1
|
Ullah A, Ullah M, Lim SI. Recent advancements in nanotechnology based drug delivery for the management of cardiovascular disease. Curr Probl Cardiol 2024; 49:102396. [PMID: 38266693 DOI: 10.1016/j.cpcardiol.2024.102396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Cardiovascular diseases (CVDs) constitute a predominant cause of both global mortality and morbidity. To address the challenges in the early diagnosis and management of CVDs, there is growing interest in the field of nanotechnology and nanomaterials to develop innovative diagnostic and therapeutic approaches. This review focuses on the recent advancements in nanotechnology-based diagnostic techniques, including cardiac immunoassays (CIA), cardiac circulating biomarkers, cardiac exosomal biomarkers, and molecular Imaging (MOI). Moreover, the article delves into the exciting developments in nanoparticles (NPs), biomimetic NPs, nanofibers, nanogels, and nanopatchs for cardiovascular applications. And discuss how these nanoscale technologies can improve the precision, sensitivity, and speed of CVD diagnosis and management. While highlighting their vast potential, we also address the limitations and challenges that must be overcome to harness these innovations successfully. Furthermore, this review focuses on the emerging opportunities for personalized and effective cardiovascular care through the integration of nanotechnology, ultimately aiming to reduce the global burden of CVDs.
Collapse
Affiliation(s)
- Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea
| | - Muneeb Ullah
- College of Pharmacy, Pusan National University, Busandaehak-ro 63 beon-gil 2, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea.
| |
Collapse
|
2
|
Tiwari A, Haj N, Elgrably B, Berihu M, Laskov V, Barash S, Zigron S, Sason H, Shamay Y, Karni-Ashkenazi S, Holdengreber M, Saar G, Vandoorne K. Cross-Modal Imaging Reveals Nanoparticle Uptake Dynamics in Hematopoietic Bone Marrow during Inflammation. ACS NANO 2024; 18:7098-7113. [PMID: 38343099 PMCID: PMC10919094 DOI: 10.1021/acsnano.3c11201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 03/06/2024]
Abstract
Nanoparticles have been employed to elucidate the innate immune cell biology and trace cells accumulating at inflammation sites. Inflammation prompts innate immune cells, the initial responders, to undergo rapid turnover and replenishment within the hematopoietic bone marrow. Yet, we currently lack a precise understanding of how inflammation affects cellular nanoparticle uptake at the level of progenitors of innate immune cells in the hematopoietic marrow. To bridge this gap, we aimed to develop imaging tools to explore the uptake dynamics of fluorescently labeled cross-linked iron oxide nanoparticles in the bone marrow niche under varying degrees of inflammation. The inflammatory models included mice that received intramuscular lipopolysaccharide injections to induce moderate inflammation and streptozotocin-induced diabetic mice with additional intramuscular lipopolysaccharide injections to intensify inflammation. In vivo magnetic resonance imaging (MRI) and fluorescence imaging revealed an elevated level of nanoparticle uptake at the bone marrow as the levels of inflammation increased. The heightened uptake of nanoparticles within the inflamed marrow was attributed to enhanced permeability and retention with increased nanoparticle intake by hematopoietic progenitor cells. Moreover, intravital microscopy showed increased colocalization of nanoparticles within slowly patrolling monocytes in these inflamed hematopoietic marrow niches. Our discoveries unveil a previously unknown role of the inflamed hematopoietic marrow in enhanced storage and rapid deployment of nanoparticles, which can specifically target innate immune cells at their production site during inflammation. These insights underscore the critical function of the hematopoietic bone marrow in distributing iron nanoparticles to innate immune cells during inflammation. Our findings offer diagnostic and prognostic value, identifying the hematopoietic bone marrow as an imaging biomarker for early detection in inflammation imaging, advancing personalized clinical care.
Collapse
Affiliation(s)
- Ashish Tiwari
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Narmeen Haj
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Betsalel Elgrably
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Maria Berihu
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Viktor Laskov
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
- Third
Faculty of Medicine, Charles University, Prague 100 00, Czech Republic
| | - Sivan Barash
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Shachar Zigron
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Hagit Sason
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Yosi Shamay
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Shiri Karni-Ashkenazi
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Maya Holdengreber
- Biomedical
Core Facility, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Galit Saar
- Biomedical
Core Facility, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Katrien Vandoorne
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
3
|
Smith BR, Edelman ER. Nanomedicines for cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:351-367. [PMID: 39195953 DOI: 10.1038/s44161-023-00232-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 01/25/2023] [Indexed: 08/29/2024]
Abstract
The leading cause of death in the world, cardiovascular disease (CVD), remains a formidable condition for researchers, clinicians and patients alike. CVD comprises a broad collection of diseases spanning the heart, the vasculature and the blood that runs through and interconnects them. Limitations in CVD therapeutic and diagnostic landscapes have generated excitement for advances in nanomedicine, a field focused on improving patient outcomes through transformative therapies, imaging agents and ex vivo diagnostics. CVD nanomedicines are fundamentally shaped by their intended clinical application, including (1) cardiac or heart-related biomaterials, which can be functionally (for example, mechanically, immunologically, electrically) improved by incorporating nanomaterials; (2) the vasculature, involving systemically injected nanotherapeutics and imaging nanodiagnostics, nano-enabled biomaterials or tissue-nanoengineered solutions; and (3) improving the sensitivity and/or specificity of ex vivo diagnostic devices for patient samples. While immunotherapy has developed into a key pillar of oncology in the past dozen years, CVD immunotherapy and immunoimaging are recently emergent and likely to factor substantially in CVD management in the coming decade. The nanomaterials in CVD-related clinical trials and many promising preclinical strategies indicate that nanomedicine is on the cusp of greatly impacting patients with CVD. Here we review these recent advances, highlighting key clinical opportunities in the rapidly emerging field of CVD nanomedicine.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Polyelectrolyte Coating of Ferumoxytol Differentially Impacts the Labeling of Inflammatory and Steady-State Dendritic Cell Subtypes. Biomedicines 2022; 10:biomedicines10123137. [PMID: 36551893 PMCID: PMC9776020 DOI: 10.3390/biomedicines10123137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Engineered magnetic nanoparticles (MNPs) are emerging as advanced tools for medical applications. The coating of MNPs using polyelectrolytes (PEs) is a versatile means to tailor MNP properties and is used to optimize MNP functionality. Dendritic cells (DCs) are critical regulators of adaptive immune responses. Functionally distinct DC subsets exist, either under steady-state or inflammatory conditions, which are explored for the specific treatment of various diseases, such as cancer, autoimmunity, and transplant rejection. Here, the impact of the PE coating of ferumoxytol for uptake into both inflammatory and steady-state DCs and the cellular responses to MNP labeling is addressed. Labeling efficiency by uncoated and PE-coated ferumoxytol is highly variable in different DC subsets, and PE coating significantly improves the labeling of steady-state DCs. Uncoated ferumoxytol results in increased cytotoxicity of steady-state DCs after labeling, which is abolished by the PE coating, while no increased cell death is observed in inflammatory DCs. Furthermore, uncoated and PE-coated ferumoxytol appear immunologically inert in inflammatory DCs, but they induce activation of steady-state DCs. These results show that the PE coating of MNPs can be applied to endow particles with desired properties for enhanced uptake and cell type-specific responses in distinct target DC populations.
Collapse
|
5
|
Ammirati E, Bizzi E, Veronese G, Groh M, Van de Heyning CM, Lehtonen J, Pineton de Chambrun M, Cereda A, Picchi C, Trotta L, Moslehi JJ, Brucato A. Immunomodulating Therapies in Acute Myocarditis and Recurrent/Acute Pericarditis. Front Med (Lausanne) 2022; 9:838564. [PMID: 35350578 PMCID: PMC8958011 DOI: 10.3389/fmed.2022.838564] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/28/2022] [Indexed: 12/15/2022] Open
Abstract
The field of inflammatory disease of the heart or "cardio-immunology" is rapidly evolving due to the wider use of non-invasive diagnostic tools able to detect and monitor myocardial inflammation. In acute myocarditis, recent data on the use of immunomodulating therapies have been reported both in the setting of systemic autoimmune disorders and in the setting of isolated forms, especially in patients with specific histology (e.g., eosinophilic myocarditis) or with an arrhythmicburden. A role for immunosuppressive therapies has been also shown in severe cases of coronavirus disease 2019 (COVID-19), a condition that can be associated with cardiac injury and acute myocarditis. Furthermore, ongoing clinical trials are assessing the role of high dosage methylprednisolone in the context of acute myocarditis complicated by heart failure or fulminant presentation or the role of anakinra to treat patients with acute myocarditis excluding patients with hemodynamically unstable conditions. In addition, the explosion of immune-mediated therapies in oncology has introduced new pathophysiological entities, such as immune-checkpoint inhibitor-associated myocarditis and new basic research models to understand the interaction between the cardiac and immune systems. Here we provide a broad overview of evolving areas in cardio-immunology. We summarize the use of new imaging tools in combination with endomyocardial biopsy and laboratory parameters such as high sensitivity troponin to monitor the response to immunomodulating therapies based on recent evidence and clinical experience. Concerning pericarditis, the normal composition of pericardial fluid has been recently elucidated, allowing to assess the actual presence of inflammation; indeed, normal pericardial fluid is rich in nucleated cells, protein, albumin, LDH, at levels consistent with inflammatory exudates in other biological fluids. Importantly, recent findings showed how innate immunity plays a pivotal role in the pathogenesis of recurrent pericarditis with raised C-reactive protein, with inflammasome and IL-1 overproduction as drivers for systemic inflammatory response. In the era of tailored medicine, anti-IL-1 agents such as anakinra and rilonacept have been demonstrated highly effective in patients with recurrent pericarditis associated with an inflammatory phenotype.
