1
|
Huang W, Zong J, Li M, Li TF, Pan S, Xiao Z. Challenges and Opportunities: Nanomaterials in Epilepsy Diagnosis. ACS NANO 2025; 19:16224-16247. [PMID: 40266286 DOI: 10.1021/acsnano.5c01203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Epilepsy is a common neurological disorder characterized by a significant rate of disability. Accurate early diagnosis and precise localization of the epileptogenic zone are essential for timely intervention, seizure prevention, and personalized treatment. However, over 30% of patients with epilepsy exhibit negative results on electroencephalography and magnetic resonance imaging (MRI), which can lead to misdiagnosis and subsequent delays in treatment. Consequently, enhancing diagnostic methodologies is imperative for effective epilepsy management. The integration of nanomaterials with biomedicine has led to the development of diagnostic tools for epilepsy. Key advancements include nanomaterial-enhanced neural electrodes, contrast agents, and biochemical sensors. Nanomaterials improve the quality of electrophysiological signals and broaden the detection range of electrodes. In imaging, functionalized magnetic nanoparticles enhance MRI sensitivity, facilitating localization of the epileptogenic zone. NIR-II nanoprobes enable tracking of seizure-related biomarkers with deep tissue penetration. Furthermore, nanomaterials enhance the sensitivity of biochemical sensors for detecting epilepsy biomarkers, which is crucial for early detection. These advancements significantly increase diagnostic sensitivity and specificity. However, challenges remain, particularly regarding biosafety, quality control, and the scalability of fabrication processes. Overcoming these obstacles is essential for successful clinical translation. Artificial-intelligence-based big data analytics can facilitate the development of diagnostic tools by screening nanomaterials with specific properties. This approach may help to address current limitations and improve both effectiveness and safety. This review explores the application of nanomaterials in the diagnosis and detection of epilepsy, with the objective of inspiring innovative ideas and strategies to enhance diagnostic effectiveness.
Collapse
Affiliation(s)
- Wanbin Huang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiabin Zong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ming Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tong-Fei Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Songqing Pan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zheman Xiao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
2
|
Batir-Marin D, Boev M, Cioanca O, Lungu II, Marin GA, Burlec AF, Mitran AM, Mircea C, Hancianu M. Exploring Oxidative Stress Mechanisms of Nanoparticles Using Zebrafish ( Danio rerio): Toxicological and Pharmaceutical Insights. Antioxidants (Basel) 2025; 14:489. [PMID: 40298867 PMCID: PMC12024358 DOI: 10.3390/antiox14040489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Nanoparticles (NPs) have revolutionized biomedical and pharmaceutical applications due to their unique physicochemical properties. However, their widespread use has raised concerns regarding their potential toxicity, particularly mediated by oxidative stress mechanisms. This redox imbalance, primarily driven by the overproduction of reactive oxygen species (ROS), plays a central role in NP-induced toxicity, leading to cellular dysfunction, inflammation, apoptosis, and genotoxicity. Zebrafish (Danio rerio) have emerged as a powerful in vivo model for nanotoxicology, offering advantages such as genetic similarity to humans, rapid development, and optical transparency, allowing real-time monitoring of oxidative damage. This review synthesizes current findings on NP-induced oxidative stress in zebrafish, highlighting key toxicity mechanisms and case studies involving metallic (gold, silver, copper), metal oxide (zinc oxide, titanium dioxide, iron oxide), polymeric, and lipid-based NPs. The influence of NP physicochemical properties, such as size, surface charge, and functionalization, on oxidative stress responses is explored. Additionally, experimental approaches used to assess ROS generation, antioxidant enzyme activity, and oxidative damage biomarkers in zebrafish models are examined. In addition to toxicity concerns, pharmaceutical applications of antioxidant-modified NPs are evaluated, particularly their potential in drug delivery, neuroprotection, and disease therapeutics. Notably, studies show that curcumin- and quercetin-loaded nanoparticles enhance antioxidant defense and reduce neurotoxicity in zebrafish models, demonstrating their promise in neuroprotective therapies. Furthermore, cerium oxide nanoparticles, which mimic catalase and SOD enzymatic activity, have shown significant efficacy in reducing ROS and protecting against oxidative damage. Challenges in zebrafish-based nanotoxicology, the need for standardized methodologies, and future directions for optimizing NP design to minimize oxidative stress-related risks are also discussed. By integrating insights from toxicity mechanisms, case studies, and pharmaceutical strategies, this review supports the development of safer and more effective nanoparticle-based therapies while addressing the challenges of oxidative stress-related toxicity.
Collapse
Affiliation(s)
- Denisa Batir-Marin
- Department of Pharmaceutical Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800008 Galati, Romania;
| | - Monica Boev
- Department of Pharmaceutical Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800008 Galati, Romania;
| | - Oana Cioanca
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-I.L.); (G.-A.M.); (A.F.B.); (A.-M.M.); (C.M.); (M.H.)
| | - Ionut-Iulian Lungu
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-I.L.); (G.-A.M.); (A.F.B.); (A.-M.M.); (C.M.); (M.H.)
| | - George-Alexandru Marin
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-I.L.); (G.-A.M.); (A.F.B.); (A.-M.M.); (C.M.); (M.H.)
| | - Ana Flavia Burlec
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-I.L.); (G.-A.M.); (A.F.B.); (A.-M.M.); (C.M.); (M.H.)
| | - Andreea-Maria Mitran
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-I.L.); (G.-A.M.); (A.F.B.); (A.-M.M.); (C.M.); (M.H.)
| | - Cornelia Mircea
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-I.L.); (G.-A.M.); (A.F.B.); (A.-M.M.); (C.M.); (M.H.)
| | - Monica Hancianu
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-I.L.); (G.-A.M.); (A.F.B.); (A.-M.M.); (C.M.); (M.H.)
| |
Collapse
|
3
|
Saba L, Cau R, Vergallo R, Kooi ME, Staub D, Faa G, Congiu T, Ntaios G, Wasserman BA, Benson J, Nardi V, Kawakami R, Lanzino G, Virmani R, Libby P. Carotid artery atherosclerosis: mechanisms of instability and clinical implications. Eur Heart J 2025; 46:904-921. [PMID: 39791527 DOI: 10.1093/eurheartj/ehae933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/25/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Cardiovascular disease remains a prominent cause of disability and premature death worldwide. Within this spectrum, carotid artery atherosclerosis is a complex and multifaceted condition, and a prominent precursor of acute ischaemic stroke and other cardiovascular events. The intricate interplay among inflammation, oxidative stress, endothelial dysfunction, lipid metabolism, and immune responses participates in the development of lesions, leading to luminal stenosis and potential plaque instability. Even non-stenotic plaques can precipitate a sudden cerebrovascular event, regardless of the degree of luminal encroachment. In this context, carotid imaging modalities have proved their efficacy in providing in vivo characterization of plaque features, contributing substantially to patient risk stratification and clinical management. This review emphasizes the importance of identifying high-risk individuals by use of current imaging modalities, biomarkers, and risk stratification tools. Such approaches inform early intervention and the implementation of personalized therapeutic strategies, ultimately enhancing patient outcomes in the realm of cardiovascular disease management.
Collapse
Affiliation(s)
- Luca Saba
- Department of Radiology, University of Cagliari, Via Università, 40, 09124 Cagliari, Italy
| | - Riccardo Cau
- Department of Radiology, University of Cagliari, Via Università, 40, 09124 Cagliari, Italy
| | - Rocco Vergallo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - M Eline Kooi
- Department of Radiology and Nuclear Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Daniel Staub
- Vascular Medicine/Angiology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Gavino Faa
- Department of Pathology, University of Cagliari, Cagliari, Italy
| | - Terenzio Congiu
- Department of Pathology, University of Cagliari, Cagliari, Italy
| | - George Ntaios
- Department of Internal Medicine, School of Health Sciences, University of Thessaly, Larissa University Hospital, Larissa 41132, Greece
| | - Bruce A Wasserman
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, MD, USA
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, MD, USA
| | - John Benson
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Valentina Nardi
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Inc., Gaithersburg, MD, USA
| | | | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Xie S, Zhu X, Han F, Wang S, Cui K, Xue J, Xi X, Shi C, Li S, Wang F, Tian J. Discussion on the comparison of Raman spectroscopy and cardiovascular disease-related imaging techniques and the future applications of Raman technology: a systematic review. Lasers Med Sci 2025; 40:116. [PMID: 39988624 PMCID: PMC11847755 DOI: 10.1007/s10103-025-04315-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/23/2025] [Indexed: 02/25/2025]
Abstract
Cardiovascular disease (CVD) is a major cause of unnatural death worldwide, so timely diagnosis of CVD is crucial for improving patient outcomes. Although the traditional diagnostic tools can locate plaque and observe inner wall of blood vessel structure, they commonly have radioactivity and cannot detect the chemical composition of the plaque accurately. Recently emerging Raman techniques can detect the plaque composition precisely, and have the advantages of being fast, high-resolution and marker-free. This makes Raman have great potential for detecting blood samples, understanding disease conditions, and real-time monitoring. This review summarizes the origin and state-of-art of Raman techniques, including the following aspects: (a) the principle and technical classification of Raman techniques; (b) the applicability of Raman techniques and its comparison with traditional diagnostic tools at different diagnosis targets; (c) the applicability of Raman spectroscopy in advanced CVD. Lastly, we highlight the possible future applications of Raman techniques in CVD diagnosis.
Collapse
Affiliation(s)
- Songcai Xie
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaotong Zhu
- Wuhan National Laboratory for Optoelectronics, Hua zhong Univeresity of Science and Technology, Wuhan, China
| | - Feiyuan Han
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shengyuan Wang
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kexin Cui
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Xue
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangwen Xi
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengyu Shi
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuo Li
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fan Wang
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China.
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jinwei Tian
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China.
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
5
|
Cau R, Anzalone N, Mannelli L, Edjlali M, Balestrieri A, Nardi V, Lanzino G, Lerman A, Suri JS, Saba L. Pericarotid Fat as a Marker of Cerebrovascular Risk. AJNR Am J Neuroradiol 2024; 45:1635-1641. [PMID: 39147585 PMCID: PMC11543090 DOI: 10.3174/ajnr.a8300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/27/2024] [Indexed: 08/17/2024]
Abstract
Vascular inflammation is widely recognized as an important factor in the atherosclerotic process, particularly in terms of plaque development and progression. Conventional tests, such as measuring circulating inflammatory biomarkers, lack the precision to identify specific areas of vascular inflammation. In this context, noninvasive imaging modalities can detect perivascular fat changes, serving as a marker of vascular inflammation. This review aims to provide a comprehensive overview of the key concepts related to perivascular carotid fat and its pathophysiology. Additionally, we examine the existing literature on the association of pericarotid fat with features of plaque vulnerability and cerebrovascular events. Finally, we scrutinize the advantages and limitations of the noninvasive assessment of pericarotid fat.
Collapse
Affiliation(s)
- Riccardo Cau
- From the Department of Radiology (R.C., A.B., L.S.), Azienda Ospedaliero Universitaria (A.O.U.), Cagliari, Italy
| | - Nicoletta Anzalone
- Vita-Salute San Raffaele University (N.A.), Milan, Italy
- Neuroradiology Unit and CERMAC (N.A.), IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Myriam Edjlali
- Department of Neuroradiology (M.E.), Université Paris-Descartes-Sorbonne-Paris-Cité, IMABRAIN-INSERM-UMR1266, DHU-Neurovasc, Centre Hospitalier Sainte-Anne, Paris, France
| | - Antonella Balestrieri
- From the Department of Radiology (R.C., A.B., L.S.), Azienda Ospedaliero Universitaria (A.O.U.), Cagliari, Italy
| | - Valentina Nardi
- Department of Neurosurgery (V.N., G.L.), Mayo Clinic, Rochester, Minnesota
| | - Giuseppe Lanzino
- Department of Neurosurgery (V.N., G.L.), Mayo Clinic, Rochester, Minnesota
| | - Amir Lerman
- Department of Cardiovascular Medicine (A.L.), Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division (J.S.S.), AtheroPoint, Roseville, California
| | - Luca Saba
- From the Department of Radiology (R.C., A.B., L.S.), Azienda Ospedaliero Universitaria (A.O.U.), Cagliari, Italy
| |
Collapse
|
6
|
Shi Z, Huang J, Chen C, Zhang X, Ma Z, Liu Q. Lipid nanoparticles encapsulating curcumin for imaging and stabilization of vulnerable atherosclerotic plaques via phagocytic "eat-me" signals. J Control Release 2024; 373:265-276. [PMID: 39019087 DOI: 10.1016/j.jconrel.2024.07.027] [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: 02/25/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
Curcumin potentiates the stabilization of atherosclerotic plaques by polarizing macrophages, but its non-specific targeting hinders its clinical application. We aim to harness multifunctional lipid nanoparticles (MLNPs) to facilitate the imaging and targeted delivery of curcumin specifically to inflammatory macrophages, counteracting vulnerable plaques and mitigating the risk of ischemic events. Cholesteryl-9-carboxynonanoate-(125I‑iron oxide nanoparticle/Curcumin)-lipid-coated nanoparticles [9-CCN-(125I-ION/Cur)-LNPs], namely MLNPs, are designed to carry hybrid imaging agents. These agents combine 125I-ION with lipids containing phagocytic 'eat-me' signals, inducing macrophages to engulf the MLNPs. Our research demonstrates that the designed MLNPs accurately accumulate at unstable plaques and are precisely visualized and highlighted by both SPECT and MRI. Furthermore, MLNPs achieve high efficiency in delivering 125I-ION and curcumin to macrophages, ultimately leading to significant M1-to-M2 macrophage polarization. These real-time imaging and polarization capabilities of plaques have immediate clinical applicability and may pave the way for novel therapies to stabilize unstable atherosclerotic plaques.
