CD. Twenty male Sprague Dawley rats with severe 2,4,6-trinitrobenzenesulfonic acid solution (TNBS)-induced proctitis were operated to generate two perianal fistulas per rat. At day 35, two inflammatory fistulas were obtained per rat and perineal magnetic resonance imaging (MRI) was performed. After a baseline MRI, a fistula tract was randomly drawn and topically treated either with saline or with USPIO for 1 min (n = 17 for each). The rats underwent a perineal MRI on postoperative days (POD) 1, 4, and 7 and were sacrificed for pathological examination. The primary outcome was the filling or closure of the fistula tract, including the external or internal openings. USPIO treatment allowed the closure and/or filling of all the treated fistulas from its application until POD 7 in comparison with the control fistulas (23%). The treatment with USPIO was safe, permanently closed the fistula along its entire length, including internal and external orifices, and paved new avenues for the treatment of perianal fistulizing Crohn's disease.
Collapse
Affiliation(s)
- Antoine Cazelles
- Département of Chirurgie Colorectale, Assistance Publique Hôpitaux de Paris, Hôpital Beaujon, CEDEX, 92110 Clichy, France; (A.C.); (M.K.C.); (C.L.); (L.M.); (Y.P.)
- Centre de Recherche sur l’Inflammation, INSERM, U1149, CNRS, ERL8252, Université Paris Cité, Team Gut Inflammation, BP 416, 75018 Paris, France; (D.C.-H.); (X.T.)
| | - Maxime K. Collard
- Département of Chirurgie Colorectale, Assistance Publique Hôpitaux de Paris, Hôpital Beaujon, CEDEX, 92110 Clichy, France; (A.C.); (M.K.C.); (C.L.); (L.M.); (Y.P.)
- Centre de Recherche sur l’Inflammation, INSERM, U1149, CNRS, ERL8252, Université Paris Cité, Team Gut Inflammation, BP 416, 75018 Paris, France; (D.C.-H.); (X.T.)
| | - Yoann Lalatonne
- Laboratory for Vascular Translational Science, Université Paris Cité, Université Sorbonne Paris Nord, LVTS, INSERM, UMR 1148, 75018 Paris, France; (Y.L.); (L.M.); (D.L.); (T.S.-Y.)
- Départements of Biochimie and de Médecine Nucléaire, Assistance Publique-Hôpitaux de Paris, Hôpital Avicenne, 93009 Bobigny, France
| | - Sabrina Doblas
- Centre de Recherche sur l’Inflammation, INSERM, U1149, CNRS, ERL8252, Laboratory of Imaging Biomarkers, Université Paris Cité, BP 416, 75018 Paris, France; (S.D.); (M.Z.)
| | - Magaly Zappa
- Centre de Recherche sur l’Inflammation, INSERM, U1149, CNRS, ERL8252, Laboratory of Imaging Biomarkers, Université Paris Cité, BP 416, 75018 Paris, France; (S.D.); (M.Z.)
- Département of Radiologie, Assistance Publique Hôpitaux de Paris, Hôpital Beaujon, CEDEX, 92110 Clichy, France
| | - Camélia Labiad
- Département of Chirurgie Colorectale, Assistance Publique Hôpitaux de Paris, Hôpital Beaujon, CEDEX, 92110 Clichy, France; (A.C.); (M.K.C.); (C.L.); (L.M.); (Y.P.)
- Centre de Recherche sur l’Inflammation, INSERM, U1149, CNRS, ERL8252, Université Paris Cité, Team Gut Inflammation, BP 416, 75018 Paris, France; (D.C.-H.); (X.T.)
| | - Dominique Cazals-Hatem
- Centre de Recherche sur l’Inflammation, INSERM, U1149, CNRS, ERL8252, Université Paris Cité, Team Gut Inflammation, BP 416, 75018 Paris, France; (D.C.-H.); (X.T.)
- Département of Pathologie, Assistance Publique Hôpitaux de Paris, Hôpital Beaujon, CEDEX, 92110 Clichy, France
| | - Léon Maggiori
- Département of Chirurgie Colorectale, Assistance Publique Hôpitaux de Paris, Hôpital Beaujon, CEDEX, 92110 Clichy, France; (A.C.); (M.K.C.); (C.L.); (L.M.); (Y.P.)
- Centre de Recherche sur l’Inflammation, INSERM, U1149, CNRS, ERL8252, Université Paris Cité, Team Gut Inflammation, BP 416, 75018 Paris, France; (D.C.-H.); (X.T.)
| | - Xavier Treton
- Centre de Recherche sur l’Inflammation, INSERM, U1149, CNRS, ERL8252, Université Paris Cité, Team Gut Inflammation, BP 416, 75018 Paris, France; (D.C.-H.); (X.T.)
- Département Gastroentérologie, Assistance Publique Hôpitaux de Paris, Hôpital Beaujon, CEDEX, 92110 Clichy, France
| | - Yves Panis
- Département of Chirurgie Colorectale, Assistance Publique Hôpitaux de Paris, Hôpital Beaujon, CEDEX, 92110 Clichy, France; (A.C.); (M.K.C.); (C.L.); (L.M.); (Y.P.)
- Centre de Recherche sur l’Inflammation, INSERM, U1149, CNRS, ERL8252, Université Paris Cité, Team Gut Inflammation, BP 416, 75018 Paris, France; (D.C.-H.); (X.T.)
| | - Ulrich Jarry
- Université Rennes, CNRS, INSERM, BIOSIT UAR 3480, US_S 018, Oncotrial, 35000 Rennes, France; (U.J.); (T.D.)
- Biotrial Pharmacology, Unité De Pharmacologie Préclinique, 35000 Rennes, France
| | - Thomas Desvallées
- Université Rennes, CNRS, INSERM, BIOSIT UAR 3480, US_S 018, Oncotrial, 35000 Rennes, France; (U.J.); (T.D.)
| | - Pierre-Antoine Eliat
- Université Rennes, CNRS, INSERM, BIOSIT UAR 3480, US_S 018, PRISM, 35000 Rennes, France;
- INRAE, INSERM, Institute NUMECAN, UMR_A 1341, Université Rennes, UMR_S 1241, 35000 Rennes, France
| | - Raphaël Pineau
- INSERM, CLCC Eugène Marquis, Oncogenesis, Stress Signaling, Université Rennes, UMR_S 1242, 35000 Rennes, France;
| | - Laurence Motte
- Laboratory for Vascular Translational Science, Université Paris Cité, Université Sorbonne Paris Nord, LVTS, INSERM, UMR 1148, 75018 Paris, France; (Y.L.); (L.M.); (D.L.); (T.S.-Y.)
| | - Didier Letourneur
- Laboratory for Vascular Translational Science, Université Paris Cité, Université Sorbonne Paris Nord, LVTS, INSERM, UMR 1148, 75018 Paris, France; (Y.L.); (L.M.); (D.L.); (T.S.-Y.)
| | - Teresa Simon-Yarza
- Laboratory for Vascular Translational Science, Université Paris Cité, Université Sorbonne Paris Nord, LVTS, INSERM, UMR 1148, 75018 Paris, France; (Y.L.); (L.M.); (D.L.); (T.S.-Y.)
| | - Eric Ogier-Denis
- Centre de Recherche sur l’Inflammation, INSERM, U1149, CNRS, ERL8252, Université Paris Cité, Team Gut Inflammation, BP 416, 75018 Paris, France; (D.C.-H.); (X.T.)
- INSERM, CLCC Eugène Marquis, Oncogenesis, Stress Signaling, Université Rennes, UMR_S 1242, 35000 Rennes, France;
- INSERM U1242, Centre Eugène Marquis, Rue de la Bataille de Flandres-Dunkerque, 35042 Rennes, France
| |
Collapse
|
4
|
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
|
5
|
Qiao R, Huang X, Qin Y, Li Y, Davis TP, Hagemeyer CE, Gao M. Recent advances in molecular imaging of atherosclerotic plaques and thrombosis. NANOSCALE 2020; 12:8040-8064. [PMID: 32239038 DOI: 10.1039/d0nr00599a] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
As the complications of atherosclerosis such as myocardial infarction and stroke are still one of the leading causes of mortality worldwide, the development of new diagnostic tools for the early detection of plaque instability and thrombosis is urgently needed. Advanced molecular imaging probes based on functional nanomaterials in combination with cutting edge imaging techniques are now paving the way for novel and unique approaches to monitor the inflammatory progress in atherosclerosis. This review focuses on the development of various molecular probes for the diagnosis of plaques and thrombosis in atherosclerosis, along with perspectives of their diagnostic applications in cardiovascular diseases. Specifically, we summarize the biological targets that can be used for atherosclerosis and thrombosis imaging. Then we describe the emerging molecular imaging techniques based on the utilization of engineered nanoprobes together with their challenges in clinical translation.
Collapse
Affiliation(s)
- Ruirui Qiao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | | | | | | | | | | | | |
Collapse
|
6
|
Hu ZP, Fang XL, Sheng B, Guo Y, Yu YQ. Melatonin inhibits macrophage infiltration and promotes plaque stabilization by upregulating anti-inflammatory HGF/c-Met system in the atherosclerotic rabbit: USPIO-enhanced MRI assessment. Vascul Pharmacol 2020; 127:106659. [DOI: 10.1016/j.vph.2020.106659] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/06/2020] [Accepted: 02/14/2020] [Indexed: 01/08/2023]
|
7
|
Shannon AH, Chordia MD, Spinosa MD, Su G, Ladd Z, Pan D, Upchurch GR, Sharma AK. Single-Photon Emission Computed Tomography Imaging Using Formyl Peptide Receptor 1 Ligand Can Diagnose Aortic Aneurysms in a Mouse Model. J Surg Res 2020; 251:239-247. [PMID: 32172010 DOI: 10.1016/j.jss.2020.01.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/10/2020] [Accepted: 01/31/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Our previous studies showed that neutrophil infiltration and activation plays an important role in the pathogenesis of abdominal aortic aneurysms (AAA). However, there is a lack of noninvasive, inflammatory cell-specific molecular imaging methods to provide early diagnosis of AAA formation. Formyl peptide receptor 1 (FPR1) is rapidly upregulated on neutrophils during inflammation. Therefore, it is hypothesized that the use of cinnamoyl-F-(D)L-F-(D)L-F-K (cFLFLF), a PEGylated peptide ligand that binds FPR1 on activated neutrophils, would permit accurate and noninvasive diagnosis of AAA via single-photon emission computed tomography (SPECT) imaging. MATERIALS AND METHODS Male C57BL/6 (wild-type) mice were treated with topical elastase (0.4 U/mL type 1 porcine pancreatic elastase) or heat-inactivated elastase (control), and aortic diameter was measured by video micrometry. Comparative histology was performed on Day 14 to assess neutrophil infiltration in aortic tissue. We performed near-infrared fluorescence imaging using c-FLFLF-Cy7 probe on Days 7 and 14 postelastase treatment and measured fluorescence intensity ex vivo in excised aortic tissue. A separate group of animals were injected with 99mTc-c-FLFLF 2 h before SPECT imaging on Day 14 using a SPECT/computed tomography/positron emission tomography trimodal scanner. Coexpression of neutrophils with c-FLFLF was also performed on aortic tissue by immunostaining on Day 14. RESULTS Aortic diameter was significantly increased in the elastase group compared with controls on Days 7 and 14. Simultaneously, a marked increase in neutrophil infiltration and elastin degradation as well as decrease in smooth muscle integrity were observed in aortic tissue after elastase treatment compared with controls. Moreover, a significant increase in fluorescence intensity of c-FLFLF-Cy7 imaging probe was also observed in elastase-treated mice on Day 7 (approximately twofold increase) and Day 14 (approximately 2.5-fold increase) compared with respective controls. SPECT imaging demonstrated a multifold increase in signal intensity for 99mTc-cFLFLF radiolabel probe in mice with AAA compared with controls on Day 14. Immunostaining of aortic tissue with c-FLFLF-Cy5 demonstrated a marked increase in coexpression with neutrophils in AAA compared with controls. CONCLUSIONS cFLFLF, a novel FPR1 ligand, enables quantifiable, noninvasive diagnosis and progression of AAAs. Clinical application of this inflammatory, cell-specific molecular probe using SPECT imaging may permit early diagnosis of AAA formation, enabling targeted therapeutic interventions and preventing impending aortic rupture.
