1
|
Pang ASR, Dinesh T, Pang NYL, Dinesh V, Pang KYL, Yong CL, Lee SJJ, Yip GW, Bay BH, Srinivasan DK. Nanoparticles as Drug Delivery Systems for the Targeted Treatment of Atherosclerosis. Molecules 2024; 29:2873. [PMID: 38930939 PMCID: PMC11206617 DOI: 10.3390/molecules29122873] [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: 04/11/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Atherosclerosis continues to be a leading cause of morbidity and mortality globally. The precise evaluation of the extent of an atherosclerotic plaque is essential for forecasting its likelihood of causing health concerns and tracking treatment outcomes. When compared to conventional methods used, nanoparticles offer clear benefits and excellent development opportunities for the detection and characterisation of susceptible atherosclerotic plaques. In this review, we analyse the recent advancements of nanoparticles as theranostics in the management of atherosclerosis, with an emphasis on applications in drug delivery. Furthermore, the main issues that must be resolved in order to advance clinical utility and future developments of NP research are discussed. It is anticipated that medical NPs will develop into complex and advanced next-generation nanobotics that can carry out a variety of functions in the bloodstream.
Collapse
Affiliation(s)
- Alexander Shao-Rong Pang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (A.S.-R.P.); (N.Y.-L.P.); (C.L.Y.)
| | - Tarini Dinesh
- Department of Medicine, Government Kilpauk Medical College, Chennai 600010, Tamilnadu, India;
| | - Natalie Yan-Lin Pang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (A.S.-R.P.); (N.Y.-L.P.); (C.L.Y.)
| | - Vishalli Dinesh
- Department of Pathology, Dhanalakshmi Srinivasan Medical College Hospital, Perambalur 621113, Tamilnadu, India;
| | - Kimberley Yun-Lin Pang
- Division of Medicine, South Australia Health, Northern Adelaide Local Health Network, Adelaide, SA 5112, Australia; (K.Y.-L.P.); (S.J.J.L.)
| | - Cai Ling Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (A.S.-R.P.); (N.Y.-L.P.); (C.L.Y.)
| | - Shawn Jia Jun Lee
- Division of Medicine, South Australia Health, Northern Adelaide Local Health Network, Adelaide, SA 5112, Australia; (K.Y.-L.P.); (S.J.J.L.)
| | - George W. Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (G.W.Y.); (B.H.B.)
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (G.W.Y.); (B.H.B.)
| | - Dinesh Kumar Srinivasan
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (G.W.Y.); (B.H.B.)
| |
Collapse
|
2
|
Abid J, Khalil FMA, Saeed S, Khan SU, Iqbal I, Khan SU, Anthony S, Shahzad R, Koerniati S, Naz F. Nano revolution in cardiovascular health: Nanoparticles (NPs) as tiny titans for diagnosis and therapeutics. Curr Probl Cardiol 2024; 49:102466. [PMID: 38369205 DOI: 10.1016/j.cpcardiol.2024.102466] [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: 02/10/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Cardiovascular diseases (CVDs) are known as life-threatening illnessescaused by severe abnormalities in the cardiovascular system. They are a leading cause of mortality and morbidity worldwide.Nanotechnology integrated substantialinnovations in cardiovascular diagnostic and therapeutic at the nanoscale. This in-depth analysis explores cutting-edge methods for diagnosing CVDs, including nanotechnological interventions and crucial components for identifying risk factors, developing treatment plans, and monitoring patients' progress with chronic CVDs.Intensive research has gone into making nano-carriers that can image and treat patients. To improve the efficiency of treating CVDs, the presentreview sheds light on a decision-tree-based solution by investigating recent and innovative approaches in CVD diagnosis by utilizing nanoparticles (NPs). Treatment choices for chronic diseases like CVD, whose etiology might take decades to manifest, are very condition-specific and disease-stage-based. Moreover, thisreview alsobenchmarks the changing landscape of employing NPs for targeted and better drug administration while examining the limitations of various NPs in CVD diagnosis, including cost, space, time, and complexity. To better understand and treatment of cardiovascular diseases, the conversation moves on to the nano-cardiovascular possibilities for medical research.We also focus on recent developments in nanoparticle applications, the ways they might be helpful, and the medical fields where they may find future use. Finally, this reviewadds to the continuing conversation on improved diagnosis and treatment approaches for cardiovascular disorders by discussing the obstacles and highlighting the revolutionary effects of nanotechnology.
Collapse
Affiliation(s)
- Junaid Abid
- Department of Food Science and Technology, University of Haripur, Pakistan; State Key Laboratory of Food nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Fatma Mohamed Ameen Khalil
- King Khalid University, College of Science and Arts, Department of Biology, MohayilAsirAbha, 61421, Saudi Arabia
| | - Sumbul Saeed
- School of Environment and Science, Griffith University, QLD, 4111, Australia
| | - Shahid Ullah Khan
- Women Medical and Dental College, Khyber Medical University, Khyber Pakhtunkhwa, Pakistan; Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and Southwest University, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China
| | - Imran Iqbal
- Department of PLR, Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513, Teltow, Germany
| | - Safir Ullah Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Stefan Anthony
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University Liaoning Provence China.
| | - Raheel Shahzad
- Research Center for Genetic Engineering, National Research and Innovation Agency (BRIN), KST-Cibinong, JI Raya Bogor KM46, Cibinong 16911, Indonesia
| | - Sri Koerniati
- Research Center for Genetic Engineering, National Research and Innovation Agency (BRIN), KST-Cibinong, JI Raya Bogor KM46, Cibinong, 16911, Indonesia
| | - Farkhanda Naz
- Biological Science Research Center, Academy for Advanced Interdisciplinary Studies, Southwest University, Chongqing, 400715, China
| |
Collapse
|
3
|
Mytych W, Bartusik-Aebisher D, Łoś A, Dynarowicz K, Myśliwiec A, Aebisher D. Photodynamic Therapy for Atherosclerosis. Int J Mol Sci 2024; 25:1958. [PMID: 38396639 PMCID: PMC10888721 DOI: 10.3390/ijms25041958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/26/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis, which currently contributes to 31% of deaths globally, is of critical cardiovascular concern. Current diagnostic tools and biomarkers are limited, emphasizing the need for early detection. Lifestyle modifications and medications form the basis of treatment, and emerging therapies such as photodynamic therapy are being developed. Photodynamic therapy involves a photosensitizer selectively targeting components of atherosclerotic plaques. When activated by specific light wavelengths, it induces localized oxidative stress aiming to stabilize plaques and reduce inflammation. The key advantage lies in its selective targeting, sparing healthy tissues. While preclinical studies are encouraging, ongoing research and clinical trials are crucial for optimizing protocols and ensuring long-term safety and efficacy. The potential combination with other therapies makes photodynamic therapy a versatile and promising avenue for addressing atherosclerosis and associated cardiovascular disease. The investigations underscore the possibility of utilizing photodynamic therapy as a valuable treatment choice for atherosclerosis. As advancements in research continue, photodynamic therapy might become more seamlessly incorporated into clinical approaches for managing atherosclerosis, providing a blend of efficacy and limited invasiveness.
Collapse
Affiliation(s)
- Wiktoria Mytych
- Students English Division Science Club, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland; (W.M.); (A.Ł.)
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland;
| | - Aleksandra Łoś
- Students English Division Science Club, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland; (W.M.); (A.Ł.)
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland; (K.D.); (A.M.)
| | - Angelika Myśliwiec
- Center for Innovative Research in Medical and Natural Sciences, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland; (K.D.); (A.M.)
| | - David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland
| |
Collapse
|
4
|
Tscheuschner L, Tzafriri AR. Cardiovascular Tissue Engineering Models for Atherosclerosis Treatment Development. Bioengineering (Basel) 2023; 10:1373. [PMID: 38135964 PMCID: PMC10740643 DOI: 10.3390/bioengineering10121373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
In the early years of tissue engineering, scientists focused on the generation of healthy-like tissues and organs to replace diseased tissue areas with the aim of filling the gap between organ demands and actual organ donations. Over time, the realization has set in that there is an additional large unmet need for suitable disease models to study their progression and to test and refine different treatment approaches. Increasingly, researchers have turned to tissue engineering to address this need for controllable translational disease models. We review existing and potential uses of tissue-engineered disease models in cardiovascular research and suggest guidelines for generating adequate disease models, aimed both at studying disease progression mechanisms and supporting the development of dedicated drug-delivery therapies. This involves the discussion of different requirements for disease models to test drugs, nanoparticles, and drug-eluting devices. In addition to realistic cellular composition, the different mechanical and structural properties that are needed to simulate pathological reality are addressed.