Collapse
Affiliation(s)
- Zhang Shi
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China; Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Huang
- Department of Radiology, The Second Naval Hospital of Southern Theater Command of PLA, Sanya, China
| | - Chao Chen
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xuefeng Zhang
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhiqiang Ma
- Pharmacy School, Naval Medical University, Shanghai, China.
| | - Qi Liu
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
7
|
Shah A, Neitzel E, Panda A, Fananapazir G. The use of ferumoxytol for high-resolution vascular imaging and troubleshooting for abdominal allografts. Abdom Radiol (NY) 2024; 49:2858-2872. [PMID: 38561553 DOI: 10.1007/s00261-024-04268-x] [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: 02/02/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 04/04/2024]
Abstract
Ferumoxytol is an ultrasmall superparamagnetic iron oxide which has been used as an off-label intravenous contrast agent for MRI. Unlike gadolinium-based contrast agents, ferumoxytol remains in the intravascular space with a long half-life of 14-21 h. During the first several hours, it acts as a blood-pool agent and has minimal parenchymal enhancement. Studies have shown adequate intravascular signal for up to 72 h after initial contrast bolus. Ferumoxytol has been shown to be safe, even in patients with renal failure. Ferumoxytol has shown promise in a variety of clinical settings. The exquisite resolution enabled by the long intravascular times and lack of background parenchymal enhancement is of particular interest in the vascular imaging of solid organ allografts. Ferumoxytol magnetic resonance angiography (MRA) may identify clinically actionable findings months before ultrasound, CT angiography, or Gadolinium-enhanced MRA. Ferumoxytol MRA is of particular benefit as a troubleshooting tool in the setting of equivocal ultrasound and CT imaging. In the following review, we highlight the use of ferumoxytol for high-resolution MR vascular imaging for abdominal solid organ allografts, with representative cases.
Collapse
Affiliation(s)
- Amar Shah
- Department of Radiology, Mayo Clinic in Arizona, Phoenix, AZ, USA.
| | - Easton Neitzel
- University of Arizona School of Medicine, Phoenix, AZ, USA
| | - Anshuman Panda
- Department of Radiology, Mayo Clinic in Arizona, Phoenix, AZ, USA
- Department of Medical Physics, Mayo Clinic in Arizona, Phoenix, AZ, USA
| | - Ghaneh Fananapazir
- Department of Radiology, Mayo Clinic in Arizona, Phoenix, AZ, USA
- Department of Radiology, University of California Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
8
|
Wang S, He H, Mao Y, Zhang Y, Gu N. Advances in Atherosclerosis Theranostics Harnessing Iron Oxide-Based Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308298. [PMID: 38368274 PMCID: PMC11077671 DOI: 10.1002/advs.202308298] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/06/2024] [Indexed: 02/19/2024]
Abstract
Atherosclerosis, a multifaceted chronic inflammatory disease, has a profound impact on cardiovascular health. However, the critical limitations of atherosclerosis management include the delayed detection of advanced stages, the intricate assessment of plaque stability, and the absence of efficacious therapeutic strategies. Nanotheranostic based on nanotechnology offers a novel paradigm for addressing these challenges by amalgamating advanced imaging capabilities with targeted therapeutic interventions. Meanwhile, iron oxide nanoparticles have emerged as compelling candidates for theranostic applications in atherosclerosis due to their magnetic resonance imaging capability and biosafety. This review delineates the current state and prospects of iron oxide nanoparticle-based nanotheranostics in the realm of atherosclerosis, including pivotal aspects of atherosclerosis development, the pertinent targeting strategies involved in disease pathogenesis, and the diagnostic and therapeutic roles of iron oxide nanoparticles. Furthermore, this review provides a comprehensive overview of theranostic nanomedicine approaches employing iron oxide nanoparticles, encompassing chemical therapy, physical stimulation therapy, and biological therapy. Finally, this review proposes and discusses the challenges and prospects associated with translating these innovative strategies into clinically viable anti-atherosclerosis interventions. In conclusion, this review offers new insights into the future of atherosclerosis theranostic, showcasing the remarkable potential of iron oxide-based nanoparticles as versatile tools in the battle against atherosclerosis.
Collapse
Affiliation(s)
- Shi Wang
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Hongliang He
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Yu Mao
- School of MedicineNanjing UniversityNanjing210093P. R. China
| | - Yu Zhang
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Ning Gu
- School of MedicineNanjing UniversityNanjing210093P. R. China
| |
Collapse
|
9
|
Haase T, Ludwig A, Stach A, Mohtashamdolatshahi A, Hauptmann R, Mundhenk L, Kratz H, Metzkow S, Kader A, Freise C, Mueller S, Stolzenburg N, Radon P, Liebl M, Wiekhorst F, Hamm B, Taupitz M, Schnorr J. Repeated Injection of Very Small Superparamagnetic Iron Oxide Particles (VSOPs) in Murine Atherosclerosis: A Safety Study. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:773. [PMID: 38727367 PMCID: PMC11085881 DOI: 10.3390/nano14090773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024]
Abstract
Citrate-coated electrostatically stabilized very small superparamagnetic iron oxide particles (VSOPs) have been successfully tested as magnetic resonance angiography (MRA) contrast agents and are promising tools for molecular imaging of atherosclerosis. Their repeated use in the background of pre-existing hyperlipidemia and atherosclerosis has not yet been studied. This study aimed to investigate the effect of multiple intravenous injections of VSOPs in atherosclerotic mice. Taurine-formulated VSOPs (VSOP-T) were repeatedly intravenously injected at 100 µmol Fe/kg in apolipoprotein E-deficient (ApoE KO) mice with diet-induced atherosclerosis. Angiographic imaging was carried out by in vivo MRI. Magnetic particle spectrometry was used to detect tissue VSOP content, and tissue iron content was quantified photometrically. Pathological changes in organs, atherosclerotic plaque development, and expression of hepatic iron-related proteins were evaluated. VSOP-T enabled the angiographic imaging of heart and blood vessels with a blood half-life of one hour. Repeated intravenous injection led to VSOP deposition and iron accumulation in the liver and spleen without affecting liver and spleen pathology, expression of hepatic iron metabolism proteins, serum lipids, or atherosclerotic lesion formation. Repeated injections of VSOP-T doses sufficient for MRA analyses had no significant effects on plaque burden, steatohepatitis, and iron homeostasis in atherosclerotic mice. These findings underscore the safety of VSOP-T and support its further development as a contrast agent and molecular imaging tool.
Collapse
Affiliation(s)
- Tobias Haase
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Antje Ludwig
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10117 Berlin, Germany
| | - Anke Stach
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Azadeh Mohtashamdolatshahi
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Ralf Hauptmann
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Lars Mundhenk
- Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Str. 15, 14163 Berlin, Germany;
| | - Harald Kratz
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Susanne Metzkow
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Avan Kader
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Christian Freise
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Susanne Mueller
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
- Charité 3R|Replace, Reduce, Refine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Nicola Stolzenburg
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Patricia Radon
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587 Berlin, Germany; (P.R.); (M.L.); (F.W.)
| | - Maik Liebl
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587 Berlin, Germany; (P.R.); (M.L.); (F.W.)
| | - Frank Wiekhorst
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587 Berlin, Germany; (P.R.); (M.L.); (F.W.)
| | - Bernd Hamm
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Matthias Taupitz
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Jörg Schnorr
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
10
|
Lungu CN, Creteanu A, Mehedinti MC. Endovascular Drug Delivery. Life (Basel) 2024; 14:451. [PMID: 38672722 PMCID: PMC11051410 DOI: 10.3390/life14040451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/12/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Drug-eluting stents (DES) and balloons revolutionize atherosclerosis treatment by targeting hyperplastic tissue responses through effective local drug delivery strategies. This review examines approved and emerging endovascular devices, discussing drug release mechanisms and their impacts on arterial drug distribution. It emphasizes the crucial role of drug delivery in modern cardiovascular care and highlights how device technologies influence vascular behavior based on lesion morphology. The future holds promise for lesion-specific treatments, particularly in the superficial femoral artery, with recent CE-marked devices showing encouraging results. Exciting strategies and new patents focus on local drug delivery to prevent restenosis, shaping the future of interventional outcomes. In summary, as we navigate the ever-evolving landscape of cardiovascular intervention, it becomes increasingly evident that the future lies in tailoring treatments to the specific characteristics of each lesion. By leveraging cutting-edge technologies and harnessing the potential of localized drug delivery, we stand poised to usher in a new era of precision medicine in vascular intervention.
Collapse
Affiliation(s)
- Claudiu N. Lungu
- Department of Functional and Morphological Science, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800010 Galati, Romania;
| | - Andreea Creteanu
- Department of Pharmaceutical Technology, University of Medicine and Pharmacy Grigore T Popa, 700115 Iași, Romania
| | - Mihaela C. Mehedinti
- Department of Functional and Morphological Science, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800010 Galati, Romania;
| |
Collapse
|
11
|
Wen L, Fu X, Zhang H, Ye P, Fu H, Zhou Z, Sun R, Xu T, Fu C, Zhu C, Guo Y, Fan H. Tailoring Zinc Ferrite Nanoparticle Surface Coating for Macrophage-Affinity Magnetic Resonance Imaging of Atherosclerosis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:13496-13508. [PMID: 38449094 DOI: 10.1021/acsami.3c17212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the formation of atherosclerotic plaques, while macrophages as key players in plaque progression and destabilization are promising targets for atherosclerotic plaque imaging. Contrast-enhanced magnetic resonance imaging (CE-MRI) has emerged as a powerful noninvasive imaging technique for the evaluation of atherosclerotic plaques within arterial walls. However, the visualization of macrophages within atherosclerotic plaques presents considerable challenges due to the intricate pathophysiology of the disease and the dynamic behavior of these cells. Biocompatible ferrite nanoparticles with diverse surface ligands possess the potential to exhibit distinct relaxivity and cellular affinity, enabling improved imaging capabilities for macrophages in atherosclerosis. In this work, we report macrophage-affinity nanoparticles for magnetic resonance imaging (MRI) of atherosclerosis via tailoring nanoparticle surface coating. The ultrasmall zinc ferrite nanoparticles (Zn0.4Fe2.6O4) as T1 contrast agents were synthesized and modified with dopamine, 3,4-dihydroxyhydrocinnamic acid, and phosphorylated polyethylene glycol to adjust their surface charges to be positively, negatively, and neutrally charged, respectively. In vitro MRI evaluation shows that the T1 relaxivity for different surface charged Zn0.4Fe2.6O4 nanoparticles was three higher than that of the clinically used Gd-DTPA. Furthermore, in vivo atherosclerotic plaque MR imaging indicates that positively charged Zn0.4Fe2.6O4 showed superior MRI efficacy on carotid atherosclerosis than the other two, which is ascribed to high affinity to macrophages of positively charged nanoparticles. This work provides improved diagnostic capability and a better understanding of the molecular imaging of atherosclerosis.
Collapse
Affiliation(s)
- Lingyi Wen
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Xiaomin Fu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Huan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, China
- School of Medicine, Northwest University, Xi'an 710069, China
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Pengfei Ye
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Hang Fu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Zhongqin Zhou
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Ran Sun
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Ting Xu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Chuan Fu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Chengcheng Zhu
- Department of Radiology, University of Washington, Seattle, Washington 98105, United States
| | - Yingkun Guo
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Haiming Fan
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, China
- School of Medicine, Northwest University, Xi'an 710069, China
| |
Collapse
|
12
|
Zou L, Zhang Y, Cheraga N, Abodunrin OD, Qu KY, Qiao L, Ma YQ, Hang Y, Huang NP, Chen LJ. M2 Macrophage Membrane-Camouflaged Fe 3 O 4 -Cy7 Nanoparticles with Reduced Immunogenicity for Targeted NIR/MR Imaging of Atherosclerosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304110. [PMID: 37806756 DOI: 10.1002/smll.202304110] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/27/2023] [Indexed: 10/10/2023]
Abstract
Atherosclerosis (AS) is the primary reason behind cardiovascular diseases, leading to approximately one-third of global deaths. Developing a novel multi-model probe to detect AS is urgently required. Macrophages are the primary cells from which AS genesis occurs. Utilizing natural macrophage membranes coated on the surface of nanoparticles is an efficient delivery method to target plaque sites. Herein, Fe3 O4 -Cy7 nanoparticles (Fe3 O4 -Cy7 NPs), functionalized using an M2 macrophage membrane and a liposome extruder for Near-infrared fluorescence and Magnetic resonance imaging, are synthesized. These macrophage membrane-coated nanoparticles (Fe3 O4 @M2 NPs) enhance the recognition and uptake using active macrophages. Moreover, they inhibit uptake using inactive macrophages and human coronary artery endothelial cells. The macrophage membrane-coated nanoparticles (Fe3 O4 @M0 NPs, Fe3 O4 @M1 NPs, Fe3 O4 @M2 NPs) can target specific sites depending on the macrophage membrane type and are related to C-C chemofactor receptor type 2 protein content. Moreover, Fe3 O4 @M2 NPs demonstrate excellent biosafety in vivo after injection, showing a significantly higher Fe concentration in the blood than Fe3 O4 -Cy7 NPs. Therefore, Fe3 O4 @M2 NPs effectively retain the physicochemical properties of nanoparticles and depict reduced immunological response in blood circulation. These NPs mainly reveal enhanced targeting imaging capability for atherosclerotic plaque lesions.
Collapse
Affiliation(s)
- Lin Zou
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Yao Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 210009, Nanjing, China
| | - Nihad Cheraga
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Oluwatosin David Abodunrin
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Kai-Yun Qu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Li Qiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Yu-Qing Ma
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Yue Hang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Ning-Ping Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Li-Juan Chen
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, 211200, Nanjing, China
| |
Collapse
|
13
|
Nadel J, Wang X, Saha P, Bongers A, Tumanov S, Giannotti N, Chen W, Vigder N, Chowdhury MM, da Cruz GL, Velasco C, Prieto C, Jabbour A, Botnar RM, Stocker R, Phinikaridou A. Molecular magnetic resonance imaging of myeloperoxidase activity identifies culprit lesions and predicts future atherothrombosis. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2024; 2:qyae004. [PMID: 38370393 PMCID: PMC10870993 DOI: 10.1093/ehjimp/qyae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 01/22/2024] [Indexed: 02/20/2024]
Abstract
Aims Unstable atherosclerotic plaques have increased activity of myeloperoxidase (MPO). We examined whether molecular magnetic resonance imaging (MRI) of intraplaque MPO activity predicts future atherothrombosis in rabbits and correlates with ruptured human atheroma. Methods and results Plaque MPO activity was assessed in vivo in rabbits (n = 12) using the MPO-gadolinium (Gd) probe at 8 and 12 weeks after induction of atherosclerosis and before pharmacological triggering of atherothrombosis. Excised plaques were used to confirm MPO activity by liquid chromatography-tandem mass spectrometry (LC-MSMS) and to determine MPO distribution by histology. MPO activity was higher in plaques that caused post-trigger atherothrombosis than plaques that did not. Among the in vivo MRI metrics, the plaques' R1 relaxation rate after administration of MPO-Gd was the best predictor of atherothrombosis. MPO activity measured in human carotid endarterectomy specimens (n = 30) by MPO-Gd-enhanced MRI was correlated with in vivo patient MRI and histological plaque phenotyping, as well as LC-MSMS. MPO-Gd retention measured as the change in R1 relaxation from baseline was significantly greater in histologic and MRI-graded American Heart Association (AHA) type VI than type III-V plaques. This association was confirmed by comparing AHA grade to MPO activity determined by LC-MSMS. Conclusion We show that elevated intraplaque MPO activity detected by molecular MRI employing MPO-Gd predicts future atherothrombosis in a rabbit model and detects ruptured human atheroma, strengthening the translational potential of this approach to prospectively detect high-risk atherosclerosis.