Collapse
Affiliation(s)
- Enrico Ammirati
- De Gasperis Cardio Center and Transplant Center, Niguarda Hospital, Milano, Italy
| | - Emanuele Bizzi
- Internal Medicine, Fatebenefratelli Hospital, Milano, Italy
| | - Giacomo Veronese
- Department of Health Sciences, University of Milano-Bicocca, Monza, Italy
| | - Matthieu Groh
- National Reference Center for Hypereosinophilic Syndromes, CEREO, Suresnes, France
- Department of Internal Medicine, Hôpital Foch, Suresnes, France
| | - Caroline M. Van de Heyning
- Department of Cardiology, Antwerp University Hospital, and GENCOR Research Group, Antwerp University, Antwerp, Belgium
| | - Jukka Lehtonen
- Department of Cardiology, Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Marc Pineton de Chambrun
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (APHP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive-Réanimation, Paris, France
- Sorbonne Université, APHP, Hôpital de la Pitié-Salpêtrière, Service de Médecine Interne 2, Centre de Référence National Lupus et SAPL et Autres Maladies Auto-immunes et Systémiques Rares, Paris, France
- Sorbonne Université, INSERM, UMRS_1166-ICAN, ICAN, Paris, France
| | - Alberto Cereda
- Cardiovascular Department, Association Socio Sanitary Territorial Santi Paolo e Carlo, Milano, Italy
| | - Chiara Picchi
- Internal Medicine, Fatebenefratelli Hospital, Milano, Italy
| | - Lucia Trotta
- Internal Medicine, Fatebenefratelli Hospital, Milano, Italy
| | - Javid J. Moslehi
- Section of Cardio-Oncology and Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Antonio Brucato
- Internal Medicine, Fatebenefratelli Hospital, Milano, Italy
- Department of Biomedical and Clinical Sciences “Luigi Sacco, ” Fatebenefratelli Hospital, University of Milano, Milano, Italy
| |
Collapse
|
6
|
Kim CW, Hwang BH, Moon H, Kang J, Park EH, Ihm SH, Chang K, Hong KS. In vivo MRI detection of intraplaque macrophages with biocompatible silica-coated iron oxide nanoparticles in murine atherosclerosis. J Appl Biomater Funct Mater 2021; 19:22808000211014751. [PMID: 34520279 DOI: 10.1177/22808000211014751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Identification of a vulnerable atherosclerotic plaque before rupture is an unmet clinical need. Integrating nanomedicine with multimodal imaging has the potential to precisely detect biological processes in atherosclerosis. We synthesized silica-coated iron oxide nanoparticles (SIONs) coated with rhodamine B isothiocyanate and polyethylene glycol and investigated their feasibility in the detection of macrophages in inflamed atherosclerotic plaques of apolipoprotein E-deficient (ApoE-/-) mice via magnetic resonance (MR) and fluorescence reflectance (FR) imaging. In vitro cellular uptake of SIONs was assessed in macrophages using confocal laser scanning microscopy (CLSM). In vivo MR imaging was performed 24 h after SION injection via the tail vein in 26-week-old ApoE-/- mice fed a high-cholesterol diet (HCD). We also performed FR imaging of the extracted aortas from four different mice: two normal-diet-fed C57BL/6 mice injected with saline or 10 mg/kg SIONs and two HCD-fed ApoE-/- mice injected with 5 or 10 mg/kg SIONs. The harvested aortas were cryosectioned and stained with immunohistochemical staining. The CLSM images at 24 h after incubation showed efficient uptake of SIONs by macrophages, with no evidence of cytotoxicity. The in vivo and ex vivo MR and FR images demonstrated SION deposition in the atheroma. Upon immunohistochemical staining of the aorta, CLSM images revealed colocalization of macrophages and SIONs in the atherosclerotic plaque. These results demonstrate that polyethylene glycosylated SIONs could be a highly effective method to identify macrophage activity in atherosclerotic plaques as a multimodal imaging agent.
Collapse
Affiliation(s)
- Chan Woo Kim
- Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Byung-Hee Hwang
- Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyeyoung Moon
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - Jongeun Kang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Republic of Korea.,Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Hye Park
- Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang-Hyun Ihm
- Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Cardiology, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Kiyuk Chang
- Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kwan Soo Hong
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Republic of Korea.,Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
7
|
Merinopoulos I, Gunawardena T, Stirrat C, Cameron D, Eccleshall SC, Dweck MR, Newby DE, Vassiliou VS. Diagnostic Applications of Ultrasmall Superparamagnetic Particles of Iron Oxide for Imaging Myocardial and Vascular Inflammation. JACC Cardiovasc Imaging 2021; 14:1249-1264. [PMID: 32861658 DOI: 10.1016/j.jcmg.2020.06.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/22/2020] [Accepted: 06/04/2020] [Indexed: 01/03/2023]
Abstract
Cardiac magnetic resonance (CMR) is at the forefront of noninvasive methods for the assessment of myocardial anatomy, function, and most importantly tissue characterization. The role of CMR is becoming even more significant with an increasing recognition that inflammation plays a major role for various myocardial diseases such as myocardial infarction, myocarditis, and takotsubo cardiomyopathy. Ultrasmall superparamagnetic particles of iron oxide (USPIO) are nanoparticles that are taken up by monocytes and macrophages accumulating at sites of inflammation. In this context, USPIO-enhanced CMR can provide valuable additional information regarding the cellular inflammatory component of myocardial and vascular diseases. Here, we will review the recent diagnostic applications of USPIO in terms of imaging myocardial and vascular inflammation, and highlight some of their future potential.
Collapse
Affiliation(s)
- Ioannis Merinopoulos
- Norwich Medical School, University of East Anglia, Norfolk and Norwich University Hospital, Norwich, United Kingdom; Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Tharusha Gunawardena
- Norwich Medical School, University of East Anglia, Norfolk and Norwich University Hospital, Norwich, United Kingdom; Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Colin Stirrat
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Donnie Cameron
- Norwich Medical School, University of East Anglia, Norfolk and Norwich University Hospital, Norwich, United Kingdom; C.J. Gorter Centre for High Field MRI, Department of Radiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Simon C Eccleshall
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Marc R Dweck
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - David E Newby
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Vassilios S Vassiliou
- Norwich Medical School, University of East Anglia, Norfolk and Norwich University Hospital, Norwich, United Kingdom; Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom.
| |
Collapse
|
8
|
Lu Y, Huang J, Neverova NV, Nguyen KL. USPIOs as targeted contrast agents in cardiovascular magnetic resonance imaging. CURRENT CARDIOVASCULAR IMAGING REPORTS 2021; 14:2. [PMID: 33824694 PMCID: PMC8021129 DOI: 10.1007/s12410-021-09552-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 01/15/2023]
Abstract
PURPOSE OF REVIEW We aim to discuss the diagnostic use of ultra-small superparamagnetic iron oxide (USPIOs) including ferumoxytol in targeted cardiovascular magnetic resonance imaging (MRI). RECENT FINDINGS Ferumoxytol is the only USPIO clinically available in the U.S. and is a negatively charged USPIO that has potential use for tracking and characterization of macrophage-infiltrated cardiovascular structures. As an iron supplement that is approved for treatment of iron deficiency anemia, the iron core of ferumoxytol is incorporated into the body once it is phagocytosed by macrophages. In organs or tissues with high inflammatory cellular infiltration, such as atherosclerotic plaques and myocardial infarction, localization of iron-laden macrophages can be visualized on delayed MRI. The iron core of ferumoxytol alters the magnetic susceptibility and results in shortening of T2* and T2 relaxation rates. Areas with high concentration appear hypointense (negative contrast) on T2 and T2* MRI. Recently, in vitro findings support the potential specificity of ferumoxytol interactions with macrophage subtypes, which has implications for therapeutic interventions. With increasing concerns about gadolinium retention in the brain and other tissues, the value of ferumoxytol-enhanced MR for targeted clinical imaging is aided by its positive safety profile in patients with impaired renal function. SUMMARY This paper discusses pharmacokinetic properties of USPIOs with a focus on ferumoxytol, and summarizes relevant in vitro, animal, and human studies investigating the diagnostic use of USPIOs in targeted contrast-enhanced imaging. We also discuss future directions for USPIOs as targeted imaging agents and associated challenges.
Collapse
Affiliation(s)
- Yi Lu
- Division of Cardiology, David Geffen School of Medicine at
UCLA and VA Greater Los Angeles Healthcare System
| | - Jenny Huang
- Division of Cardiology, David Geffen School of Medicine at
UCLA and VA Greater Los Angeles Healthcare System
- Diagnostic Cardiovascular Imaging Research Laboratory,
Department of Radiology, David Geffen School of Medicine at UCLA
| | - Natalia V. Neverova
- Division of Cardiology, David Geffen School of Medicine at
UCLA and VA Greater Los Angeles Healthcare System
| | - Kim-Lien Nguyen
- Division of Cardiology, David Geffen School of Medicine at
UCLA and VA Greater Los Angeles Healthcare System
- Physics and Biology in Medicine Graduate Program,
University of California, Los Angeles
- Diagnostic Cardiovascular Imaging Research Laboratory,
Department of Radiology, David Geffen School of Medicine at UCLA
| |
Collapse
|
9
|
Li H, Zeng Y, Zhang H, Gu Z, Gong Q, Luo K. Functional gadolinium-based nanoscale systems for cancer theranostics. J Control Release 2020; 329:482-512. [PMID: 32898594 DOI: 10.1016/j.jconrel.2020.08.064] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/25/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023]
Abstract
Cancer theranostics is a new strategy for combating cancer that integrates cancer imaging and treatment through theranostic agents to provide an efficient and safe way to improve cancer prognosis. Design and synthesis of these cancer theranostic agents are crucial since these agents are required to be biocompatible, tumor-specific, imaging distinguishable and therapeutically efficacious. In this regard, several types of gadolinium (Gd)-based nanomaterials have been introduced to combine different therapeutic agents with Gd to enhance the efficacy of therapeutic agents. At the same time, the entire treatment procedure could be monitored via imaging tools due to incorporation of Gd ions, Gd chelates and Gd/other imaging probes in the theranostic agents. This review aims to overview recent advances in the Gd-based nanomaterials for cancer theranostics and perspectives for Gd nanomaterial-based cancer theranostics are provided.