Collapse
Affiliation(s)
- Linnea Tscheuschner
- Department of Vascular Surgery, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Abraham R. Tzafriri
- Department of Research and Innovation, CBSET Inc., Lexington, MA 02421, USA;
| |
Collapse
|
5
|
Li F, Shao H, Zhou G, Wang B, Xu Y, Liang W, Chen L. The recent applications of nanotechnology in the diagnosis and treatment of common cardiovascular diseases. Vascul Pharmacol 2023; 152:107200. [PMID: 37500029 DOI: 10.1016/j.vph.2023.107200] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Almost a third of all fatalities may be attributed to cardiovascular disease (CVD), making it a primary cause of mortalities worldwide. Better diagnostic tools and secure, non-invasive imaging techniques are needed to offer accurate information on CVD progression. Several elements contribute to the success of CVD personalized therapy, and two of the most crucial are accurate diagnosis and early detection. The therapy options available for conditions with a pathogenesis that unfold over decades, such as CVD, are very condition-specific and disease-stage based. Nanotechnology is increasingly being used as a therapeutic tool in the biomedical area, where they are used in various contexts, including diagnostics, biosensing, and drug administration. This review article provides an overview of the most recent applications of nanotechnology in the detection and management of prevalent CVDs.
Collapse
Affiliation(s)
- Feize Li
- Department of Cardiology, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China.
| | - Haibin Shao
- Department of Cardiology, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China
| | - Guoer Zhou
- Department of Pharmacy, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China
| | - Bingzhu Wang
- Internal Medicine of Integrated Traditional Chinese and Western Medicine, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China
| | - Yan Xu
- Intensive Care Unit, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China
| | - Wenqing Liang
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China
| | - Lin Chen
- Department of Cardiology, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China.
| |
Collapse
|
6
|
Wickline SA, Hou KK, Pan H. Peptide-Based Nanoparticles for Systemic Extrahepatic Delivery of Therapeutic Nucleotides. Int J Mol Sci 2023; 24:ijms24119455. [PMID: 37298407 DOI: 10.3390/ijms24119455] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Peptide-based nanoparticles (PBN) for nucleotide complexation and targeting of extrahepatic diseases are gaining recognition as potent pharmaceutical vehicles for fine-tuned control of protein production (up- and/or down-regulation) and for gene delivery. Herein, we review the principles and mechanisms underpinning self-assembled formation of PBN, cellular uptake, endosomal release, and delivery to extrahepatic disease sites after systemic administration. Selected examples of PBN that have demonstrated recent proof of concept in disease models in vivo are summarized to offer the reader a comparative view of the field and the possibilities for clinical application.
Collapse
Affiliation(s)
- Samuel A Wickline
- Division of Cardiology, Department of Medical Engineering, University of South Florida, Tampa, FL 33602, USA
| | - Kirk K Hou
- Department of Ophthalmology, Stein and Doheny Eye Institutes, University of California, Los Angeles, CA 90095, USA
| | - Hua Pan
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
7
|
Vargas I, Grabau RP, Chen J, Weinheimer C, Kovacs A, Dominguez-Viqueira W, Mitchell A, Wickline SA, Pan H. Simultaneous Inhibition of Thrombosis and Inflammation Is Beneficial in Treating Acute Myocardial Infarction. Int J Mol Sci 2023; 24:7333. [PMID: 37108494 PMCID: PMC10138953 DOI: 10.3390/ijms24087333] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Myocardial ischemia reperfusion injury (IRI) in acute coronary syndromes is a condition in which ischemic/hypoxic injury to cells subtended by the occluded vessel continues despite successful resolution of the thrombotic obstruction. For decades, most efforts to attenuate IRI have focused on interdicting singular molecular targets or pathways, but none have successfully transitioned to clinical use. In this work, we investigate a nanoparticle-based therapeutic strategy for profound but local thrombin inhibition that may simultaneously mitigate both thrombosis and inflammatory signaling pathways to limit myocardial IRI. Perfluorocarbon nanoparticles (PFC NP) were covalently coupled with an irreversible thrombin inhibitor, PPACK (Phe[D]-Pro-Arg-Chloromethylketone), and delivered intravenously to animals in a single dose prior to ischemia reperfusion injury. Fluorescent microscopy of tissue sections and 19F magnetic resonance images of whole hearts ex vivo demonstrated abundant delivery of PFC NP to the area at risk. Echocardiography at 24 h after reperfusion demonstrated preserved ventricular structure and improved function. Treatment reduced thrombin deposition, suppressed endothelial activation, inhibited inflammasome signaling pathways, and limited microvascular injury and vascular pruning in infarct border zones. Accordingly, thrombin inhibition with an extraordinarily potent but locally acting agent suggested a critical role for thrombin and a promising therapeutic strategy in cardiac IRI.
Collapse
Affiliation(s)
- Ian Vargas
- University of South Florida Heart Institute, University of South Florida, Tampa, FL 33602, USA
| | - Ryan P. Grabau
- University of South Florida Heart Institute, University of South Florida, Tampa, FL 33602, USA
| | - Junjie Chen
- Consortium for Translational Research in Advanced Imaging and Nanomedicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla Weinheimer
- Cardiovascular Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Attila Kovacs
- Cardiovascular Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Adam Mitchell
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel A. Wickline
- University of South Florida Heart Institute, University of South Florida, Tampa, FL 33602, USA
| | - Hua Pan
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63105, USA
| |
Collapse
|
8
|
Zhou Q, Quirk JD, Hu Y, Yan H, Gaut JP, Pham CTN, Wickline SA, Pan H. Rapamycin Perfluorocarbon Nanoparticle Mitigates Cisplatin-Induced Acute Kidney Injury. Int J Mol Sci 2023; 24:6086. [PMID: 37047059 PMCID: PMC10093942 DOI: 10.3390/ijms24076086] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
For nearly five decades, cisplatin has played an important role as a standard chemotherapeutic agent and been prescribed to 10-20% of all cancer patients. Although nephrotoxicity associated with platinum-based agents is well recognized, treatment of cisplatin-induced acute kidney injury is mainly supportive and no specific mechanism-based prophylactic approach is available to date. Here, we postulated that systemically delivered rapamycin perfluorocarbon nanoparticles (PFC NP) could reach the injured kidneys at sufficient and sustained concentrations to mitigate cisplatin-induced acute kidney injury and preserve renal function. Using fluorescence microscopic imaging and fluorine magnetic resonance imaging/spectroscopy, we illustrated that rapamycin-loaded PFC NP permeated and were retained in injured kidneys. Histologic evaluation and blood urea nitrogen (BUN) confirmed that renal structure and function were preserved 48 h after cisplatin injury. Similarly, weight loss was slowed down. Using western blotting and immunofluorescence staining, mechanistic studies revealed that rapamycin PFC NP significantly enhanced autophagy in the kidney, reduced the expression of intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1), as well as decreased the expression of the apoptotic protein Bax, all of which contributed to the suppression of apoptosis that was confirmed with TUNEL staining. In summary, the delivery of an approved agent such as rapamycin in a PFC NP format enhances local delivery and offers a novel mechanism-based prophylactic therapy for cisplatin-induced acute kidney injury.
Collapse
Affiliation(s)
- Qingyu Zhou
- Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA
| | - James D. Quirk
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ying Hu
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Huimin Yan
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph P. Gaut
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christine T. N. Pham
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel A. Wickline
- Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Hua Pan
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
9
|
Zhang W, Lv Z, Zhang Y, Gopinath SCB, Yuan Y, Huang D, Miao L. Targeted Diagnosis, Therapeutic Monitoring, and Assessment of Atherosclerosis Based on Mesoporous Silica Nanoparticles Coated with cRGD-Platelets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6006601. [PMID: 36211824 PMCID: PMC9537012 DOI: 10.1155/2022/6006601] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/20/2022] [Indexed: 12/07/2022]
Abstract
Objective The off-target effects and severe side effects of PPARα and LXRα agonists greatly limit their application in atherosclerosis (AS). Therefore, this study intended to use mesoporous silica nanoparticles as carriers to generate MnO nanoparticles in situ with T1WI-MRI in mesoporous pores and simultaneously load PPARα and LXRα agonists. Afterward, cRGD-chelated platelet membranes can be used for coating to construct a new nanotheranostic agent. Methods cRGD-platelet@MnO/MSN@PPARα/LXRα nanoparticles were synthesized by a chemical method. Dynamic light scattering (DLS) was utilized to detect the size distribution and polydispersity index (PDI) of the nanoparticles. The safety of the nanoparticles was detected by CCK8 in vitro and HE staining and kidney function in vivo. Cell apoptosis was detected by flow cytometry detection and TUNEL staining. Oxidative stress responses (ROS, SOD, MDA, and NOX levels) were tested via a DCFH-DA assay and commercial kits. Immunofluorescence and phagocytosis experiments were used to detect the targeting of nanoparticles. Magnetic resonance imaging (MRI) was used to detect the imaging performance of cRGD-platelet@MnO/MSN@PPARα/LXRα nanoparticles. Using western blotting, the expression changes in LXRα and ABCA1 were identified. Results cRGD-platelet@MnO/MSN@PPARα/LXRα nanoparticles were successfully established, with a particle size of approximately 150 nm and PDI less than 0.3, and showed high safety both in vitro and in vivo. cRGD-platelet@MnO/MSN@PPARα/LXRα nanoparticles showed good targeting properties and better MRI imaging performance in AS. cRGD-platelet@MnO/MSN@PPARα/LXRα nanoparticles showed better antioxidative capacities, MRI imaging performance, and diagnostic and therapeutic effects on AS by regulating the expression of LXRα and ABCA1. Conclusion In the present study, cRGD-platelet@MnO/MSN@PPARα/LXRα nanoparticles with high safety and the capacity to target vulnerable plaques of AS were successfully established. They showed better performance on MRI images and treatment effects on AS by promoting cholesterol efflux through the regulation of ABCA1. These findings might address the problems of off-target effects and side effects of nanoparticle-mediated drug delivery, which will enhance the efficiency of AS treatment and provide new ideas for the clinical treatment of AS.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Radiology, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi 545006, China
| | - Zheng Lv
- Department of Radiology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, China
| | - Yupeng Zhang
- Department of Radiology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, China
| | - Subash C. B. Gopinath
- Faculty of Chemical Engineering & Technology, Micro System Technology, Centre of Excellence (CoE), and Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), Perlis, Malaysia
| | - Yi Yuan
- Institute of Life Sciences, Jiangsu University, Zhengjiang, Jiangsu 212013, China
| | - Deyou Huang
- Department of Radiology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China
| | - Liu Miao
- Department of Cardiology, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi 545006, China
| |
Collapse
|
10
|
Tackling inflammation in atherosclerosis: Are we there yet and what lies beyond? Curr Opin Pharmacol 2022; 66:102283. [PMID: 36037627 DOI: 10.1016/j.coph.2022.102283] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/21/2022] [Accepted: 07/19/2022] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is a lipid-driven disease of the artery characterized by chronic non-resolving inflammation. Despite availability of excellent lipid-lowering therapies, atherosclerosis remains the leading cause of disability and death globally. The demonstration that suppressing inflammation prevents the adverse clinical manifestations of atherosclerosis in recent clinical trials has led to heightened interest in anti-inflammatory therapies. In this review, we briefly highlight some key anti-inflammatory and pro-resolution pathways, which could be targeted to modulate pathogenesis and stall atherosclerosis progression. We also highlight key challenges that must be overcome to turn the concept of inflammation targeting therapies into clinical reality for atherosclerotic heart disease.