Collapse
Affiliation(s)
- James Nadel
- Heart Research Institute, Arterial Inflammation and Redox Biology Group, 7 Eliza St, Newtown, Sydney, NSW 2042, Australia
- Department of Cardiology, St Vincent’s Hospital, Sydney, NSW, Australia
- Department of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Xiaoying Wang
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Prakash Saha
- Academic Department of Surgery, Cardiovascular Division, King’s College London, London, UK
| | - André Bongers
- Biological Resources Imaging Laboratory, University of New South Wales, Sydney, NSW, Australia
| | - Sergey Tumanov
- Heart Research Institute, Arterial Inflammation and Redox Biology Group, 7 Eliza St, Newtown, Sydney, NSW 2042, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Nicola Giannotti
- Medical Imaging Science, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Weiyu Chen
- Heart Research Institute, Arterial Inflammation and Redox Biology Group, 7 Eliza St, Newtown, Sydney, NSW 2042, Australia
| | - Niv Vigder
- Heart Research Institute, Arterial Inflammation and Redox Biology Group, 7 Eliza St, Newtown, Sydney, NSW 2042, Australia
| | | | | | - Carlos Velasco
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Claudia Prieto
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
- Pontificia Universidad Católica de Chile, Institute for Biological and Medical Engineering, Santiago, Chile
| | - Andrew Jabbour
- Department of Cardiology, St Vincent’s Hospital, Sydney, NSW, Australia
- Department of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - René M Botnar
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
- Pontificia Universidad Católica de Chile, Institute for Biological and Medical Engineering, Santiago, Chile
- King’s BHF Centre of Research Excellence, London, UK
| | - Roland Stocker
- Heart Research Institute, Arterial Inflammation and Redox Biology Group, 7 Eliza St, Newtown, Sydney, NSW 2042, Australia
| | - Alkystis Phinikaridou
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
- King’s BHF Centre of Research Excellence, London, UK
| |
Collapse
|
14
|
Aalbregt E, Rijken L, Nederveen A, van Ooij P, Yeung KK, Jongkind V. Quantitative Magnetic Resonance Imaging to Assess Progression and Rupture Risk of Aortic Aneurysms: A Scoping Review. J Endovasc Ther 2023:15266028231204830. [PMID: 37853734 DOI: 10.1177/15266028231204830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
PURPOSE In current practice, the diameter of an aortic aneurysm is utilized to estimate the rupture risk and decide upon timing of elective repair, although it is known to be imprecise and not patient-specific. Quantitative magnetic resonance imaging (MRI) enables the visualization of several biomarkers that provide information about processes within the aneurysm and may therefore facilitate patient-specific risk stratification. We performed a scoping review of the literature on quantitative MRI techniques to assess aortic aneurysm progression and rupture risk, summarized these findings, and identified knowledge gaps. METHODS Literature concerning primary research was of interest and the medical databases PubMed, Scopus, Embase, and Cochrane were systematically searched. This study used the PRISMA protocol extension for scoping reviews. Articles published between January 2010 and February 2023 involving animals and/or humans were included. Data were extracted by 2 authors using a predefined charting method. RESULTS A total of 1641 articles were identified, of which 21 were included in the scoping review. Quantitative MRI-derived biomarkers were categorized into hemodynamic (8 studies), wall (5 studies) and molecular biomarkers (8 studies). Fifteen studies included patients and/or healthy human subjects. Animal models were investigated in the other 6 studies. A cross-sectional study design was the most common, whereas 5 animal studies had a longitudinal component and 2 studies including patients had a prospective design. A promising hemodynamic biomarker is wall shear stress (WSS), which is estimated based on 4D-flow MRI. Molecular biomarkers enable the assessment of inflammatory and wall deterioration processes. The ADAMTS4-specific molecular magnetic resonance (MR) probe showed potential to predict abdominal aortic aneurysm (AAA) formation and rupture in a murine model. Wall biomarkers assessed using dynamic contrast-enhanced (DCE) MRI showed great potential for assessing AAA progression independent of the maximum diameter. CONCLUSION This scoping review provides an overview of quantitative MRI techniques studied and the biomarkers derived from them to assess aortic aneurysm progression and rupture risk. Longitudinal studies are needed to validate the causal relationships between the identified biomarkers and aneurysm growth, rupture, or repair. In the future, quantitative MRI could play an important role in the personalized risk assessment of aortic aneurysm rupture. CLINICAL IMPACT The currently used maximum aneurysm diameter fails to accurately assess the multifactorial pathology of an aortic aneurysm and precisely predicts rupture in a patient-specific manner. Quantitative magnetic resonance imaging (MRI) enables the detection of various quantitative parameters involved in aneurysm progression and subsequent rupture. This scoping review provides an overview of the studied quantitative MRI techniques, the biomarkers derived from them, and recommendations for future research needed for the implementation of these biomarkers. Ultimately, quantitative MRI could facilitate personalized risk assessment for patients with aortic aneurysms, thereby reducing untimely repairs and improving rupture prevention.
Collapse
Affiliation(s)
- Eva Aalbregt
- Department of Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Lotte Rijken
- Department of Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Aart Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Pim van Ooij
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Vincent Jongkind
- Department of Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Kopyto E, Czeczelewski M, Mikos E, Stępniak K, Kopyto M, Matuszek M, Nieoczym K, Czarnecki A, Kuczyńska M, Cheda M, Drelich-Zbroja A, Jargiełło T. Contrast-Enhanced Ultrasound Feasibility in Assessing Carotid Plaque Vulnerability-Narrative Review. J Clin Med 2023; 12:6416. [PMID: 37835061 PMCID: PMC10573420 DOI: 10.3390/jcm12196416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
The risk assessment for carotid atherosclerotic lesions involves not only determining the degree of stenosis but also plaque morphology and its composition. Recently, carotid contrast-enhanced ultrasound (CEUS) has gained importance for evaluating vulnerable plaques. This review explores CEUS's utility in detecting carotid plaque surface irregularities and ulcerations as well as intraplaque neovascularization and its alignment with histology. Initial indications suggest that CEUS might have the potential to anticipate cerebrovascular incidents. Nevertheless, there is a need for extensive, multicenter prospective studies that explore the relationships between CEUS observations and patient clinical outcomes in cases of carotid atherosclerotic disease.
Collapse
Affiliation(s)
- Ewa Kopyto
- Students’ Scientific Society, Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (E.K.); (E.M.); (K.S.); (M.K.); (M.M.); (K.N.); (A.C.)
| | - Marcin Czeczelewski
- Students’ Scientific Society, Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (E.K.); (E.M.); (K.S.); (M.K.); (M.M.); (K.N.); (A.C.)
| | - Eryk Mikos
- Students’ Scientific Society, Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (E.K.); (E.M.); (K.S.); (M.K.); (M.M.); (K.N.); (A.C.)
| | - Karol Stępniak
- Students’ Scientific Society, Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (E.K.); (E.M.); (K.S.); (M.K.); (M.M.); (K.N.); (A.C.)
| | - Maja Kopyto
- Students’ Scientific Society, Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (E.K.); (E.M.); (K.S.); (M.K.); (M.M.); (K.N.); (A.C.)
| | - Małgorzata Matuszek
- Students’ Scientific Society, Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (E.K.); (E.M.); (K.S.); (M.K.); (M.M.); (K.N.); (A.C.)
| | - Karolina Nieoczym
- Students’ Scientific Society, Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (E.K.); (E.M.); (K.S.); (M.K.); (M.M.); (K.N.); (A.C.)
| | - Adam Czarnecki
- Students’ Scientific Society, Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (E.K.); (E.M.); (K.S.); (M.K.); (M.M.); (K.N.); (A.C.)
| | - Maryla Kuczyńska
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (M.K.); (M.C.); (A.D.-Z.); (T.J.)
| | - Mateusz Cheda
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (M.K.); (M.C.); (A.D.-Z.); (T.J.)
| | - Anna Drelich-Zbroja
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (M.K.); (M.C.); (A.D.-Z.); (T.J.)
| | - Tomasz Jargiełło
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-594 Lublin, Poland; (M.K.); (M.C.); (A.D.-Z.); (T.J.)
| |
Collapse
|
16
|
Burlec AF, Corciova A, Boev M, Batir-Marin D, Mircea C, Cioanca O, Danila G, Danila M, Bucur AF, Hancianu M. Current Overview of Metal Nanoparticles' Synthesis, Characterization, and Biomedical Applications, with a Focus on Silver and Gold Nanoparticles. Pharmaceuticals (Basel) 2023; 16:1410. [PMID: 37895881 PMCID: PMC10610223 DOI: 10.3390/ph16101410] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/23/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Metal nanoparticles (NPs) have garnered considerable attention, due to their unique physicochemical properties, that render them promising candidates for various applications in medicine and industry. This article offers a comprehensive overview of the most recent advancements in the manufacturing, characterization, and biomedical utilization of metal NPs, with a primary focus on silver and gold NPs. Their potential as effective anticancer, anti-inflammatory, and antimicrobial agents, drug delivery systems, and imaging agents in the diagnosis and treatment of a variety of disorders is reviewed. Moreover, their translation to therapeutic settings, and the issue of their inclusion in clinical trials, are assessed in light of over 30 clinical investigations that concentrate on administering either silver or gold NPs in conditions ranging from nosocomial infections to different types of cancers. This paper aims not only to examine the biocompatibility of nanomaterials but also to emphasize potential challenges that may limit their safe integration into healthcare practices. More than 100 nanomedicines are currently on the market, which justifies ongoing study into the use of nanomaterials in medicine. Overall, the present review aims to highlight the potential of silver and gold NPs as innovative and effective therapeutics in the field of biomedicine, citing some of their most relevant current applications.
Collapse
Affiliation(s)
- Ana Flavia Burlec
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (A.F.B.); (A.C.); (C.M.); (O.C.); (M.H.)
| | - Andreia Corciova
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (A.F.B.); (A.C.); (C.M.); (O.C.); (M.H.)
| | - Monica Boev
- Research Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.D.); (M.D.); (A.F.B.)
| | - Denisa Batir-Marin
- Research Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.D.); (M.D.); (A.F.B.)
| | - Cornelia Mircea
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (A.F.B.); (A.C.); (C.M.); (O.C.); (M.H.)
| | - Oana Cioanca
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (A.F.B.); (A.C.); (C.M.); (O.C.); (M.H.)
| | - Gabriela Danila
- Research Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.D.); (M.D.); (A.F.B.)
| | - Marius Danila
- Research Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.D.); (M.D.); (A.F.B.)
| | - Anca Florentina Bucur
- Research Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.D.); (M.D.); (A.F.B.)
| | - Monica Hancianu
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (A.F.B.); (A.C.); (C.M.); (O.C.); (M.H.)
| |
Collapse
|
17
|
Baj J, Kowalska B, Barbachowska A, Forma A, Flieger M, Majerek D, Teresiński G, Flieger W, Portincasa P, Buszewicz G, Radzikowska-Büchner E, Flieger J. Linking Metallic Micronutrients and Toxic Xenobiotics to Atherosclerosis and Fatty Liver Disease-Postmortem ICP-MS Analysis of Selected Human Tissues. Nutrients 2023; 15:3458. [PMID: 37571395 PMCID: PMC10420647 DOI: 10.3390/nu15153458] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Dyslipidaemia is a disorder of the lipid metabolism, caused mainly by poor eating habits. The most severe consequence of an inappropriate diet is the development of atherosclerosis and hepatic steatosis. It is generally believed that a change in nutrition, and increased physical activity can eliminate these health problems. The contemporary research and therapies used to treat dyslipidemia mainly focus on lowering the triglyceride and cholesterol levels. However, disturbances in trace element homeostasis or the accumulation of toxic elements can also affect physiological processes, and be involved in the development of metabolically mediated diseases. The present study aimed to determine the mineral profiles of liver and brain tissues collected at autopsy (n = 39) in groups of people with hepatic steatosis (n = 5), atherosclerosis (n = 9), hepatic steatosis, and atherosclerosis (n = 16), and others without the selected disorders (n = 9). Concentrations of 51 elements were analysed via inductively coupled plasma mass spectrometry (ICP-MS) after the initial wet mineralisation of the samples with nitric acid. The results obtained allow us to conclude that the hepatic steatosis group suffers from a deficiency of important trace elements, such as copper, zinc, and molybdenum (p < 0.05), whereas the group with atherosclerosis is characterised by elevated levels of cadmium in the liver tissue (p = 0.01). Analysing the mean values of the element concentrations measured in 11 brain areas, statistically significant higher levels of calcium and copper (p < 0.001) were found in the atherosclerosis group, compared to the hepatic steatosis group, confirming the involvement of these elements in the pathogenesis of atherosclerosis. In addition, an accumulation of cadmium, lead, titanium, and strontium in the brain tissue was observed in the atherosclerosis group. While the accumulation of individual elements differs in different parts of the brain, the differences in the cadmium content (p < 0.05) between the study groups apply to the whole brain, except for the nucleus accumbens septi area, where a statistically significant titanium accumulation occurs in the atherosclerosis and steatosis groups, compared to the others (p < 0.05). In addition, the disruption of elemental homeostasis in the brain of a single case with bipolar disorder, and a case with hip replacement was observed. Our results confirm the involvement of chemical elements in the pathogenesis of selected metabolic diseases, and the need for further studies in larger populations.