Collapse
Affiliation(s)
- Haonan Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yujun Zeng
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA 91711, USA
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
10
|
Wang P, Kim T, Harada M, Contag C, Huang X, Smith BR. Nano-immunoimaging. NANOSCALE HORIZONS 2020; 5:628-653. [PMID: 32226975 DOI: 10.1039/c9nh00514e] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Immunoimaging is a rapidly growing field stoked in large part by the intriguing triumphs of immunotherapy. On the heels of immunotherapy's successes, there exists a growing need to evaluate tumor response to therapy particularly immunotherapy, stratify patients into responders vs. non-responders, identify inflammation, and better understand the fundamental roles of immune system components to improve both immunoimaging and immunotherapy. Innovative nanomaterials have begun to provide novel opportunities for immunoimaging, in part due to their sensitivity, modularity, capacity for many potentially varied ligands (high avidity), and potential for multifunctionality/multimodality imaging. This review strives to comprehensively summarize the integration of nanotechnology and immunoimaging, and the field's potential for clinical applications.
Collapse
Affiliation(s)
- Ping Wang
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Precision Health Program, Michigan State University, East Lansing, MI 488824, USA
| | - Taeho Kim
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA
| | - Masako Harada
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA
| | - Christopher Contag
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Precision Health Program, Michigan State University, East Lansing, MI 488824, USA and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI 488824, USA
| | - Xuefei Huang
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Chemistry, Michigan State University, East Lansing, MI 488824, USA
| | - Bryan Ronain Smith
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Radiology, Stanford University, Stanford, CA 94306, USA
| |
Collapse
|
11
|
Choi S, Chung JH, Nam MH, Bang E, Hong KS, Kim YH, Seo JB, Chi SG. Elevated aldolase 1A, retrogene 1 expression induces cardiac apoptosis in rat experimental autoimmune myocarditis model. Can J Physiol Pharmacol 2020; 98:373-382. [PMID: 31999472 DOI: 10.1139/cjpp-2019-0539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Acute myocarditis is an unpredictable heart disease that is caused by inflammation-associated cell death. Although viral infection and drug exposure are known to induce acute myocarditis, the molecular basis for its development remains undefined. Using proteomics and molecular analyses in myosin-induced rat experimental autoimmune myocarditis (EAM), we identified that elevated expression of aldolase 1A, retrogene 1 (Aldoart1) is critical to induce mitochondrial dysfunction and acute myocarditis development. Here, we demonstrate that cardiac cell death is associated with increased expressions of proapoptotic genes in addition to high levels of glucose, lactate, and triglyceride in metabolite profiling. The functional protein association network analysis also suggests that Aldoart1 upregulation correlates with high levels of dihydroxyacetone kinase and triglyceride. In H9c2 cardiac cells, lipopolysaccharides (LPS) or high glucose exposure significantly increases the cytochrome c release and the conversion of pro-caspase 3 into the cleaved form of caspase 3. We also found that LPS- or glucose-induced toxicities are almost completely reversed by siRNA-mediated knockdown of Aldoartl, which consequently increases cell viability. Together, our study strongly suggests that Aldoart1 may be involved in inducing mitochondrial apoptotic processes and can be a novel therapeutic target to prevent the onset of acute myocarditis or cardiac apoptosis.
Collapse
Affiliation(s)
- Seungmin Choi
- Department of Life Sciences, Korea University, Seoul 02841, Korea.,Korea Basic Science Institute, Seoul Center, Seoul 02841, Korea
| | - Joo Hee Chung
- Korea Basic Science Institute, Seoul Center, Seoul 02841, Korea
| | - Myung-Hee Nam
- Korea Basic Science Institute, Seoul Center, Seoul 02841, Korea
| | - Eunjung Bang
- Korea Basic Science Institute, Western Seoul Center, Seoul 03759, Korea
| | - Kwan Soo Hong
- Korea Basic Science Institute, Bioimaging Research Team, Cheongju 28123, Korea
| | - Yong-Hwan Kim
- Department of Biological Sciences, Delaware State University, Dover, DE 19901, USA
| | - Jong Bok Seo
- Korea Basic Science Institute, Seoul Center, Seoul 02841, Korea
| | - Sung-Gil Chi
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| |
Collapse
|
12
|
Tada Y, Tachibana A, Heidary S, Yang PC, McConnell MV, Dash R. Ferumoxytol-enhanced cardiovascular magnetic resonance detection of early stage acute myocarditis. J Cardiovasc Magn Reson 2019; 21:77. [PMID: 31842900 PMCID: PMC6913003 DOI: 10.1186/s12968-019-0587-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 11/21/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The diagnostic utility of cardiovascular magnetic resonance (CMR) is limited during the early stages of myocarditis. This study examined whether ferumoxytol-enhanced CMR (FE-CMR) could detect an earlier stage of acute myocarditis compared to gadolinium-enhanced CMR. METHODS Lewis rats were induced to develop autoimmune myocarditis. CMR (3 T, GE Signa) was performed at the early- (day 14, n = 7) and the peak-phase (day 21, n = 8) of myocardial inflammation. FE-CMR was evaluated as % myocardial dephasing signal loss on gradient echo images at 6 and 24 h (6 h- & 24 h-FE-CMR) following the administration of ferumoxytol (300μmolFe/kg). Pre- and post-contrast T2* mapping was also performed. Early (EGE) and late (LGE) gadolinium enhancement was obtained after the administration of gadolinium-DTPA (0.5 mmol/kg) on day 14 and 21. Healthy rats were used as control (n = 6). RESULTS Left ventricular ejection fraction (LVEF) was preserved at day 14 with inflammatory cells but no fibrosis seen on histology. EGE and LGE at day 14 both showed limited myocardial enhancement (EGE: 11.7 ± 15.5%; LGE: 8.7 ± 8.7%; both p = ns vs. controls). In contrast, 6 h-FE-CMR detected extensive myocardial signal loss (33.2 ± 15.0%, p = 0.02 vs. EGE and p < 0.01 vs. LGE). At day 21, LVEF became significantly decreased (47.4 ± 16.4% vs control: 66.2 ± 6.1%, p < 0.01) with now extensive myocardial involvement detected on EGE, LGE, and 6 h-FE-CMR (41.6 ± 18.2% of LV). T2* mapping also detected myocardial uptake of ferumoxytol both at day 14 (6 h R2* = 299 ± 112 s- 1vs control: 125 ± 26 s- 1, p < 0.01) and day 21 (564 ± 562 s- 1, p < 0.01 vs control). Notably, the myocardium at peak-phase myocarditis also showed significantly higher pre-contrast T2* (27 ± 5 ms vs control: 16 ± 1 ms, p < 0.001), and the extent of myocardial necrosis had a strong positive correlation with T2* (r = 0.86, p < 0.001). CONCLUSIONS FE-CMR acquired at 6 h enhance detection of early stages of myocarditis before development of necrosis or fibrosis, which could potentially enable appropriate therapeutic intervention.
Collapse
Affiliation(s)
- Yuko Tada
- Department of Medicine (Cardiovascular Medicine), Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305 USA
| | - Atsushi Tachibana
- Department of Medicine (Cardiovascular Medicine), Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305 USA
| | - Shahriar Heidary
- Department of Medicine (Cardiovascular Medicine), Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305 USA
| | - Phillip C. Yang
- Department of Medicine (Cardiovascular Medicine), Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305 USA
| | - Michael V. McConnell
- Department of Medicine (Cardiovascular Medicine), Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305 USA
| | - Rajesh Dash
- Department of Medicine (Cardiovascular Medicine), Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305 USA
| |
Collapse
|
13
|
Vacchini M, Edwards R, Guizzardi R, Palmioli A, Ciaramelli C, Paiotta A, Airoldi C, La Ferla B, Cipolla L. Glycan Carriers As Glycotools for Medicinal Chemistry Applications. Curr Med Chem 2019; 26:6349-6398. [DOI: 10.2174/0929867326666190104164653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 11/07/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022]
Abstract
Carbohydrates are one of the most powerful and versatile classes of biomolecules that nature
uses to regulate organisms’ biochemistry, modulating plenty of signaling events within cells, triggering
a plethora of physiological and pathological cellular behaviors. In this framework, glycan carrier
systems or carbohydrate-decorated materials constitute interesting and relevant tools for medicinal
chemistry applications. In the last few decades, efforts have been focused, among others, on the development
of multivalent glycoconjugates, biosensors, glycoarrays, carbohydrate-decorated biomaterials
for regenerative medicine, and glyconanoparticles. This review aims to provide the reader with a general
overview of the different carbohydrate carrier systems that have been developed as tools in different
medicinal chemistry approaches relying on carbohydrate-protein interactions. Given the extent of
this topic, the present review will focus on selected examples that highlight the advancements and potentialities
offered by this specific area of research, rather than being an exhaustive literature survey of
any specific glyco-functionalized system.
Collapse
Affiliation(s)
- Mattia Vacchini
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Rana Edwards
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Roberto Guizzardi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Alessandro Palmioli
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Carlotta Ciaramelli
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Alice Paiotta
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Cristina Airoldi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Barbara La Ferla
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Laura Cipolla
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| |
Collapse
|
14
|
Early Detection of Localized Immunity in Experimental Autoimmune Myocarditis Using [ 99mTc]Fucoidan SPECT. Mol Imaging Biol 2019; 22:643-652. [PMID: 31432389 DOI: 10.1007/s11307-019-01420-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE The aim of the study was to evaluate the ability of technetium-99m-fucoidan ([99mTc]fucoidan), a molecular imaging agent specific for selectins, in the assessment of early localized immunity in a rat model of experimental autoimmune myocarditis (EAM). PROCEDURES EAM was induced in Lewis rats and troponin T; brain natriuretic peptide (BNP) and anti-myosin antibodies were measured in plasma. Separately, [99mTc]fucoidan single-photon emission computed tomography (SPECT)/x-ray computed tomography (CT) was performed in the very early phase of myocarditis at 10, 15, and 21 days after immunization. Then, hearts were collected and used for autoradiography, well counting, histology, and flow cytometry analysis. RESULTS The EAM acute phase is characterized by extensive myocardial necrosis, release of troponin and BNP, and pericardial effusion. [99mTc]Fucoidan uptake was significantly increased in EAM compared with controls starting from D15. There was a close relationship between uptake of the tracer and myocardial content in CD45+, CD8+, CD11b+, and CD31+ cells. CONCLUSIONS [99mTc]Fucoidan SPECT/CT accurately diagnosed the autoimmune attack in the early steps of EAM and could be used to monitor disease evolution and therapy efficiency.