Collapse
|
11
|
Wang W, Ma P, Zhao Q, Goorani S. Beneficial properties of the biosynthesized silver/chitosan nanoparticles mediated by Mentha piperita in rats with heart failure following myocardial infarction. INORG CHEM COMMUN 2022. [DOI: 10.1016/j.inoche.2022.109581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
12
|
Shah P, Chandra S. Review on emergence of nanomaterial coatings in bio-engineered cardiovascular stents. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Joseph JM, Gigliobianco MR, Firouzabadi BM, Censi R, Di Martino P. Nanotechnology as a Versatile Tool for 19F-MRI Agent's Formulation: A Glimpse into the Use of Perfluorinated and Fluorinated Compounds in Nanoparticles. Pharmaceutics 2022; 14:382. [PMID: 35214114 PMCID: PMC8874484 DOI: 10.3390/pharmaceutics14020382] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simultaneously being a non-radiative and non-invasive technique makes magnetic resonance imaging (MRI) one of the highly sought imaging techniques for the early diagnosis and treatment of diseases. Despite more than four decades of research on finding a suitable imaging agent from fluorine for clinical applications, it still lingers as a challenge to get the regulatory approval compared to its hydrogen counterpart. The pertinent hurdle is the simultaneous intrinsic hydrophobicity and lipophobicity of fluorine and its derivatives that make them insoluble in any liquids, strongly limiting their application in areas such as targeted delivery. A blossoming technique to circumvent the unfavorable physicochemical characteristics of perfluorocarbon compounds (PFCs) and guarantee a high local concentration of fluorine in the desired body part is to encapsulate them in nanosystems. In this review, we will be emphasizing different types of nanocarrier systems studied to encapsulate various PFCs and fluorinated compounds, headway to be applied as a contrast agent (CA) in fluorine-19 MRI (19F MRI). We would also scrutinize, especially from studies over the last decade, the different types of PFCs and their specific applications and limitations concerning the nanoparticle (NP) system used to encapsulate them. A critical evaluation for future opportunities would be speculated.
Collapse
Affiliation(s)
- Joice Maria Joseph
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (J.M.J.); (B.M.F.); (P.D.M.)
| | | | | | - Roberta Censi
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (J.M.J.); (B.M.F.); (P.D.M.)
| | - Piera Di Martino
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (J.M.J.); (B.M.F.); (P.D.M.)
- Dipartimento di Farmacia, Università “G. D’Annunzio” Chieti e Pescara, 66100 Chieti, Italy
| |
Collapse
|
14
|
Friedrich RP, Cicha I, Alexiou C. Iron Oxide Nanoparticles in Regenerative Medicine and Tissue Engineering. NANOMATERIALS 2021; 11:nano11092337. [PMID: 34578651 PMCID: PMC8466586 DOI: 10.3390/nano11092337] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022]
Abstract
In recent years, many promising nanotechnological approaches to biomedical research have been developed in order to increase implementation of regenerative medicine and tissue engineering in clinical practice. In the meantime, the use of nanomaterials for the regeneration of diseased or injured tissues is considered advantageous in most areas of medicine. In particular, for the treatment of cardiovascular, osteochondral and neurological defects, but also for the recovery of functions of other organs such as kidney, liver, pancreas, bladder, urethra and for wound healing, nanomaterials are increasingly being developed that serve as scaffolds, mimic the extracellular matrix and promote adhesion or differentiation of cells. This review focuses on the latest developments in regenerative medicine, in which iron oxide nanoparticles (IONPs) play a crucial role for tissue engineering and cell therapy. IONPs are not only enabling the use of non-invasive observation methods to monitor the therapy, but can also accelerate and enhance regeneration, either thanks to their inherent magnetic properties or by functionalization with bioactive or therapeutic compounds, such as drugs, enzymes and growth factors. In addition, the presence of magnetic fields can direct IONP-labeled cells specifically to the site of action or induce cell differentiation into a specific cell type through mechanotransduction.
Collapse
|
15
|
MacRitchie N, Di Francesco V, Ferreira MFMM, Guzik TJ, Decuzzi P, Maffia P. Nanoparticle theranostics in cardiovascular inflammation. Semin Immunol 2021; 56:101536. [PMID: 34862118 PMCID: PMC8811479 DOI: 10.1016/j.smim.2021.101536] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/30/2022]
Abstract
Theranostics, literally derived from the combination of the words diagnostics and therapy, is an emerging field of clinical and preclinical research, where contrast agents, drugs and diagnostic techniques are combined to simultaneously diagnose and treat pathologies. Nanoparticles are extensively employed in theranostics due to their potential to target specific organs and their multifunctional capacity. In this review, we will discuss the current state of theranostic nanomedicine, providing key examples of its application in the imaging and treatment of cardiovascular inflammation.
Collapse
Affiliation(s)
- Neil MacRitchie
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
| | - Valentina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | | | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
16
|
Guo B, Li Z, Tu P, Tang H, Tu Y. Molecular Imaging and Non-molecular Imaging of Atherosclerotic Plaque Thrombosis. Front Cardiovasc Med 2021; 8:692915. [PMID: 34291095 PMCID: PMC8286992 DOI: 10.3389/fcvm.2021.692915] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/08/2021] [Indexed: 12/11/2022] Open
Abstract
Thrombosis in the context of atherosclerosis typically results in life-threatening consequences, including acute coronary events and ischemic stroke. As such, early detection and treatment of thrombosis in atherosclerosis patients is essential. Clinical diagnosis of thrombosis in these patients is typically based upon a combination of imaging approaches. However, conventional imaging modalities primarily focus on assessing the anatomical structure and physiological function, severely constraining their ability to detect early thrombus formation or the processes underlying such pathology. Recently, however, novel molecular and non-molecular imaging strategies have been developed to assess thrombus composition and activity at the molecular and cellular levels more accurately. These approaches have been successfully used to markedly reduce rates of atherothrombotic events in patients suffering from acute coronary syndrome (ACS) by facilitating simultaneous diagnosis and personalized treatment of thrombosis. Moreover, these modalities allow monitoring of plaque condition for preventing plaque rupture and associated adverse cardiovascular events in such patients. Sustained developments in molecular and non-molecular imaging technologies have enabled the increasingly specific and sensitive diagnosis of atherothrombosis in animal studies and clinical settings, making these technologies invaluable to patients' health in the future. In the present review, we discuss current progress regarding the non-molecular and molecular imaging of thrombosis in different animal studies and atherosclerotic patients.