Collapse
Affiliation(s)
- Jacek Baj
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (W.F.)
| | - Beata Kowalska
- Department of Water Supply and Wastewater Disposal, Lublin University of Technology, 20-618 Lublin, Poland;
| | - Aleksandra Barbachowska
- Department of Plastic, Reconstructive and Burn Surgery, ul. Krasnystawska, 21-010 Łęczna, Poland;
| | - Alicja Forma
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (W.F.)
| | - Michał Flieger
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Dariusz Majerek
- Department of Applied Mathematics, University of Technology, 20-618 Lublin, Poland;
| | - Grzegorz Teresiński
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Wojciech Flieger
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (W.F.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Meical School, 70124 Bari, Italy;
| | - Grzegorz Buszewicz
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | | | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
18
|
Li Y, Wang J, Xie J. Biomimetic nanoparticles targeting atherosclerosis for diagnosis and therapy. SMART MEDICINE 2023; 2:e20230015. [PMID: 39188346 PMCID: PMC11236035 DOI: 10.1002/smmd.20230015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/28/2023] [Indexed: 08/28/2024]
Abstract
Atherosclerosis is a typical chronic inflammatory vascular disease that seriously endangers human health. At present, oral lipid-lowering or anti-inflammatory drugs are clinically used to inhibit the development of atherosclerosis. However, traditional oral drug treatments have problems such as low utilization, slow response, and serious side effects. Traditional nanodrug delivery systems are difficult to interactively recognize by normal biological organisms, and it is difficult to target the delivery of drugs to target lesions. Therefore, building a biomimetic nanodrug delivery system with targeted drug delivery based on the pathological characteristics of atherosclerosis is the key to achieving efficient and safe treatment of atherosclerosis. In this review, various nanodrug delivery systems that can target atherosclerosis are summarized and discussed. In addition, the future prospects and challenges of its clinical translation are also discussed.
Collapse
Affiliation(s)
- Yuyu Li
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Key Laboratory of Remodeling‐Related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical UniversityBeijingChina
- Beijing Institute of Heart, Lung, and Blood Vessel DiseasesBeijing Anzhen Hospital Affiliated to Capital Medical UniversityBeijingChina
| | - Jifang Wang
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Department of CardiologyDrum Tower HospitalMedical School of Nanjing UniversityNanjingChina
| | - Jun Xie
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| |
Collapse
|
19
|
Cau R, Gupta A, Kooi ME, Saba L. Pearls and Pitfalls of Carotid Artery Imaging: Ultrasound, Computed Tomography Angiography, and MR Imaging. Radiol Clin North Am 2023; 61:405-413. [PMID: 36931758 DOI: 10.1016/j.rcl.2023.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Stroke represents a major cause of morbidity and mortality worldwide with carotid atherosclerosis responsible for a large proportion of ischemic strokes. Given the high burden of the disease , early diagnosis and optimal secondary prevention are essential elements in clinical practice. For a long time, the degree of stenosis had been considered the parameter to judge the severity of carotid atherosclerosis. Over the last 30 years, literature has shifted attention from stenosis to structural characteristics of atherosclerotic lesion, eventually leading to the "vulnerable plaque" model. These "vulnerable plaques" frequently demonstrate high-risk imaging features that can be assessed by various non-invasive imaging modalities.
Collapse
Affiliation(s)
- Riccardo Cau
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari - Polo di Monserrato, s.s. 554, Monserrato, Cagliari 09045, Italy
| | - Ajay Gupta
- Department of Radiology Weill Cornell Medical College, New York, NY, USA
| | - Marianne Eline Kooi
- Department of Radiology and Nuclear Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari - Polo di Monserrato, s.s. 554, Monserrato, Cagliari 09045, Italy.
| |
Collapse
|
20
|
Chen S, Su Y, Zhang M, Zhang Y, Xiu P, Luo W, Zhang Q, Zhang X, Liang H, Lee APW, Shao L, Xiu J. Insights into the toxicological effects of nanomaterials on atherosclerosis: mechanisms involved and influence factors. J Nanobiotechnology 2023; 21:140. [PMID: 37118804 PMCID: PMC10148422 DOI: 10.1186/s12951-023-01899-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/16/2023] [Indexed: 04/30/2023] Open
Abstract
Atherosclerosis is one of the most common types of cardiovascular disease and is driven by lipid accumulation and chronic inflammation in the arteries, which leads to stenosis and thrombosis. Researchers have been working to design multifunctional nanomedicines with the ability to target, diagnose, and treat atherosclerosis, but recent studies have also identified that nanomaterials can cause atherosclerosis. Therefore, this review aims to outline the molecular mechanisms and physicochemical properties of nanomaterials that promote atherosclerosis. By analyzing the toxicological effects of nanomaterials on cells involved in the pathogenesis of atherosclerosis such as vascular endothelial cells, vascular smooth muscle cells and immune cells, we aim to provide new perspectives for the prevention and treatment of atherosclerosis, and raise awareness of nanotoxicology to advance the clinical translation and sustainable development of nanomaterials.
Collapse
Affiliation(s)
- Siyu Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuan Su
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, 528300, China
| | - Manjin Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Yulin Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Peiming Xiu
- Guangdong Medical University, Dongguan, 523808, China
| | - Wei Luo
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiuxia Zhang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinlu Zhang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongbin Liang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Alex Pui-Wai Lee
- Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| | - Jiancheng Xiu
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
21
|
Toljan K, Ashok A, Labhasetwar V, Hussain MS. Nanotechnology in Stroke: New Trails with Smaller Scales. Biomedicines 2023; 11:biomedicines11030780. [PMID: 36979759 PMCID: PMC10045028 DOI: 10.3390/biomedicines11030780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Stroke is a leading cause of death, long-term disability, and socioeconomic costs, highlighting the urgent need for effective treatment. During acute phase, intravenous administration of recombinant tissue plasminogen activator (tPA), a thrombolytic agent, and endovascular thrombectomy (EVT), a mechanical intervention to retrieve clots, are the only FDA-approved treatments to re-establish cerebral blood flow. Due to a short therapeutic time window and high potential risk of cerebral hemorrhage, a limited number of acute stroke patients benefit from tPA treatment. EVT can be performed within an extended time window, but such intervention is performed only in patients with occlusion in a larger, anatomically more proximal vasculature and is carried out at specialty centers. Regardless of the method, in case of successful recanalization, ischemia-reperfusion injury represents an additional challenge. Further, tPA disrupts the blood-brain barrier integrity and is neurotoxic, aggravating reperfusion injury. Nanoparticle-based approaches have the potential to circumvent some of the above issues and develop a thrombolytic agent that can be administered safely beyond the time window for tPA treatment. Different attributes of nanoparticles are also being explored to develop a multifunctional thrombolytic agent that, in addition to a thrombolytic agent, can contain therapeutics such as an anti-inflammatory, antioxidant, neuro/vasoprotective, or imaging agent, i.e., a theragnostic agent. The focus of this review is to highlight these advances as they relate to cerebrovascular conditions to improve clinical outcomes in stroke patients.
Collapse
Affiliation(s)
- Karlo Toljan
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Anushruti Ashok
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Vinod Labhasetwar
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Correspondence: (V.L.); (M.S.H.)
| | - M. Shazam Hussain
- Cerebrovascular Center, Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Correspondence: (V.L.); (M.S.H.)
| |
Collapse
|
22
|
Saba L, Loewe C, Weikert T, Williams MC, Galea N, Budde RPJ, Vliegenthart R, Velthuis BK, Francone M, Bremerich J, Natale L, Nikolaou K, Dacher JN, Peebles C, Caobelli F, Redheuil A, Dewey M, Kreitner KF, Salgado R. State-of-the-art CT and MR imaging and assessment of atherosclerotic carotid artery disease: the reporting-a consensus document by the European Society of Cardiovascular Radiology (ESCR). Eur Radiol 2023; 33:1088-1101. [PMID: 36194266 PMCID: PMC9889425 DOI: 10.1007/s00330-022-09025-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023]
Abstract
The European Society of Cardiovascular Radiology (ESCR) is the European specialist society of cardiac and vascular imaging. This society's highest priority is the continuous improvement, development, and standardization of education, training, and best medical practice, based on experience and evidence. The present intra-society consensus is based on the existing scientific evidence and on the individual experience of the members of the ESCR writing group on carotid diseases, the members of the ESCR guidelines committee, and the members of the executive committee of the ESCR. The recommendations published herein reflect the evidence-based society opinion of ESCR. The purpose of this second document is to discuss suggestions for standardized reporting based on the accompanying consensus document part I. KEY POINTS: • CT and MR imaging-based evaluation of carotid artery disease provides essential information for risk stratification and prediction of stroke. • The information in the report must cover vessel morphology, description of stenosis, and plaque imaging features. • A structured approach to reporting ensures that all essential information is delivered in a standardized and consistent way to the referring clinician.
Collapse
Affiliation(s)
- Luca Saba
- Department of Radiology, University of Cagliari, Cagliari, Italy
| | - Christian Loewe
- Division of Cardiovascular and Interventional Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Thomas Weikert
- Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Michelle C Williams
- BHF Centre for Cardiovascular Science, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH164SB, UK
- Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| | - Nicola Galea
- Policlinico Umberto I, Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Ricardo P J Budde
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Rozemarijn Vliegenthart
- Department of Radiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, The Netherlands
| | - Birgitta K Velthuis
- Department of Radiology, Utrecht University Medical Center, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Marco Francone
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Jens Bremerich
- Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Luigi Natale
- Department of Radiological Sciences - Institute of Radiology, Catholic University of Rome, "A. Gemelli" University Hospital, Rome, Italy
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, University of Tuebingen, Tübingen, Germany
| | - Jean-Nicolas Dacher
- Department of Radiology, Normandie University, UNIROUEN, INSERM U1096 - Rouen University Hospital, F 76000, Rouen, France
| | - Charles Peebles
- Department of Cardiothoracic Radiology, University Hospital Southampton, Southampton, UK
| | - Federico Caobelli
- University Clinic of Nuclear Medicine Inselspital Bern, University of Bern, Bern, Switzerland
| | - Alban Redheuil
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Department of Cardiovascular and Thoracic, Imaging and Interventional Radiology, Institute of Cardiology, APHP, Pitié-Salpêtrière University Hospital, Paris, France
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM 1146, CNRS 7371, Paris, France
| | - Marc Dewey
- Department of Radiology, Charité - Universitätsmedizin Berlin, Charitéplatz 1371, 10117 Berlin, Germany
| | - Karl-Friedrich Kreitner
- Department of Diagnostic and Interventional Radiology, University Medical Center, Mainz Langenbeckstraße 1, 55131, Mainz, Germany
| | - Rodrigo Salgado
- Department of Radiology, Antwerp University Hospital & Antwerp University, Holy Heart Lier, Berlaar, Belgium.
| |
Collapse
|
23
|
Saba L, Loewe C, Weikert T, Williams MC, Galea N, Budde RPJ, Vliegenthart R, Velthuis BK, Francone M, Bremerich J, Natale L, Nikolaou K, Dacher JN, Peebles C, Caobelli F, Redheuil A, Dewey M, Kreitner KF, Salgado R. State-of-the-art CT and MR imaging and assessment of atherosclerotic carotid artery disease: standardization of scanning protocols and measurements-a consensus document by the European Society of Cardiovascular Radiology (ESCR). Eur Radiol 2023; 33:1063-1087. [PMID: 36194267 PMCID: PMC9889495 DOI: 10.1007/s00330-022-09024-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023]
Abstract
The European Society of Cardiovascular Radiology (ESCR) is the European specialist society of cardiac and vascular imaging. This society's highest priority is the continuous improvement, development, and standardization of education, training, and best medical practice, based on experience and evidence. The present intra-society consensus is based on the existing scientific evidence and on the individual experience of the members of the ESCR writing group on carotid diseases, the members of the ESCR guidelines committee, and the members of the executive committee of the ESCR. The recommendations published herein reflect the evidence-based society opinion of ESCR. We have produced a twin-papers consensus, indicated through the documents as respectively "Part I" and "Part II." The first document (Part I) begins with a discussion of features, role, indications, and evidence for CT and MR imaging-based diagnosis of carotid artery disease for risk stratification and prediction of stroke (Section I). It then provides an extensive overview and insight into imaging-derived biomarkers and their potential use in risk stratification (Section II). Finally, detailed recommendations about optimized imaging technique and imaging strategies are summarized (Section III). The second part of this consensus paper (Part II) is focused on structured reporting of carotid imaging studies with CT/MR. KEY POINTS: • CT and MR imaging-based evaluation of carotid artery disease provides essential information for risk stratification and prediction of stroke. • Imaging-derived biomarkers and their potential use in risk stratification are evolving; their correct interpretation and use in clinical practice must be well-understood. • A correct imaging strategy and scan protocol will produce the best possible results for disease evaluation.
Collapse
Affiliation(s)
- Luca Saba
- Department of Radiology, University of Cagliari, Cagliari, Italy
| | - Christian Loewe
- Division of Cardiovascular and Interventional Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Thomas Weikert
- Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Michelle C Williams
- BHF Centre for Cardiovascular Science, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH164SB, UK
- Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| | - Nicola Galea
- Policlinico Umberto I, Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Ricardo P J Budde
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Rozemarijn Vliegenthart
- Department of Radiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Birgitta K Velthuis
- Department of Radiology, Utrecht University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Marco Francone
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Jens Bremerich
- Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Luigi Natale
- Department of Radiological Sciences - Institute of Radiology, Catholic University of Rome, "A. Gemelli" University Hospital, Rome, Italy
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, University of Tuebingen, Tübingen, Germany
| | - Jean-Nicolas Dacher
- Department of Radiology, Normandie University, UNIROUEN, INSERM U1096 - Rouen University Hospital, F 76000, Rouen, France
| | - Charles Peebles
- Department of Cardiothoracic Radiology, University Hospital Southampton, Southampton, UK
| | - Federico Caobelli
- University Clinic of Nuclear Medicine Inselspital Bern, University of Bern, Bern, Switzerland
| | - Alban Redheuil
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Department of Cardiovascular and Thoracic, Imaging and Interventional Radiology, Institute of Cardiology, APHP, Pitié-Salpêtrière University Hospital, Paris, France
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM 1146, CNRS 7371, Paris, France
| | - Marc Dewey
- Department of Radiology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Karl-Friedrich Kreitner
- Department of Diagnostic and Interventional Radiology, University Medical Center, Mainz; Langenbeckstraße 1, 55131, Mainz, Germany
| | - Rodrigo Salgado
- Department of Radiology, Antwerp University Hospital & Antwerp University, Holy Heart Lier, Belgium.
| |
Collapse
|
24
|
Zhang K, Xu H, Li K. Molecular Imaging for Early-Stage Disease Diagnosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1199:39-58. [PMID: 37460726 DOI: 10.1007/978-981-32-9902-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
With the development of cellular biology, molecular biology, and other subjects, targeted molecular probe was combined with medical imaging technologies to launch a new scientific discipline of molecular imaging that is a research discipline to visualize, characterize, and analyze biological process at the cellular and molecular levels for real-time tracking and precision therapy, also termed as the medical imaging in the twenty-first century. An array of imaging techniques has been developed to image specific targets of living cells or tissues by molecular probes, including optical molecular imaging (OI), magnetic resonance molecular imaging, ultrasound (US) molecular imaging, nuclear medicine molecular imaging, X-ray molecular imaging, and multi-mode molecular imaging. These imaging techniques make the early diagnosis of various diseases possible by means of visualization of gene expression, interactions between proteins, signal transduction, cell metabolism, cell traces, and other physiological or pathological processes in the living system, which bridge the gap between molecular biology and clinical medicine. This chapter will lay the emphasis on the early-stage diagnosis of fatal diseases, such as malignant tumors, cardio- or cerebrovascular diseases, digestive system disease, central nervous system disease, and other diseases employing molecular imaging in a real-time visualized manner.