Collapse
|
15
|
Stirrat CG, Alam SR, MacGillivray TJ, Gray CD, Dweck MR, Dibb K, Spath N, Payne JR, Prasad SK, Gardner RS, Mirsadraee S, Henriksen PA, Semple SI, Newby DE. Ferumoxytol-enhanced magnetic resonance imaging in acute myocarditis. Heart 2018; 104:300-305. [PMID: 28986407 PMCID: PMC5861394 DOI: 10.1136/heartjnl-2017-311688] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/24/2017] [Accepted: 06/12/2017] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Ultrasmall superparamagnetic particles of iron oxide (USPIO)-enhanced MRI can detect tissue-resident macrophage activity and identify cellular inflammation within tissues. We hypothesised that USPIO-enhanced MRI would provide a non-invasive imaging technique that would improve the diagnosis and management of patients with acute myocarditis. METHODS Ten volunteers and 14 patients with suspected acute myocarditis underwent T2, T2* and late gadolinium enhancement (LGE) 3T MRI, with further T2* imaging at 24 hours after USPIO (ferumoxytol, 4 mg/kg) infusion, at baseline and 3 months. Myocardial oedema and USPIO enhancement were determined within areas of LGE as well as throughout the myocardium. RESULTS Myocarditis was confirmed in nine of the 14 suspected cases of myocarditis. There was greater myocardial oedema in regions of LGE in patients with myocarditis when compared with healthy volunteer myocardium (T2 value, 57.1±5.3 vs 46.7±1.6 ms, p<0.0001). There was no demonstrable difference in USPIO enhancement between patients and volunteers even within regions displaying LGE (change in R2*, 35.0±15.0 vs 37.2±9.6 s-1, p>0.05). Imaging after 3 months in patients with myocarditis revealed a reduction in volume of LGE, a reduction in oedema measures within regions displaying LGE and improvement in ejection fraction (mean -19.7 mL, 95% CI (-0.5 to -40.0)), -5.8 ms (-0.9 to -10.7) and +6% (0.5% to 11.5%), respectively, p<0.05 for all). CONCLUSION In patients with acute myocarditis, USPIO-enhanced MRI does not provide additional clinically relevant information to LGE and T2 mapping MRI. This suggests that tissue-resident macrophages do not provide a substantial contribution to the myocardial inflammation in this condition.Clinical trial registration NCT02319278; Results.
Collapse
Affiliation(s)
- Colin G Stirrat
- British Heart Foundation/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Shirjel R Alam
- British Heart Foundation/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Thomas J MacGillivray
- Clinical Research Facility, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging QMRI Facility, University of Edinburgh, Edinburgh, UK
| | - Calum D Gray
- Clinical Research Facility, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging QMRI Facility, University of Edinburgh, Edinburgh, UK
| | - Marc R Dweck
- British Heart Foundation/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Kevin Dibb
- British Heart Foundation/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Nick Spath
- British Heart Foundation/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - John R Payne
- Department of Cardiology, Golden Jubilee National Hospital, Clydebank, UK
| | - Sanjay K Prasad
- Department of Cardiology, Royal Brompton Hospital, London, UK
| | - Roy S Gardner
- Department of Cardiology, Golden Jubilee National Hospital, Clydebank, UK
| | | | - Peter A Henriksen
- British Heart Foundation/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Scott Ik Semple
- British Heart Foundation/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging QMRI Facility, University of Edinburgh, Edinburgh, UK
| | - David E Newby
- British Heart Foundation/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging QMRI Facility, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
16
|
MR Assessment of Acute Pathologic Process after Myocardial Infarction in a Permanent Ligation Mouse Model: Role of Magnetic Nanoparticle-Contrasted MRI. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:2870802. [PMID: 29114174 PMCID: PMC5664276 DOI: 10.1155/2017/2870802] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/27/2017] [Accepted: 09/14/2017] [Indexed: 01/17/2023]
Abstract
We evaluated the relationship between myocardial infarct size and inflammatory response using cardiac magnetic resonance imaging (CMR) in an acute myocardial infarction (AMI) mouse model. Myocardial infarction (MI) was induced in 14 mice by permanent ligation of the left anterior descending artery. Late gadolinium enhancement (LGE), manganese-enhanced MRI (MEMRI), and magnetofluorescent nanoparticle MRI (MNP-MRI) were performed 1, 2, and 3 days after MI, respectively. The size of the enhanced lesion was quantitatively determined using Otsu's thresholding method in area-based and sector-based approaches and was compared statistically. Linear correlation between the enhanced lesion sizes was evaluated by Pearson's correlation coefficients. Differences were compared using Bland-Altman analysis. The size of the inflammatory area determined by MNP-MRI (57.1 ± 10.1%) was significantly larger than that of the infarct area measured by LGE (40.8 ± 11.7%, P < 0.0001) and MEMRI (44.1 ± 14.9%, P < 0.0001). There were significant correlations between the sizes of the infarct and inflammatory lesions (MNP-MRI versus LGE: r = 0.3418, P = 0.0099; MNP-MRI versus MEMRI: r = 0.4764, P = 0.0002). MNP-MRI provides information about inflammatory responses in a mouse model of AMI. Thus, MNP-MRI associated with LGE and MEMRI may play an important role in monitoring the disease progression in MI.
Collapse
|
17
|
Park HS, Kim J, Cho MY, Lee H, Nam SH, Suh YD, Hong KS. Convenient and effective ICGylation of magnetic nanoparticles for biomedical applications. Sci Rep 2017; 7:8831. [PMID: 28821875 PMCID: PMC5562755 DOI: 10.1038/s41598-017-09627-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/27/2017] [Indexed: 01/25/2023] Open
Abstract
Nanoprobes used for biomedical applications usually require surface modifications with amphiphilic surfactants or inorganic coating materials to enhance their biocompatibility. We proposed a facile synthetic approach for the phase transfer of hydrophobic magnetic nanoparticles by the direct adherence of fluorescent probes, without any chemical modifications, for use as a magnetic resonance (MR)/near-infrared (NIR) fluorescence bimodal imaging contrast agent. Indocyanine green (ICG) was used not only as an optical component for NIR imaging, but also as a surfactant for phase transfer with no superfluous moiety: we therefore called the process "ICGylation". Cell labeling and tracking in vivo with ICGylated magnetic nanoparticles were successfully performed by MR/NIR dual-mode imaging for three days, which showed remarkable biostability without any additional surface functionalization. We expect that this novel MR/NIR contrast agent demonstrating sensitive detection and simultaneous imaging capability can be used in diverse fields, such as the imaging and tracking of immune cells to confirm immunotherapeutic efficacy. The approach used could also be applied to other kinds of nanoparticles, and it would promote the development of advanced functional multimodal nanobioprobes.
Collapse
Affiliation(s)
- Hye Sun Park
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju, 28119, Korea
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Korea
| | - Jongwoo Kim
- Laboratory for Advanced Molecular Probing (LAMP), Research Center for Convergence NanoRaman Technology, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea
| | - Mi Young Cho
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju, 28119, Korea
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Korea
| | - Hyunseung Lee
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju, 28119, Korea
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Korea
| | - Sang Hwan Nam
- Laboratory for Advanced Molecular Probing (LAMP), Research Center for Convergence NanoRaman Technology, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea
| | - Yung Doug Suh
- Laboratory for Advanced Molecular Probing (LAMP), Research Center for Convergence NanoRaman Technology, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea.
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Korea.
| | - Kwan Soo Hong
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju, 28119, Korea.
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Korea.
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Korea.
| |
Collapse
|
18
|
Mavrogeni SI, Markousis-Mavrogenis G, Kolovou G. Ferumoxytol in acute myocarditis: ‘keep your head in the clouds and your feet on the earth’. Heart 2017; 104:278-279. [DOI: 10.1136/heartjnl-2017-312004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
19
|
Seth A, Park HS, Hong KS. Current Perspective on In Vivo Molecular Imaging of Immune Cells. Molecules 2017; 22:molecules22060881. [PMID: 28587110 PMCID: PMC6152742 DOI: 10.3390/molecules22060881] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/19/2017] [Indexed: 12/31/2022] Open
Abstract
Contemporaneous development of improved immune cell-based therapies, and powerful imaging tools, has prompted growth in technologies for immune cell tracking in vivo. Over the past couple of decades, imaging tools such as magnetic resonance imaging (MRI) and optical imaging have successfully monitored the trafficking patterns of therapeutic immune cells and assisted the evaluation of the success or failure of immunotherapy. Recent advancements in imaging technology have made imaging an indispensable module of immune cell-based therapies. In this review, emerging applications of non-radiation imaging modalities for the tracking of a range of immune cells are discussed. Applications of MRI, NIR, and other imaging tools have demonstrated the potential of non-invasively surveying the fate of both phagocytic and non-phagocytic immune cells in vivo.
Collapse
Affiliation(s)
- Anushree Seth
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju 28119, Korea.
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
| | - Hye Sun Park
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju 28119, Korea.
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
| | - Kwan Soo Hong
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju 28119, Korea.
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Korea.
| |
Collapse
|
20
|
Park SM, Aalipour A, Vermesh O, Yu JH, Gambhir SS. Towards clinically translatable in vivo nanodiagnostics. NATURE REVIEWS. MATERIALS 2017; 2:17014. [PMID: 29876137 PMCID: PMC5985817 DOI: 10.1038/natrevmats.2017.14] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Nanodiagnostics as a field makes use of fundamental advances in nanobiotechnology to diagnose, characterize and manage disease at the molecular scale. As these strategies move closer to routine clinical use, a proper understanding of different imaging modalities, relevant biological systems and physical properties governing nanoscale interactions is necessary to rationally engineer next-generation bionanomaterials. In this Review, we analyse the background physics of several clinically relevant imaging modalities and their associated sensitivity and specificity, provide an overview of the materials currently used for in vivo nanodiagnostics, and assess the progress made towards clinical translation. This work provides a framework for understanding both the impressive progress made thus far in the nanodiagnostics field as well as presenting challenges that must be overcome to obtain widespread clinical adoption.