Collapse
Affiliation(s)
- Bingchen Guo
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhaoyue Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peiyang Tu
- College of Clinical Medicine, Hubei University of Science and Technology, Xianning, China
| | - Hao Tang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingfeng Tu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Zhu C, Ma J, Ji Z, Shen J, Wang Q. Recent Advances of Cell Membrane Coated Nanoparticles in Treating Cardiovascular Disorders. Molecules 2021; 26:3428. [PMID: 34198794 PMCID: PMC8201295 DOI: 10.3390/molecules26113428] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/30/2021] [Accepted: 06/03/2021] [Indexed: 01/13/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide, causing approximately 17.9 million deaths annually, an estimated 31% of all deaths, according to the WHO. CVDs are essentially rooted in atherosclerosis and are clinically classified into coronary heart disease, stroke and peripheral vascular disorders. Current clinical interventions include early diagnosis, the insertion of stents, and long-term preventive therapy. However, clinical diagnostic and therapeutic tools are subject to a number of limitations including, but not limited to, potential toxicity induced by contrast agents and unexpected bleeding caused by anti-platelet drugs. Nanomedicine has achieved great advancements in biomedical area. Among them, cell membrane coated nanoparticles, denoted as CMCNPs, have acquired enormous expectations due to their biomimetic properties. Such membrane coating technology not only helps avoid immune clearance, but also endows nanoparticles with diverse cellular and functional mimicry. In this review, we will describe the superiorities of CMCNPs in treating cardiovascular diseases and their potentials in optimizing current clinical managements.
Collapse
Affiliation(s)
- Chaojie Zhu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Chu Kochen Honors College, Zhejiang University, Hangzhou 310058, China; (J.M.); (Z.J.)
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Junkai Ma
- Chu Kochen Honors College, Zhejiang University, Hangzhou 310058, China; (J.M.); (Z.J.)
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhiheng Ji
- Chu Kochen Honors College, Zhejiang University, Hangzhou 310058, China; (J.M.); (Z.J.)
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jie Shen
- Department of Pharmacy, School of Medicine, Zhejiang University City College, Hangzhou 310015, China
| | - Qiwen Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| |
Collapse
|
18
|
Karantas ID, Okur ME, Okur NÜ, Siafaka PI. Dyslipidemia Management in 2020: An Update on Diagnosis and Therapeutic Perspectives. Endocr Metab Immune Disord Drug Targets 2021; 21:815-834. [PMID: 32778041 DOI: 10.2174/1871530320666200810144004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/10/2020] [Accepted: 06/19/2020] [Indexed: 11/22/2022]
Abstract
Cardiovascular diseases are the leading cause of death in the modern world and dyslipidemia is one of the major risk factors. The current therapeutic strategies for cardiovascular diseases involve the management of risk factors, especially dyslipidemia and hypertension. Recently, the updated guidelines of dyslipidemia management were presented, and the newest data were included in terms of diagnosis, imaging, and treatment. In this targeted literature review, the researchers presented the most recent evidence on dyslipidemia management by including the current therapeutic goals for it. In addition, the novel diagnostic tools based on theranostics are shown. Finally, the future perspectives on treatment based on novel drug delivery systems and their potential to be used in clinical trials were also analyzed. It should be noted that dyslipidemia management can be achieved by the strict lifestyle change, i.e., by adopting a healthy life, and choosing the most suitable medication. This review can help medical professionals as well as specialists of other sciences to update their knowledge on dyslipidemia management, which can lead to better therapeutic outcomes and newer drug developments.
Collapse
Affiliation(s)
| | - Mehmet E Okur
- University of Health Sciences, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey
| | - Neslihan Ü Okur
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Health Sciences, Istanbul, Turkey
| | - Panoraia I Siafaka
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
19
|
Imaging of Inflammation in Spinal Cord Injury: Novel Insights on the Usage of PFC-Based Contrast Agents. Biomedicines 2021; 9:biomedicines9040379. [PMID: 33916774 PMCID: PMC8065995 DOI: 10.3390/biomedicines9040379] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/04/2022] Open
Abstract
Labeling of macrophages with perfluorocarbon (PFC)-based compounds allows the visualization of inflammatory processes by 19F-magnetic resonance imaging (19F-MRI), due to the absence of endogenous background. Even if PFC-labeling of monocytes/macrophages has been largely investigated and used, information is lacking about the impact of these agents over the polarization towards one of their cell subsets and on the best way to image them. In the present work, a PFC-based nanoemulsion was developed to monitor the course of inflammation in a model of spinal cord injury (SCI), a pathology in which the understanding of immunological events is of utmost importance to select the optimal therapeutic strategies. The effects of PFC over macrophage polarization were studied in vitro, on cultured macrophages, and in vivo, in a mouse SCI model, by testing and comparing various cell tracking protocols, including single and multiple administrations, the use of MRI or Point Resolved Spectroscopy (PRESS), and application of pre-saturation of Kupffer cells. The blood half-life of nanoemulsion was also investigated by 19F Magnetic Resonance Spectroscopy (MRS). In vitro and in vivo results indicate the occurrence of a switch towards the M2 (anti-inflammatory) phenotype, suggesting a possible theranostic function of these nanoparticles. The comparative work presented here allows the reader to select the most appropriate protocol according to the research objectives (quantitative data acquisition, visual monitoring of macrophage recruitment, theranostic purpose, rapid MRI acquisition, etc.). Finally, the method developed here to determine the blood half-life of the PFC nanoemulsion can be extended to other fluorinated compounds.
Collapse
|
20
|
Mohanta Z, Gaonkar SK, Kumar M, Saini J, Tiwari V, Srivastava C, Atreya HS. Influence of Oxidation Degree of Graphene Oxide on Its Nuclear Relaxivity and Contrast in MRI. ACS OMEGA 2020; 5:22131-22139. [PMID: 32923771 PMCID: PMC7482091 DOI: 10.1021/acsomega.0c02220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/14/2020] [Indexed: 06/11/2023]
Abstract
Graphene oxide (GO) serves as a versatile platform for various applications, with the oxygen content of GO playing an important role in governing its properties. In the present study, different GO types covering a wide range of oxidation degree were prepared using our newly developed two-step method involving ball milling of graphite followed by its oxidation to GO. In addition to the variations in their physicochemical properties, the different GO types exhibited differences in proton relaxivity due to their paramagnetic nature. Nuclear magnetic resonance spectroscopy studies showed that the degree of oxidation of GO perturbs its nuclear relaxation properties and, together with intercalated Mn2+ ions, provides large contrast variation in magnetic resonance imaging (MRI). The study for the first time reveals that the surface chemistry of GO affects its relaxivity and opens up new avenues for developing tunable GO-based contrast agents in magnetic resonance imaging for diagnostics and therapies.
Collapse
Affiliation(s)
- Zinia Mohanta
- Centre
for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Sumana K. Gaonkar
- Nuclear
Magnetic Resonance Research Centre, Indian
Institute of Science, Bengaluru 560012, India
| | - Manoj Kumar
- Department
of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences, Bengaluru 560029, India
| | - Jitender Saini
- Department
of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences, Bengaluru 560029, India
| | - Vivek Tiwari
- Centre
for Brain Research, Indian Institute of
Science, Bengaluru 560012, India
| | - Chandan Srivastava
- Department
of Materials Engineering, Indian Institute
of Science, Bengaluru 560012, India
| | - Hanudatta S. Atreya
- Nuclear
Magnetic Resonance Research Centre, Indian
Institute of Science, Bengaluru 560012, India
| |
Collapse
|
21
|
Pala R, Anju VT, Dyavaiah M, Busi S, Nauli SM. Nanoparticle-Mediated Drug Delivery for the Treatment of Cardiovascular Diseases. Int J Nanomedicine 2020; 15:3741-3769. [PMID: 32547026 PMCID: PMC7266400 DOI: 10.2147/ijn.s250872] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are one of the foremost causes of high morbidity and mortality globally. Preventive, diagnostic, and treatment measures available for CVDs are not very useful, which demands promising alternative methods. Nanoscience and nanotechnology open a new window in the area of CVDs with an opportunity to achieve effective treatment, better prognosis, and less adverse effects on non-target tissues. The application of nanoparticles and nanocarriers in the area of cardiology has gathered much attention due to the properties such as passive and active targeting to the cardiac tissues, improved target specificity, and sensitivity. It has reported that more than 50% of CVDs can be treated effectively through the use of nanotechnology. The main goal of this review is to explore the recent advancements in nanoparticle-based cardiovascular drug carriers. This review also summarizes the difficulties associated with the conventional treatment modalities in comparison to the nanomedicine for CVDs.
Collapse
Affiliation(s)
- Rajasekharreddy Pala
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA92618, USA
- Department of Medicine, University of California Irvine, Irvine, CA92868, USA
| | - V T Anju
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Madhu Dyavaiah
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Siddhardha Busi
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Surya M Nauli
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA92618, USA
- Department of Medicine, University of California Irvine, Irvine, CA92868, USA
| |
Collapse
|
22
|
Qiao R, Huang X, Qin Y, Li Y, Davis TP, Hagemeyer CE, Gao M. Recent advances in molecular imaging of atherosclerotic plaques and thrombosis. NANOSCALE 2020; 12:8040-8064. [PMID: 32239038 DOI: 10.1039/d0nr00599a] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
As the complications of atherosclerosis such as myocardial infarction and stroke are still one of the leading causes of mortality worldwide, the development of new diagnostic tools for the early detection of plaque instability and thrombosis is urgently needed. Advanced molecular imaging probes based on functional nanomaterials in combination with cutting edge imaging techniques are now paving the way for novel and unique approaches to monitor the inflammatory progress in atherosclerosis. This review focuses on the development of various molecular probes for the diagnosis of plaques and thrombosis in atherosclerosis, along with perspectives of their diagnostic applications in cardiovascular diseases. Specifically, we summarize the biological targets that can be used for atherosclerosis and thrombosis imaging. Then we describe the emerging molecular imaging techniques based on the utilization of engineered nanoprobes together with their challenges in clinical translation.
