1
|
Yang X, Hu J, Gao Q, Deng Y, Liu Y, He X, Li C, Yu X, Wan Y, Pi C, Wei Y, Li C. Advances in nano-delivery systems based on diagnosis and theranostics strategy for atherosclerosis. J Drug Target 2025; 33:492-507. [PMID: 39601425 DOI: 10.1080/1061186x.2024.2433560] [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: 08/05/2024] [Revised: 10/21/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
Atherosclerosis (AS) is a chronic systemic inflammatory disease, where early diagnosis and theranostics strategy for AS are crucial for improving outcomes. However, conventional diagnostic techniques are limited in identifying early AS lesions, failing to stop the progression of AS in time. Nano-delivery systems have shown significant potential in AS diagnosis and treatment, offering distinct advantages in plaque identification and enhancing drugs concentration at lesion sites, thereby advancing new-generation theranostics strategy. This review discusses the application of nano-delivery systems based on imaging technology in AS diagnosis, and we further explore recent advancements in combining different imaging technologies with emerging theranostics strategy. In addition, we also discuss the challenges faced by nano-delivery systems for AS diagnosis and theranostics in clinical translation, such as nanoparticle targeting efficiency, cytotoxicity and long-term accumulation, immune clearance and inaccurate disease modelling. Finally, we also provide prospects on nano-delivery systems based on diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jian Hu
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Quanle Gao
- Department of Neurology, Geriatrics, Hejiang County People's Hospital, Luzhou, Sichuan, China
| | - Yiping Deng
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yilin Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xinghui He
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chuang Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chao Pi
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yumeng Wei
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
2
|
Gu X, Du L, Lin R, Ding Z, Guo Z, Wei J, Li Y. How Advanced Is Nanomedicine for Atherosclerosis? Int J Nanomedicine 2025; 20:3445-3470. [PMID: 40125442 PMCID: PMC11928726 DOI: 10.2147/ijn.s508757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in nanotechnology have opened new avenues for precision therapy, personalized medicine, and multifunctional theranostics in atherosclerosis (AS). This review provides a comprehensive overview of the role of nanoparticles (NPs) in precision medicine for AS, discussing their applications, challenges, and future prospects. The review first analyzes the current treatment landscape of AS and outlines potential biological targets for therapy. Various nanocarriers, including organic, inorganic, and hybrid systems, are evaluated for their therapeutic potential, with a focus on targeted drug delivery, anti-inflammatory therapy, vascular repair, plaque stabilization, and lipid clearance. Additionally, the review explores NP preparation methods, emphasizing strategies to enhance drug loading, stability, and controlled release. Finally, the translational challenges of NP-based therapies, including biocompatibility, large-scale production, regulatory hurdles, and clinical implementation, are critically analyzed. Future directions highlight the importance of interdisciplinary collaboration and technological innovation in advancing nanoparticle-based precision medicine for AS.