Collapse
Affiliation(s)
- Seung-Min Park
- Department of Radiology, Stanford University School of Medicine
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California 94305, USA
| | - Amin Aalipour
- Department of Radiology, Stanford University School of Medicine
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California 94305, USA
| | - Ophir Vermesh
- Department of Radiology, Stanford University School of Medicine
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California 94305, USA
| | - Jung Ho Yu
- Department of Radiology, Stanford University School of Medicine
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California 94305, USA
| | - Sanjiv S Gambhir
- Department of Radiology, Stanford University School of Medicine
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California 94305, USA
- Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, California 94304, USA
| |
Collapse
|
21
|
Liang G, Vo D, Nguyen PK. Fundamentals of Cardiovascular Molecular Imaging: a Review of Concepts and Strategies. CURRENT CARDIOVASCULAR IMAGING REPORTS 2017. [DOI: 10.1007/s12410-017-9403-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
22
|
Tian B, Liu R, Chen S, Chen L, Liu F, Jia G, Dong Y, Li J, Chen H, Lu J. Mannose-coated gadolinium liposomes for improved magnetic resonance imaging in acute pancreatitis. Int J Nanomedicine 2017; 12:1127-1141. [PMID: 28260882 PMCID: PMC5325132 DOI: 10.2147/ijn.s123290] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Acute pancreatitis (AP) is an acute inflammatory condition of the pancreas. The symptoms, treatment, and prognosis of mild and severe AP are different, and severe AP is a potentially life-threatening disease with a high incidence of complications and high mortality rate. Thus, it is urgent to develop an effective approach to reliably discriminate between mild and severe AP. Methods We have developed novel gadolinium-diethylenetriaminepentaacetic (Gd-DTPA)-loaded mannosylated liposomes (named thereafter M-Gd-NL) that preferably target macrophages in AP. The targeting ability of M-Gd-NL toward macrophages in AP and its ability to discriminate between mild and severe AP were evaluated. Results The liposomes were of desired particle size (~100 nm), Gd-DTPA encapsulation efficiency (~85%), and stability. M-Gd-NL and non-targeted Gd-DTPA-loaded liposomes (Gd-NL) exhibited increased relaxivity compared with Gd-DTPA. Compared with Gd-NL and Gd-DTPA, M-Gd-NL showed increased uptake in macrophages, resulting in increased T1 imaging ability both in vitro (macrophage cell line) and in vivo (severe AP model). Importantly, M-Gd-NL had the ability to discriminate between mild and severe AP, as reflected by a significantly higher T1 magnetic resonance imaging signal in severe AP than in mild AP. M-Gd-NL did not show severe organ toxicity in rats. Conclusion Our data suggest that M-Gd-NL had enhanced magnetic resonance imaging ability by targeting macrophages in AP and good ability to discriminate between mild and severe AP. We believe that M-Gd-NL could shed new light on the diagnosis of AP in the near future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Huaiwen Chen
- Center of Clinical and Translational Medicine, Shanghai Changhai Hospital, The Second Military Medical University; Sunlipo Biotech Research Center for Nanomedicine, Shanghai, People's Republic of China
| | | |
Collapse
|
23
|
Choo EH, Lee JH, Park EH, Park HE, Jung NC, Kim TH, Koh YS, Kim E, Seung KB, Park C, Hong KS, Kang K, Song JY, Seo HG, Lim DS, Chang K. Infarcted Myocardium-Primed Dendritic Cells Improve Remodeling and Cardiac Function After Myocardial Infarction by Modulating the Regulatory T Cell and Macrophage Polarization. Circulation 2017; 135:1444-1457. [PMID: 28174192 DOI: 10.1161/circulationaha.116.023106] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 01/20/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Inflammatory responses play a critical role in left ventricular remodeling after myocardial infarction (MI). Tolerogenic dendritic cells (tDCs) can modulate immune responses, inducing regulatory T cells in a number of inflammatory diseases. METHODS We generated tDCs by treating bone marrow-derived dendritic cells with tumor necrosis factor-α and cardiac lysate from MI mice. We injected MI mice, induced by a ligation of the left anterior descending coronary artery in C57BL/6 mice, twice with tDCs within 24 hours and at 7 days after the ligation. RESULTS In vivo cardiac magnetic resonance imaging and ex vivo histology confirmed the beneficial effect on postinfarct left ventricular remodeling in MI mice treated with tDCs. Subcutaneously administered infarct lysate-primed tDCs near the inguinal lymph node migrated to the regional lymph node and induced infarct tissue-specific regulatory T-cell populations in the inguinal and mediastinal lymph nodes, spleen, and infarcted myocardium, indicating that a local injection of tDCs induces a systemic activation of MI-specific regulatory T cells. These events elicited an inflammatory-to-reparative macrophage shift. The altered immune environment in the infarcted heart resulted in a better wound remodeling, preserved left ventricular systolic function after myocardial tissue damage, and improved survival. CONCLUSIONS This study showed that tDC therapy in a preclinical model of MI was potentially translatable into an antiremodeling therapy for ischemic tissue repair.
Collapse
Affiliation(s)
- Eun Ho Choo
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Jun-Ho Lee
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Eun-Hye Park
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Hyo Eun Park
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Nam-Chul Jung
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Tae-Hoon Kim
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Yoon-Seok Koh
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Eunmin Kim
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Ki-Bae Seung
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Cheongsoo Park
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Kwan-Soo Hong
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Kwonyoon Kang
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Jie-Young Song
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Han Geuk Seo
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Dae-Seog Lim
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.)
| | - Kiyuk Chang
- From Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea (E.H.C., E.-H.P., H.E.P., T.-H.K., Y.-S.K., E.K., K.-B.S., K.K., K.C.); Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea (J.-H.L., D.-S.L.); Pharos Vaccine Inc, Seongnam-si, Gyeonggido, Korea (J.-H.L., N.-C.J.); Division of Magnetic Resonance Research, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk- do, Korea (C.P., K.-S.H.); Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, Seoul (J.-Y.S.); and Department of Animal Biotechnology, Konkuk University, Seoul, Korea (H.G.S.).
| |
Collapse
|
24
|
Park KE, Noh YW, Kim A, Lim YT. Hyaluronic acid-coated nanoparticles for targeted photodynamic therapy of cancer guided by near-infrared and MR imaging. Carbohydr Polym 2017; 157:476-483. [DOI: 10.1016/j.carbpol.2016.10.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 08/29/2016] [Accepted: 10/06/2016] [Indexed: 12/16/2022]
|
25
|
Lee SP, Im HJ, Kang S, Chung SJ, Cho YS, Kang H, Park HS, Hwang DW, Park JB, Paeng JC, Cheon GJ, Lee YS, Jeong JM, Kim YJ. Noninvasive Imaging of Myocardial Inflammation in Myocarditis using 68Ga-tagged Mannosylated Human Serum Albumin Positron Emission Tomography. Am J Cancer Res 2017; 7:413-424. [PMID: 28042344 PMCID: PMC5197074 DOI: 10.7150/thno.15712] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 10/27/2016] [Indexed: 12/23/2022] Open
Abstract
The diagnosis of myocarditis traditionally relies on invasive endomyocardial biopsy but none of the imaging studies so far are specific for infiltration of the inflammatory cells itself. We synthesized 68Ga-2-(p-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) mannosylated human serum albumin (MSA) by conjugating human serum albumin with mannose, followed by conjugation with NOTA and labeling it with 68Ga. The efficacy of 68Ga-NOTA-MSA positron emission tomography (PET) for imaging myocardial inflammation was tested in a rat myocarditis model. A significant number of mannose receptor-positive inflammatory cells infiltrated the myocardium in both human and rat myocarditis tissue. 68Ga-NOTA-MSA uptake was upregulated in organs of macrophage accumulation, such as liver, spleen, bone marrow and myocardium (0.32 (0.31~0.33) for normal versus 1.02 (0.86~1.06) for myocarditis (median (range), SUV); n=4~6 per group, p-value=0.01). 68Ga-NOTA-MSA uptake in the left ventricle was upregulated in myocarditis compared with normal rats (2.29 (1.42~3.40) for normal versus 4.18 (3.43~6.15) for myocarditis (median (range), average standard uptake value ratio against paraspinal muscle); n=6 per group, p-value<0.01), which was downregulated in rats with cyclosporine-A treated myocarditis (3.69 (2.59~3.86) for myocarditis versus 2.28 (1.76~2.60) for cyclosporine-A treated myocarditis; n=6 per group, p-value<0.01). The specificity of the tracer was verified by administration of excess non-labeled MSA. 68Ga-NOTA-MSA uptake was significantly enhanced earlier in the evolution of myocarditis before any signs of inflammation could be seen on echocardiography. These results demonstrate the potential utility of visualizing infiltration of mannose receptor-positive macrophages with 68Ga-NOTA-MSA PET in the early diagnosis of as well as in the monitoring of treatment response of myocarditis.
Collapse
|
26
|
Bietenbeck M, Florian A, Faber C, Sechtem U, Yilmaz A. Remote magnetic targeting of iron oxide nanoparticles for cardiovascular diagnosis and therapeutic drug delivery: where are we now? Int J Nanomedicine 2016; 11:3191-203. [PMID: 27486321 PMCID: PMC4957681 DOI: 10.2147/ijn.s110542] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Magnetic resonance imaging (MRI) allows for an accurate assessment of both functional and structural cardiac parameters, and thereby appropriate diagnosis and validation of cardiovascular diseases. The diagnostic yield of cardiovascular MRI examinations is often increased by the use of contrast agents that are almost exclusively based on gadolinium compounds. Another clinically approved contrast medium is composed of superparamagnetic iron oxide nanoparticles (IONs). These particles may expand the field of contrast-enhanced cardiovascular MRI as recently shown in clinical studies focusing on acute myocardial infarction (AMI) and atherosclerosis. Furthermore, IONs open up new research opportunities such as remote magnetic drug targeting (MDT). The approach of MDT relies on the coupling of bioactive molecules and magnetic nanoparticles to form an injectable complex. This complex, in turn, can be attracted to and retained at a desired target inside the body with the help of applied magnetic fields. In comparison to common systemic drug applications, MDT techniques promise both higher concentrations at the target site and lower concentrations elsewhere in the body. Moreover, concurrent or subsequent MRI can be used for noninvasive monitoring of drug distribution and successful delivery to the desired organ in vivo. This review does not only illustrate the basic conceptual and biophysical principles of IONs, but also focuses on new research activities and achievements in the cardiovascular field, mainly in the management of AMI. Based on the presentation of successful MDT applications in preclinical models of AMI, novel approaches and the translational potential of MDT are discussed.