Collapse
Affiliation(s)
- Ruirui Qiao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Molecular imaging of inflammation - Current and emerging technologies for diagnosis and treatment. Pharmacol Ther 2020; 211:107550. [PMID: 32325067 DOI: 10.1016/j.pharmthera.2020.107550] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022]
Abstract
Inflammation is a key factor in multiple diseases including primary immune-mediated inflammatory diseases e.g. rheumatoid arthritis but also, less obviously, in many other common conditions, e.g. cardiovascular disease and diabetes. Together, chronic inflammatory diseases contribute to the majority of global morbidity and mortality. However, our understanding of the underlying processes by which the immune response is activated and sustained is limited by a lack of cellular and molecular information obtained in situ. Molecular imaging is the visualization, detection and quantification of molecules in the body. The ability to reveal information on inflammatory biomarkers, pathways and cells can improve disease diagnosis, guide and monitor therapeutic intervention and identify new targets for research. The optimum molecular imaging modality will possess high sensitivity and high resolution and be capable of non-invasive quantitative imaging of multiple disease biomarkers while maintaining an acceptable safety profile. The mainstays of current clinical imaging are computed tomography (CT), magnetic resonance imaging (MRI), ultrasound (US) and nuclear imaging such as positron emission tomography (PET). However, none of these have yet progressed to routine clinical use in the molecular imaging of inflammation, therefore new approaches are required to meet this goal. This review sets out the respective merits and limitations of both established and emerging imaging modalities as clinically useful molecular imaging tools in addition to potential theranostic applications.
Collapse
|
24
|
Vigne J, Cabella C, Dézsi L, Rustique E, Couffin AC, Aid R, Anizan N, Chauvierre C, Letourneur D, Le Guludec D, Rouzet F, Hyafil F, Mészáros T, Fülöp T, Szebeni J, Cordaro A, Oliva P, Mourier V, Texier I. Nanostructured lipid carriers accumulate in atherosclerotic plaques of ApoE -/- mice. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 25:102157. [PMID: 31982616 DOI: 10.1016/j.nano.2020.102157] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/04/2019] [Accepted: 01/18/2020] [Indexed: 12/20/2022]
Abstract
Nanostructured lipid carriers (NLC) might represent an interesting approach for the identification and targeting of rupture-prone atherosclerotic plaques. In this study, we evaluated the biodistribution, targeting ability and safety of 64Cu-fonctionalized NLC in atherosclerotic mice. 64Cu-chelating-NLC (51.8±3.1 nm diameter) with low dispersity index (0.066±0.016) were produced by high pressure homogenization at tens-of-grams scale. 24 h after injection of 64Cu-chelated particles in ApoE-/- mice, focal regions of the aorta showed accumulation of particles on autoradiography that colocalized with Oil Red O lipid mapping. Signal intensity was significantly greater in aortas isolated from ApoE-/- mice compared to wild type (WT) control (8.95 [7.58, 10.16]×108 vs 4.59 [3.11, 5.03]×108 QL/mm2, P < 0.05). Moreover, NLC seemed safe in relevant biocompatibility studies. NLC could constitute an interesting platform with high clinical translation potential for targeted delivery and imaging purposes in atherosclerosis.
Collapse
Affiliation(s)
- Jonathan Vigne
- Université de Paris, LVTS, INSERM U1148, Paris, France; Nuclear Medicine Department, X. Bichat Hospital, APHP and DHU FIRE, Paris, France; Université de Paris, UMS34 FRIM, Paris, France
| | - Claudia Cabella
- Centro Ricerche Bracco, Bracco Imaging SpA, Colleretto Giacosa, Italy
| | - László Dézsi
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | | | | | - Rachida Aid
- Université de Paris, UMS34 FRIM, Paris, France
| | | | | | | | - Dominique Le Guludec
- Université de Paris, LVTS, INSERM U1148, Paris, France; Nuclear Medicine Department, X. Bichat Hospital, APHP and DHU FIRE, Paris, France; Université de Paris, UMS34 FRIM, Paris, France
| | - François Rouzet
- Université de Paris, LVTS, INSERM U1148, Paris, France; Nuclear Medicine Department, X. Bichat Hospital, APHP and DHU FIRE, Paris, France; Université de Paris, UMS34 FRIM, Paris, France
| | - Fabien Hyafil
- Université de Paris, LVTS, INSERM U1148, Paris, France; Nuclear Medicine Department, X. Bichat Hospital, APHP and DHU FIRE, Paris, France; Université de Paris, UMS34 FRIM, Paris, France
| | - Tamás Mészáros
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - Tamás Fülöp
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - János Szebeni
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - Alessia Cordaro
- Centro Ricerche Bracco, Bracco Imaging SpA, Colleretto Giacosa, Italy
| | - Paolo Oliva
- Centro Ricerche Bracco, Bracco Imaging SpA, Colleretto Giacosa, Italy
| | | | | |
Collapse
|
25
|
Biodistribution of Nanostructured Lipid Carriers in Mice Atherosclerotic Model. Molecules 2019; 24:molecules24193499. [PMID: 31561608 PMCID: PMC6803849 DOI: 10.3390/molecules24193499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 01/08/2023] Open
Abstract
Atherosclerosis is a major cardiovascular disease worldwide, that could benefit from innovative nanomedicine imaging tools and treatments. In this perspective, we here studied, by fluorescence imaging in ApoE-/- mice, the biodistribution of non-functionalized and RXP470.1-targeted nanostructured lipid carriers (NLC) loaded with DiD dye. RXP470.1 specifically binds to MMP12, a metalloprotease that is over-expressed by macrophages residing in atherosclerotic plaques. Physico-chemical characterizations showed that RXP-NLC (about 105 RXP470.1 moieties/particle) displayed similar features as non-functionalized NLC in terms of particle diameter (about 60-65 nm), surface charge (about −5 — −10 mV), and colloidal stability. In vitro inhibition assays demonstrated that RXP-NLC conserved a selectivity and affinity profile, which favored MMP-12. In vivo data indicated that NLC and RXP-NLC presented prolonged blood circulation and accumulation in atherosclerotic lesions in a few hours. Twenty-four hours after injection, particle uptake in atherosclerotic plaques of the brachiocephalic artery was similar for both nanoparticles, as assessed by ex vivo imaging. This suggests that the RXP470.1 coating did not significantly induce an active targeting of the nanoparticles within the plaques. Overall, NLCs appeared to be very promising nanovectors to efficiently and specifically deliver imaging agents or drugs in atherosclerotic lesions, opening avenues for new nanomedicine strategies for cardiovascular diseases.
Collapse
|
26
|
Cattaneo M, Froio A, Gallino A. Cardiovascular Imaging and Theranostics in Cardiovascular Pharmacotherapy. Eur Cardiol 2019; 14:62-64. [PMID: 31131039 PMCID: PMC6523052 DOI: 10.15420/ecr.2019.6.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Imaging plays a pivotal role in the diagnostic and prognostic assessment of cardiovascular diseases. During the past two decades, there has been an expansion of the available imaging techniques, some of which are now part of routine clinical practice. Cardiovascular imaging of atherosclerosis is a useful instrument, and it can corroborate and expand pathophysiological evidence on cardiovascular disease, providing proof of concept for medical therapy and can predict its responsiveness, and it may be able to be used as surrogate endpoints for clinical trials. Theranostics is an emerging therapy that combines imaging and therapeutic functions, using imaging-based therapeutic delivery systems. Theranostics could partially overcome current imaging limitations and translate experimental evidence and large-scale trials assessing clinical endpoints, rationalising cardiovascular drug development and paving the way to personalised medicine. The medical community cannot overlook the use of cardiovascular imaging as a complementary and supportive adjunct to trials investigating clinical endpoints, which remain the mainstay for investigating the efficacy and safety of cardiovascular pharmacotherapy.