Collapse
Affiliation(s)
| | - Mahendra D Chordia
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia
| | - Michael D Spinosa
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Gang Su
- Department of Surgery, University of Florida, Gainesville, Florida
| | - Zachary Ladd
- Department of Surgery, University of Florida, Gainesville, Florida
| | - Dongfeng Pan
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia
| | | | - Ashish K Sharma
- Department of Surgery, University of Florida, Gainesville, Florida.
| |
Collapse
|
8
|
Dual-probe molecular MRI for the in vivo characterization of atherosclerosis in a mouse model: Simultaneous assessment of plaque inflammation and extracellular-matrix remodeling. Sci Rep 2019; 9:13827. [PMID: 31554825 PMCID: PMC6761132 DOI: 10.1038/s41598-019-50100-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/03/2019] [Indexed: 01/07/2023] Open
Abstract
Molecular MRI is a promising in-vivo modality to detect and quantify morphological and molecular vessel-wall changes in atherosclerosis. The combination of different molecular biomarkers may improve the risk stratification of patients. This study aimed to investigate the feasibility of simultaneous visualization and quantification of plaque-burden and inflammatory activity by dual-probe molecular MRI in a mouse-model of progressive atherosclerosis and in response-to-therapy. Homozygous apolipoprotein E knockout mice (ApoE−/−) were fed a high-fat-diet (HFD) for up to four-months prior to MRI of the brachiocephalic-artery. To assess response-to-therapy, a statin was administered for the same duration. MR imaging was performed before and after administration of an elastin-specific gadolinium-based and a macrophage-specific iron-oxide-based probe. Following in-vivo MRI, samples were analyzed using histology, immunohistochemistry, inductively-coupled-mass-spectrometry and laser-inductively-coupled-mass-spectrometry. In atherosclerotic-plaques, intraplaque expression of elastic-fibers and inflammatory activity were not directly linked. While the elastin-specific probe demonstrated the highest accumulation in advanced atherosclerotic-plaques after four-months of HFD, the iron-oxide-based probe showed highest accumulation in early atherosclerotic-plaques after two-months of HFD. In-vivo measurements for the elastin and iron-oxide-probe were in good agreement with ex-vivo histopathology (Elastica-van-Giesson stain: y = 298.2 + 5.8, R2 = 0.83, p < 0.05; Perls‘ Prussian-blue-stain: y = 834.1 + 0.67, R2 = 0.88, p < 0.05). Contrast-to-noise-ratio (CNR) measurements of the elastin probe were in good agreement with ICP-MS (y = 0.11x-11.3, R² = 0.73, p < 0.05). Late stage atherosclerotic-plaques displayed the strongest increase in both CNR and gadolinium concentration (p < 0.05). The gadolinium probe did not affect the visualization of the iron-oxide-probe and vice versa. This study demonstrates the feasibility of simultaneous assessment of plaque-burden and inflammatory activity by dual-probe molecular MRI of progressive atherosclerosis. The in-vivo detection and quantification of different MR biomarkers in a single scan could be useful to improve characterization of atherosclerotic-lesions.
Collapse
|
9
|
Vigne J, Thackeray J, Essers J, Makowski M, Varasteh Z, Curaj A, Karlas A, Canet-Soulas E, Mulder W, Kiessling F, Schäfers M, Botnar R, Wildgruber M, Hyafil F. Current and Emerging Preclinical Approaches for Imaging-Based Characterization of Atherosclerosis. Mol Imaging Biol 2019; 20:869-887. [PMID: 30250990 DOI: 10.1007/s11307-018-1264-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Atherosclerotic plaques can remain quiescent for years, but become life threatening upon rupture or disruption, initiating clot formation in the vessel lumen and causing acute myocardial infarction and ischemic stroke. Whether and how a plaque ruptures is determined by its macroscopic structure and microscopic composition. Rupture-prone plaques usually consist of a thin fibrous cap with few smooth muscle cells, a large lipid core, a dense infiltrate of inflammatory cells, and neovessels. Such lesions, termed high-risk plaques, can remain asymptomatic until the thrombotic event. Various imaging technologies currently allow visualization of morphological and biological characteristics of high-risk atherosclerotic plaques. Conventional protocols are often complex and lack specificity for high-risk plaque. Conversely, new imaging approaches are emerging which may overcome these limitations. Validation of these novel imaging techniques in preclinical models of atherosclerosis is essential for effective translational to clinical practice. Imaging the vessel wall, as well as its biological milieu in small animal models, is challenging because the vessel wall is a small structure that undergoes continuous movements imposed by the cardiac cycle as it is adjacent to circulating blood. The focus of this paper is to provide a state-of-the-art review on techniques currently available for preclinical imaging of atherosclerosis in small animal models and to discuss the advantages and limitations of each approach.
Collapse
Affiliation(s)
- Jonathan Vigne
- Department of Nuclear Medicine, Bichat University Hospital, AP-HP; INSERM, U-1148, DHU FIRE, University Diderot, Paris, France
| | - James Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Jeroen Essers
- Departments of Vascular Surgery, Molecular Genetics, Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - Marcus Makowski
- Department of Radiology, Charité-University Medicine Berlin, Berlin, Germany
| | - Zoreh Varasteh
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Adelina Curaj
- Institute for Molecular Cardiovascular Research (IMCAR), Institute for Experimental Molecular Imaging (ExMI), University Hospital Aachen, RWTH, Aachen, Germany
| | - Angelos Karlas
- Institute for Biological and Medical Imaging, Helmholtz Zentrum München, Oberschleissheim, Germany
| | - Emmanuel Canet-Soulas
- Laboratoire CarMeN, INSERM U-1060, Lyon/Hospices Civils Lyon, IHU OPERA Cardioprotection, Université de Lyon, Bron, France
| | - Willem Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, Mount Sinai, New York, USA
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging (ExMI), University Hospital Aachen, RWTH, Aachen, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - René Botnar
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Moritz Wildgruber
- Translational Research Imaging Center, Institut für Klinische Radiologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Fabien Hyafil
- Department of Nuclear Medicine, Bichat University Hospital, AP-HP; INSERM, U-1148, DHU FIRE, University Diderot, Paris, France. .,Département de Médecine Nucléaire, Centre Hospitalier Universitaire Bichat, 46 rue Henri Huchard, 75018, Paris, France.
| | | |
Collapse
|
10
|
Dweck MR, Robson PM, Rudd JH, Fayad ZA. Atherosclerotic Plaque Imaging. CARDIOVASCULAR MAGNETIC RESONANCE 2019:335-342.e3. [DOI: 10.1016/b978-0-323-41561-3.00028-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Sadat U, Usman A, Gillard JH. Imaging pathobiology of carotid atherosclerosis with ultrasmall superparamagnetic particles of iron oxide: an update. Curr Opin Cardiol 2018; 32:437-440. [PMID: 28463893 PMCID: PMC5617556 DOI: 10.1097/hco.0000000000000413] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Purpose of review To provide brief overview of the developments regarding use of ultrasmall superparamagnetic particles of iron oxide in imaging pathobiology of carotid atherosclerosis. Recent findings MRI is a promising technique capable of providing morphological and functional information about atheromatous plaques. MRI using iron oxide particles, called ultrasmall superparamagnetic iron oxide (USPIO) particles, allows detection of macrophages in atherosclerotic tissue. Ferumoxytol has emerged as a new USPIO agent, which has an excellent safety profile. Based on the macrophage-selective properties of ferumoxytol, there is increasing number of recent reports suggesting its effectiveness to detect pathological inflammation. Summary USPIO particles allow magnetic resonance detection of macrophages in atherosclerotic tissue. Ferumoxytol has emerged as a new USPIO agent, with an excellent safety profile. This has the potential to be used for MRI of the pathobiology of atherosclerosis.
Collapse
Affiliation(s)
- Umar Sadat
- aCambridge Vascular Unit bUniversity Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
12
|
Chan JMS, Monaco C, Wylezinska-Arridge M, Tremoleda JL, Cole JE, Goddard M, Cheung MSH, Bhakoo KK, Gibbs RGJ. Imaging vulnerable plaques by targeting inflammation in atherosclerosis using fluorescent-labeled dual-ligand microparticles of iron oxide and magnetic resonance imaging. J Vasc Surg 2018; 67:1571-1583.e3. [PMID: 28648478 DOI: 10.1016/j.jvs.2017.04.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/01/2017] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Identification of patients with high-risk asymptomatic carotid plaques remains an elusive but essential step in stroke prevention. Inflammation is a key process in plaque destabilization and a prelude to clinical sequelae. There are currently no clinical imaging tools to assess the inflammatory activity within plaques. This study characterized inflammation in atherosclerosis using dual-targeted microparticles of iron oxide (DT-MPIO) as a magnetic resonance imaging (MRI) probe. METHODS DT-MPIO were used to detect and characterize inflammatory markers, vascular cell adhesion molecule 1 (VCAM-1). and P-selectin on (1) tumor necrosis factor-α-treated cells by immunocytochemistry and (2) aortic root plaques of apolipoprotein-E deficient mice by in vivo MRI. Furthermore, apolipoprotein E-deficient mice with focal carotid plaques of different phenotypes were developed by means of periarterial cuff placement to allow in vivo molecular MRI using these probes. The association between biomarkers and the magnetic resonance signal in different contrast groups was assessed longitudinally in these models. RESULTS Immunocytochemistry confirmed specificity and efficacy of DT-MPIO to VCAM-1 and P-selectin. Using this in vivo molecular MRI strategy, we demonstrated (1) the DT-MPIO-induced magnetic resonance signal tracked with VCAM-1 (r = 0.69; P = .014), P-selectin (r = 0.65; P = .022), and macrophage content (r = 0.59; P = .045) within aortic root plaques and (2) high-risk inflamed plaques were distinguished from noninflamed plaques in the murine carotid artery within a practical clinical imaging time frame. CONCLUSIONS These molecular MRI probes constitute a novel imaging tool for in vivo characterization of plaque vulnerability and inflammatory activity in atherosclerosis. Further development and translation into the clinical arena will facilitate more accurate risk stratification in carotid atherosclerotic disease in the future.
Collapse
Affiliation(s)
- Joyce M S Chan
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, United Kingdom; Regional Vascular Unit, St Mary's Hospital, Imperial College Healthcare National Health Service Trust, Imperial College London, London, United Kingdom; The Laboratory of Molecular Imaging, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (ASTAR), Singapore.