Collapse
Affiliation(s)
- Kuo Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China.
| | - Haiyan Xu
- School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China
| | - Kai Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
25
|
Parry R, Majeed K, Pixley F, Hillis GS, Francis RJ, Schultz CJ. Unravelling the role of macrophages in cardiovascular inflammation through imaging: a state-of-the-art review. Eur Heart J Cardiovasc Imaging 2022; 23:e504-e525. [PMID: 35993316 PMCID: PMC9671294 DOI: 10.1093/ehjci/jeac167] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 07/31/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular disease remains the leading cause of death and disability for patients across the world. Our understanding of atherosclerosis as a primary cholesterol issue has diversified, with a significant dysregulated inflammatory component that largely remains untreated and continues to drive persistent cardiovascular risk. Macrophages are central to atherosclerotic inflammation, and they exist along a functional spectrum between pro-inflammatory and anti-inflammatory extremes. Recent clinical trials have demonstrated a reduction in major cardiovascular events with some, but not all, anti-inflammatory therapies. The recent addition of colchicine to societal guidelines for the prevention of recurrent cardiovascular events in high-risk patients with chronic coronary syndromes highlights the real-world utility of this class of therapies. A highly targeted approach to modification of interleukin-1-dependent pathways shows promise with several novel agents in development, although excessive immunosuppression and resulting serious infection have proven a barrier to implementation into clinical practice. Current risk stratification tools to identify high-risk patients for secondary prevention are either inadequately robust or prohibitively expensive and invasive. A non-invasive and relatively inexpensive method to identify patients who will benefit most from novel anti-inflammatory therapies is required, a role likely to be fulfilled by functional imaging methods. This review article outlines our current understanding of the inflammatory biology of atherosclerosis, upcoming therapies and recent landmark clinical trials, imaging modalities (both invasive and non-invasive) and the current landscape surrounding functional imaging including through targeted nuclear and nanobody tracer development and their application.
Collapse
Affiliation(s)
- Reece Parry
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Royal Perth Hospital, 197 Wellington Street, Perth, WA 6000, Australia
| | - Kamran Majeed
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Waikato District Health Board, Hamilton 3204, New Zealand
| | - Fiona Pixley
- School of Biomedical Sciences, Pharmacology and Toxicology, University of Western Australia, Perth 6009, Australia
| | - Graham Scott Hillis
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Royal Perth Hospital, 197 Wellington Street, Perth, WA 6000, Australia
| | - Roslyn Jane Francis
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Perth 6009, Australia
| | - Carl Johann Schultz
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Royal Perth Hospital, 197 Wellington Street, Perth, WA 6000, Australia
| |
Collapse
|
26
|
Sato H, Fujimoto S, Kawaguchi YO, Nozaki YO, Tomizawa N, Kogure Y, Minamino T. Dual-energy CT imaging of atherosclerotic plaque using novel ultrasmall superparamagnetic iron oxide in hyperlipidemic rabbits. Acta Radiol 2022; 64:1718-1724. [PMID: 36226361 DOI: 10.1177/02841851221131904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND A study using magnetic resonance imaging (MRI) revealed that ultra-small superparamagnetic iron oxide is phagocytosed by macrophages. However, MRI has limitations in obtaining clear images due to its poor spatial and temporal resolutions. PURPOSE To examine whether the use of dual-energy computed tomography (DECT) facilitated the visualization of carboxymethyl-diethylaminoethyl dextran magnetite ultra-small superparamagnetic iron oxide (CMEADM-U) accumulation in arteriosclerotic lesions using hyperlipidemic rabbits. MATERIAL AND METHODS CMEADM-U at 0.5 mmol Fe/kg was administered to Watanabe hereditary atherosclerotic (WHHL) rabbits (n = 6, 24 sections) and New Zealand white (NZW) rabbits (n = 2, 6 sections). After 72 h, DECT was performed to prepare virtual monochromatic images (35 keV, 70 keV) and an iron-based map. Subsequently, the aorta was collected along with hematoxylin and eosin staining, Berlin blue (BB) staining, and RAM11 immunostaining. RESULTS In the WHHL rabbits, CMEADM-U accumulation was not observed at 70 keV. However, CMEADM-U accumulation consistent with an arteriosclerotic lesion was observed at 35 keV and the iron-based map. On the other hand, in the NZW rabbits, there was no accumulation of CMEADM-U in any images. Further, there were significant differences in the iron-based map value at the site of accumulation among the grades of expression on BB staining and RAM11 immunostaining. In addition, there was a good correlation at 35 kev and iron-based map value (r = 0.42; P < 0.05). CONCLUSION DECT imaging for CMEADM-U facilitated the assessment of macrophage accumulation in atherosclerotic lesions in an in vivo study using a rabbit model of induced aortic atherosclerosis.
Collapse
Affiliation(s)
- Hideyuki Sato
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Radiological Technology, Juntendo University Hospital, Tokyo, Japan
| | - Shinichiro Fujimoto
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuko O Kawaguchi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yui O Nozaki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobuo Tomizawa
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yosuke Kogure
- Department of Radiological Technology, Juntendo University Hospital, Tokyo, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
27
|
Li X, Qi H, Cui W, Wang Z, Fu X, Li T, Ma H, Yang Y, Yu T. Recent advances in targeted delivery of non-coding RNA-based therapeutics for atherosclerosis. Mol Ther 2022; 30:3118-3132. [PMID: 35918894 PMCID: PMC9552813 DOI: 10.1016/j.ymthe.2022.07.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 10/16/2022] Open
Abstract
Cardiovascular disease (CVD) has overtaken infectious illnesses as the leading cause of mortality and disability worldwide. The pathology that underpins CVD is atherosclerosis, characterized by chronic inflammation caused by the accumulation of plaques in the arteries. As our knowledge about the microenvironment of blood vessel walls deepens, there is an opportunity to fine-tune treatments to target the mechanisms driving atherosclerosis more directly. The application of non-coding RNAs (ncRNAs) as biomarkers or intervention targets is increasing. Although these ncRNAs play an important role in driving atherosclerosis and vascular dysfunction, the cellular and extracellular environments pose a challenge for targeted transmission and therapeutic regulation of ncRNAs. Specificity, delivery, and tolerance have hampered the clinical translation of ncRNA-based therapeutics. Nanomedicine is an emerging field that uses nanotechnology for targeted drug delivery and advanced imaging. Recently, nanoscale carriers have shown promising results and have introduced new possibilities for nucleic acid targeted drug delivery, particularly for atherosclerosis. In this review, we discuss the latest developments in nanoparticles to aid ncRNA-based drug development, particularly miRNA, and we analyze the current challenges in ncRNA targeted delivery. In particular, we highlight the emergence of various kinds of nanotherapeutic approaches based on ncRNAs, which can improve treatment options for atherosclerosis.
Collapse
Affiliation(s)
- Xiaoxin Li
- Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China
| | - Hongzhao Qi
- Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China
| | - Weigang Cui
- Department of Cardiology, People's Hospital of Rizhao, No. 126 Taian Road, Rizhao 276827, People's Republic of China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266000, China
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266000, China
| | - Tianxiang Li
- Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China
| | - Huibo Ma
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao 266021, People's Republic of China.
| | - Tao Yu
- Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China; Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266000, China.
| |
Collapse
|
28
|
Hu PP, Luo SX, Fan XQ, Li D, Tong XY. Macrophage-targeted nanomedicine for the diagnosis and management of atherosclerosis. Front Pharmacol 2022; 13:1000316. [PMID: 36160452 PMCID: PMC9501673 DOI: 10.3389/fphar.2022.1000316] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/23/2022] [Indexed: 12/04/2022] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular diseases, such as myocardial infarction and stroke, which account for the highest death toll worldwide. Macrophage is the major contributor to atherosclerosis progression, and therefore, macrophage-associated pathological process is considered an extremely important target for the diagnosis and treatment of atherosclerosis. However, the existing clinical strategies still have many bottlenecks and challenges in atherosclerosis’s early detection and management. Nanomedicine, using various nanoparticles/nanocarriers for medical purposes, can effectively load therapeutic agents, significantly improve their stability and accurately deliver them to the atherosclerotic plaques. In this review, we summarized the latest progress of the macrophage-targeted nanomedicine in the diagnosis and treatment of atherosclerosis, and their potential applications and clinical benefits are also discussed.
Collapse
Affiliation(s)
- Ping Ping Hu
- Chongqing Engineering Research Center for Pharmacodynamics Evaluation, College of Pharmacy, Chongqing Medical University, Chongqing, China
- *Correspondence: Ping Ping Hu, ; Xiao Yong Tong,
| | - Shuang Xue Luo
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiao Qing Fan
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Di Li
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Yong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
- *Correspondence: Ping Ping Hu, ; Xiao Yong Tong,
| |
Collapse
|
29
|
Cai D, Gao W, Li Z, Zhang Y, Xiao L, Xiao Y. Current Development of Nano-Drug Delivery to Target Macrophages. Biomedicines 2022; 10:1203. [PMID: 35625939 PMCID: PMC9139084 DOI: 10.3390/biomedicines10051203] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages are the most important innate immune cells that participate in various inflammation-related diseases. Therefore, macrophage-related pathological processes are essential targets in the diagnosis and treatment of diseases. Since nanoparticles (NPs) can be preferentially taken up by macrophages, NPs have attracted most attention for specific macrophage-targeting. In this review, the interactions between NPs and the immune system are introduced to help understand the pharmacokinetics and biodistribution of NPs in immune cells. The current design and strategy of NPs modification for specific macrophage-targeting are investigated and summarized.
Collapse
Affiliation(s)
- Donglin Cai
- Centre for Biomedical Technologies, School of Mechanical, Medical & Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (D.C.); (W.G.); (Z.L.)
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China;
| | - Wendong Gao
- Centre for Biomedical Technologies, School of Mechanical, Medical & Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (D.C.); (W.G.); (Z.L.)
| | - Zhelun Li
- Centre for Biomedical Technologies, School of Mechanical, Medical & Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (D.C.); (W.G.); (Z.L.)
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China;
| | - Yufeng Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China;
| | - Lan Xiao
- Centre for Biomedical Technologies, School of Mechanical, Medical & Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (D.C.); (W.G.); (Z.L.)
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, 60 Musk Ave., Kelvin Grove, Brisbane, QLD 4059, Australia
| | - Yin Xiao
- Centre for Biomedical Technologies, School of Mechanical, Medical & Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (D.C.); (W.G.); (Z.L.)
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, 60 Musk Ave., Kelvin Grove, Brisbane, QLD 4059, Australia
| |
Collapse
|
30
|
Chen W, Schilperoort M, Cao Y, Shi J, Tabas I, Tao W. Macrophage-targeted nanomedicine for the diagnosis and treatment of atherosclerosis. Nat Rev Cardiol 2022; 19:228-249. [PMID: 34759324 PMCID: PMC8580169 DOI: 10.1038/s41569-021-00629-x] [Citation(s) in RCA: 270] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
Nanotechnology could improve our understanding of the pathophysiology of atherosclerosis and contribute to the development of novel diagnostic and therapeutic strategies to further reduce the risk of cardiovascular disease. Macrophages have key roles in atherosclerosis progression and, therefore, macrophage-associated pathological processes are important targets for both diagnostic imaging and novel therapies for atherosclerosis. In this Review, we highlight efforts in the past two decades to develop imaging techniques and to therapeutically manipulate macrophages in atherosclerotic plaques with the use of rationally designed nanoparticles. We review the latest progress in nanoparticle-based imaging modalities that can specifically target macrophages. Using novel molecular imaging technology, these modalities enable the identification of advanced atherosclerotic plaques and the assessment of the therapeutic efficacy of medical interventions. Additionally, we provide novel perspectives on how macrophage-targeting nanoparticles can deliver a broad range of therapeutic payloads to atherosclerotic lesions. These nanoparticles can suppress pro-atherogenic macrophage processes, leading to improved resolution of inflammation and stabilization of plaques. Finally, we propose future opportunities for novel diagnostic and therapeutic strategies and provide solutions to challenges in this area for the purpose of accelerating the clinical translation of nanomedicine for the treatment of atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Wei Chen
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maaike Schilperoort
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Yihai Cao
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Wei Tao
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Segers FME, Ruder AV, Westra MM, Lammers T, Dadfar SM, Roemhild K, Lam TS, Kooi ME, Cleutjens KBJM, Verheyen FK, Schurink GWH, Haenen GR, van Berkel TJC, Bot I, Halvorsen B, Sluimer JC, Biessen EAL. Magnetic resonance imaging contrast-enhancement with superparamagnetic iron oxide nanoparticles amplifies macrophage foam cell apoptosis in human and murine atherosclerosis. Cardiovasc Res 2022; 118:3346-3359. [PMID: 35325057 PMCID: PMC9847560 DOI: 10.1093/cvr/cvac032] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 01/28/2022] [Accepted: 02/23/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS (Ultra) Small superparamagnetic iron oxide nanoparticles, (U)SPIO, are widely used as magnetic resonance imaging contrast media and assumed to be safe for clinical applications in cardiovascular disease. As safety tests largely relied on normolipidaemic models, not fully representative of the clinical setting, we investigated the impact of (U)SPIOs on disease-relevant endpoints in hyperlipidaemic models of atherosclerosis. METHODS AND RESULTS RAW264.7 foam cells, exposed in vitro to ferumoxide (dextran-coated SPIO), ferumoxtran (dextran-coated USPIO), or ferumoxytol [carboxymethyl (CM) dextran-coated USPIO] (all 1 mg Fe/mL) showed increased apoptosis and reactive oxygen species accumulation for ferumoxide and ferumoxtran, whereas ferumoxytol was tolerated well. Pro-apoptotic (TUNEL+) and pro-oxidant activity of ferumoxide (0.3 mg Fe/kg) and ferumoxtran (1 mg Fe/kg) were confirmed in plaque, spleen, and liver of hyperlipidaemic ApoE-/- (n = 9/group) and LDLR-/- (n = 9-16/group) mice that had received single IV injections compared with saline-treated controls. Again, ferumoxytol treatment (1 mg Fe/kg) failed to induce apoptosis or oxidative stress in these tissues. Concomitant antioxidant treatment (EUK-8/EUK-134) largely prevented these effects in vitro (-68%, P < 0.05) and in plaques from LDLR-/- mice (-60%, P < 0.001, n = 8/group). Repeated ferumoxtran injections of LDLR-/- mice with pre-existing atherosclerosis enhanced plaque inflammation and apoptosis but did not alter plaque size. Strikingly, carotid artery plaques of endarterectomy patients who received ferumoxtran (2.6 mg Fe/kg) before surgery (n = 9) also showed five-fold increased apoptosis (18.2 vs. 3.7%, respectively; P = 0.004) compared with controls who did not receive ferumoxtran. Mechanistically, neither coating nor particle size seemed accountable for the observed cytotoxicity of ferumoxide and ferumoxtran. CONCLUSIONS Ferumoxide and ferumoxtran, but not ferumoxytol, induced apoptosis of lipid-laden macrophages in human and murine atherosclerosis, potentially impacting disease progression in patients with advanced atherosclerosis.