Collapse
Affiliation(s)
- Mattia Cattaneo
- Cardiovascular Research Unit, Ospedale Regionale di Bellinzona e Valli Bellinzona, Switzerland.,Department of Cardiovascular Intensive Care, Cardiocentro Ticino Lugano, Switzerland
| | - Alberto Froio
- Department of Surgery and Interdisciplinary Medicine, University of Milano-Bicocca Milan, Italy
| | - Augusto Gallino
- Cardiovascular Research Unit, Ospedale Regionale di Bellinzona e Valli Bellinzona, Switzerland.,University of Zurich Zurich, Switzerland
| |
Collapse
|
27
|
Fluorinated MRI contrast agents and their versatile applications in the biomedical field. Future Med Chem 2019; 11:1157-1175. [DOI: 10.4155/fmc-2018-0463] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
MRI has been recognized as one of the most applied medical imaging techniques in clinical practice. However, the presence of background signal coming from water protons in surrounding tissues makes sometimes the visualization of local contrast agents difficult. To remedy this, fluorine has been introduced as a reliable perspective, thanks to its magnetic properties being relatively close to those of protons. In this review, we aim to give an overall description of fluorine incorporation in contrast agents for MRI. The different kinds of fluorinated probes such as perfluorocarbons, fluorinated dendrimers, polymers and paramagnetic probes will be described, as will their imaging applications such as chemical exchange saturation transfer (CEST) imaging, physico-chemical changes detection, drug delivery, cell tracking and inflammation or tumors detection.
Collapse
|
28
|
Han X, Xu K, Taratula O, Farsad K. Applications of nanoparticles in biomedical imaging. NANOSCALE 2019; 11:799-819. [PMID: 30603750 PMCID: PMC8112886 DOI: 10.1039/c8nr07769j] [Citation(s) in RCA: 289] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
An urgent need for early detection and diagnosis of diseases continuously pushes the advancements of imaging modalities and contrast agents. Current challenges remain for fast and detailed imaging of tissue microstructures and lesion characterization that could be achieved via development of nontoxic contrast agents with longer circulation time. Nanoparticle technology offers this possibility. Here, we review nanoparticle-based contrast agents employed in most common biomedical imaging modalities, including fluorescence imaging, MRI, CT, US, PET and SPECT, addressing their structure related features, advantages and limitations. Furthermore, their applications in each imaging modality are also reviewed using commonly studied examples. Future research will investigate multifunctional nanoplatforms to address safety, efficacy and theranostic capabilities. Nanoparticles as imaging contrast agents have promise to greatly benefit clinical practice.
Collapse
Affiliation(s)
- Xiangjun Han
- Department of Radiology, First Hospital of China Medical University, Shenyang, Liaoning, 110001 P. R. China.
| | | | | | | |
Collapse
|
29
|
Strobel HA, Qendro EI, Alsberg E, Rolle MW. Targeted Delivery of Bioactive Molecules for Vascular Intervention and Tissue Engineering. Front Pharmacol 2018; 9:1329. [PMID: 30519186 PMCID: PMC6259603 DOI: 10.3389/fphar.2018.01329] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 10/29/2018] [Indexed: 01/25/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death in the United States. Treatment often requires surgical interventions to re-open occluded vessels, bypass severe occlusions, or stabilize aneurysms. Despite the short-term success of such interventions, many ultimately fail due to thrombosis or restenosis (following stent placement), or incomplete healing (such as after aneurysm coil placement). Bioactive molecules capable of modulating host tissue responses and preventing these complications have been identified, but systemic delivery is often harmful or ineffective. This review discusses the use of localized bioactive molecule delivery methods to enhance the long-term success of vascular interventions, such as drug-eluting stents and aneurysm coils, as well as nanoparticles for targeted molecule delivery. Vascular grafts in particular have poor patency in small diameter, high flow applications, such as coronary artery bypass grafting (CABG). Grafts fabricated from a variety of approaches may benefit from bioactive molecule incorporation to improve patency. Tissue engineering is an especially promising approach for vascular graft fabrication that may be conducive to incorporation of drugs or growth factors. Overall, localized and targeted delivery of bioactive molecules has shown promise for improving the outcomes of vascular interventions, with technologies such as drug-eluting stents showing excellent clinical success. However, many targeted vascular drug delivery systems have yet to reach the clinic. There is still a need to better optimize bioactive molecule release kinetics and identify synergistic biomolecule combinations before the clinical impact of these technologies can be realized.
Collapse
Affiliation(s)
- Hannah A. Strobel
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Elisabet I. Qendro
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA, United States
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Marsha W. Rolle
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States
| |
Collapse
|
30
|
Poon C, Gallo J, Joo J, Chang T, Bañobre-López M, Chung EJ. Hybrid, metal oxide-peptide amphiphile micelles for molecular magnetic resonance imaging of atherosclerosis. J Nanobiotechnology 2018; 16:92. [PMID: 30442135 PMCID: PMC6238287 DOI: 10.1186/s12951-018-0420-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 11/07/2018] [Indexed: 01/17/2023] Open
Abstract
Background Atherosclerosis, a major source of cardiovascular disease, is asymptomatic for decades until the activation of thrombosis and the rupture of enlarged plaques, resulting in acute coronary syndromes and sudden cardiac arrest. Magnetic resonance imaging (MRI) is a noninvasive nuclear imaging technique to assess the degree of atherosclerotic plaque with high spatial resolution and excellent soft tissue contrast. However, MRI lacks sensitivity for preventive medicine, which limits the ability to observe the onset of vulnerable plaques. In this study, we engineered hybrid metal oxide-peptide amphiphile micelles (HMO-Ms) that combine an inorganic, magnetic iron oxide or manganese oxide inner core with organic, fibrin-targeting peptide amphiphiles, consisting of the sequence CREKA, for potential MRI imaging of thrombosis on atherosclerotic plaques. Results Hybrid metal oxide-peptide amphiphile micelles, consisting of an iron oxide (Fe-Ms) or manganese oxide (Mn-Ms) core with CREKA peptides, were self-assembled into 20–30 nm spherical nanoparticles, as confirmed by dynamic light scattering and transmission electron microscopy. These hybrid nanoparticles were found to be biocompatible with human aortic endothelial cells in vitro, and HMO-Ms bound to human clots three to five times more efficiently than its non-targeted counterparts. Relaxivity studies showed ultra-high r2 value of 457 mM−1 s−1 and r1 value of 0.48 mM−1 s−1 for Fe-Ms and Mn-Ms, respectively. In vitro, MR imaging studies demonstrated the targeting capability of CREKA-functionalized hybrid nanoparticles with twofold enhancement of MR signals. Conclusion This novel hybrid class of MR agents has potential as a non-invasive imaging method that specifically detects thrombosis during the pathogenesis of atherosclerosis. Electronic supplementary material The online version of this article (10.1186/s12951-018-0420-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher Poon
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, Los Angeles, CA, 90089, USA
| | - Juan Gallo
- Advanced (Magnetic) Theranostic Nanostructures Lab, Department of Life Sciences, International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga, Portugal
| | - Johan Joo
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, Los Angeles, CA, 90089, USA
| | - Timothy Chang
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, Los Angeles, CA, 90089, USA
| | - Manuel Bañobre-López
- Advanced (Magnetic) Theranostic Nanostructures Lab, Department of Life Sciences, International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga, Portugal.
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, Los Angeles, CA, 90089, USA. .,Department of Materials Science and Chemical Engineering, University of Southern California, 925 Bloom Walk, Los Angeles, CA, 90089, USA. .,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA. .,Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA. .,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
31
|
Antithrombin Perfluorocarbon Nanoparticles Improve Renal Allograft Function in a Murine Deceased Criteria Donor Model. Transplant Direct 2018; 4:e384. [PMID: 30234153 PMCID: PMC6133403 DOI: 10.1097/txd.0000000000000817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 06/19/2018] [Indexed: 11/26/2022] Open
Abstract
Background Over 100 000 patients await renal transplantation and 4000 die per year. Compounding this mismatch between supply and demand is delayed graft function which contributes to short-term and long-term graft failures. Previously, we reported that thrombin-targeted perfluorocarbon nanoparticles (PFC-NP) protect kidneys from ischemic renal injury after transient arterial occlusion. Here we hypothesize that perfusion of renal allografts with PFC-NP similarly can protect graft function after an ischemic interval. Methods After 60 minutes of warm ischemia, male Lewis rats underwent left renal explantation followed by renal perfusion with 5 mL of standard perfusate alone (N = 3) or with 0.3 mL of untargeted PFC-NP (N = 5) or 0.3 mL thrombin-targeted of PFC NP functionalized with phenylalanine-proline-arginine-chloromethylketone (PPACK) (PFC-PPACK), an irreversible thrombin inhibitor (N = 5). Kidneys underwent 6 hours of cold storage, followed by transplantation into recipients and native nephrectomy. Animals were euthanized at 24 hours for tissue collection or at 48 hours for blood and renal tissue collection. A survival experiment was performed using the same protocol with saline control (N = 3), PFC-NP (N = 3) or PFC-PPACK (N = 6). Results Serum creatinine was improved for the PFC-PPACK groups as compared with control groups (P < 0.04). Kaplan-Meier survival curves also indicated increased longevity (P < 0.05). Blinded histologic scoring revealed markedly attenuated renal damage in the PFC-PPACK group compared to untreated animals (2.75 ± 1.60 versus 0.83 ± 3.89; P = 0.0001) and greater preservation of renal vasculature. Conclusions These results validate an NP-based approach to improve renal graft function as antithrombin NPs improved allograft function, decreased renal damage, protected vasculature, and improved longevity.