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Marzena Wylezinska-Arridge
- Neuroradiological Academic Unit, University of College London Institute of Neurology, University College London, London, United Kingdom
| | - Jordi L Tremoleda
- Medical Research Council-Clinical Sciences Centre, Imperial College London, and Centre for Trauma Sciences, Queen Mary University of London, London, United Kingdom
| | - Jennifer E Cole
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Michael Goddard
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Maggie S H Cheung
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, United Kingdom
| | - Kishore K Bhakoo
- The Laboratory of Molecular Imaging, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (ASTAR), Singapore
| | - Richard G J Gibbs
- Regional Vascular Unit, St Mary's Hospital, Imperial College Healthcare National Health Service Trust, Imperial College London, London, United Kingdom
| |
Collapse
|
13
|
Nasr SH, Tonson A, El-Dakdouki MH, Zhu DC, Agnew D, Wiseman R, Qian C, Huang X. Effects of Nanoprobe Morphology on Cellular Binding and Inflammatory Responses: Hyaluronan-Conjugated Magnetic Nanoworms for Magnetic Resonance Imaging of Atherosclerotic Plaques. ACS APPLIED MATERIALS & INTERFACES 2018; 10:11495-11507. [PMID: 29558108 PMCID: PMC5995107 DOI: 10.1021/acsami.7b19708] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Atherosclerosis is an inflammatory disease of arterial walls and the rupturing of atherosclerotic plaques is a major cause of heart attack and stroke. Imaging techniques that can enable the detection of atherosclerotic plaques before clinical manifestation are urgently needed. Magnetic resonance imaging (MRI) is a powerful technique to image the morphology of atherosclerotic plaques. In order to better analyze molecular processes in plaques, contrast agents that can selectively bind to plaque receptors will prove invaluable. CD44 is a cell surface protein overexpressed in plaque tissues, the level of which can be correlated with the risks of plaque rupture. Thus, targeting CD44 is an attractive strategy for detection of atherosclerotic plaques. Herein, we report the synthesis of hyaluronan-conjugated iron oxide nanoworms (HA-NWs). A new purification and gel electrophoresis protocol was developed to ensure the complete removal of free HA from HA-NWs. Compared to the more traditional spherical HA-bearing nanoparticles, HA-NWs had an elongated shape, which interacted much stronger with CD44-expressing cells in CD44- and HA-dependent manners. Furthermore, the HA-NWs did not induce much inflammatory response compared to the spherical HA nanoparticles. When assessed in vivo, HA-NWs enabled successful imaging of atherosclerotic plaques in a clinically relevant model of ApoE knockout transgenic mice for noninvasive plaque detection by MRI. Thus, nanoprobe shape engineering can be a useful strategy to significantly enhance their desired biological properties.
Collapse
Affiliation(s)
| | - Anne Tonson
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Mohammad H. El-Dakdouki
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Chemistry, Beirut Arab University, P.O. Box 11-5020, Riad El Solh 11072809, Beirut, Lebanon
| | - David C. Zhu
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Dalen Agnew
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan 48824, United States
| | - Robert Wiseman
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Chunqi Qian
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
14
|
Nguyen LT, Muktabar A, Tang J, Dravid VP, Thaxton CS, Venkatraman S, Ng KW. Engineered nanoparticles for the detection, treatment and prevention of atherosclerosis: how close are we? Drug Discov Today 2017; 22:1438-1446. [DOI: 10.1016/j.drudis.2017.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 06/12/2017] [Accepted: 07/10/2017] [Indexed: 01/09/2023]
|
15
|
Delbosc S, Bayles RG, Laschet J, Ollivier V, Ho-Tin-Noé B, Touat Z, Deschildre C, Morvan M, Louedec L, Gouya L, Guedj K, Nicoletti A, Michel JB. Erythrocyte Efferocytosis by the Arterial Wall Promotes Oxidation in Early-Stage Atheroma in Humans. Front Cardiovasc Med 2017; 4:43. [PMID: 28824922 PMCID: PMC5539175 DOI: 10.3389/fcvm.2017.00043] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 06/26/2017] [Indexed: 01/21/2023] Open
Abstract
Background Since red blood cells (RBCs) are the predominant cellular blood component interacting with the arterial wall, we explored the role of RBCs efferocytosis by vascular smooth muscle cells (vSMCs) in the initiation of human atheroma. Methods and results The comparison of human healthy aortas with aortic fatty streaks or fibroatheromas revealed that RBC angiophagy is implicated from the earliest stages of atherogenesis, as documented by the concomitant detection of redox-active iron, hemoglobin, glycophorin A, and ceroids. RBCs infiltration in the arterial wall was associated with local lipid and protein oxidation, as well as vascular response (expression of heme oxygenase-1 and of genes related to iron metabolism as well as those encoding for phagocytosis). These effects were recapitulated in vitro when vSMCs were co-cultured with phosphatidyl-exposing senescent (s) RBCs but not with fresh RBCs. VSMCs engulfing sRBC increased their intracellular iron content, accumulated hemoglobin, lipids, and activated their phagolysosomes. Strikingly, injections of sRBCs into rats promoted iron accumulation in the aortic wall. In rabbits, hypercholesterolemia increased circulating senescent RBCs and induced the subendothelial accumulation of iron-rich phagocytic foam cells. RBCs bring cholesterol and iron/heme into the vascular wall and interact with vSMCs that phagocytize them. Conclusion This study presents a previously unforeseen mechanism of plaque formation that implicates intimal RBC infiltration as one of the initial triggers for foam cell formation and intimal oxidation. Pathogenic effects exerted by several metabolic and hemodynamic factors may rely on their effect on RBC biology, thereby impacting how RBCs interact with the vascular wall.
Collapse
Affiliation(s)
- Sandrine Delbosc
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Richard Graham Bayles
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Jamila Laschet
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Veronique Ollivier
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Benoit Ho-Tin-Noé
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Ziad Touat
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Catherine Deschildre
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Marion Morvan
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Liliane Louedec
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Laurent Gouya
- Département Hospitalo-Universitaire DHU "FIRE", Paris, France.,UMRS 1149, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France
| | - Kevin Guedj
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Antonino Nicoletti
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| | - Jean-Baptiste Michel
- UMRS 1148, INSERM, Paris 7-Denis Diderot University, Hôpital Xavier Bichat, Paris, France.,Département Hospitalo-Universitaire DHU "FIRE", Paris, France
| |
Collapse
|
16
|
Miller MA, Arlauckas S, Weissleder R. Prediction of Anti-cancer Nanotherapy Efficacy by Imaging. Nanotheranostics 2017; 1:296-312. [PMID: 29071194 PMCID: PMC5646731 DOI: 10.7150/ntno.20564] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/27/2017] [Indexed: 12/17/2022] Open
Abstract
Anticancer nanotherapeutics have shown mixed results in clinical trials, raising the questions of whether imaging should be used to i) identify patients with a higher likelihood of nanoparticle accumulation, ii) assess nanotherapeutic efficacy before traditional measures show response, and iii) guide adjuvant treatments to enhance therapeutic nanoparticle (TNP) delivery. Here we review the use of a clinically approved MRI nanoparticle (ferumoxytol, FMX) to predict TNP delivery and efficacy. It is becoming increasingly apparent that nanoparticles used for imaging, despite clearly distinct physicochemical properties, often co-localize with TNP in tumors. This evidence offers the possibility of using FMX as a generic “companion diagnostic” nanoparticle for multiple TNP formulations, thus potentially allowing many of the complex regulatory and cost challenges of other approaches to be avoided.
Collapse
Affiliation(s)
- Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital, USA.,Department of Radiology, Massachusetts General Hospital, USA
| | - Sean Arlauckas
- Center for Systems Biology, Massachusetts General Hospital, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, USA.,Department of Radiology, Massachusetts General Hospital, USA.,Department of Systems Biology, Harvard Medical School, USA
| |
Collapse
|
17
|
Stein-Merlob AF, Hara T, McCarthy JR, Mauskapf A, Hamilton JA, Ntziachristos V, Libby P, Jaffer FA. Atheroma Susceptible to Thrombosis Exhibit Impaired Endothelial Permeability In Vivo as Assessed by Nanoparticle-Based Fluorescence Molecular Imaging. Circ Cardiovasc Imaging 2017; 10:CIRCIMAGING.116.005813. [PMID: 28487316 DOI: 10.1161/circimaging.116.005813] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/28/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND The role of local alterations in endothelial functional integrity in atherosclerosis remains incompletely understood. This study used nanoparticle-enhanced optical molecular imaging to probe in vivo mechanisms involving impaired endothelial barrier function in experimental atherothrombosis. METHODS AND RESULTS Atherosclerosis was induced in rabbits (n=31) using aortic balloon injury and high-cholesterol diet. Rabbits received ultrasmall superparamagnetic iron oxide nanoparticles (CLIO) derivatized with a near-infrared fluorophore (CyAm7) 24 hours before near-infrared fluorescence imaging. Rabbits were then either euthanized (n=9) or underwent a pharmacological triggering protocol to induce thrombosis (n=22). CLIO-CyAm7 nanoparticles accumulated in areas of atheroma (P<0.05 versus reference areas). On near-infrared fluorescence microscopy, CLIO-CyAm7 primarily deposited in the superficial intima within plaque macrophages, endothelial cells, and smooth muscle cells. Nanoparticle-positive areas further exhibited impaired endothelial barrier function as illuminated by Evans blue leakage. Deeper nanoparticle deposition occurred in areas of plaque neovascularization. In rabbits subject to pharmacological triggering, plaques that thrombosed exhibited significantly higher CLIO-CyAm7 accumulation compared with nonthrombosed plaques (P<0.05). In thrombosed plaques, nanoparticles accumulated preferentially at the plaque-thrombus interface. Intravascular 2-dimensional near-infrared fluorescence imaging detected nanoparticles in human coronary artery-sized atheroma in vivo (P<0.05 versus reference segments). CONCLUSIONS Plaques that exhibit impaired in vivo endothelial permeability in cell-rich areas are susceptible to subsequent thrombosis. Molecular imaging of nanoparticle deposition may help to identify biologically high-risk atheroma.