Collapse
Affiliation(s)
| | | | - Cornelius Faber
- Department of Clinical Radiology, University Hospital Münster, Münster
| | - Udo Sechtem
- Division of Cardiology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | | |
Collapse
|
27
|
Maya Y, Werner RA, Schütz C, Wakabayashi H, Samnick S, Lapa C, Zechmeister C, Jahns R, Jahns V, Higuchi T. 11C-Methionine PET of Myocardial Inflammation in a Rat Model of Experimental Autoimmune Myocarditis. J Nucl Med 2016; 57:1985-1990. [PMID: 27390159 DOI: 10.2967/jnumed.116.174045] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/17/2016] [Indexed: 12/30/2022] Open
Abstract
Myocarditis represents a major cause of dilated cardiomyopathy and sudden cardiac death in younger adults. Currently, definitive diagnosis of myocarditis requires endomyocardial biopsy, which is highly invasive and has the drawback of variable sensitivity due to inherent sampling error. Therefore, reliable noninvasive methods to detect and monitor cardiac inflammation are clinically relevant. In this study, we explored the potential of radiolabeled methionine to assess myocardial inflammatory activity in a rat model of experimental autoimmune myocarditis (EAM). METHODS Autoimmune myocarditis was induced by immunizing Lewis rats twice with porcine cardiac myosin and Freund complete adjuvant. Control animals were treated with adjuvant alone. Dual-tracer autoradiography was performed to assess 14C-methionine uptake and to compare the distributions of 14C-methionine versus 18F-FDG. Hematoxylin and eosin staining and anti-CD68 macrophage staining were performed for histologic analysis. Additionally, cardiac 11C-methionine PET was performed to evaluate the feasibility of in vivo imaging. 18F-FDG PET was also conducted to compare the in vivo uptake of 11C-methionine and 18F-FDG. RESULTS Multiple focal cardiac inflammatory lesions were histologically identified in myosin-immunized rats, whereas no cardiac lesions were observed in the controls. Autoradiographic images clearly showed a high-density accumulation of 14C-methionine in inflammatory lesions of EAM rats, whereas no significant uptake was observed in the control animals. 14C-methionine uptake was significantly higher in inflammatory lesions than in remote noninflammatory areas and control rat hearts. The distribution of 14C-methionine correlated well with that of 18F-FDG and with macrophage density. The contrast between inflammatory and noninflammatory areas was higher for 18F-FDG than for 14C-methionine (3.45 ± 0.68 vs. 2.07 ± 0.21, respectively; P < 0.05). In the PET imaging study, the regional 11C-methionine uptake (percentage injected dose per cubic centimeter) observed in EAM rats was significantly higher than the values obtained for control animals (0.64 ± 0.09 vs. 0.28 ± 0.02, respectively; P < 0.001). A good positive correlation between 11C-methionine and 18F-FDG uptake was found. CONCLUSION In a rat model of autoimmune myocarditis, we demonstrated the colocalization of radiolabeled methionine accumulation with 18F-FDG uptake in histologically proven inflammatory lesions. These data suggest that 11C-methionine might represent a promising candidate for the noninvasive detection and monitoring of myocarditis.
Collapse
Affiliation(s)
- Yoshifumi Maya
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany.,Research Centre, Nihon Medi-Physics Co., Ltd., Chiba, Japan
| | - Rudolf A Werner
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Else-Kröner-Forschungskolleg, Interdisciplinary Center for Clinical Research, University of Würzburg,Würzburg, Germany
| | - Claudia Schütz
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Department of Pharmacology, University of Würzburg, Würzburg, Germany; and
| | | | - Samuel Samnick
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - Christina Zechmeister
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Department of Pharmacology, University of Würzburg, Würzburg, Germany; and
| | - Roland Jahns
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Interdisciplinary Bank of Biomaterials and Data Würzburg (IBDW), University of Würzburg, Würzburg, Germany
| | - Valérie Jahns
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Department of Pharmacology, University of Würzburg, Würzburg, Germany; and
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany .,Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
28
|
Jeuthe S, Wassilew K, O h-Ici D, da Silva TF, Münch F, Berger F, Kuehne T, Pieske B, Messroghli DR. Myocardial T1 maps reflect histological findings in acute and chronic stages of myocarditis in a rat model. J Cardiovasc Magn Reson 2016; 18:19. [PMID: 27084492 PMCID: PMC4833926 DOI: 10.1186/s12968-016-0241-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/31/2016] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Cardiovascular magnetic resonance offers both diagnostic and prognostic information in myocarditis. Using an established animal model of myocarditis, the aim of this study was to measure myocardial T1 before the onset, in the acute and in the chronic phases of the disease and to compare its course with histological and immunohistochemistry findings. METHODS Male young Lewis rats were immunized with 0.25 mg porcine myocardial myosin into the rear footpads on day 0. Native and contrast-enhanced ECG-triggered cardiac MRI examinations were performed before immunization on day 0 and on days 14, 21 and 35. Left ventricular function, pre- and post- contrast T1 parameters and LGE images were assessed using Small animal look-locker inversion recovery (SALLI). For each of the indicated time points a minimum of 4 rats were randomly sacrificed for pathological investigations including conventional histology (HE and Sirius-Red staining) and immunohistochemistry (CD 68) investigations. RESULTS All immunized rats developed myocarditis (morbidity 100%). Histologically we observed increased wall thickness with biventricular macrophage-rich mixed inflammatory infiltrates. All rats with a histologically severe myocarditis showed increased native T1 and decreased post-contrast T1 of the myocardium. CONCLUSIONS The assessment of native T1 and post-contrast T1 allows accurate differentiation between healthy myocardium and myocardium with inflammation and also between the acute and chronic phases of the disease.
Collapse
MESH Headings
- Acute Disease
- Animals
- Antigens, CD/immunology
- Antigens, Differentiation, Myelomonocytic/immunology
- Autoimmune Diseases/chemically induced
- Autoimmune Diseases/immunology
- Autoimmune Diseases/pathology
- Autoimmune Diseases/physiopathology
- Cardiomyopathy, Dilated/chemically induced
- Cardiomyopathy, Dilated/immunology
- Cardiomyopathy, Dilated/pathology
- Cardiomyopathy, Dilated/physiopathology
- Chronic Disease
- Disease Models, Animal
- Immunohistochemistry
- Magnetic Resonance Imaging
- Male
- Myocarditis/chemically induced
- Myocarditis/immunology
- Myocarditis/pathology
- Myocarditis/physiopathology
- Myocardium/immunology
- Myocardium/pathology
- Myosins
- Predictive Value of Tests
- Rats, Inbred Lew
- Time Factors
- Ventricular Function, Left
Collapse
Affiliation(s)
- Sarah Jeuthe
- />Internal Medicine/Cardiology, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Katharina Wassilew
- />Cardiovascular Pathology, Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Darach O h-Ici
- />Congenital Heart Disease and Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Tiago Ferreira da Silva
- />Congenital Heart Disease and Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Frédéric Münch
- />Congenital Heart Disease and Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Felix Berger
- />Congenital Heart Disease and Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Titus Kuehne
- />Congenital Heart Disease and Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Burkert Pieske
- />Internal Medicine/Cardiology, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Daniel R. Messroghli
- />Internal Medicine/Cardiology, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
29
|
Amsallem M, Saito T, Tada Y, Dash R, McConnell MV. Magnetic Resonance Imaging and Positron Emission Tomography Approaches to Imaging Vascular and Cardiac Inflammation. Circ J 2016; 80:1269-77. [DOI: 10.1253/circj.cj-16-0224] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Myriam Amsallem
- Division of Cardiovascular Medicine, Stanford University School of Medicine
| | - Toshinobu Saito
- Division of Cardiovascular Medicine, Stanford University School of Medicine
| | - Yuko Tada
- Division of Cardiovascular Medicine, Stanford University School of Medicine
| | - Rajesh Dash
- Division of Cardiovascular Medicine, Stanford University School of Medicine
| | | |
Collapse
|
30
|
Visualizing myocardial inflammation in a rat model of type 4 cardiorenal syndrome by dual-modality molecular imaging. Biomaterials 2015; 68:67-76. [DOI: 10.1016/j.biomaterials.2015.07.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 07/27/2015] [Accepted: 07/31/2015] [Indexed: 12/21/2022]
|
31
|
Bietenbeck M, Florian A, Sechtem U, Yilmaz A. The diagnostic value of iron oxide nanoparticles for imaging of myocardial inflammation--quo vadis? J Cardiovasc Magn Reson 2015; 17:54. [PMID: 26152269 PMCID: PMC4495803 DOI: 10.1186/s12968-015-0165-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/24/2015] [Indexed: 01/23/2023] Open
Abstract
Cardiovascular magnetic resonance (CMR) is an integral part in the diagnostic work-up of cardiac inflammatory diseases. In this context, superparamagnetic iron oxide-based contrast agents can provide additional diagnostic information regarding the assessment of myocardial infarction and myocarditis. After intravenous administration, these nanoparticles are taken up by activated monocytes and macrophages, which predominantly accumulate in regions associated with inflammation as was successfully shown in recent preclinical studies. Furthermore, first clinical studies with a new iron oxide-complex that was clinically approved for the treatment of iron deficiency anaemia recently demonstrated a superior diagnostic value of iron oxide nanoparticles compared to gadolinium-based compounds for imaging of myocardial inflammation in patients with acute myocardial infarction. In this article, we outline the basic features of superparamagnetic iron oxide-based contrast agents and review recent studies using such nanoparticles for cardiac imaging in case of acute myocardial infarction as well as acute myocarditis. Moreover, we highlight the translational potential of these agents and possible research applications with regard to imaging and therapy.