Collapse
|
32
|
Chandarana M, Curtis A, Hoskins C. The use of nanotechnology in cardiovascular disease. APPLIED NANOSCIENCE 2018. [DOI: 10.1007/s13204-018-0856-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
33
|
Yu M, Bouley BS, Xie D, Enriquez JS, Que EL. 19F PARASHIFT Probes for Magnetic Resonance Detection of H2O2 and Peroxidase Activity. J Am Chem Soc 2018; 140:10546-10552. [DOI: 10.1021/jacs.8b05685] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Meng Yu
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712-1224, United States
| | - Bailey S. Bouley
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712-1224, United States
| | - Da Xie
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712-1224, United States
| | - José S. Enriquez
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712-1224, United States
| | - Emily L. Que
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712-1224, United States
| |
Collapse
|
34
|
Woodside DG, Tanifum EA, Ghaghada KB, Biediger RJ, Caivano AR, Starosolski ZA, Khounlo S, Bhayana S, Abbasi S, Craft JW, Maxwell DS, Patel C, Stupin IV, Bakthavatsalam D, Market RV, Willerson JT, Dixon RAF, Vanderslice P, Annapragada AV. Magnetic Resonance Imaging of Atherosclerotic Plaque at Clinically Relevant Field Strengths (1T) by Targeting the Integrin α4β1. Sci Rep 2018; 8:3733. [PMID: 29487319 PMCID: PMC5829217 DOI: 10.1038/s41598-018-21893-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 02/12/2018] [Indexed: 02/07/2023] Open
Abstract
Inflammation drives the degradation of atherosclerotic plaque, yet there are no non-invasive techniques available for imaging overall inflammation in atherosclerotic plaques, especially in the coronary arteries. To address this, we have developed a clinically relevant system to image overall inflammatory cell burden in plaque. Here, we describe a targeted contrast agent (THI0567-targeted liposomal-Gd) that is suitable for magnetic resonance (MR) imaging and binds with high affinity and selectivity to the integrin α4β1(very late antigen-4, VLA-4), a key integrin involved in recruiting inflammatory cells to atherosclerotic plaques. This liposomal contrast agent has a high T1 relaxivity (~2 × 105 mM-1s-1 on a particle basis) resulting in the ability to image liposomes at a clinically relevant MR field strength. We were able to visualize atherosclerotic plaques in various regions of the aorta in atherosclerosis-prone ApoE-/- mice on a 1 Tesla small animal MRI scanner. These enhanced signals corresponded to the accumulation of monocyte/macrophages in the subendothelial layer of atherosclerotic plaques in vivo, whereas non-targeted liposomal nanoparticles did not demonstrate comparable signal enhancement. An inflammatory cell-targeted method that has the specificity and sensitivity to measure the inflammatory burden of a plaque could be used to noninvasively identify patients at risk of an acute ischemic event.
Collapse
Affiliation(s)
- Darren G Woodside
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA.
| | - Eric A Tanifum
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Ketan B Ghaghada
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Ronald J Biediger
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Amy R Caivano
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Zbigniew A Starosolski
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Sayadeth Khounlo
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Saakshi Bhayana
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Shahrzad Abbasi
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - John W Craft
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA.,Department of Biology and Chemistry, University of Houston, 4800 Calhoun Road, Houston, Texas, 77004, USA
| | - David S Maxwell
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas, 77030, USA.,Department of Institutional Analytics and Informatics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chandreshkumar Patel
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Igor V Stupin
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | | | - Robert V Market
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - James T Willerson
- Division of Cardiology Research, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Richard A F Dixon
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Peter Vanderslice
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Ananth V Annapragada
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA.
| |
Collapse
|
35
|
Martinez JO, Molinaro R, Hartman KA, Boada C, Sukhovershin R, De Rosa E, Kuri D, Zhang S, Evangelopoulos M, Carter AM, Bibb JA, Cooke JP, Tasciotti E. Biomimetic nanoparticles with enhanced affinity towards activated endothelium as versatile tools for theranostic drug delivery. Theranostics 2018; 8:1131-1145. [PMID: 29464004 PMCID: PMC5817115 DOI: 10.7150/thno.22078] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/09/2017] [Indexed: 12/30/2022] Open
Abstract
Activation of the vascular endothelium is characterized by increased expression of vascular adhesion molecules and chemokines. This activation occurs early in the progression of several diseases and triggers the recruitment of leukocytes. Inspired by the tropism of leukocytes, we investigated leukocyte-based biomimetic nanoparticles (i.e., leukosomes) as a novel theranostic platform for inflammatory diseases. Methods: Leukosomes were assembled by combining phospholipids and membrane proteins from leukocytes. For imaging applications, phospholipids modified with rhodamine and gadolinium were used. Leukosomes incubated with antibodies blocking lymphocyte function-associated antigen 1 (LFA-1) and CD45 were administered to explore their roles in targeting inflammation. In addition, relaxometric assessment of NPs was evaluated. Results: Liposomes and leukosomes were both spherical in shape with sizes ranging from 140-170 nm. Both NPs successfully integrated 8 and 13 µg of rhodamine and gadolinium, respectively, and demonstrated less than 4% variation in physicochemical features. Leukosomes demonstrated a 16-fold increase in breast tumor accumulation relative to liposomes. Furthermore, quantification of leukosomes in tumor vessels demonstrated a 4.5-fold increase in vessel lumens and a 14-fold increase in vessel walls. Investigating the targeting mechanism of action revealed that blockage of LFA-1 on leukosomes resulted in a 95% decrease in tumor accumulation. Whereas blockage of CD45 yielded a 60% decrease in targeting and significant increases in liver and spleen accumulation. In addition, when administered in mice with atherosclerotic plaques, leukosomes exhibited a 4-fold increase in the targeting of inflammatory vascular lesions. Lastly, relaxometric assessment of NPs demonstrated that the incorporation of membrane proteins into leukosomes did not impact the r1 and r2 relaxivities of the NPs, demonstrating 6 and 30 mM-1s-1, respectively. Conclusion: Our study demonstrates the ability of leukosomes to target activated vasculature and exhibit superior accumulation in tumors and vascular lesions. The versatility of the phospholipid backbone within leukosomes permits the incorporation of various contrast agents. Furthermore, leukosomes can potentially be loaded with therapeutics possessing diverse physical properties and thus warrant further investigation toward the development of powerful theranostic agents.
Collapse
|
36
|
Yuan N, Zhang X, Cao Y, Jiang X, Zhao S, Feng Y, Fan Y, Lu Z, Gao H. Contrast-enhanced computerized tomography combined with a targeted nanoparticle contrast agent for screening for early-phase non-small cell lung cancer. Exp Ther Med 2017; 14:5063-5068. [PMID: 29201215 DOI: 10.3892/etm.2017.5140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 05/11/2017] [Indexed: 12/13/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a major cause of morbidity and mortality, and patients with NSCLC are frequently diagnosed at an advanced stage. This is primarily due to a lack of advanced and sensitive protocols for the detection of early stage NSCLC. Therefore, methods for the accurate diagnosis of early stage NSCLC are urgently required to improve survival rates. The present study investigated the use of contrast-enhanced computerized tomography (CECT) combined with a targeted nanoparticle contrast agent (TNCA) to diagnose early-stage NSCLC in a mice xenograft model. The TNCA used was lenvatinib, a multi-target tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor 1-3, fibroblast growth factor receptor 1-4, platelet-derived growth factor receptor β, proto-oncogene tyrosine-protein kinase receptor Ret and mast/stem cell growth factor receptor Kit. Xenograft NSCLC mice were established and used to analyze the efficacy of CECT-TNCA compared with CT scanning alone. The TNCA was inhaled with the use of an atomizer. The results demonstrated that CECT-TNCA improved the sensitivity of the diagnosis of early stage NSCLC. In addition, imaging using the TNCA enabled the visualization of nodules in the lung in mice with early stage NSCLC. In addition, lung nodule signal enhancement was increased in CECT-TNCA compared with CT, suggesting a high accurate accumulation of the TNCA in tumor nodules. Mice diagnosed with early stage NSCLC exhibited a higher eradication rate of NSCLC after treatment with cisplatin compared with mice with advanced stage NSCLC. These data indicate that the sensitivity and accuracy of CT imaging for the diagnosis of early stage NSCLC was improved through combination with the liposome-encapsulated TNCA.