Collapse
Affiliation(s)
- Filip M E Segers
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands,Faculty of Medicine, Research Institute of Internal Medicine, University Hospital Oslo, Oslo, Norway
| | - Adele V Ruder
- Department of Pathology, CARIM School for Cardiovascular Sciences, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Marijke M Westra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, RWTH Aachen University, Aachen, Germany
| | | | - Karolin Roemhild
- Department of Nanomedicine and Theranostics, RWTH Aachen University, Aachen, Germany,Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Tin Sing Lam
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Marianne Eline Kooi
- Department of Radiology and Nuclear Medicine, CARIM School for Cardiovascular Sciences, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Kitty B J M Cleutjens
- Department of Pathology, CARIM School for Cardiovascular Sciences, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Fons K Verheyen
- Molecular Cell Biology and Electron Microscopy (CRISP), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Geert W H Schurink
- Department of Surgery, CARIM School for Cardiovascular Sciences, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Guido R Haenen
- Department of Toxicology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Theo J C van Berkel
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Bente Halvorsen
- Faculty of Medicine, Research Institute of Internal Medicine, University Hospital Oslo, Oslo, Norway
| | - Judith C Sluimer
- Corresponding author. Tel: +31 43 3877675; Fax: +31 43 3874613, E-mail: (J.C.S.); E-mail: (E.A.L.B.)
| | - Erik A L Biessen
- Corresponding author. Tel: +31 43 3877675; Fax: +31 43 3874613, E-mail: (J.C.S.); E-mail: (E.A.L.B.)
| |
Collapse
|
32
|
Zhang M, Xie Z, Long H, Ren K, Hou L, Wang Y, Xu X, Lei W, Yang Z, Ahmed S, Zhang H, Zhao G. Current advances in the imaging of atherosclerotic vulnerable plaque using nanoparticles. Mater Today Bio 2022; 14:100236. [PMID: 35341094 PMCID: PMC8943324 DOI: 10.1016/j.mtbio.2022.100236] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/13/2022] [Accepted: 03/05/2022] [Indexed: 01/29/2023]
Abstract
Vulnerable atherosclerotic plaques of the artery wall that pose a significant risk of cardio-cerebral vascular accidents remain the global leading cause of morbidity and mortality. Thus, early delineation of vulnerable atherosclerotic plaques is of clinical importance for prevention and treatment. The currently available imaging technologies mainly focus on the structural assessment of the vascular wall. Unfortunately, several disadvantages in these strategies limit the improvement in imaging effect. Nanoparticle technology is a novel diagnostic strategy for targeting and imaging pathological biomarkers. New functionalized nanoparticles that detect hallmarks of vulnerable plaques are promising for advance further control of this critical illness. The review aims to address the current opportunities and challenges for the use of nanoparticle technology in imagining vulnerable plaques.
Collapse
|
33
|
Izquierdo-Garcia D, Diyabalanage H, Ramsay IA, Rotile NJ, Mauskapf A, Choi JK, Witzel T, Humblet V, Jaffer FA, Brownell AL, Tawakol A, Catana C, Conrad MF, Caravan P, Ay I. Imaging High-Risk Atherothrombosis Using a Novel Fibrin-Binding Positron Emission Tomography Probe. Stroke 2022; 53:595-604. [PMID: 34965737 PMCID: PMC8792326 DOI: 10.1161/strokeaha.121.035638] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND PURPOSE High-risk atherosclerosis is an underlying cause of cardiovascular events, yet identifying the specific patient population at immediate risk is still challenging. Here, we used a rabbit model of atherosclerotic plaque rupture and human carotid endarterectomy specimens to describe the potential of molecular fibrin imaging as a tool to identify thrombotic plaques. METHODS Atherosclerotic plaques in rabbits were induced using a high-cholesterol diet and aortic balloon injury (N=13). Pharmacological triggering was used in a group of rabbits (n=9) to induce plaque disruption. Animals were grouped into thrombotic and nonthrombotic plaque groups based on gross pathology (gold standard). All animals were injected with a novel fibrin-specific probe 68Ga-CM246 followed by positron emission tomography (PET)/magnetic resonance imaging 90 minutes later. 68Ga-CM246 was quantified on the PET images using tissue-to-background (back muscle) ratios and standardized uptake value. RESULTS Both tissue-to-background (back muscle) ratios and standardized uptake value were significantly higher in the thrombotic versus nonthrombotic group (P<0.05). Ex vivo PET and autoradiography of the abdominal aorta correlated positively with in vivo PET measurements. Plaque disruption identified by 68Ga-CM246 PET agreed with gross pathology assessment (85%). In ex vivo surgical specimens obtained from patients undergoing elective carotid endarterectomy (N=12), 68Ga-CM246 showed significantly higher binding to carotid plaques compared to a D-cysteine nonbinding control probe. CONCLUSIONS We demonstrated that molecular fibrin PET imaging using 68Ga-CM246 could be a useful tool to diagnose experimental and clinical atherothrombosis. Based on our initial results using human carotid plaque specimens, in vivo molecular imaging studies are warranted to test 68Ga-CM246 PET as a tool to stratify risk in atherosclerotic patients.
Collapse
Affiliation(s)
- David Izquierdo-Garcia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA,Harvard-MIT Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA
| | | | - Ian A. Ramsay
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA,Collagen Medical, LLC, Belmont, MA,The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA
| | - Nicholas J. Rotile
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA,The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA
| | - Adam Mauskapf
- Cardiovascular Research Center, Division of Cardiology, Department of Medicine Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ji-Kyung Choi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | - Thomas Witzel
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | | | - Farouc A. Jaffer
- Cardiovascular Research Center, Division of Cardiology, Department of Medicine Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Anna-Liisa Brownell
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | - Ahmed Tawakol
- Nuclear Cardiology, Division of Cardiology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ciprian Catana
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA,The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA
| | - Mark F. Conrad
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA,The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA
| | - Ilknur Ay
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| |
Collapse
|
34
|
Tiwari A, Elgrably B, Saar G, Vandoorne K. Multi-Scale Imaging of Vascular Pathologies in Cardiovascular Disease. Front Med (Lausanne) 2022; 8:754369. [PMID: 35071257 PMCID: PMC8766766 DOI: 10.3389/fmed.2021.754369] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Cardiovascular disease entails systemic changes in the vasculature. The endothelial cells lining the blood vessels are crucial in the pathogenesis of cardiovascular disease. Healthy endothelial cells direct the blood flow to tissues as vasodilators and act as the systemic interface between the blood and tissues, supplying nutrients for vital organs, and regulating the smooth traffic of leukocytes into tissues. In cardiovascular diseases, when inflammation is sensed, endothelial cells adjust to the local or systemic inflammatory state. As the inflamed vasculature adjusts, changes in the endothelial cells lead to endothelial dysfunction, altered blood flow and permeability, expression of adhesion molecules, vessel wall inflammation, thrombosis, angiogenic processes, and extracellular matrix production at the endothelial cell level. Preclinical multi-scale imaging of these endothelial changes using optical, acoustic, nuclear, MRI, and multimodal techniques has progressed, due to technical advances and enhanced biological understanding on the interaction between immune and endothelial cells. While this review highlights biological processes that are related to changes in the cardiac vasculature during cardiovascular diseases, it also summarizes state-of-the-art vascular imaging techniques. The advantages and disadvantages of the different imaging techniques are highlighted, as well as their principles, methodologies, and preclinical and clinical applications with potential future directions. These multi-scale approaches of vascular imaging carry great potential to further expand our understanding of basic vascular biology, to enable early diagnosis of vascular changes and to provide sensitive diagnostic imaging techniques in the management of cardiovascular disease.
Collapse
Affiliation(s)
- Ashish Tiwari
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Betsalel Elgrably
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Galit Saar
- Biomedical Core Facility, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Katrien Vandoorne
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
35
|
Huang Y, Hsu JC, Koo H, Cormode DP. Repurposing ferumoxytol: Diagnostic and therapeutic applications of an FDA-approved nanoparticle. Am J Cancer Res 2022; 12:796-816. [PMID: 34976214 PMCID: PMC8692919 DOI: 10.7150/thno.67375] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/12/2021] [Indexed: 02/07/2023] Open
Abstract
Ferumoxytol is an intravenous iron oxide nanoparticle formulation that has been approved by the U.S. Food and Drug Administration (FDA) for treating anemia in patients with chronic kidney disease. In recent years, ferumoxytol has also been demonstrated to have potential for many additional biomedical applications due to its excellent inherent physical properties, such as superparamagnetism, biocatalytic activity, and immunomodulatory behavior. With good safety and clearance profiles, ferumoxytol has been extensively utilized in both preclinical and clinical studies. Here, we first introduce the medical needs and the value of current iron oxide nanoparticle formulations in the market. We then focus on ferumoxytol nanoparticles and their physicochemical, diagnostic, and therapeutic properties. We include examples describing their use in various biomedical applications, including magnetic resonance imaging (MRI), multimodality imaging, iron deficiency treatment, immunotherapy, microbial biofilm treatment and drug delivery. Finally, we provide a brief conclusion and offer our perspectives on the current limitations and emerging applications of ferumoxytol in biomedicine. Overall, this review provides a comprehensive summary of the developments of ferumoxytol as an agent with diagnostic, therapeutic, and theranostic functionalities.
Collapse
|
36
|
Pillai SC, Borah A, Jacob EM, Kumar DS. Nanotechnological approach to delivering nutraceuticals as promising drug candidates for the treatment of atherosclerosis. Drug Deliv 2021; 28:550-568. [PMID: 33703990 PMCID: PMC7954496 DOI: 10.1080/10717544.2021.1892241] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/25/2022] Open
Abstract
Atherosclerosis is Caesar's sword, which poses a huge risk to the present generation. Understanding the atherosclerotic disease cycle would allow ensuring improved diagnosis, better care, and treatment. Unfortunately, a highly effective and safe way of treating atherosclerosis in the medical community remains a continuous challenge. Conventional treatments have shown considerable success, but have some adverse effects on the human body. Natural derived medications or nutraceuticals have gained immense popularity in the treatment of atherosclerosis due to their decreased side effects and toxicity-related issues. In hindsight, the contribution of nutraceuticals in imparting enhanced clinical efficacy against atherosclerosis warrants more experimental evidence. On the other hand, nanotechnology and drug delivery systems (DDS) have revolutionized the way therapeutics are performed and researchers have been constantly exploring the positive effects that DDS brings to the field of therapeutic techniques. It could be as exciting as ever to apply nano-mediated delivery of nutraceuticals as an additional strategy to target the atherosclerotic sites boasting high therapeutic efficiency of the nutraceuticals and fewer side effects.
Collapse
Affiliation(s)
- Sindhu C. Pillai
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary New Science, Toyo University, Saitama, Japan
| | - Ankita Borah
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary New Science, Toyo University, Saitama, Japan
| | - Eden Mariam Jacob
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary New Science, Toyo University, Saitama, Japan
| | - D. Sakthi Kumar
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary New Science, Toyo University, Saitama, Japan
| |
Collapse
|
37
|
Prigent K, Vigne J. Advances in Radiopharmaceutical Sciences for Vascular Inflammation Imaging: Focus on Clinical Applications. Molecules 2021; 26:molecules26237111. [PMID: 34885690 PMCID: PMC8659223 DOI: 10.3390/molecules26237111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/09/2021] [Accepted: 11/19/2021] [Indexed: 01/18/2023] Open
Abstract
Biomedical imaging technologies offer identification of several anatomic and molecular features of disease pathogenesis. Molecular imaging techniques to assess cellular processes in vivo have been useful in advancing our understanding of several vascular inflammatory diseases. For the non-invasive molecular imaging of vascular inflammation, nuclear medicine constitutes one of the best imaging modalities, thanks to its high sensitivity for the detection of probes in tissues. 2-[18F]fluoro-2-deoxy-d-glucose ([18F]FDG) is currently the most widely used radiopharmaceutical for molecular imaging of vascular inflammatory diseases such as atherosclerosis and large-vessel vasculitis. The combination of [18F]FDG and positron emission tomography (PET) imaging has become a powerful tool to identify and monitor non-invasively inflammatory activities over time but suffers from several limitations including a lack of specificity and avid background in different localizations. The use of novel radiotracers may help to better understand the underlying pathophysiological processes and overcome some limitations of [18F]FDG PET for the imaging of vascular inflammation. This review examines how [18F]FDG PET has given us deeper insight into the role of inflammation in different vascular pathologies progression and discusses perspectives for alternative radiopharmaceuticals that could provide a more specific and simple identification of pathologies where vascular inflammation is implicated. Use of these novel PET tracers could lead to a better understanding of underlying disease mechanisms and help inform the identification and stratification of patients for newly emerging immune-modulatory therapies. Future research is needed to realize the true clinical translational value of PET imaging in vascular inflammatory diseases.