Collapse
Affiliation(s)
- Michael Bietenbeck
- Department of Cardiology and Angiology, Albert-Schweitzer-Campus 1, building A1, 48149, Münster, Germany
| | - Anca Florian
- Department of Cardiology and Angiology, Albert-Schweitzer-Campus 1, building A1, 48149, Münster, Germany
| | - Udo Sechtem
- Division of Cardiology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Ali Yilmaz
- Department of Cardiology and Angiology, Albert-Schweitzer-Campus 1, building A1, 48149, Münster, Germany.
| |
Collapse
|
32
|
Benali K, Louedec L, Azzouna RB, Merceron O, Nassar P, Al Shoukr F, Petiet A, Barbato D, Michel JB, Sarda-Mantel L, Le Guludec D, Rouzet F. Preclinical Validation of99mTc–Annexin A5–128 in Experimental Autoimmune Myocarditis and Infective Endocarditis: Comparison with99mTc–HYNIC–Annexin A5. Mol Imaging 2015; 13. [DOI: 10.2310/7290.2014.00049] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Khadija Benali
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| | - Liliane Louedec
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| | - Rana Ben Azzouna
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| | - Olivier Merceron
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| | - Pierre Nassar
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| | - Faisal Al Shoukr
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| | - Anne Petiet
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| | - Donato Barbato
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| | - Jean-Baptiste Michel
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| | - Laure Sarda-Mantel
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| | - Dominique Le Guludec
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| | - Francois Rouzet
- From Inserm, U1148, and Paris Diderot University, Paris, France; Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale, Paris Diderot University, Paris, France; and Advanced Accelerator Applications - via Ribes 5 - 10010 - Colleretto Giacosa, Turin, Italy
| |
Collapse
|
33
|
Clinically relevant strategies for lowering cardiomyocyte glucose uptake for 18F-FDG imaging of myocardial inflammation in mice. Eur J Nucl Med Mol Imaging 2014; 42:771-80. [PMID: 25389013 DOI: 10.1007/s00259-014-2956-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 10/29/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE Myocardial inflammation is an emerging target for novel therapies and thus for molecular imaging. Positron emission tomography (PET) with (18)F-fluorodeoxyglucose (FDG) has been employed, but requires an approach for suppression of cardiomyocyte uptake. We tested clinically viable strategies for their suitability in mouse models in order to optimize preclinical imaging protocols. METHODS C57BL/6 mice (n = 56) underwent FDG PET under various conditions. In healthy animals, the effect of low-dose (5 units/kg) or high-dose (500 units/kg, 15 min prior) intravenous heparin, extended fasting (18 h) and the impact of conscious injection with limited, late application of isoflurane anaesthesia after 40 min of conscious uptake were examined in comparison to ketamine/xylazine anaesthesia. Conscious injection/uptake strategies were further evaluated at 3 days after permanent coronary artery occlusion. RESULTS Under continuous isoflurane anaesthesia, neither heparin administration nor extended fasting significantly impacted myocardial (18)F-FDG accumulation. Injection with 40 min uptake in awake mice resulted in a marked reduction of global myocardial (18)F-FDG uptake compared to standard isoflurane anaesthesia (5.7 ± 1.1 %ID/g vs 30.2 ± 7.9 %ID/g, p < 0.01). Addition of heparin and fasting further reduced uptake compared to conscious injection alone (3.8 ± 1.5 %ID/g, p < 0.01) similar to ketamine/xylazine (2.4 ± 2.2 %ID/g, p < 0.001). In the inflammatory phase, 3 days after myocardial infarction, conscious injection/uptake with and without heparin/fasting identified a marked increase in myocardial (18)F-FDG accumulation that was similar to that observed under ketamine/xylazine. CONCLUSION Continuous isoflurane anaesthesia obscures any suppressive effect of heparin or fasting on cardiomyocyte glucose utilization. Conscious injection of FDG in rodents significantly reduces cardiomyocyte uptake and enables further suppression by heparin and fasting, similar to clinical observations. In contrast to ketamine/xylazine, this represents a more physiological, translatable strategy for suppression of cardiomyocyte (18)F-FDG uptake when targeting myocardial inflammation.
Collapse
|
34
|
Leuschner F, Courties G, Dutta P, Mortensen LJ, Gorbatov R, Sena B, Novobrantseva TI, Borodovsky A, Fitzgerald K, Koteliansky V, Iwamoto Y, Bohlender M, Meyer S, Lasitschka F, Meder B, Katus HA, Lin C, Libby P, Swirski FK, Anderson DG, Weissleder R, Nahrendorf M. Silencing of CCR2 in myocarditis. Eur Heart J 2014; 36:1478-88. [PMID: 24950695 DOI: 10.1093/eurheartj/ehu225] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 05/13/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Myocarditis is characterized by inflammatory cell infiltration of the heart and subsequent deterioration of cardiac function. Monocytes are the most prominent population of accumulating leucocytes. We investigated whether in vivo administration of nanoparticle-encapsulated siRNA targeting chemokine (C-C motif) receptor 2 (CCR2)-a chemokine receptor crucial for leucocyte migration in humans and mice--reduces inflammation in autoimmune myocarditis. METHODS AND RESULTS In myocardium of patients with myocarditis, CCL2 mRNA levels and CCR2(+) cells increased (P < 0.05), motivating us to pursue CCR2 silencing. Flow cytometric analysis showed that siRNA silencing of CCR2 (siCCR2) reduced the number of Ly6C(high) monocytes in hearts of mice with acute autoimmune myocarditis by 69% (P < 0.05), corroborated by histological assessment. The nanoparticle-delivered siRNA was not only active in monocytes but also in bone marrow haematopoietic progenitor cells. Treatment with siCCR2 reduced the migration of bone marrow granulocyte macrophage progenitors into the blood. Cellular magnetic resonance imaging (MRI) after injection of macrophage-avid magnetic nanoparticles detected myocarditis and therapeutic effects of RNAi non-invasively. Mice with acute myocarditis showed enhanced macrophage MRI contrast, which was prevented by siCCR2 (P < 0.05). Follow-up MRI volumetry revealed that siCCR2 treatment improved ejection fraction (P < 0.05 vs. control siRNA-treated mice). CONCLUSION This study highlights the importance of CCR2 in the pathogenesis of myocarditis. In addition, we show that siCCR2 affects leucocyte progenitor trafficking. The data also point to a novel therapeutic strategy for the treatment of myocarditis.
Collapse
Affiliation(s)
- Florian Leuschner
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, Heidelberg D-69120, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Gabriel Courties
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Partha Dutta
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Luke J Mortensen
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Rostic Gorbatov
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Brena Sena
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | | | - Anna Borodovsky
- Alnylam Pharmaceuticals, 300 3rd Street, Cambridge, MA 02142, USA
| | - Kevin Fitzgerald
- Alnylam Pharmaceuticals, 300 3rd Street, Cambridge, MA 02142, USA
| | - Victor Koteliansky
- Department of Chemical Engineering, Massachusetts Institute of Technology, University Hospital Heidelberg, Heidelberg, Germany
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Marina Bohlender
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, Heidelberg D-69120, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Soeren Meyer
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, Heidelberg D-69120, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 220/221, Heidelberg 69120, Germany
| | - Benjamin Meder
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, Heidelberg D-69120, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, Heidelberg D-69120, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Charles Lin
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Daniel G Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, University Hospital Heidelberg, Heidelberg, Germany David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA Division of Health Science Technology, Massachusetts Institute of Technology, Boston, MA, USA Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| |
Collapse
|
35
|
Podrouzkova H, Feitova V, Panovsky R, Meluzin J, Orban M. Superparamagnetic iron oxide-enhanced magnetic resonance for imaging cardiac inflammation. A minireview. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2014; 159:378-81. [PMID: 24993740 DOI: 10.5507/bp.2014.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 05/29/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Advances in nanotechnology have lead to the development of a novel contrast media for Magnetic Resonance Imaging (MRI) - the superparamagnetic iron oxide nanoparticle (SPIO). SPIO nanoparticles are used to image inflammation on the cellular level in various settings. This review covers the physicochemical characteristics of SPIO particles as well as relevant animal and clinical studies and discusses the potential of SPIO particles to image cardiac inflammation including cardiac graft rejection. METHODS We searched the scientific biomedical databases Medline/PubMed, BioMedCentral, Google Scholar, Ovid and, ProQuest from to 2000 to 2013 for publications relevant to the topic. CONCLUSIONS SPIO nanoparticles due to their unique properties could become a useful tool in imaging cardiac inflammation. However, the task is to find a suitable particle size and coating with corresponding pharmacokinetics, establish the right dose and MRI scan timing for individual applications.
Collapse
Affiliation(s)
- Helena Podrouzkova
- Department of Cardiovascular Diseases, ICRC, St. Anne's University Hospital in Brno, Czech Republic.,ICRC, St. Anne's University Hospital in Brno
| | - Vera Feitova
- Department of Imaging Methods, ICRC, St. Anne's University Hospital in Brno.,ICRC, St. Anne's University Hospital in Brno
| | - Roman Panovsky
- Department of Cardiovascular Diseases, ICRC, St. Anne's University Hospital in Brno, Czech Republic.,ICRC, St. Anne's University Hospital in Brno
| | - Jaroslav Meluzin
- Department of Cardiovascular Diseases, ICRC, St. Anne's University Hospital in Brno, Czech Republic.,ICRC, St. Anne's University Hospital in Brno
| | - Marek Orban
- ICRC, St. Anne's University Hospital in Brno
| |
Collapse
|
36
|
Doltra A, Amundsen BH, Gebker R, Fleck E, Kelle S. Emerging concepts for myocardial late gadolinium enhancement MRI. Curr Cardiol Rev 2014; 9:185-90. [PMID: 23909638 PMCID: PMC3780343 DOI: 10.2174/1573403x113099990030] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 02/24/2013] [Indexed: 12/25/2022] Open
Abstract
Late gadolinium enhancement is a useful tool for scar detection, based on differences in the volume of distribution of gadolinium, an extracellular agent. The presence of fibrosis in the myocardium amenable to be detected with late gadolinium enhancement MRI is found not only in ischemic cardiomyopathy, in which it offers information regarding viability and prognosis, but also in a wide variety of non-ischemic cardiomyopathies. In the following review we will discuss the methodological aspects of gadolinium-based imaging, as well as its applications and anticipated future developments.