Collapse
Affiliation(s)
- Ashley F Stein-Merlob
- From the Cardiovascular Research Center, Cardiology Division (A.F.S., T.H., A.M., F.A.J.) and Center for Systems Biology (J.R.M.), Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Physiology and Biophysics, Boston University School of Medicine, MA (J.A.H.); Department of Biomedical Engineering, Boston University, MA (J.A.H.); Institute of Biological and Medical Imaging, Chair of Biological Imaging, Technical University of Munich, Germany (V.N.); and Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.)
| | - Tetsuya Hara
- From the Cardiovascular Research Center, Cardiology Division (A.F.S., T.H., A.M., F.A.J.) and Center for Systems Biology (J.R.M.), Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Physiology and Biophysics, Boston University School of Medicine, MA (J.A.H.); Department of Biomedical Engineering, Boston University, MA (J.A.H.); Institute of Biological and Medical Imaging, Chair of Biological Imaging, Technical University of Munich, Germany (V.N.); and Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.)
| | - Jason R McCarthy
- From the Cardiovascular Research Center, Cardiology Division (A.F.S., T.H., A.M., F.A.J.) and Center for Systems Biology (J.R.M.), Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Physiology and Biophysics, Boston University School of Medicine, MA (J.A.H.); Department of Biomedical Engineering, Boston University, MA (J.A.H.); Institute of Biological and Medical Imaging, Chair of Biological Imaging, Technical University of Munich, Germany (V.N.); and Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.)
| | - Adam Mauskapf
- From the Cardiovascular Research Center, Cardiology Division (A.F.S., T.H., A.M., F.A.J.) and Center for Systems Biology (J.R.M.), Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Physiology and Biophysics, Boston University School of Medicine, MA (J.A.H.); Department of Biomedical Engineering, Boston University, MA (J.A.H.); Institute of Biological and Medical Imaging, Chair of Biological Imaging, Technical University of Munich, Germany (V.N.); and Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.)
| | - James A Hamilton
- From the Cardiovascular Research Center, Cardiology Division (A.F.S., T.H., A.M., F.A.J.) and Center for Systems Biology (J.R.M.), Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Physiology and Biophysics, Boston University School of Medicine, MA (J.A.H.); Department of Biomedical Engineering, Boston University, MA (J.A.H.); Institute of Biological and Medical Imaging, Chair of Biological Imaging, Technical University of Munich, Germany (V.N.); and Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.)
| | - Vasilis Ntziachristos
- From the Cardiovascular Research Center, Cardiology Division (A.F.S., T.H., A.M., F.A.J.) and Center for Systems Biology (J.R.M.), Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Physiology and Biophysics, Boston University School of Medicine, MA (J.A.H.); Department of Biomedical Engineering, Boston University, MA (J.A.H.); Institute of Biological and Medical Imaging, Chair of Biological Imaging, Technical University of Munich, Germany (V.N.); and Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.)
| | - Peter Libby
- From the Cardiovascular Research Center, Cardiology Division (A.F.S., T.H., A.M., F.A.J.) and Center for Systems Biology (J.R.M.), Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Physiology and Biophysics, Boston University School of Medicine, MA (J.A.H.); Department of Biomedical Engineering, Boston University, MA (J.A.H.); Institute of Biological and Medical Imaging, Chair of Biological Imaging, Technical University of Munich, Germany (V.N.); and Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.)
| | - Farouc A Jaffer
- From the Cardiovascular Research Center, Cardiology Division (A.F.S., T.H., A.M., F.A.J.) and Center for Systems Biology (J.R.M.), Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Physiology and Biophysics, Boston University School of Medicine, MA (J.A.H.); Department of Biomedical Engineering, Boston University, MA (J.A.H.); Institute of Biological and Medical Imaging, Chair of Biological Imaging, Technical University of Munich, Germany (V.N.); and Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.).
| |
Collapse
|
18
|
Emeto TI, Alele FO, Smith AM, Smith FM, Dougan T, Golledge J. Use of Nanoparticles As Contrast Agents for the Functional and Molecular Imaging of Abdominal Aortic Aneurysm. Front Cardiovasc Med 2017; 4:16. [PMID: 28386544 PMCID: PMC5362602 DOI: 10.3389/fcvm.2017.00016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/09/2017] [Indexed: 01/19/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a degenerative disease of the aorta common in adults older than 65 years of age. AAA is usually imaged using ultrasound or computed tomography. Molecular imaging technologies employing nanoparticles (NPs) have been proposed as novel ways to quantify pathological processes, such as inflammation, within AAAs as a means to identify the risk of rapid progression or rupture. This article reviews the current evidence supporting the role of NP-based imaging in the management of AAA. Currently, ultrasmall superparamagnetic NPs enhanced magnetic resonance imaging appears to hold the greatest potential for imaging macrophage-mediated inflammation in human AAA.
Collapse
Affiliation(s)
- Theophilus I Emeto
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia; Queensland Research Centre for Peripheral Vascular Diseases, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia
| | - Faith O Alele
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University , Townsville, QLD , Australia
| | - Amy M Smith
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University , Townsville, QLD , Australia
| | - Felicity M Smith
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University , Townsville, QLD , Australia
| | - Tammy Dougan
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Addenbrookes Hospital , Cambridge , UK
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Diseases, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia; Department of Vascular and Endovascular Surgery, The Townsville Hospital, Townsville, QLD, Australia
| |
Collapse
|
19
|
Chan JMS, Cheung MSH, Gibbs RGJ, Bhakoo KK. MRI detection of endothelial cell inflammation using targeted superparamagnetic particles of iron oxide (SPIO). Clin Transl Med 2017; 6:1. [PMID: 28044245 PMCID: PMC5206220 DOI: 10.1186/s40169-016-0134-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 12/21/2016] [Indexed: 01/10/2023] Open
Abstract
Background There is currently no clinical imaging technique available to assess the degree of inflammation associated with atherosclerotic plaques. This study aims to develop targeted superparamagnetic particles of iron oxide (SPIO) as a magnetic resonance imaging (MRI) probe for detecting inflamed endothelial cells. Methods The in vitro study consists of the characterisation and detection of inflammatory markers on activated endothelial cells by immunocytochemistry and MRI using biotinylated anti-P-selectin and anti-VCAM-1 (vascular cell adhesion molecule 1) antibody and streptavidin conjugated SPIO. Results Established an in vitro cellular model of endothelial inflammation induced with TNF-α (tumor necrosis factor alpha). Inflammation of endothelial cells was confirmed with both immunocytochemistry and MRI. These results revealed both a temporal and dose dependent expression of the inflammatory markers, P-selectin and VCAM-1, on exposure to TNF-α. Conclusion This study has demonstrated the development of an in vitro model to characterise and detect inflamed endothelial cells by immunocytochemistry and MRI. This will allow the future development of contrast agents and protocols for imaging vascular inflammation in atherosclerosis. This work may form the basis for a translational study to provide clinicians with a novel tool for the in vivo assessment of atherosclerosis.
Collapse
Affiliation(s)
- Joyce M S Chan
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China. .,Regional Vascular Unit, St Mary's Hospital, Imperial College Healthcare NHS Trust, Imperial College London, London, UK. .,Translational Molecular Imaging Group, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios, 138667, Singapore.
| | - Maggie S H Cheung
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Richard G J Gibbs
- Regional Vascular Unit, St Mary's Hospital, Imperial College Healthcare NHS Trust, Imperial College London, London, UK
| | - Kishore K Bhakoo
- Translational Molecular Imaging Group, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios, 138667, Singapore
| |
Collapse
|
20
|
Li B, Jiao Y, Fu C, Xie B, Ma G, Teng G, Yao Y. Contralateral artery enlargement predicts carotid plaque progression based on machine learning algorithm models in apoE -/- mice. Biomed Eng Online 2016; 15:146. [PMID: 28155719 PMCID: PMC5259854 DOI: 10.1186/s12938-016-0265-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND This study specifically focused on anatomical MRI characterization of the low shear stress-induced atherosclerotic plaque in mice. We used machine learning algorithms to analyze multiple correlation factors of plaque to generate predictive models and to find the predictive factor for vulnerable plaque. METHODS Branches of the left carotid artery in apoE-/- and C57BL/6J mice were ligated to produce the partial left carotid artery model. Before surgery, and 7, 14, and 28 days after surgery, in vivo serial MRI measurements of carotid artery diameter were obtained. Meanwhile, proximal blood flow was evaluated. After image acquisition and animal sacrifice, carotid arteries were harvested for histological analysis. Support vector machine (SVM) and decision tree (DT) were used to select features and generate predictive models of vulnerable plaque progression. RESULT Seven days after surgery, neointima formation was visualized on micro-MRI in both apoE-/- and C57BL/6J mice. Ultrasonography showed that blood flow had significantly decreased compared to that in the contralateral artery. Partial ligation of the carotid artery for 4 weeks in apoE-/- mice induced vulnerable plaque; however, in C57BL/6J mice this same technique performed for 4 weeks induced arterial stenosis. Contralateral carotid artery diameter at 7 days after surgery was the most reliable predictive factor in plaque progression. We achieved over 87.5% accuracy, 80% sensitivity, and 95% specificity with SVM. The accuracy, sensitivity, and specificity for the DT classifier were 90, 90, and 90%, respectively. CONCLUSIONS This study is the first to demonstrate that SVM and DT methods could be suitable models for identifying vulnerable plaque progression in mice. And contralateral artery enlargement can predict the vulnerable plaque in carotid artery at the very early stage. It may be a valuable tool which helps to optimize the clinical work flow process by providing more decision in selecting patients for treatment.
Collapse
Affiliation(s)
- Bing Li
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Yun Jiao
- Jiangsu Key Lab of Molecular and Function Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Cong Fu
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Bo Xie
- Jiangsu Key Lab of Molecular and Function Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Gaojun Teng
- Jiangsu Key Lab of Molecular and Function Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
21
|
Chhour P, Naha PC, O'Neill SM, Litt HI, Reilly MP, Ferrari VA, Cormode DP. Labeling monocytes with gold nanoparticles to track their recruitment in atherosclerosis with computed tomography. Biomaterials 2016; 87:93-103. [PMID: 26914700 DOI: 10.1016/j.biomaterials.2016.02.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 01/26/2016] [Accepted: 02/07/2016] [Indexed: 01/21/2023]
Abstract
Monocytes are actively recruited from the circulation into developing atherosclerotic plaques. In the plaque, monocytes differentiate into macrophages and eventually form foam cells. Continued accumulation of foam cells can lead to plaque rupture and subsequent myocardial infarction. X-ray computed tomography (CT) is the best modality to image the coronary arteries non-invasively, therefore we have sought to track the accumulation of monocytes into atherosclerotic plaques using CT. Gold nanoparticles were synthesized and stabilized with a variety of ligands. Select formulations were incubated with an immortalized monocyte cell line in vitro and evaluated for cytotoxicity, effects on cytokine release, and cell uptake. These data identified a lead formulation, 11-MUDA capped gold nanoparticles, to test for labeling primary monocytes. The formulation did not the affect the viability or cytokine release of primary monocytes and was highly taken up by these cells. Gold labeled primary monocytes were injected into apolipoprotein E deficient mice kept on Western diet for 10 weeks. Imaging was done with a microCT scanner. A significant increase in attenuation was measured in the aorta of mice receiving the gold labeled cells as compared to control animals. Following the experiment, the biodistribution of gold was evaluated in major organs. Additionally, plaques were sectioned and examined with electron microscopy. The results showed that gold nanoparticles were present inside monocytes located within plaques. This study demonstrates the feasibility of using gold nanoparticles as effective cell labeling contrast agents for non-invasive imaging of monocyte accumulation within plaques with CT.