Collapse
Affiliation(s)
- Kevin Prigent
- CHU de Caen Normandie, Department of Nuclear Medicine, Normandie Université, UNICAEN, 14000 Caen, France;
| | - Jonathan Vigne
- CHU de Caen Normandie, Department of Nuclear Medicine, Normandie Université, UNICAEN, 14000 Caen, France;
- CHU de Caen Normandie, Department of Pharmacy, Normandie Université, UNICAEN, 14000 Caen, France
- UNICAEN, INSERM U1237, Etablissement Français du Sang, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, 14000 Caen, France
- Correspondence:
| |
Collapse
|
38
|
Collettini F, Brangsch J, Reimann C, Chapiro J, Savic LJ, Buchholz R, Keller S, Hamm B, Goldberg SN, Makowski MR. Hepatic Radiofrequency Ablation: Monitoring of Ablation-Induced Macrophage Recruitment in the Periablational Rim Using SPION-Enhanced Macrophage-Specific Magnetic Resonance Imaging. Invest Radiol 2021; 56:591-598. [PMID: 33787536 DOI: 10.1097/rli.0000000000000777] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Macrophages accumulating in the periablational rim play a pivotal role in initiating and sustaining the perifocal inflammatory reaction, which has been shown to be at least 1 of the mechanisms responsible for the systemic pro-oncogenic effects of focal hepatic radiofrequency ablation (RFA). Herein, we tested the hypothesis to use superparamagnetic iron oxide nanoparticle (SPION)-enhanced magnetic resonance imaging (MRI) for noninvasive quantification of iron-loaded macrophages in the periablational rim of VX2 tumor-bearing rabbits. MATERIALS AND METHODS Twelve VX2 tumor-bearing rabbits underwent MRI immediately after and up to 3 weeks after focal hepatic RFA. For noninvasive quantification of macrophage accumulation in the periablational rim, animals were scanned before and 24 hours after SPION injection. T2*-weighted images were analyzed and correlated with histopathological and immunohistochemical findings. Furthermore, correlations with quantitative measurements (ICP-MS [inductively coupled plasma-mass spectrometry] and LA-ICP-MS [laser ablation-ICP-MS]) were performed. RESULTS SPION-enhanced T2*-weighted MRI scans displayed a progressive increase in the areas of signal intensity (SI) loss within the periablational rim peaking 3 weeks after RFA. Accordingly, quantitative analysis of SI changes demonstrated a significant decline in the relative SI ratio reflecting a growing accumulation of iron-loaded macrophages in the rim. Histological analyses confirmed a progressive accumulation of iron-loaded macrophages in the periablational rim. The ICP-MS and LA-ICP-MS confirmed a progressive increase of iron concentration in the periablational rim. CONCLUSIONS SPION-enhanced MRI enables noninvasive monitoring and quantification of ablation-induced macrophage recruitment in the periablational rim. Given the close interplay between ablation-induced perifocal inflammation and potential unwanted tumorigenic effects of RFA, SPION-enhanced MRI may serve as a valuable tool to guide and modulate adjuvant therapies after hepatic RFA.
Collapse
Affiliation(s)
| | | | | | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Lynn Jeanette Savic
- From the Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health
| | - Rebecca Buchholz
- Institute of Inorganic and Analytical Chemistry, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Sarah Keller
- From the Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health
| | - Bernd Hamm
- From the Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health
| | - S Nahum Goldberg
- Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | |
Collapse
|
39
|
Della Camera G, Madej M, Ferretti AM, La Spina R, Li Y, Corteggio A, Heinzl T, Swartzwelter BJ, Sipos G, Gioria S, Ponti A, Boraschi D, Italiani P. Personalised Profiling of Innate Immune Memory Induced by Nano-Imaging Particles in Human Monocytes. Front Immunol 2021; 12:692165. [PMID: 34421901 PMCID: PMC8377278 DOI: 10.3389/fimmu.2021.692165] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/14/2021] [Indexed: 11/25/2022] Open
Abstract
Engineered nanoparticles used for medical purposes must meet stringent safety criteria, which include immunosafety, i.e., the inability to activate possibly detrimental immune/inflammatory effects. Even medical nanomaterials devoid of direct immunotoxic or inflammatory effects may have an impact on human health if able to modify innate memory, which is the ability to “prime” future immune responses towards a different, possibly more detrimental reactivity. Although innate memory is usually protective, anomalous innate memory responses may be at the basis of immune pathologies. In this study, we have examined the ability of two nanomaterials commonly used for diagnostic imaging purposes, gold and iron oxide nanoparticles, to induce or modulate innate memory, using an in vitro model based on human primary monocytes. Monocytes were exposed in culture to nanoparticles alone or together with the bacterial agent LPS (priming phase/primary response), then rested for six days (extinction phase), and eventually challenged with LPS (memory/secondary response). The memory response to the LPS challenge was measured as changes in the production of inflammatory (TNFα, IL-6) and anti-inflammatory cytokines (IL-10, IL-1Ra), as compared to unprimed monocytes. The results show that both types of nanoparticles can have an effect in the induction of memory, with changes observed in the cytokine production. By comparing nanomaterials of different shapes (spherical vs. rod-shaped gold particles) and different size (17 vs. 22 nm diameter spherical iron oxide particles), it was evident that innate memory could be differentially induced and modulated depending on size, shape and chemical composition. However, the main finding was that the innate memory effect of the particles was strongly donor-dependent, with monocytes from each donor showing a distinct memory profile upon priming with the same particles, thereby making impossible to draw general conclusions on the particle effects. Thus, in order to predict the effect of imaging nanoparticles on the innate memory of patients, a personalised profiling would be required, able to take in consideration the peculiarities of the individual innate immune reactivity.
Collapse
Affiliation(s)
- Giacomo Della Camera
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Mariusz Madej
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Anna Maria Ferretti
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" (SCITEC), National Research Council (CNR), Milano, Italy
| | - Rita La Spina
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Yang Li
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Annunziata Corteggio
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Tommaso Heinzl
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Benjamin J Swartzwelter
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Gergö Sipos
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Sabrina Gioria
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Alessandro Ponti
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" (SCITEC), National Research Council (CNR), Milano, Italy
| | - Diana Boraschi
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy.,Stazione Zoologica Anton Dohrn, Napoli, Italy
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| |
Collapse
|
40
|
Nakamura M, Kosuge H, Oyane A, Kuroiwa K, Shimizu Y, Aonuma K. In vivostudy of iron oxide-calcium phosphate composite nanoparticles for delivery to atherosclerosis. NANOTECHNOLOGY 2021; 32:345101. [PMID: 34057430 DOI: 10.1088/1361-6528/ac007d] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/12/2021] [Indexed: 06/12/2023]
Abstract
Atherosclerosis is a macrophage-related inflammatory disease that remains a leading cause of death worldwide. Magnetic iron oxide (IO) nanocrystals are clinically used as magnetic resonance imaging contrast agents and their application as a detection agent for macrophages in arterial lesions has been studied extensively. We recently fabricated heparin-modified calcium phosphate (CaP) nanoparticles loaded with a large number of IO nanocrystals via coprecipitation from a supersaturated CaP solution supplemented with heparin and ferucarbotran (IO nanocrystals coated with carboxydextran). In this study, we further increased the content of IO nanocrystals in the heparin-modified IO-CaP composite nanoparticles by increasing the ferucarbotran concentration in the supersaturated CaP solution. The increase in nanoparticle IO content caused a decrease in particle diameter without impairing its dispersibility; the nanoparticles remained dispersed in water for up to 2 h due to electrostatic repulsion between particles due to the surface modification with heparin. The nanoparticles were more effectively taken up by murine RAW264.7 macrophages compared to free ferucarbotran without showing significant cytotoxicity. A preliminaryin vivostudy showed that the nanoparticles injected intravenously into mice delivered more IO nanocrystals to macrophage-rich carotid arterial lesions than free ferucarbotran. Our nanoparticles have potential as a delivery agent of IO nanocrystals to macrophages in arterial lesions.
Collapse
Affiliation(s)
- Maki Nakamura
- Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Hisanori Kosuge
- Department of Cardiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Ayako Oyane
- Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Kiyoko Kuroiwa
- Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Yoshiki Shimizu
- Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Kazutaka Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
41
|
Pan C, Lin J, Zheng J, Liu C, Yuan B, Akakuru OU, Zubair Iqbal M, Fang Q, Hu J, Chen J, Lin J, Dai Q, Guo X, Li Z, Zhang T, Xu C, Ma X, Chen T, Wu A, Jin Y. An intelligent T 1-T 2 switchable MRI contrast agent for the non-invasive identification of vulnerable atherosclerotic plaques. NANOSCALE 2021; 13:6461-6474. [PMID: 33885526 DOI: 10.1039/d0nr08039j] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Unlike stable atherosclerotic plaques, vulnerable plaques are very likely to cause serious cardio-cerebrovascular diseases. Meanwhile, how to non-invasively identify vulnerable plaques at early stages has been an urgent but challenging problem in clinical practices. Here, we propose a macrophage-targeted and in situ stimuli-triggered T1-T2 switchable magnetic resonance imaging (MRI) nanoprobe for the non-invasive diagnosis of vulnerable plaques. Precisely, single-dispersed iron oxide nanoparticles (IONPs) modified with hyaluronic acid (HA), denoted as IONP-HP, show macrophage targetability and T1 MRI enhancement (r2/r1 = 3.415). Triggered by the low pH environment of macrophage lysosomes, the single-dispersed IONP-HP transforms into a cluster analogue, which exhibits T2 MRI enhancement (r2/r1 = 13.326). Furthermore, an in vivo switch of T1-T2 enhancement modes shows that the vulnerable plaques exhibit strong T1 enhancement after intravenous administration of the nanoprobe, followed by a switch to T2 enhancement after 9 h. In contrast, stable plaques show only slight T1 enhancement but without T2 enhancement. It is therefore imperative that the intelligent and novel nanoplatform proposed in this study achieves a substantial non-invasive diagnosis of vulnerable plaques by means of a facile but effective T1-T2 switchable process, which will significantly contribute to the application of materials science in solving clinical problems.
Collapse
Affiliation(s)
- Chunshu Pan
- Department of Radiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Carter TJ, Agliardi G, Lin FY, Ellis M, Jones C, Robson M, Richard-Londt A, Southern P, Lythgoe M, Zaw Thin M, Ryzhov V, de Rosales RTM, Gruettner C, Abdollah MRA, Pedley RB, Pankhurst QA, Kalber TL, Brandner S, Quezada S, Mulholland P, Shevtsov M, Chester K. Potential of Magnetic Hyperthermia to Stimulate Localized Immune Activation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2005241. [PMID: 33734595 DOI: 10.1002/smll.202005241] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/20/2021] [Indexed: 05/27/2023]
Abstract
Magnetic hyperthermia (MH) harnesses the heat-releasing properties of superparamagnetic iron oxide nanoparticles (SPIONs) and has potential to stimulate immune activation in the tumor microenvironment whilst sparing surrounding normal tissues. To assess feasibility of localized MH in vivo, SPIONs are injected intratumorally and their fate tracked by Zirconium-89-positron emission tomography, histological analysis, and electron microscopy. Experiments show that an average of 49% (21-87%, n = 9) of SPIONs are retained within the tumor or immediately surrounding tissue. In situ heating is subsequently generated by exposure to an externally applied alternating magnetic field and monitored by thermal imaging. Tissue response to hyperthermia, measured by immunohistochemical image analysis, reveals specific and localized heat-shock protein expression following treatment. Tumor growth inhibition is also observed. To evaluate the potential effects of MH on the immune landscape, flow cytometry is used to characterize immune cells from excised tumors and draining lymph nodes. Results show an influx of activated cytotoxic T cells, alongside an increase in proliferating regulatory T cells, following treatment. Complementary changes are found in draining lymph nodes. In conclusion, results indicate that biologically reactive MH is achievable in vivo and can generate localized changes consistent with an anti-tumor immune response.
Collapse
Affiliation(s)
- Thomas J Carter
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Giulia Agliardi
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Fang-Yu Lin
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
| | - Matthew Ellis
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Cancer Sciences Unit, Cancer Research UK Centre, University of Southampton, Somers Building, Southampton, SO16 6YD, UK
| | - Clare Jones
- School of Biomedical Engineering and Imaging Sciences, King's College London (KCL), St Thomas' Hospital, London, SE1 7EH, UK
| | - Mathew Robson
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Angela Richard-Londt
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Paul Southern
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
- Resonant Circuits Limited (RCL), London, W1S 4BS, UK
| | - Mark Lythgoe
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - May Zaw Thin
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Vyacheslav Ryzhov
- NRC "Kurchatov Institute", Petersburg Nuclear Physics Institute, Gatchina, 188300, Russia
| | - Rafael T M de Rosales
- School of Biomedical Engineering and Imaging Sciences, King's College London (KCL), St Thomas' Hospital, London, SE1 7EH, UK
| | - Cordula Gruettner
- Micromod Partikeltechnologie GmbH, Friedrich-Barnewitz-Str. 4, Rostock, D-18119, Germany
| | - Maha R A Abdollah
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt (BUE), El Shorouk City, Misr- Ismalia Desert Road, 11873, Cairo, Egypt
| | - R Barbara Pedley
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Quentin A Pankhurst
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
- Resonant Circuits Limited (RCL), London, W1S 4BS, UK
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Sebastian Brandner
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Sergio Quezada
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Paul Mulholland
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Maxim Shevtsov
- NRC "Kurchatov Institute", Petersburg Nuclear Physics Institute, Gatchina, 188300, Russia
- Technical University of Munich, Klinikum Rechts der Isar, Ismaninger str. 22, Munich, 81675, Germany
| | - Kerry Chester
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| |
Collapse
|
43
|
Chen J, Zhang X, Millican R, Sherwood J, Martin S, Jo H, Yoon YS, Brott BC, Jun HW. Recent advances in nanomaterials for therapy and diagnosis for atherosclerosis. Adv Drug Deliv Rev 2021; 170:142-199. [PMID: 33428994 PMCID: PMC7981266 DOI: 10.1016/j.addr.2021.01.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/02/2021] [Accepted: 01/03/2021] [Indexed: 12/18/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease driven by lipid accumulation in arteries, leading to narrowing and thrombosis. It affects the heart, brain, and peripheral vessels and is the leading cause of mortality in the United States. Researchers have strived to design nanomaterials of various functions, ranging from non-invasive imaging contrast agents, targeted therapeutic delivery systems to multifunctional nanoagents able to target, diagnose, and treat atherosclerosis. Therefore, this review aims to summarize recent progress (2017-now) in the development of nanomaterials and their applications to improve atherosclerosis diagnosis and therapy during the preclinical and clinical stages of the disease.