Collapse
Affiliation(s)
- Adelina Doltra
- Thorax Institute, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Spain
| | | | | | | | | |
Collapse
|
37
|
Schmerler P, Jeuthe S, O h-Ici D, Wassilew K, Lauer D, Kaschina E, Kintscher U, Müller S, Muench F, Kuehne T, Berger F, Unger T, Steckelings UM, Paulis L, Messroghli D. Mortality and morbidity in different immunization protocols for experimental autoimmune myocarditis in rats. Acta Physiol (Oxf) 2014; 210:889-98. [PMID: 24410878 DOI: 10.1111/apha.12227] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 10/04/2013] [Accepted: 01/06/2014] [Indexed: 01/22/2023]
Abstract
AIM We aimed to investigate the histological and clinical presentations of experimental autoimmune myocarditis (EAM) induced by different immunization schemes. METHODS Male young Lewis rats were divided into five groups immunized by porcine myocardial myosin: subcutaneously (SC) 2 mg (in two 1-mg doses on day 0 and 7), 0 mg (sham group) subcutaneously into rear footpads (RF), 0.25 mg RF, 0.5 mg RF or 1 mg RF (all RF once on day 0). On day 21, left ventricular (LV) function was assessed by cardiac magnetic resonance imaging and cardiac catheterization. The type and degree of myocardial inflammatory infiltrates were determined by conventional histology and immunohistochemistry. RESULTS In the SC immunized rats and in the RF sham group, we observed 0% mortality, while in the actively RF immunized rats, mortality was 20, 20 and 44% for the 0.25 mg, 0.5 mg and 1 mg myosin doses respectively. Morbidity as defined by inflammatory infiltrates on haematoxylin and eosin (HE) staining was 22% in the SC immunized rats, 0% in the RF sham group and 100% in all actively RF immunized groups. We observed augmented relative ventricle weight and spleen weight, increased LV end-diastolic pressure, reduced LV developed pressure and reduced LV ejection fraction in all with myosin-immunized RF groups without any systematic dose effect. CONCLUSION Subcutaneous immunization to the neck and flanks did not induce a reproducible EAM, while RF myosin administration reliably led to EAM. Lower myosin doses seem to induce the complete histological and clinical picture of EAM while being associated with lower mortality, non-specific symptoms and animal distress.
Collapse
Affiliation(s)
- P. Schmerler
- Center for Cardiovascular Research; Charité-University Medicine; Berlin Germany
| | - S. Jeuthe
- Congenital Heart Disease and Pediatric Cardiology; German Heart Institute; Berlin Germany
| | - D. O h-Ici
- Congenital Heart Disease and Pediatric Cardiology; German Heart Institute; Berlin Germany
| | - K. Wassilew
- Department of Pathology; German Heart Institute; Berlin Germany
| | - D. Lauer
- Center for Cardiovascular Research; Charité-University Medicine; Berlin Germany
| | - E. Kaschina
- Center for Cardiovascular Research; Charité-University Medicine; Berlin Germany
| | - U. Kintscher
- Center for Cardiovascular Research; Charité-University Medicine; Berlin Germany
| | - S. Müller
- Experimental Neurology; Charité-University Medicine; Berlin Germany
| | - F. Muench
- Congenital Heart Disease and Pediatric Cardiology; German Heart Institute; Berlin Germany
| | - T. Kuehne
- Congenital Heart Disease and Pediatric Cardiology; German Heart Institute; Berlin Germany
| | - F. Berger
- Congenital Heart Disease and Pediatric Cardiology; German Heart Institute; Berlin Germany
| | - T. Unger
- CARIM-School for Cardiovascular Diseases; Maastricht University; Maastricht the Netherlands
| | - U. M. Steckelings
- Center for Cardiovascular Research; Charité-University Medicine; Berlin Germany
- Department of Cardiovascular and Renal Research; University of Southern Denmark; Odense Denmark
| | - L. Paulis
- Center for Cardiovascular Research; Charité-University Medicine; Berlin Germany
- Institute of Pathophysiology; Faculty of Medicine; Comenius University; Bratislava Slovak Republic
| | - D. Messroghli
- Congenital Heart Disease and Pediatric Cardiology; German Heart Institute; Berlin Germany
| |
Collapse
|
38
|
Lee JY, Choi DY, Cho MY, Park KE, Lee SH, Hun Cho S, Hong KS, Lim YT. Targeted theranostic nanoparticles: receptor-mediated entry into cells, pH-induced signal generation and cytosolic delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:901-906. [PMID: 24106164 DOI: 10.1002/smll.201302136] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 08/27/2013] [Indexed: 06/02/2023]
Abstract
Virus-like theranostic nanoparticles: virus-like poly(amino acid) nanoparticles are synthesized that can be internalized via receptor-mediated endocytosis, resulting in encapsulated pH-activatable fluorescence probes that can be turned on in acidic environments but otherwise remain undetectable. The encapsulated anticancer drugs are also released into cytosol by endosome disruption.
Collapse
Affiliation(s)
- Ji Yeon Lee
- Graduate School and Department of Analytical Science and Technology, Chungnam National University, Daejeon, 305-764, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Stirrat CG, Newby DE, Robson JMJ, Jansen MA. The Use of Superparamagnetic Iron Oxide Nanoparticles to Assess Cardiac Inflammation. CURRENT CARDIOVASCULAR IMAGING REPORTS 2014. [DOI: 10.1007/s12410-014-9263-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
40
|
Milani-Nejad N, Janssen PML. Small and large animal models in cardiac contraction research: advantages and disadvantages. Pharmacol Ther 2014; 141:235-49. [PMID: 24140081 PMCID: PMC3947198 DOI: 10.1016/j.pharmthera.2013.10.007] [Citation(s) in RCA: 308] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 08/15/2013] [Indexed: 12/22/2022]
Abstract
The mammalian heart is responsible for not only pumping blood throughout the body but also adjusting this pumping activity quickly depending upon sudden changes in the metabolic demands of the body. For the most part, the human heart is capable of performing its duties without complications; however, throughout many decades of use, at some point this system encounters problems. Research into the heart's activities during healthy states and during adverse impacts that occur in disease states is necessary in order to strategize novel treatment options to ultimately prolong and improve patients' lives. Animal models are an important aspect of cardiac research where a variety of cardiac processes and therapeutic targets can be studied. However, there are differences between the heart of a human being and an animal and depending on the specific animal, these differences can become more pronounced and in certain cases limiting. There is no ideal animal model available for cardiac research, the use of each animal model is accompanied with its own set of advantages and disadvantages. In this review, we will discuss these advantages and disadvantages of commonly used laboratory animals including mouse, rat, rabbit, canine, swine, and sheep. Since the goal of cardiac research is to enhance our understanding of human health and disease and help improve clinical outcomes, we will also discuss the role of human cardiac tissue in cardiac research. This review will focus on the cardiac ventricular contractile and relaxation kinetics of humans and animal models in order to illustrate these differences.
Collapse
Affiliation(s)
- Nima Milani-Nejad
- Department of Physiology and Cell Biology and D. Davis Heart Lung Institute, College of Medicine, The Ohio State University, OH, USA
| | - Paul M L Janssen
- Department of Physiology and Cell Biology and D. Davis Heart Lung Institute, College of Medicine, The Ohio State University, OH, USA.
| |
Collapse
|
41
|
Weissleder R, Nahrendorf M, Pittet MJ. Imaging macrophages with nanoparticles. NATURE MATERIALS 2014; 13:125-38. [PMID: 24452356 DOI: 10.1038/nmat3780] [Citation(s) in RCA: 567] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 09/17/2013] [Indexed: 05/02/2023]
Abstract
Nanomaterials have much to offer, not only in deciphering innate immune cell biology and tracking cells, but also in advancing personalized clinical care by providing diagnostic and prognostic information, quantifying treatment efficacy and designing better therapeutics. This Review presents different types of nanomaterial, their biological properties and their applications for imaging macrophages in human diseases, including cancer, atherosclerosis, myocardial infarction, aortic aneurysm, diabetes and other conditions. We anticipate that future needs will include the development of nanomaterials that are specific for immune cell subsets and can be used as imaging surrogates for nanotherapeutics. New in vivo imaging clinical tools for noninvasive macrophage quantification are thus ultimately expected to become relevant to predicting patients' clinical outcome, defining treatment options and monitoring responses to therapy.
Collapse
Affiliation(s)
- Ralph Weissleder
- 1] Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, USA [2] Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, Massachusetts 02115, USA [3] Department of Radiology, Massachusetts General Hospital, 32 Fruit Street, Boston, Massachusetts 02114, USA
| | - Matthias Nahrendorf
- 1] Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, USA [2] Department of Radiology, Massachusetts General Hospital, 32 Fruit Street, Boston, Massachusetts 02114, USA
| | - Mikael J Pittet
- 1] Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, USA [2] Department of Radiology, Massachusetts General Hospital, 32 Fruit Street, Boston, Massachusetts 02114, USA
| |
Collapse
|
42
|
Circulation: Cardiovascular Imaging
Editors’ Picks. Circ Cardiovasc Imaging 2013. [DOI: 10.1161/circimaging.113.001335] [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: 11/16/2022]
|
43
|
Cardiac Magnetic Resonance Imaging: Recent Advances and New Insights in Cardiovascular Disease. CURRENT CARDIOVASCULAR IMAGING REPORTS 2012. [DOI: 10.1007/s12410-012-9176-y] [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]
|