Collapse
Affiliation(s)
- Ninglu Yuan
- Department of Radiology, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaohe Zhang
- Department of Cardiothoracic Surgery, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Yonghui Cao
- Department of Radiology, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaojie Jiang
- Department of Computerized Tomography, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Si Zhao
- Department of Radiology, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Yingying Feng
- Department of Radiology, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Yimeng Fan
- Department of Computerized Tomography, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Zhitao Lu
- Department of Radiology, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Hongmei Gao
- Department of Radiology, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
37
|
Moore JK, Chen J, Pan H, Gaut JP, Jain S, Wickline SA. Quantification of vascular damage in acute kidney injury with fluorine magnetic resonance imaging and spectroscopy. Magn Reson Med 2017; 79:3144-3153. [PMID: 29148253 DOI: 10.1002/mrm.26985] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/02/2017] [Accepted: 10/03/2017] [Indexed: 12/28/2022]
Abstract
PURPOSE To design a fluorine MRI/MR spectroscopy approach to quantify renal vascular damage after ischemia-reperfusion injury, and the therapeutic response to antithrombin nanoparticles (NPs) to protect kidney function. METHODS A total of 53 rats underwent 45 min of bilateral renal artery occlusion and were treated at reperfusion with either plain perfluorocarbon NPs or NPs functionalized with a direct thrombin inhibitor (PPACK:phenyalanine-proline-arginine-chloromethylketone). Three hours after reperfusion, kidneys underwent ex vivo fluorine MRI/MR spectroscopy at 4.7 T to quantify the extent and volume of trapped NPs, as an index of vascular damage and ischemia-reperfusion injury. Microscopic evaluation of structural damage and NP trapping in non-reperfused renal segments was performed. Serum creatinine was quantified serially over 7 days. RESULTS The damaged renal cortico-medullary junction trapped a significant volume of NPs (P = 0.04), which correlated linearly (r = 0.64) with the severity of kidney injury 3 h after reperfusion. Despite global large vessel reperfusion, non-reperfusion in medullary peritubular capillaries was confirmed by MRI and microscopy, indicative of continuing hypoxia due to vascular compromise. Treatment of animals with PPACK NPs after acute kidney injury did not accelerate kidney functional recovery. CONCLUSIONS Quantification of ischemia-reperfusion injury after acute kidney injury with fluorine MRI/MR spectroscopy of perfluorocarbon NPs objectively depicts the extent and severity of vascular injury and its linear relationship to renal dysfunction. The lack of kidney function improvement after early posttreatment thrombin inhibition confirms the rapid onset of ischemia-reperfusion injury as a consequence of vascular damage and non-reperfusion. The prolongation of medullary ischemia renders cortico-medullary tubular structures susceptible to continued necrosis despite restoration of large vessel flow, which suggests limitations to acute interventions after acute kidney injury, designed to interdict renal tubular damage. Magn Reson Med 79:3144-3153, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Jeremy K Moore
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Junjie Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hua Pan
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joseph P Gaut
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sanjay Jain
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Samuel A Wickline
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA.,Department of Cardiovascular Science, University of South Florida, Tampa, Florida, USA.,Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
38
|
Craciun I, Gunkel-Grabole G, Belluati A, Palivan CG, Meier W. Expanding the potential of MRI contrast agents through multifunctional polymeric nanocarriers. Nanomedicine (Lond) 2017; 12:811-817. [PMID: 28322116 DOI: 10.2217/nnm-2016-0413] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
MRI is a sought-after, noninvasive tool in medical diagnostics, yet the direct application of contrast agents to tissue suffers from several drawbacks. Hosting the contrast agents in polymeric nanocarriers can solve many of these issues while creating additional benefit through exploitation of the intrinsic characteristics of the polymeric carriers. In this report, the versatility is highlighted with recent examples of dendritic and hyperbranched polymers, polymer nanoparticles and micelles, and polymersomes as multifunctional bioresponsive nanocarriers for MRI contrast agents.
Collapse
Affiliation(s)
- Ioana Craciun
- Department of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Gesine Gunkel-Grabole
- Department of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Andrea Belluati
- Department of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Wolfgang Meier
- Department of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| |
Collapse
|
39
|
Belosludov RV, Rhoda HM, Zhdanov RK, Belosludov VR, Kawazoe Y, Nemykin VN. Conceptual design of tetraazaporphyrin- and subtetraazaporphyrin-based functional nanocarbon materials: electronic structures, topologies, optical properties, and methane storage capacities. Phys Chem Chem Phys 2016; 18:13503-18. [PMID: 27128697 DOI: 10.1039/c5cp07552a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A large variety of conceptual three- and fourfold tetraazaporphyrin- and subtetraazaporphyrin-based functional 3D nanocage and nanobarrel structures have been proposed on the basis of in silico design. The designed structures differ in their sizes, topology, porosity, and conjugation properties. The stability of nanocages of Oh symmetry and nanobarrels of D4h symmetry was revealed on the basis of DFT and MD calculations, whereas their optical properties were assessed using a TDDFT approach and a long-range corrected LC-wPBE exchange-correlation functional. It was shown that the electronic structures and vertical excitation energies of the functional nanocage and nanobarrel structures could be easily tuned via their size, topology, and the presence of bridging sp(3) carbon atoms. TDDFT calculations suggest significantly lower excitation energies in fully conjugated nanocages and nanobarrels compared with systems with bridging sp(3) carbon fragments. Based on DFT and TDDFT calculations, the optical properties of the new materials can rival those of known quantum dots and are superior to those of monomeric phthalocyanines and their analogues. The methane gas adsorption properties of the new nanostructures and nanotubes generated by conversion from nanobarrels were studied using an MD simulation approach. The ability to store large quantities of methane (106-216 cm(3) (STP) cm(-3)) was observed in all cases with several compounds being close to or exceeding the DOE target of 180 cm(3) (STP) cm(-3) for material-based methane storage at a pressure of 3.5 MPa and room temperature.
Collapse
Affiliation(s)
- Rodion V Belosludov
- Institute for Materials Research, Tohoku University, Sendai, 980-85577, Japan.
| | - Hannah M Rhoda
- Department of Chemistry and Biochemistry, University of Minnesota Duluth, Duluth, Minnesota 55812, USA.
| | - Ravil K Zhdanov
- Nikolaev Institute of Inorganic Chemistry, SB RAS, Lavrentiev 3, Novosibirsk 630090, Russia
| | - Vladimir R Belosludov
- Nikolaev Institute of Inorganic Chemistry, SB RAS, Lavrentiev 3, Novosibirsk 630090, Russia
| | - Yoshiyuki Kawazoe
- New Industry Creation Hatchery Center, Tohoku University, 6-6-4 Aoba, Aramaki, Sendai 980-8579, Japan
| | - Victor N Nemykin
- Department of Chemistry and Biochemistry, University of Minnesota Duluth, Duluth, Minnesota 55812, USA.
| |
Collapse
|
40
|
Nie S, Zhang J, Martinez-Zaguilan R, Sennoune S, Hossen MN, Lichtenstein AH, Cao J, Meyerrose GE, Paone R, Soontrapa S, Fan Z, Wang S. Detection of atherosclerotic lesions and intimal macrophages using CD36-targeted nanovesicles. J Control Release 2015; 220:61-70. [PMID: 26450668 DOI: 10.1016/j.jconrel.2015.10.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 09/24/2015] [Accepted: 10/01/2015] [Indexed: 10/23/2022]
Abstract
Current approaches to the diagnosis and therapy of atherosclerosis cannot target lesion-determinant cells in the artery wall. Intimal macrophage infiltration promotes atherosclerotic lesion development by facilitating the accumulation of oxidized low-density lipoproteins (oxLDL) and increasing inflammatory responses. The presence of these cells is positively associated with lesion progression, severity and destabilization. Hence, they are an important diagnostic and therapeutic target. The objective of this study was to noninvasively assess the distribution and accumulation of intimal macrophages using CD36-targeted nanovesicles. Soy phosphatidylcholine was used to synthesize liposome-like nanovesicles. 1-(Palmitoyl)-2-(5-keto-6-octene-dioyl) phosphatidylcholine was incorporated on their surface to target the CD36 receptor. All in vitro data demonstrate that these targeted nanovesicles had a high binding affinity for the oxLDL binding site of the CD36 receptor and participated in CD36-mediated recognition and uptake of nanovesicles by macrophages. Intravenous administration into LDL receptor null mice of targeted compared to non-targeted nanovesicles resulted in higher uptake in aortic lesions. The nanovesicles co-localized with macrophages and their CD36 receptors in aortic lesions. This molecular target approach may facilitate the in vivo noninvasive imaging of atherosclerotic lesions in terms of intimal macrophage accumulation and distribution and disclose lesion features related to inflammation and possibly vulnerability thereby facilitate early lesion detection and targeted delivery of therapeutic compounds to intimal macrophages.
Collapse
Affiliation(s)
- Shufang Nie
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Jia Zhang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Raul Martinez-Zaguilan
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX 79416, USA
| | - Souad Sennoune
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX 79416, USA
| | - Md Nazir Hossen
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Alice H Lichtenstein
- Cardiovascular Nutrition Laboratory, Jean Mayer Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Jun Cao
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, Jiangxi, China
| | - Gary E Meyerrose
- Division of Cardiology, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Ralph Paone
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Suthipong Soontrapa
- Division of Cardiology, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Zhaoyang Fan
- Department of Electrical and Computer Engineering and Nano Tech Center, Texas Tech University, Lubbock, TX 79409, USA
| | - Shu Wang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA.
| |
Collapse
|