Collapse
Affiliation(s)
- Peter Chhour
- Department of Radiology, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
| | - Pratap C Naha
- Department of Radiology, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
| | - Sean M O'Neill
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
| | - Harold I Litt
- Department of Radiology, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA; Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
| | - Muredach P Reilly
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
| | - Victor A Ferrari
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
| | - David P Cormode
- Department of Radiology, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA; Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA.
| |
Collapse
|
22
|
Wang C, Wen J, Zhou Y, Li L, Cui X, Wang J, Pan L, Ye Z, Liu P, Wu L. Apelin induces vascular smooth muscle cells migration via a PI3K/Akt/FoxO3a/MMP-2 pathway. Int J Biochem Cell Biol 2015; 69:173-82. [PMID: 26494002 DOI: 10.1016/j.biocel.2015.10.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 10/04/2015] [Accepted: 10/13/2015] [Indexed: 11/19/2022]
Abstract
Apelin is an adipokine that has a critical role in the development of atherosclerosis, which may offer potential for therapy. Because migration of vascular smooth muscle cells (VSMCs) is a key event in the development of atherosclerosis, understanding its effect on the atherosclerotic vasculature is needed. Here we investigated the effect of apelin on VSMC migration and the possible signaling mechanism. In cultured rat VSMCs, apelin dose- and time-dependently promoted VSMC migration. Apelin increased the phosphorylation of Akt, whereas LY294002, an inhibitor of phosphatidylinositol 3-kinase (PI3K), and an Akt1/2 kinase inhibitor blocked the apelin-induced VSMC migration. Apelin dose-dependently induced phosphorylation of Forkhead box O3a (FoxO3a) and promoted its translocation from the nucleus to cytoplasm, which were blocked by LY294002 and Akt1/2 kinase inhibitor. Furthermore, apelin increased matrix metalloproteinase 2 (MMP-2) expression and gelatinolytic activity. Overexpression of a constitutively active, phosphorylation-resistant mutant, TM-FoxO3a, in VSMCs abrogated the effect of apelin on MMP-2 expression and VSMC migration. ARP101, an inhibitor of MMP-2, suppressed apelin-induced VSMC migration. Moreover, the levels of apelin, phosphorylated Akt, FoxO3a, and MMP-2 were higher in human carotid-artery atherosclerotic plaque than in adjacent normal vessels. We demonstrate that PI3K/Akt/FoxO3a signaling may be involved in apelin inducing VSMC migration. Phosphorylation of FoxO3a plays a central role in mediating the apelin-induced MMP-2 activation and VSMC migration.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Jianyan Wen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yun Zhou
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Li Li
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Xiaobing Cui
- Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing 100191, China
| | - Jinyu Wang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Lin Pan
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Zhidong Ye
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Peng Liu
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Liling Wu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China.
| |
Collapse
|
23
|
Usman A, Sadat U, Patterson AJ, Tang TY, Varty K, Boyle JR, Armon MP, Hayes PD, Graves MJ, Gillard JH. Use of ultrasmall superparamagnetic iron oxide particles for imaging carotid atherosclerosis. Nanomedicine (Lond) 2015; 10:3077-3087. [PMID: 26420349 DOI: 10.2217/nnm.15.136] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Based on the results of histopathological studies, inflammation within atherosclerotic tissue is now widely accepted as a key determinant of the disease process. Conventional imaging methods can highlight the location and degree of luminal stenosis but not the inflammatory activity of the plaque. Iron oxide-based MRI contrast media particularly ultrasmall supermagnetic particles of iron oxide have shown potential in assessing atheromatous plaque inflammation and in determining efficacy of antiatherosclerosis pharmacological treatments. In this paper, we review current data on the use of ultrasmall superparamagnetic iron oxides in atherosclerosis imaging with focus on ferumoxtran-10 and ferumoxytol. The basic chemistry, pharmacokinetics and dynamics, potential applications, limitations and future perspectives of these contrast media nanoparticles are discussed.
Collapse
Affiliation(s)
- Ammara Usman
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Box 218, Level 5, Hills Road, Cambridge, CB2 0QQ, UK
| | - Umar Sadat
- Cambridge Vascular Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Andrew J Patterson
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Box 218, Level 5, Hills Road, Cambridge, CB2 0QQ, UK
| | - Tjun Y Tang
- Department of Surgery, Changi General Hospital, Singapore
| | - Kevin Varty
- Cambridge Vascular Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Jonathan R Boyle
- Cambridge Vascular Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Mathew P Armon
- Department of Vascular Surgery, Norfolk & Norwich University Hospital, Norwich, NR4 7UY, UK
| | - Paul D Hayes
- Cambridge Vascular Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Martin J Graves
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Box 218, Level 5, Hills Road, Cambridge, CB2 0QQ, UK
| | - Jonathan H Gillard
- Department of Radiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Box 218, Level 5, Hills Road, Cambridge, CB2 0QQ, UK
| |
Collapse
|
24
|
Alam SR, Stirrat C, Richards J, Mirsadraee S, Semple SIK, Tse G, Henriksen P, Newby DE. Vascular and plaque imaging with ultrasmall superparamagnetic particles of iron oxide. J Cardiovasc Magn Reson 2015; 17:83. [PMID: 26381872 PMCID: PMC4574723 DOI: 10.1186/s12968-015-0183-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 08/16/2015] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular Magnetic Resonance (CMR) has become a primary tool for non-invasive assessment of cardiovascular anatomy, pathology and function. Existing contrast agents have been utilised for the identification of infarction, fibrosis, perfusion deficits and for angiography. Novel ultrasmall superparamagnetic particles of iron oxide (USPIO) contrast agents that are taken up by inflammatory cells can detect cellular inflammation non-invasively using CMR, potentially aiding the diagnosis of inflammatory medical conditions, guiding their treatment and giving insight into their pathophysiology. In this review we describe the utilization of USPIO as a novel contrast agent in vascular disease.
Collapse
Affiliation(s)
- Shirjel R Alam
- Centre for Cardiovascular Science, The University of Edinburgh, The Chancellor's Building, Little France Crescent, Edinburgh, EH16 5SA, UK.
- Department of Cardiology, Royal Infirmary of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - Colin Stirrat
- Centre for Cardiovascular Science, The University of Edinburgh, The Chancellor's Building, Little France Crescent, Edinburgh, EH16 5SA, UK.
- Department of Cardiology, Royal Infirmary of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - Jennifer Richards
- Centre for Cardiovascular Science, The University of Edinburgh, The Chancellor's Building, Little France Crescent, Edinburgh, EH16 5SA, UK.
| | - Saeed Mirsadraee
- Clinical Research Imaging Centre, University of Edinburgh, Edinburgh, EH16 5SA, UK.
- Department of Radiology, Royal Infirmary of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - Scott I K Semple
- Clinical Research Imaging Centre, University of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - George Tse
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - Peter Henriksen
- Centre for Cardiovascular Science, The University of Edinburgh, The Chancellor's Building, Little France Crescent, Edinburgh, EH16 5SA, UK.
- Department of Cardiology, Royal Infirmary of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - David E Newby
- Centre for Cardiovascular Science, The University of Edinburgh, The Chancellor's Building, Little France Crescent, Edinburgh, EH16 5SA, UK.
- Department of Cardiology, Royal Infirmary of Edinburgh, Edinburgh, EH16 5SA, UK.
| |
Collapse
|
25
|
Kaneko C, Nitta N, Tsuchiya K, Watanabe S, Nitta-Seko A, Ohta S, Otani H, Sonoda A, Murata K, Shiomi M. MRI study of atherosclerotic plaque progression using ultrasmall superparamagnetic iron oxide in Watanabe heritable hyperlipidemic rabbits. Br J Radiol 2015; 88:20150167. [PMID: 26083261 DOI: 10.1259/bjr.20150167] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE The purpose of this study was to evaluate plaque progression by using MRI with ultrasmall superparamagnetic iron oxide (USPIO) and by histopathological studies. METHODS We divided 12 Watanabe heritable hyperlipidemic (WHHL) rabbits into 4 groups based on their age (3, 9, 14 and 26 months) and injected them intravenously with 0.8 mmol (Fe) kg(-1) of USPIO (size, 32 nm; concentration, 15 mg dl(-1)). On the fifth post-injection day, they were again given an intravenous injection with 40 μmol kg(-1) of the same USPIO, and MR angiography (MRA) was performed. The signal-to-noise ratio (SNR) in regions of interest in the wall of the upper abdominal aorta was calculated on coronal images. Specimens from the same level of the aorta were subjected to iron staining and RAM-11 immunostaining and used for histopathological study. For statistical analysis of the MRA and histopathological findings, we used analysis of variance [Tukey's honest significant difference (HSD) test]. RESULTS In 9-month-old rabbits, the SNR was significantly lower than in rabbits of the other ages (p < 0.01), and the area of RAM-11 (DAKO Corporation, Glostrup, Denmark) and iron uptake in the aortic wall was significantly larger (RAM-11, p < 0.01; iron, p < 0.05). These areas were the smallest in 3-month-old rabbits. CONCLUSION Histopathologically, the number of macrophages was the greatest in 9-month-old rabbits. Our findings indicate that the SNR on MRI scans reflects the number of macrophages in the aortic wall of WHHL rabbits. ADVANCES IN KNOWLEDGE USPIO-enhanced MRI visualized the accumulation of macrophages in early atherosclerotic plaques of WHHL rabbits in the course of natural progression.
Collapse
Affiliation(s)
- C Kaneko
- 1 Department of Radiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - N Nitta
- 1 Department of Radiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - K Tsuchiya
- 1 Department of Radiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - S Watanabe
- 1 Department of Radiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - A Nitta-Seko
- 1 Department of Radiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - S Ohta
- 1 Department of Radiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - H Otani
- 1 Department of Radiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - A Sonoda
- 1 Department of Radiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - K Murata
- 1 Department of Radiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - M Shiomi
- 2 Institute for Experimental Animals, Kobe University School of Medicine, Kobe, Hyogo, Japan
| |
Collapse
|
26
|
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: 85] [Impact Index Per Article: 8.5] [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
|
27
|
Thukkani AK, Jaffer FA. Molecular Imaging. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
28
|
Suzuki M, Bachelet-Violette L, Rouzet F, Beilvert A, Autret G, Maire M, Menager C, Louedec L, Choqueux C, Saboural P, Haddad O, Chauvierre C, Chaubet F, Michel JB, Serfaty JM, Letourneur D. Ultrasmall superparamagnetic iron oxide nanoparticles coated with fucoidan for molecular MRI of intraluminal thrombus. Nanomedicine (Lond) 2014; 10:73-87. [PMID: 24960075 DOI: 10.2217/nnm.14.51] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIM We have designed ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles associated with fucoidan (USPOI-FUCO), a natural sulfated polysaccharide with high affinity for activated platelets, to visualize by MRI arterial thrombi. MATERIALS & METHODS USPIOs were prepared and sizes, zeta-potentials and relaxivities were measured. Elastase perfusion in the infrarenal aorta of Wistar rats induced intraluminal thrombus. They were scanned on 4.7 T MRI before and after injection of USPIO-FUCO or USPIO coated with anionic dextran. RESULTS Surface plasmon resonance evidenced that fucoidan and USPIO-FUCO bind in vitro to immobilized P-selectin. All intraluminal hyposignals detected by MRI after injection of USPIO-FUCO on animals (13 out of 13) were correlated by histology with thrombi, whereas none could be identified with control USPIOs (0 out of 7). No signal was seen in absence of thrombus. Thrombi by MRI were correlated with P-selectin immunostaining and USPIO detection by electron microscopy. CONCLUSION In vivo thrombi can thus be evidenced by MRI with USPIO-FUCO.
Collapse
Affiliation(s)
- Michimasa Suzuki
- French Institute of Health & Medical Research (Inserm) U1148, Laboratory for Vascular Translational Science, CHU X Bichat, University Paris 7, 46 rue H Huchard, F-75877, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Silva AKA, Letourneur D, Chauvierre C. Polysaccharide nanosystems for future progress in cardiovascular pathologies. Theranostics 2014; 4:579-91. [PMID: 24723980 PMCID: PMC3982129 DOI: 10.7150/thno.7688] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/16/2013] [Indexed: 11/09/2022] Open
Abstract
Natural polysaccharides have received a lot of attention in the biomedical field. Indeed, sources of polysaccharides, extracted or produced from plants, bacteria, fungi or algae, are diverse and renewable. Moreover, recent progresses in polysaccharide chemistry and nanotechnologies allow elaborating new dedicated nanosystems. Polysaccharide-based nanosystems may be designed for interacting in several biological processes. In particular, the atherothrombotic pathology is highly concerned by polysaccharide-mediated recognition. Atherothrombotic diseases, regardless of the anatomical localization, remain the main causes of morbidity and mortality in the industrialized world. This review intends to provide an overview on polysaccharide-based nanosystems as drug delivery systems and targeted contrast agents for molecular imaging with an emphasis on the treatment and imaging of cardiovascular pathologies.