Collapse
Affiliation(s)
- Xiang Gu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Lixin Du
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ruifang Lin
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zehui Ding
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhihua Guo
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jiaming Wei
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ya Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| |
Collapse
|
3
|
Lu Y, Wang Y, Li Y, Li Y, Jiang YW, Li J. Golden Tandem of Photothermal Ablation and Simultaneous Anti-Inflammation in One Nanoparticle for Activated Macrophage-Targeted Atherosclerosis Treatment. Int J Nanomedicine 2025; 20:1731-1746. [PMID: 39936024 PMCID: PMC11812566 DOI: 10.2147/ijn.s503774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
Introduction Photothermal therapy (PTT) is attracting increasing attention in treating atherosclerotic plaques. However, PTT can induce inflammatory responses, in turn stimulating the regeneration of atherosclerosis and hindering subsequent therapy. Methods In this paper, a multifunctional nanoparticle (Au NR@SiO2/RSNO/DS, GSNPD) for the synergistic treatment of atherosclerosis through PTT and anti-inflammation effects was developed. The preparation and characterization of GSNPD, their cellular toxicity, photothermal conversion and targeted ablation efficiency, anti-inflammation and ROS scavenging effect, as well as the inhibition of foam cell formation were studied in vitro. Results The experimental results showed that the fabricated GSNPD NPs displayed positive effects on anti-atherosclerosis by pro-inflammatory macrophages ablation, NO production and ROS scavenging. Discussion GSNPD NPs were designed to effectively and accurately ablate pro-inflammatory macrophages by recognizing and targeting to SR-A overexpressed on the activated macrophages of arterial plaques via PTT, and simultaneous inhibit the PTT-induced inflammation through the laser-activated NO release in situ. This match of therapeutic agents and inhibitors not only achieves good therapeutic effects but also minimizes side effects as much as possible, which may provide an effective way for PTT-based treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yuqing Lu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, People’s Republic of China
| | - Yan Wang
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, People’s Republic of China
| | - Yize Li
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, People’s Republic of China
| | - Yunan Li
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, People’s Republic of China
| | - Yao-Wen Jiang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, People’s Republic of China
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, People’s Republic of China
| | - Jingjing Li
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, People’s Republic of China
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, People’s Republic of China
| |
Collapse
|
4
|
Wang X, Mu D, Liang J, Xin R, Zhang Y, Liu R, Yao M, Zhang B. Emerging nanoprobes for the features visualization of vulnerable atherosclerotic plaques. SMART MEDICINE 2024; 3:e20240033. [PMID: 39776593 PMCID: PMC11669784 DOI: 10.1002/smmd.20240033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/28/2024] [Indexed: 01/11/2025]
Abstract
Atherosclerosis (AS) is a major cause of cardiovascular disease. In particular, the unpredictable rupture of vulnerable atherosclerotic plaques (VASPs) can cause serious cardiovascular events such as myocardial infarction, stroke, and even sudden death. Therefore, early evaluation of the vulnerability of atherosclerotic plaques is of great importance. However, clinical imaging techniques are only marginally useful in the presence of severe anatomical structural changes, making it difficult to evaluate plaque vulnerability at an early stage. With the development of molecular imaging and nanotechnology, specific nanoprobes constructed for the pathological features of VASPs have attracted much attention for their ability to visualize VASPs early and noninvasively at the cellular and molecular levels. Here, we outline the pathological features of VASPs, analyze the superiority and limitations of current clinical imaging techniques, introduce the rational design principles of nanoprobes, and systematically summarize the application of nanoprobes to visualize the features of VASPs at the cellular and molecular levels. In addition, we discussed the prospects and urgent challenges in this field, and we believe it will provide new ideas for the early and accurate diagnosis of cardiovascular diseases.
Collapse
Affiliation(s)
- Xin Wang
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Dan Mu
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Jing Liang
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Ruijing Xin
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Yukun Zhang
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Renyuan Liu
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Mei Yao
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Bing Zhang
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
- Medical Imaging CenterAffiliated Drum Tower HospitalMedical School of Nanjing UniversityNanjingChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjingChina
- Department of RadiologyDrum Tower HospitalClinical College of Nanjing Medical UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular MedicineNanjingChina
- Institute of Brain ScienceNanjing UniversityNanjingChina
| |
Collapse
|
5
|
Luo Y, He X, Du Q, Xu L, Xu J, Wang J, Zhang W, Zhong Y, Guo D, Liu Y, Chen X. Metal-based smart nanosystems in cancer immunotherapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230134. [PMID: 39713201 PMCID: PMC11655314 DOI: 10.1002/exp.20230134] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/12/2024] [Indexed: 12/24/2024]
Abstract
Metals are an emerging topic in cancer immunotherapy that have shown great potential in modulating cancer immunity cycle and promoting antitumor immunity by activating the intrinsic immunostimulatory mechanisms which have been identified in recent years. The main challenge of metal-assisted immunotherapy lies in the fact that the free metals as ion forms are easily cleared during circulation, and even cause systemic metal toxicity due to the off-target effects. With the rapid development of nanomedicine, metal-based smart nanosystems (MSNs) with unique controllable structure become one of the most promising delivery carriers to solve the issue, owing to their various endogenous/external stimuli-responsiveness to release free metal ions for metalloimmunotherapy. In this review, the state-of-the-art research progress in metal-related immunotherapy is comprehensively summarized. First, the mainstream mechanisms of MSNs-assisted immunotherapy will be delineated. The immunological effects of certain metals and categorization of MSNs with different characters and compositions are then provided, followed by the representative exemplar applications of MSNs in cancer treatment, and synergistic combination immunotherapy. Finally, we conclude this review with a summary of the remaining challenges associated with MSNs and provide the authors' perspective on their further advances.