Collapse
Affiliation(s)
- Jun Chen
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Xixi Zhang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | | | | | - Sean Martin
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States; Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Young-Sup Yoon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Brigitta C Brott
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ho-Wook Jun
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
44
|
Moonen RPM, Coolen BF, Sluimer JC, Daemen MJAP, Strijkers GJ. Iron Oxide Nanoparticle Uptake in Mouse Brachiocephalic Artery Atherosclerotic Plaque Quantified by T 2-Mapping MRI. Pharmaceutics 2021; 13:pharmaceutics13020279. [PMID: 33669667 PMCID: PMC7922981 DOI: 10.3390/pharmaceutics13020279] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/29/2022] Open
Abstract
The purpose of our study was to monitor the iron oxide contrast agent uptake in mouse brachiocephalic artery (BCA) atherosclerotic plaques in vivo by quantitative T2-mapping magnetic resonance imaging (MRI). Female ApoE−/− mice (n = 32) on a 15-week Western-type diet developed advanced plaques in the BCA and were injected with ultra-small superparamagnetic iron oxides (USPIOs). Quantitative in vivo MRI at 9.4 T was performed with a Malcolm-Levitt (MLEV) prepared T2-mapping sequence to monitor the nanoparticle uptake in the atherosclerotic plaque. Ex vivo histology and particle electron paramagnetic resonance (pEPR) were used for validation. Longitudinal high-resolution in vivo T2-value maps were acquired with consistent quality. Average T2 values in the plaque decreased from a baseline value of 34.5 ± 0.6 ms to 24.0 ± 0.4 ms one day after injection and partially recovered to an average T2 of 27 ± 0.5 ms after two days. T2 values were inversely related to iron levels in the plaque as determined by ex vivo particle electron paramagnetic resonance (pEPR). We concluded that MRI T2 mapping facilitates a robust quantitative readout for USPIO uptake in atherosclerotic plaques in arteries near the mouse heart.
Collapse
Affiliation(s)
- Rik P. M. Moonen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands;
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands;
| | - Bram F. Coolen
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Judith C. Sluimer
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands;
- Department of Pathology, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands
| | - Mat J. A. P. Daemen
- Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands;
| | - Gustav J. Strijkers
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
- Correspondence: ; Tel.: +31-20-566-52-02
| |
Collapse
|
45
|
Andelovic K, Winter P, Jakob PM, Bauer WR, Herold V, Zernecke A. Evaluation of Plaque Characteristics and Inflammation Using Magnetic Resonance Imaging. Biomedicines 2021; 9:185. [PMID: 33673124 PMCID: PMC7917750 DOI: 10.3390/biomedicines9020185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis is an inflammatory disease of large and medium-sized arteries, characterized by the growth of atherosclerotic lesions (plaques). These plaques often develop at inner curvatures of arteries, branchpoints, and bifurcations, where the endothelial wall shear stress is low and oscillatory. In conjunction with other processes such as lipid deposition, biomechanical factors lead to local vascular inflammation and plaque growth. There is also evidence that low and oscillatory shear stress contribute to arterial remodeling, entailing a loss in arterial elasticity and, therefore, an increased pulse-wave velocity. Although altered shear stress profiles, elasticity and inflammation are closely intertwined and critical for plaque growth, preclinical and clinical investigations for atherosclerosis mostly focus on the investigation of one of these parameters only due to the experimental limitations. However, cardiovascular magnetic resonance imaging (MRI) has been demonstrated to be a potent tool which can be used to provide insights into a large range of biological parameters in one experimental session. It enables the evaluation of the dynamic process of atherosclerotic lesion formation without the need for harmful radiation. Flow-sensitive MRI provides the assessment of hemodynamic parameters such as wall shear stress and pulse wave velocity which may replace invasive and radiation-based techniques for imaging of the vascular function and the characterization of early plaque development. In combination with inflammation imaging, the analyses and correlations of these parameters could not only significantly advance basic preclinical investigations of atherosclerotic lesion formation and progression, but also the diagnostic clinical evaluation for early identification of high-risk plaques, which are prone to rupture. In this review, we summarize the key applications of magnetic resonance imaging for the evaluation of plaque characteristics through flow sensitive and morphological measurements. The simultaneous measurements of functional and structural parameters will further preclinical research on atherosclerosis and has the potential to fundamentally improve the detection of inflammation and vulnerable plaques in patients.
Collapse
Affiliation(s)
- Kristina Andelovic
- Institute of Experimental Biomedicine, University Hospital Würzburg, 97080 Würzburg, Germany
- Experimental Physics V, University of Würzburg, 97074 Würzburg, Germany; (P.W.); (P.M.J.); (V.H.)
| | - Patrick Winter
- Experimental Physics V, University of Würzburg, 97074 Würzburg, Germany; (P.W.); (P.M.J.); (V.H.)
- Internal Medicine I, Cardiology, University Hospital Würzburg, 97080 Würzburg, Germany;
| | - Peter Michael Jakob
- Experimental Physics V, University of Würzburg, 97074 Würzburg, Germany; (P.W.); (P.M.J.); (V.H.)
| | - Wolfgang Rudolf Bauer
- Internal Medicine I, Cardiology, University Hospital Würzburg, 97080 Würzburg, Germany;
| | - Volker Herold
- Experimental Physics V, University of Würzburg, 97074 Würzburg, Germany; (P.W.); (P.M.J.); (V.H.)
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
46
|
Dhanasekara CS, Zhang J, Nie S, Li G, Fan Z, Wang S. Nanoparticles target intimal macrophages in atherosclerotic lesions. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 32:102346. [PMID: 33259961 PMCID: PMC8514141 DOI: 10.1016/j.nano.2020.102346] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 10/25/2020] [Accepted: 11/22/2020] [Indexed: 10/22/2022]
Abstract
Oxidized phosphatidylcholines (oxPCs) enriched on the oxidized LDL (oxLDL) surface are responsible ligands for binding oxLDL to the CD36 receptor of intimal macrophages in atherosclerotic lesions. We synthesized liposome-like nanoparticles (NPs) using soy phosphatidylcholine and incorporated 1-palmitoyl-2-(4-keto-dodec-3-enedioyl) phosphatidylcholine, a type of oxPCs, on their surface to make ligand-NP (L-NPs). The objectives of this study were to measure and compare their binding affinity to and uptake by primary mouse and THP-1 derived macrophages, and to determine their target specificity to intimal macrophages in aortic lesions in LDL receptor null (LDLr-/-) mice. All in vitro data demonstrate that L-NPs had a high binding affinity to macrophage CD36 receptor. L-NPs had 1.4-fold higher accumulation in aortic lesion areas than NPs. L-NPs co-localized with intimal macrophages and CD36 receptors in the aortic lesions. This target delivery approach may portend a breakthrough in molecular imaging and targeted treatment of atherosclerosis.
Collapse
Affiliation(s)
| | - Jia Zhang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Shufang Nie
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Guigen Li
- Department of Chemistry, Texas Tech University, Lubbock, TX, USA
| | - Zhaoyang Fan
- Department of Electrical & Computer Engineering and Nano Tech Center, Texas Tech University, Lubbock, TX, USA
| | - Shu Wang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; College of Health Solutions, Arizona State University, Phoenix, AZ, USA.
| |
Collapse
|
47
|
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: 1.0] [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
|
48
|
Custom, spray coated receive coils for magnetic resonance imaging. Sci Rep 2021; 11:2635. [PMID: 33514816 PMCID: PMC7846777 DOI: 10.1038/s41598-021-81833-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 12/17/2020] [Indexed: 11/08/2022] Open
Abstract
We have developed a process for fabricating patient specific Magnetic Resonance Imaging (MRI) Radio-frequency (RF) receive coil arrays using additive manufacturing. Our process involves spray deposition of silver nanoparticle inks and dielectric materials onto 3D printed substrates to form high-quality resonant circuits. In this paper, we describe the material selection and characterization, process optimization, and design and testing of a prototype 4-channel neck array for carotid imaging. We show that sprayed polystyrene can form a low loss dielectric layer in a parallel plate capacitor. We also demonstrate that by using sprayed silver nanoparticle ink as conductive traces, our devices are still dominated by sample noise, rather than material losses. These results are critical for maintaining high Signal-to-Noise-Ratio (SNR) in clinical settings. Finally, our prototype patient specific coil array exhibits higher SNR (5 × in the periphery, 1.4 × in the center) than a commercially available array designed to fit the majority of subjects when tested on our custom neck phantom. 3D printed substrates ensure an optimum fit to complex body parts, improve diagnostic image quality, and enable reproducible placement on subjects.
Collapse
|
49
|
Caixeta MB, Araújo PS, Rodrigues CC, Gonçalves BB, Araújo OA, Bevilaqua GB, Malafaia G, Silva LD, Rocha TL. Risk assessment of iron oxide nanoparticles in an aquatic ecosystem: A case study on Biomphalaria glabrata. JOURNAL OF HAZARDOUS MATERIALS 2021; 401:123398. [PMID: 32763694 DOI: 10.1016/j.jhazmat.2020.123398] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 06/11/2023]
Abstract
Iron oxide nanoparticles (IONPs) have been applied in several sectors in the environmental field, such as aquatic nanoremediation, due to their unique superparamagnetic and nanospecific properties. However, the knowledge of chronic toxicity of IONPs on aquatic invertebrate remains limited. Thus, the present study aimed to analyze the chronic toxicity of gluconic acid-functionalized IONPs (GLA-IONPs) and their dissolved counterpart (FeCl3) to freshwater snail Biomphalaria glabrata. GLA-IONPs were synthesized and characterized by multiple techniques, and the snails were exposed to both Fe forms at environmentally relevant concentrations (1.0-15.6 mg L-1) for 28 days. The bioaccumulation, mortality rate, behavior impairments, morphological alterations, fecundity and fertility of snails were analyzed. Results showed that GLA-IONPs induced high iron bioaccumulation in the entire soft tissue portion. Chronic exposure to GLA-IONP increased the behavioral impairments of snails compared to iron ions and control groups. Both Fe forms reduced the fecundity, while the mortality and reduced fertility were observed only after the exposure to GLA-IONPs at 15.6 mg L-1. Overall results indicated the behavioral impairments and reproductive toxicity associated, possibly, to bioaccumulation of GLA-IONPs in the B. glabrata. These results can be useful for the development of eco-friendly nanotechnologies.
Collapse
Affiliation(s)
- Maxwell Batista Caixeta
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Paula Sampaio Araújo
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Cândido Carvalho Rodrigues
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Bruno Bastos Gonçalves
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Olacir Alves Araújo
- Laboratory of Chemistry and Molecular Modelling, Chemistry Institute, State University of Goiás, Anápolis, Goiás, Brazil
| | - Giovanni Bonatti Bevilaqua
- Laboratory of Chemistry and Molecular Modelling, Chemistry Institute, State University of Goiás, Anápolis, Goiás, Brazil
| | - Guilherme Malafaia
- Biological Research Laboratory, Goiano Federal Institute - Urutaí Campus, Goiás, Brazil
| | - Luciana Damacena Silva
- Laboratory of Host-Parasite Interactions, State University of Goiás, Anápolis, Goiás, Brazil
| | - Thiago Lopes Rocha
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil.
| |
Collapse
|
50
|
Sangha GS, Goergen CJ, Prior SJ, Ranadive SM, Clyne AM. Preclinical techniques to investigate exercise training in vascular pathophysiology. Am J Physiol Heart Circ Physiol 2021; 320:H1566-H1600. [PMID: 33385323 PMCID: PMC8260379 DOI: 10.1152/ajpheart.00719.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Atherosclerosis is a dynamic process starting with endothelial dysfunction and inflammation and eventually leading to life-threatening arterial plaques. Exercise generally improves endothelial function in a dose-dependent manner by altering hemodynamics, specifically by increased arterial pressure, pulsatility, and shear stress. However, athletes who regularly participate in high-intensity training can develop arterial plaques, suggesting alternative mechanisms through which excessive exercise promotes vascular disease. Understanding the mechanisms that drive atherosclerosis in sedentary versus exercise states may lead to novel rehabilitative methods aimed at improving exercise compliance and physical activity. Preclinical tools, including in vitro cell assays, in vivo animal models, and in silico computational methods, broaden our capabilities to study the mechanisms through which exercise impacts atherogenesis, from molecular maladaptation to vascular remodeling. Here, we describe how preclinical research tools have and can be used to study exercise effects on atherosclerosis. We then propose how advanced bioengineering techniques can be used to address gaps in our current understanding of vascular pathophysiology, including integrating in vitro, in vivo, and in silico studies across multiple tissue systems and size scales. Improving our understanding of the antiatherogenic exercise effects will enable engaging, targeted, and individualized exercise recommendations to promote cardiovascular health rather than treating cardiovascular disease that results from a sedentary lifestyle.
Collapse
Affiliation(s)
- Gurneet S Sangha
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Steven J Prior
- Department of Kinesiology, University of Maryland School of Public Health, College Park, Maryland.,Baltimore Veterans Affairs Geriatric Research, Education, and Clinical Center, Baltimore, Maryland
| | - Sushant M Ranadive
- Department of Kinesiology, University of Maryland School of Public Health, College Park, Maryland
| | - Alisa M Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|