Collapse
Affiliation(s)
| | | | - Cédric Chauvierre
- Inserm, U698, Cardiovascular Bio-Engineering; X. Bichat hospital, 46 rue H. Huchard, F-75018, Paris, France; Université Paris 13, Sorbonne Paris Cité, F-93430, Villetaneuse, France
| |
Collapse
|
30
|
Wildgruber M, Swirski FK, Zernecke A. Molecular imaging of inflammation in atherosclerosis. Am J Cancer Res 2013; 3:865-84. [PMID: 24312156 PMCID: PMC3841337 DOI: 10.7150/thno.5771] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 04/29/2013] [Indexed: 01/13/2023] Open
Abstract
Acute rupture of vulnerable plaques frequently leads to myocardial infarction and stroke. Within the last decades, several cellular and molecular players have been identified that promote atherosclerotic lesion formation, maturation and plaque rupture. It is now widely recognized that inflammation of the vessel wall and distinct leukocyte subsets are involved throughout all phases of atherosclerotic lesion development. The mechanisms that render a stable plaque unstable and prone to rupture, however, remain unknown and the identification of the vulnerable plaque remains a major challenge in cardiovascular medicine. Imaging technologies used in the clinic offer minimal information about the underlying biology and potential risk for rupture. New imaging technologies are therefore being developed, and in the preclinical setting have enabled new and dynamic insights into the vessel wall for a better understanding of this complex disease. Molecular imaging has the potential to track biological processes, such as the activity of cellular and molecular biomarkers in vivo and over time. Similarly, novel imaging technologies specifically detect effects of therapies that aim to stabilize vulnerable plaques and silence vascular inflammation. Here we will review the potential of established and new molecular imaging technologies in the setting of atherosclerosis, and discuss the cumbersome steps required for translating molecular imaging approaches into the clinic.
Collapse
|
31
|
Chalouhi N, Jabbour P, Magnotta V, Hasan D. Molecular imaging of cerebrovascular lesions. Transl Stroke Res 2013; 5:260-8. [PMID: 24323714 DOI: 10.1007/s12975-013-0291-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 09/23/2013] [Accepted: 09/26/2013] [Indexed: 01/19/2023]
Abstract
Inflammation is a key component in the pathogenesis of cerebrovascular lesions. Two agents have emerged as promising possibilities for imaging cerebrovascular lesions. These agents are ferumoxytol and myeloperoxidase (MPO)-specific paramagnetic magnetic resonance (MR) contrast agent. Ferumoxytol is an iron oxide nanoparticle coated by a carbohydrate shell that is used in MRI studies as an inflammatory marker as it is cleared by macrophages. Ferumoxytol-enhanced MRI allows noninvasive assessment of the inflammatory status of cerebral aneurysms and arteriovenous malformations and, possibly, may differentiate "unstable" lesions that require early intervention from "stable" lesions that can be safely observed. Several pilot studies have also suggested that MPO-specific paramagnetic MR contrast agent, di-5-hydroxytryptamide of gadopentetate dimeglumine, may allow imaging of inflammation in the wall of saccular aneurysms in animal models. However, studies in human subjects have yet to be performed. In this paper, we review current data regarding ferumoxytol-enhanced MRI and MPO-specific paramagnetic MR contrast agent and discuss current and future applications.
Collapse
Affiliation(s)
- Nohra Chalouhi
- Department of Neurosurgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
32
|
Chalouhi N, Jabbour P, Magnotta V, Hasan D. The emerging role of ferumoxytol-enhanced MRI in the management of cerebrovascular lesions. Molecules 2013; 18:9670-83. [PMID: 23945642 PMCID: PMC6270297 DOI: 10.3390/molecules18089670] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/08/2013] [Indexed: 12/22/2022] Open
Abstract
Inflammation is increasingly being understood to be a key component to the pathophysiology of cerebrovascular lesions. Ferumoxytol, an iron oxide nanoparticle coated by a carbohydrate shell, has been used in MRI studies as an inflammatory marker because it is cleared by macrophages. Ferumoxytol-enhanced MRI has emerged as an important tool for noninvasive assessment of the inflammatory status of cerebrovascular lesions, namely aneurysms and arteriovenous malformations. Moreover, preliminary evidence suggests that ferumoxytol-enhanced MRI could be applied as a non-invasive tool to differentiate “unstable” lesions that require early intervention from “stable” lesions in which observation may be safe. Assessment of the effects of anti-inflammatory pharmacological interventions on cerebrovascular lesions is also a potentially crucial application of the technique. Future improvements in technique and MRI signal quantification will certainly pave the way for widespread and efficient use of ferumoxytol-enhanced MRI in clinical practice. In this paper, we review current data regarding ferumoxytol-enhanced MRI and discuss its current/potential applications and future perspectives.
Collapse
Affiliation(s)
- Nohra Chalouhi
- Department of Neurosurgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, PA 19107, USA; E-Mails: (N.C.); (P.J.)
| | - Pascal Jabbour
- Department of Neurosurgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, PA 19107, USA; E-Mails: (N.C.); (P.J.)
| | - Vincent Magnotta
- Department of Neurosurgery, University of Iowa, Iowa City, IA 52242, USA; E-Mail:
| | - David Hasan
- Department of Neurosurgery, University of Iowa, Iowa City, IA 52242, USA; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-319-384-8669; Fax: +1-319-356-2237
| |
Collapse
|
33
|
CD44 targeting magnetic glyconanoparticles for atherosclerotic plaque imaging. Pharm Res 2013; 31:1426-37. [PMID: 23568520 DOI: 10.1007/s11095-013-1021-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 03/04/2013] [Indexed: 12/21/2022]
Abstract
PURPOSE The cell surface adhesion molecule CD44 plays important roles in the initiation and development of atherosclerotic plaques. We aim to develop nanoparticles that can selectively target CD44 for the non-invasive detection of atherosclerotic plaques by magnetic resonance imaging. METHODS Magnetic glyconanoparticles with hyaluronan immobilized on the surface have been prepared. The binding of these nanoparticles with CD44 was evaluated in vitro by enzyme linked immunosorbent assay, flow cytometry and confocal microscopy. In vivo magnetic resonance imaging of plaques was performed on an atherosclerotic rabbit model. RESULTS The magnetic glyconanoparticles can selectively bind CD44. In T2* weighted magnetic resonance images acquired in vivo, significant contrast changes in aorta walls were observed with a very low dose of the magnetic nanoparticles, allowing the detection of atherosclerotic plaques. Furthermore, imaging could be performed without significant delay after probe administration. The selectivity of hyaluronan nanoparticles in plaque imaging was established by several control experiments. CONCLUSIONS Magnetic nanoparticles bearing surface hyaluronan enabled the imaging of atherosclerotic plaques in vivo by magnetic resonance imaging. The low dose of nanoparticles required, the possibility to image without much delay and the high biocompatibility are the advantages of these nanoparticles as contrast agents for plaque imaging.
Collapse
|
34
|
Imaging Atherosclerotic Plaques with MRI: Role of Contrast Agents. CURRENT CARDIOVASCULAR IMAGING REPORTS 2013. [DOI: 10.1007/s12410-012-9179-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
35
|
Hyafil F, Feldman L, Le Guludec D, Fayad ZA. Evaluating Efficacy of Pharmaceutical Interventions in Atherosclerosis: Role of Magnetic Resonance Imaging and Positron Emission Tomography. ACTA ACUST UNITED AC 2012; 79:689-704. [DOI: 10.1002/msj.21349] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
36
|
Hasan D, Chalouhi N, Jabbour P, Dumont AS, Kung DK, Magnotta VA, Young WL, Hashimoto T, Winn HR, Heistad D. Early change in ferumoxytol-enhanced magnetic resonance imaging signal suggests unstable human cerebral aneurysm: a pilot study. Stroke 2012; 43:3258-65. [PMID: 23138441 DOI: 10.1161/strokeaha.112.673400] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE The clinical significance of early (ie, within the first 24 hours) uptake of ferumoxytol by macrophages in the wall of human cerebral aneurysms is not clear. The purpose of this study was to determine whether early uptake of ferumoxytol suggests unstable cerebral aneurysm. METHODS Thirty unruptured aneurysms in 22 patients were imaged with magnetic resonance imaging 24 hours after infusion of ferumoxytol. Eighteen aneurysms were also imaged 72 hours after infusion of ferumoxytol. Aneurysm dome tissue was collected from 4 patients with early magnetic resonance imaging signal changes, 5 patients with late signal changes, and 5 other patients with ruptured aneurysms. The tissue was immunostained for expression of cyclooxygenase-1, cyclooxygenase-2, microsomal prostaglandin E2 synthase-1, and macrophages. RESULTS In 23% (7/30) of aneurysms, there was pronounced early uptake of ferumoxytol. Four aneurysms were clipped. The remaining 3 aneurysms were managed conservatively; all 3 ruptured within 6 months. In 53% (16 of 30) of aneurysms, there was pronounced uptake of ferumoxytol at 72 hours. Eight aneurysms were surgically clipped, and 8 were managed conservatively; none ruptured or increased in size after 6 months. Expression of cyclooxygenase-2, microsomal prostaglandin E2 synthase-1, and macrophages was similar in unruptured aneurysms with early uptake of ferumoxytol and ruptured aneurysms. Expression of these inflammatory molecules was significantly higher in aneurysms with early uptake of ferumoxytol versus aneurysms with late uptake. CONCLUSIONS Uptake of ferumoxytol in aneurysm walls within the first 24 hours strongly suggests aneurysm instability and probability of rupture within 6 months, and may warrant urgent intervention.
Collapse
Affiliation(s)
- David Hasan
- Department of Neurosurgery, Carver College of Medicine, University of Iowa, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, JCP 1616, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Phinikaridou A, Andia ME, Shah AM, Botnar RM. Advances in molecular imaging of atherosclerosis and myocardial infarction: shedding new light on in vivo cardiovascular biology. Am J Physiol Heart Circ Physiol 2012; 303:H1397-410. [PMID: 23064836 DOI: 10.1152/ajpheart.00583.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Molecular imaging of the cardiovascular system heavily relies on the development of new imaging probes and technologies to facilitate visualization of biological processes underlying or preceding disease. Molecular imaging is a highly active research discipline that has seen tremendous growth over the past decade. It has broadened our understanding of oncologic, neurologic, and cardiovascular diseases by providing new insights into the in vivo biology of disease progression and therapeutic interventions. As it allows for the longitudinal evaluation of biological processes, it is ideally suited for monitoring treatment response. In this review, we will concentrate on the major accomplishments and advances in the field of molecular imaging of atherosclerosis and myocardial infarction with a special focus on magnetic resonance imaging.