Collapse
Affiliation(s)
- Ying Luo
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Xiaojing He
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
- Clinical Imaging Research CentreCentre for Translational MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Qianying Du
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Lian Xu
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Jie Xu
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Junrui Wang
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Wenli Zhang
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Yixin Zhong
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Dajing Guo
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Yun Liu
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Clinical Imaging Research CentreCentre for Translational MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research ProgramNUS Center for NanomedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of SurgeryChemical and Biomolecular Engineeringand Biomedical EngineeringYong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingaporeSingapore
- Institute of Molecular and Cell BiologyAgency for Science, Technology, and Research (A*STAR)SingaporeSingapore
| |
Collapse
|
6
|
Li Y, Li M, Zheng J, Ma Z, Yu T, Zhu Y, Li P, Nie F. Ultrasound-Responsive Nanocarriers Delivering siRNA and Fe 3O 4 Nanoparticles Reprogram Macrophages and Inhibit M2 Polarization for Enhanced NSCLC Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56634-56652. [PMID: 39378273 DOI: 10.1021/acsami.4c10036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Lung cancer has emerged as the second most common type of malignant tumor worldwide, and it has the highest mortality rate. The overall 5-year survival rate stands at less than 20%, which is primarily related to the limited therapeutic options and the complexity of the tumor immune microenvironment. In the tumor microenvironment, M1 macrophages are known for their tumor-killing capabilities. Although they are less numerous, they play an important role in tumor immunity. Therefore, increasing M1 macrophages' presence is considered a strategy to enhance targeted phagocytosis and antitumor efficacy in nonsmall cell lung cancer (NSCLC). This study introduces the development of folic acid (FA)-conjugated liposomal nanobubbles for precise delivery of PFH, STAT3 siRNA, and Fe3O4 to the tumor microenvironment. These encapsulated PFH liposomal nanobubbles exhibit significant visualization potential and underwent phase transition when exposed to low-intensity focused ultrasound (LIFU). The release of Fe3O4 activates the IRF5 signaling pathway, converting M2-like macrophages to M1. In addition, STAT3 siRNA effectively interrupts the JAK-STAT3 pathway, inhibiting the polarization of M2-like macrophages in tumor-associated macrophages (TAMs). This dual-action therapy facilitates T-cell activation and proliferation, thereby enhancing the immune response against NSCLC.
Collapse
Affiliation(s)
- Yuanyuan Li
- Ultrasound Medical Center, Gansu Province Clinical Research Center for Ultrasonography, Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Ming Li
- Ultrasound Medical Center, Gansu Province Clinical Research Center for Ultrasonography, Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Jun Zheng
- The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Zhen Ma
- Peking University Third Hospital, Beijing 100191, China
| | - Tingting Yu
- Ultrasound Medical Center, Gansu Province Clinical Research Center for Ultrasonography, Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Yangyang Zhu
- Ultrasound Medical Center, Gansu Province Clinical Research Center for Ultrasonography, Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Pan Li
- The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Fang Nie
- Ultrasound Medical Center, Gansu Province Clinical Research Center for Ultrasonography, Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou University Second Hospital, Lanzhou 730000, China
| |
Collapse
|
7
|
Yu C, Zhong L, Zhou Y, Jiang N, Chen J, Cao S. Identification of the Vulnerability of Atherosclerotic Plaques by a Photoacoustic/Ultrasonic Dual-Modal cRGD Nanomolecular Probe. Int J Nanomedicine 2024; 19:9395-9410. [PMID: 39282572 PMCID: PMC11402356 DOI: 10.2147/ijn.s476236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