Collapse
Affiliation(s)
- Alkystis Phinikaridou
- Division of Imaging Science and Biomedical Engineering, King's College London, United Kingdom.
| | | | | | | |
Collapse
|
38
|
Ouimet T, Lancelot E, Hyafil F, Rienzo M, Deux F, Lemaître M, Duquesnoy S, Garot J, Roques BP, Michel JB, Corot C, Ballet S. Molecular and cellular targets of the MRI contrast agent P947 for atherosclerosis imaging. Mol Pharm 2012; 9:850-61. [PMID: 22352457 DOI: 10.1021/mp2003863] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
P947 (DOTA-Gd-peptide) was recently identified as an MRI contrast agent for the detection and characterization of the matrix metalloproteinases (MMP)-rich atherosclerotic plaques. Because this product displays a broad spectrum affinity for the MMP family, we hypothesized that it may also recognize other metalloproteinases overactivated in vulnerable atherosclerotic plaques. Therefore, this study aimed at describing, at the molecular and cellular level, the interactions between P947 and proteases of atherosclerotic plaques. Fluorimetric assays were used to measure the in vitro affinity of P947 toward recombinant and purified MMPs, angiotensin-converting enzyme (ACE), endothelin-converting enzyme (ECE-1), neutral endopeptidase (NEP), and both aminopeptidases A and N (APA and APN). Using similar fluorimetric assays associated with specific substrates, enzymatic activities were measured in vulnerable and stable plaques collected from human atherosclerotic carotid arteries. Ex vivo affinity of P947 for metalloproteinases in vulnerable lesions was subsequently determined. Interaction between P947 and major cell types present in atherosclerotic plaques was also investigated in different cell lines: PMA-1-differentiated THP-1 (macrophage), Ox-LDL-treated THP-1 (foam cell), Jurkat cell line (lymphocyte), and human umbilical vein endothelial cell (HUVEC, endothelial cell). Molecular targeting of P947 was confirmed by fluorimetry, ICP-MS, and in vitro MRI approaches. Potential application of P947 for detecting atherosclerotic plaques by in vivo MRI was tested in a rabbit model of atherosclerosis. In vitro, P947 displayed affinities for purified MMPs, ACE, ECE-1, NEP, APA, and APN in the micromolar range. Interestingly, MMPs, ACE, and APN exhibited higher activities in vulnerable plaques from human atherosclerotic carotid samples, as compared to stable plaques. ECE-1, NEP, and APA had either no activity or the same low activity in both vulnerable and stable plaques. P947 showed micromolar affinities for MMPs, ACE, and APN secreted by plaque samples. Moreover, P947 bound to THP-1 macrophages and THP-1 foam cells in a concentration-dependent manner and with a higher intensity than the control contrast agents DOTA-Gd or P1135 (DOTA-Gd coupled to a scrambled peptide). In THP-1 macrophages, P947 inhibited largely (70%) and almost completely (95%) MMP and APN activities, respectively, which strongly suggested an MMP- and APN-dependent binding of P947 to these cells. This enzyme-specific binding was confirmed with in vitro MRI. Indeed, the T1 value of THP-1 cells decreased from 2.094 s (macrophages w/o P947) to 2.004 s (macrophages with 1 mM of P947). In addition, the Gd content measured by ICP-MS was 11.01 ± 1.05 fg Gd/macrophage when cells were incubated in the presence of P947 and only 5.18 ± 0.43 fg Gd/macrophage with the control product P1135. The difference of Gd concentration between both contrast agents corresponded to a specific accumulation of 5.83 fg Gd/cell, which may be detected by MRI. MR imaging in the atherosclerosis rabbit model showed enhancement of the aortic wall after P947 injection with a significant increase of CNR values from 0.21 ± 0.02 (before injection) to 0.37 ± 0.07 (after injection), demonstrating the efficacy of the contrast agent to detect the atherosclerotic plaques in vivo. Taken together, these data suggest that P947 may be an interesting contrast agent for in vivo molecular MR imaging of MMPs, ACE, and APN activities present in vulnerable atherosclerotic plaques.
Collapse
|
39
|
Hasan DM, Mahaney KB, Magnotta VA, Kung DK, Lawton MT, Hashimoto T, Winn HR, Saloner D, Martin A, Gahramanov S, Dósa E, Neuwelt E, Young WL. Macrophage imaging within human cerebral aneurysms wall using ferumoxytol-enhanced MRI: a pilot study. Arterioscler Thromb Vasc Biol 2012; 32:1032-8. [PMID: 22328774 DOI: 10.1161/atvbaha.111.239871] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Macrophages play a critical role in cerebral aneurysm formation and rupture. The purpose of this study is to demonstrate the feasibility and optimal parameters of imaging macrophages within human cerebral aneurysm wall using ferumoxytol-enhanced MRI. METHODS AND RESULTS Nineteen unruptured aneurysms in 11 patients were imaged using T2*-GE-MRI sequence. Two protocols were used. Protocol A was an infusion of 2.5 mg/kg of ferumoxytol and imaging at day 0 and 1. Protocol B was an infusion of 5 mg/kg of ferumoxytol and imaging at day 0 and 3. All images were reviewed independently by 2 neuroradiologists to assess for ferumoxytol-associated loss of MRI signal intensity within aneurysm wall. Aneurysm tissue was harvested for histological analysis. Fifty percent (5/10) of aneurysms in protocol A showed ferumoxytol-associated signal changes in aneurysm walls compared to 78% (7/9) of aneurysms in protocol B. Aneurysm tissue harvested from patients infused with ferumoxytol stained positive for both CD68+, demonstrating macrophage infiltration, and Prussian blue, demonstrating uptake of iron particles. Tissue harvested from controls stained positive for CD68 but not Prussian blue. CONCLUSIONS Imaging with T2*-GE-MRI at 72 hours postinfusion of 5 mg/kg of ferumoxytol establishes a valid and useful approximation of optimal dose and timing parameters for macrophages imaging within aneurysm wall. Further studies are needed to correlate these imaging findings with risk of intracranial aneurysm rupture.
Collapse
Affiliation(s)
- David M Hasan
- Department of Neurosurgery, Carver College of Medicine, University of Iowa, Iowa City, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Botnar RM, Makowski MR. Molecular MRI of Atherosclerosis Burden. CURRENT CARDIOVASCULAR IMAGING REPORTS 2012. [DOI: 10.1007/s12410-011-9116-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
41
|
Su HS, Nahrendorf M, Panizzi P, Breckwoldt MO, Rodriguez E, Iwamoto Y, Aikawa E, Weissleder R, Chen JW. Vasculitis: molecular imaging by targeting the inflammatory enzyme myeloperoxidase. Radiology 2011; 262:181-90. [PMID: 22084204 DOI: 10.1148/radiol.11110040] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To determine if a molecular imaging approach targeting the highly oxidative enzyme myeloperoxidase (MPO) can help noninvasively identify and confirm sites of vascular wall inflammation in a murine model of vasculitis. MATERIALS AND METHODS Animal experiments were approved by the institutional animal care committee. Twenty-six mice were studied, including eight MPO-deficient and six sham-operated mice as controls. Vasculitis was induced with intraperitoneal injection of Candida albicans water-soluble fraction (CAWS). Aortic root magnetic resonance imaging was performed after intravenous injection of the activatable MPO sensor (bis-5-hydroxytryptamide-diethylenetriaminepentatacetate gadolinium) (n = 23), referred to as MPO-Gd, or gadopentetate dimeglumine (n = 10). Seven mice were randomly assigned to receive either MPO-Gd or gadopentetate dimeglumine first. Aortic root specimens were collected for biochemical and histopathologic analyses to validate imaging findings. Statistical significance was calculated for contrast-to-noise ratios (CNRs) by using the paired t test. RESULTS In the aortic root, the mean MPO-Gd CNRs after agent injection (CNR = 28.1) were more than 2.5-fold higher than those of sham-operated mice imaged with MPO-Gd and vasculitis mice imaged with gadopentetate dimeglumine (CNR = 10.6) (P < .05). MPO-Gd MR imaging helped identify areas of vasculitis that were not seen at unenhanced and contrast material-enhanced imaging with gadopentetate dimeglumine. Histopathologic and biochemical analyses for MPO and myeloid cells confirmed imaging findings. In MPO-deficient mice, injection of CAWS did not result in a vasculitis phenotype, implying a key role of the imaging target in disease cause. CONCLUSION Molecular imaging targeting MPO can be a useful biomarker to noninvasively detect and confirm inflammation in vasculitis by using a murine model of Kawasaki disease.
Collapse
Affiliation(s)
- Henry S Su
- Center for Molecular Imaging Research, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hibino N, Yi T, Duncan DR, Rathore A, Dean E, Naito Y, Dardik A, Kyriakides T, Madri J, Pober JS, Shinoka T, Breuer CK. A critical role for macrophages in neovessel formation and the development of stenosis in tissue-engineered vascular grafts. FASEB J 2011; 25:4253-63. [PMID: 21865316 DOI: 10.1096/fj.11-186585] [Citation(s) in RCA: 174] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The primary graft-related complication during the first clinical trial evaluating the use of tissue-engineered vascular grafts (TEVGs) was stenosis. We investigated the role of macrophages in the formation of TEVG stenosis in a murine model. We analyzed the natural history of TEVG macrophage infiltration at critical time points and evaluated the role of cell seeding on neovessel formation. To assess the function of infiltrating macrophages, we implanted TEVGs into mice that had been macrophage depleted using clodronate liposomes. To confirm this, we used a CD11b-diphtheria toxin-receptor (DTR) transgenic mouse model. Monocytes infiltrated the scaffold within the first few days and initially transformed into M1 macrophages. As the scaffold degraded, the macrophage infiltrate disappeared. Cell seeding decreased the incidence of stenosis (32% seeded, 64% unseeded, P=0.024) and the degree of macrophage infiltration at 2 wk. Unseeded TEVGs demonstrated conversion from M1 to M2 phenotype, whereas seeded grafts did not. Clodronate and DTR inhibited macrophage infiltration and decreased stenosis but blocked formation of vascular neotissue, evidenced by the absence of endothelial and smooth muscle cells and collagen. These findings suggest that macrophage infiltration is critical for neovessel formation and provides a strategy for predicting, detecting, and inhibiting stenosis in TEVGs.
Collapse
Affiliation(s)
- Narutoshi Hibino
- Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Atherosclerosis is an inflammatory disease that causes most myocardial infarctions, strokes and acute coronary syndromes. Despite the identification of multiple risk factors and widespread use of drug therapies, it still remains a global health concern with associated costs. Although angiography is established as the gold standard means of detecting coronary artery stenosis, it does not image the vessel wall itself, reporting only on its consequences such as luminal narrowing and obstruction. MRI and computed tomography provide more information about the plaque structure, but recently positron emission tomography (PET) imaging using [(18) F]-fluorodeoxyglucose (FDG) has been advocated as a means of measuring arterial inflammation. This results from the ability of FDG-PET to highlight areas of high glucose metabolism, a feature of macrophages within atherosclerosis, particularly in high-risk plaques. It is suggested that the degree of FDG accumulation in the vessel wall reflects underlying inflammation levels and that tracking any changes in FDG uptake over time or with drug therapy might be a way of getting an early efficacy readout for novel anti-atherosclerotic drugs. Early reports also demonstrate that FDG uptake is correlated with the number of cardiovascular risk factors and possibly even the risk of future cardiovascular events. This review will outline the evidence base, shortcomings and emerging applications for FDG-PET in vascular imaging. Alternative PET tracers and other candidate imaging modalities for measuring vascular inflammation will also be discussed.
Collapse
Affiliation(s)
- F Joshi
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | | | | | | |
Collapse
|
44
|
Lefevre S, Ruimy D, Jehl F, Neuville A, Robert P, Sordet C, Ehlinger M, Dietemann JL, Bierry G. Septic arthritis: monitoring with USPIO-enhanced macrophage MR imaging. Radiology 2011; 258:722-8. [PMID: 21339348 DOI: 10.1148/radiol.10101272] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To prospectively evaluate in vivo noninvasive monitoring of antibiotic therapy in experimental infectious arthritis by imaging macrophages by using magnetic resonance (MR) imaging enhanced with ultrasmall superparamagnetic iron oxide (USPIO) particles. MATERIALS AND METHODS The institutional review committee on animal care approved the experimental protocol. Unilateral knee infection was induced by intra-articular injection of Staphylococcus aureus in 12 rabbits. Each rabbit underwent MR imaging before and after injection of USPIO particles, as well as before and after injection of gadoterate meglumine. All 12 of the animals were imaged during the acute phase of infection. Half were then sacrificed to obtain histopathologic samples, and the other half were imaged a second time after antibiotic treatment. MR imaging data were analyzed and compared with bacteriologic and histopathologic findings. RESULTS In acute infections, intense synovitis with marked signal intensity increase of the synovium on gadoterate dimeglumine-enhanced fat-suppressed T1-weighted images was observed in all animals and was associated with areas of signal intensity loss within the infected synovium on USPIO-enhanced T2*-weighted gradient-echo images, reflecting an intense infiltration of USPIO-loaded macrophages. After antibiotic treatment and histologic evidence of healing infection, less synovial signal intensity loss was seen (P = .03). In contradistinction, the signal intensity increase on gadoterate dimeglumine-enhanced fat-suppressed T1-weighted images remained unchanged. CONCLUSION In contrast to conventional MR imaging performed by using extracellular contrast agents, USPIO-enhanced macrophage MR imaging can demonstrate resolution of experimental bacterial joint infection.
Collapse
|
45
|
Maiseyeu A, Mihai G, Roy S, Kherada N, Simonetti OP, Sen CK, Sun Q, Parthasarathy S, Rajagopalan S. Detection of macrophages via paramagnetic vesicles incorporating oxidatively tailored cholesterol ester: an approach for atherosclerosis imaging. Nanomedicine (Lond) 2011; 5:1341-56. [PMID: 21128718 DOI: 10.2217/nnm.10.87] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
AIM Macrophages play a key role in the initiation, progression and complications of atherosclerosis. In this article we describe the synthesis of biocompatible, paramagnetic, fluorescent phosphatidylserine vesicles containing cholesterol ester with a free carboxylic acid function and its use for targeted imaging of macrophages. METHODS & RESULTS We synthesized anionic vesicles containing a combination of phosphatidylserine and a novel synthetic oxidized cholesterol ester derivative (cholesterol-9-carboxynonanoate [9-CCN]). In vitro studies to characterize particle size, MRI relaxation times and stability were performed. Vesicles containing 9-CCN demonstrated enhanced ability to bind human low-density lipoprotein and to be internalized by macrophages. Experiments in cultured macrophages with 9-CCN vesicles, alone and in the presence of low-density lipoprotein, indicated uptake of vesicles through scavenger receptor and integrin-dependent pathways. In vivo MRI using 9-CCN vesicles containing gadolinium in a rabbit model of atherosclerosis revealed protracted enhancement of 9-CCN vesicles and colocalization with arterial macrophages not seen with control vesicles. Pharmacokinetic experiments demonstrated prolonged plasma residence time of 9-CCN vesicles, perhaps due to its capacity to bind to low-density lipoprotein. CONCLUSION Vesicles containing 9-CCN demonstrate prolonged plasma and plaque retention in experimental atherosclerosis. Such a strategy may represent a simple yet clinically relevant approach for macrophage imaging.
Collapse
Affiliation(s)
- Andrei Maiseyeu
- Davis Heart & Lung Research Institute, Room 110, 473 W 12th Avenue, Columbus, OH 43210-1252, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mahmoudi M, Sahraian MA, Shokrgozar MA, Laurent S. Superparamagnetic iron oxide nanoparticles: promises for diagnosis and treatment of multiple sclerosis. ACS Chem Neurosci 2011; 2:118-40. [PMID: 22778862 PMCID: PMC3369738 DOI: 10.1021/cn100100e] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 12/08/2010] [Indexed: 12/15/2022] Open
Abstract
Smart superparamagnetic iron oxide nanoparticles (SPIONs) are the most promising candidate for theragnosis (i.e., diagnosis and treatment) of multiple sclerosis. A deep understanding of the dynamics of the in vivo neuropathology of multiple sclerosis can be achieved by improving the efficiency of various medical techniques (e.g., positron emission tomography and magnetic resonance imaging) using multimodal SPIONs. In this Review, recent advances and challenges in the development of smart SPIONs for theragnostic applications are comprehensively described. In addition, critical outlines of emerging developments are provided from the points of view of both clinicians and nanotechnologists.
Collapse
Affiliation(s)
- Morteza Mahmoudi
- National Cell Bank, Pasteur Institute of Iran, Tehran, 11365-8639, Iran.
| | | | | | | |
Collapse
|
47
|
Kosuge H, Sherlock SP, Kitagawa T, Terashima M, Barral JK, Nishimura DG, Dai H, McConnell MV. FeCo/graphite nanocrystals for multi-modality imaging of experimental vascular inflammation. PLoS One 2011; 6:e14523. [PMID: 21264237 PMCID: PMC3021517 DOI: 10.1371/journal.pone.0014523] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 12/16/2010] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND FeCo/graphitic-carbon nanocrystals (FeCo/GC) are biocompatible, high-relaxivity, multi-functional nanoparticles. Macrophages represent important cellular imaging targets for assessing vascular inflammation. We evaluated FeCo/GC for vascular macrophage uptake and imaging in vivo using fluorescence and MRI. METHODS AND RESULTS Hyperlipidemic and diabetic mice underwent carotid ligation to produce a macrophage-rich vascular lesion. In situ and ex vivo fluorescence imaging were performed at 48 hours after intravenous injection of FeCo/GC conjugated to Cy5.5 (n = 8, 8 nmol of Cy5.5/mouse). Significant fluorescence signal from FeCo/GC-Cy5.5 was present in the ligated left carotid arteries, but not in the control (non-ligated) right carotid arteries or sham-operated carotid arteries (p = 0.03 for ligated vs. non-ligated). Serial in vivo 3T MRI was performed at 48 and 72 hours after intravenous FeCo/GC (n = 6, 270 µg Fe/mouse). Significant T2* signal loss from FeCo/GC was seen in ligated left carotid arteries, not in non-ligated controls (p = 0.03). Immunofluorescence staining showed colocalization of FeCo/GC and macrophages in ligated carotid arteries. CONCLUSIONS FeCo/GC accumulates in vascular macrophages in vivo, allowing fluorescence and MR imaging. This multi-functional high-relaxivity nanoparticle platform provides a promising approach for cellular imaging of vascular inflammation.
Collapse
Affiliation(s)
- Hisanori Kosuge
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
| | - Sarah P. Sherlock
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Toshiro Kitagawa
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
| | - Masahiro Terashima
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
| | - Joëlle K. Barral
- Electrical Engineering, Stanford University, Stanford, California, United States of America
| | - Dwight G. Nishimura
- Electrical Engineering, Stanford University, Stanford, California, United States of America
| | - Hongjie Dai
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Michael V. McConnell
- Division of Cardiovascular Medicine, Stanford University, Stanford, California, United States of America
- Electrical Engineering, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
48
|
Molecular imaging of atherosclerosis in translational medicine. Eur J Nucl Med Mol Imaging 2010; 38:969-75. [DOI: 10.1007/s00259-010-1697-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 11/29/2010] [Indexed: 10/18/2022]
|
49
|
Hyafil F, Vucic E, Cornily JC, Sharma R, Amirbekian V, Blackwell F, Lancelot E, Corot C, Fuster V, Galis ZS, Feldman LJ, Fayad ZA. Monitoring of arterial wall remodelling in atherosclerotic rabbits with a magnetic resonance imaging contrast agent binding to matrix metalloproteinases. Eur Heart J 2010; 32:1561-71. [PMID: 21118852 DOI: 10.1093/eurheartj/ehq413] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
AIMS P947 is a gadolinium-based magnetic resonance imaging (MRI) contrast agent with high affinity for several matrix metalloproteinases (MMPs) involved in arterial wall remodelling. We tested whether the intensity of enhancement detected in vivo in the arterial wall with P947 and MRI correlates with actual tissue MMP-related enzymatic activity measured in a rabbit atherosclerotic model subjected to dietary manipulations. METHODS AND RESULTS Aortas of 15 rabbits in which atherosclerotic lesions were induced by balloon angioplasty and 4 months of hypercholesterolaemic diet were imaged at 'baseline' with P947-enhanced MRI. Atherosclerotic rabbits were divided into three groups: five rabbits were sacrificed ('baseline' group); five rabbits continued to be fed a lipid-supplemented diet ('high-fat' group); and five rabbits were switched from atherogenic to a purified chow diet ('low-fat' group). Four months later, a second P947-enhanced MRI was acquired in the 10 remaining rabbits. A significantly lower signal was detected in the aortic wall of rabbits from the 'low-fat' group as compared with rabbits from the 'high-fat' group (21 ± 6 vs. 46 ± 3%, respectively; P = 0.04). Such differences were not detected with the contrast agent P1135, which lacks the MMP-specific peptide sequence. In addition, the intensity of aortic wall enhancement detected with MRI after injection of P947 strongly correlated with actual MMP-2 gelatinolytic activity measured in corresponding aortic segments using zymography (r = 0.87). CONCLUSION P947-enhanced MRI can distinguish dietary-induced variations in MMP-related enzymatic activity within plaques in an experimental atherosclerotic model, supporting its utility as a clinical imaging tool for in vivo detection of arterial wall remodelling.
Collapse
Affiliation(s)
- Fabien Hyafil
- Translational and Molecular Imaging Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Chen W, Cormode DP, Fayad ZA, Mulder WJM. Nanoparticles as magnetic resonance imaging contrast agents for vascular and cardiac diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2010; 3:146-161. [PMID: 20967875 DOI: 10.1002/wnan.114] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Advances in nanoparticle contrast agents for molecular imaging have made magnetic resonance imaging a promising modality for noninvasive visualization and assessment of vascular and cardiac disease processes. This review provides a description of the various nanoparticles exploited for imaging cardiovascular targets. Nanoparticle probes detecting inflammation, apoptosis, extracellular matrix, and angiogenesis may provide tools for assessing the risk of progressive vascular dysfunction and heart failure. The utility of nanoparticles as multimodal probes and/or theranostic agents has also been investigated. Although clinical application of these nanoparticles is largely unexplored, the potential for enhancing disease diagnosis and treatment is considerable.
Collapse
Affiliation(s)
- Wei Chen
- Translational and Molecular Imaging Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - David P Cormode
- Translational and Molecular Imaging Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Mount Sinai School of Medicine, New York, NY, USA.,Department of Radiology, Mount Sinai School of Medicine, New York, NY, USA
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Mount Sinai School of Medicine, New York, NY, USA.,Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|