Objective To explore the feasibility of using cRGD-GNR-PFP-NPs to assess plaque vulnerability in an atherosclerotic plaque mouse model by dual-modal photoacoustic/ultrasonic imaging. Methods A nanomolecular probe containing gold nanorods (GNRs) and perfluoropentane (PFP) coated with the cyclic Arg-Gly-Asp (cRGD) peptide were prepared by double emulsion solvent evaporation and carbodiimide methods. The morphology, particle size, potential, cRGD conjugation and absorption features of the nanomolecular probe were characterized, along with its in vitro phase transformation and photoacoustic/ultrasonic dual-modal imaging properties. In vivo fluorescence imaging was used to determine the distribution of cRGD-GNR-PFP-NPs in vivo in apolipoprotein E-deficient (ApoE-/-) atherosclerotic plaque model mice, the optimal imaging time was determined, and photoacoustic/ultrasonic dual-modal molecular imaging of integrin αvβ3 expressed in atherosclerotic plaques was performed. Pathological assessments verified the imaging results in terms of integrin αvβ3 expression and plaque vulnerability. Results cRGD-GNR-PFP-NPs were spherical with an appropriate particle size (average of approximately 258.03±6.75 nm), a uniform dispersion, and a potential of approximately -9.36±0.53 mV. The probe had a characteristic absorption peak at 780~790 nm, and the surface conjugation of the cRGD peptide reached 92.79%. cRGD-GNR-PFP-NPs were very stable in the non-excited state but very easily underwent phase transformation under low-intensity focused ultrasound (LIFU) and had excellent photoacoustic/ultrasonic dual-modal imaging capability. Mice fed a high-fat diet for 20 weeks had obvious hyperlipidemia with larger, more vulnerable plaques. These plaques could be specifically targeted by cRGD-GNR-PFP-NPs as determined by in vivo fluorescence imaging, and the enrichment of nanomolecular probe increased with the increasing of plaque vulnerability; the photoacoustic/ultrasound signals of the plaques in the high-fat group were stronger. The pathological assessments were in good agreement with the cRGD-GNR-PFP-NPs plaque accumulation, integrin αvβ3 expression and plaque vulnerability results. Conclusion A phase variant photoacoustic/ultrasonic dual-modal cRGD nanomolecular probe was successfully prepared and can be used to identify plaque vulnerability safely and effectively.
Collapse
Affiliation(s)
- Caigui Yu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Lu Zhong
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Yanxiang Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Nan Jiang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Jinling Chen
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Sheng Cao
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| |
Collapse
|
8
|
Su Y, Huang L, Zhang D, Zeng Z, Hong S, Lin X. Recent Advancements in Ultrasound Contrast Agents Based on Nanomaterials for Imaging. ACS Biomater Sci Eng 2024; 10:5496-5512. [PMID: 39246058 DOI: 10.1021/acsbiomaterials.4c00890] [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] [Indexed: 09/10/2024]
Abstract
Ultrasound (US) is a type of mechanical wave that is capable of transmitting energy through biological tissues. By utilization of various frequencies and intensities, it can elicit specific biological effects. US imaging (USI) technology has been continuously developed with the advantages of safety and the absence of radiation. The advancement of nanotechnology has led to the utilization of various nanomaterials composed of both organic and inorganic compounds as ultrasound contrast agents (UCAs). These UCAs enhance USI, enabling real-time monitoring, diagnosis, and treatment of diseases, thereby facilitating the widespread adoption of UCAs in precision medicine. In this review, we introduce various UCAs based on nanomaterials for USI. Their principles can be roughly divided into the following categories: carrying and transporting gases, endogenous gas production, and the structural characteristics of the nanomaterial itself. Furthermore, the synergistic benefits of US in conjunction with various imaging modalities and their combined application in disease monitoring and diagnosis are introduced. In addition, the challenges and prospects for the development of UCAs are also discussed.
Collapse
Affiliation(s)
- Yina Su
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Linjie Huang
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Dongdong Zhang
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Zheng Zeng
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Shanni Hong
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Xiahui Lin
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| |
Collapse
|
9
|
Shen L, Bi Y, Yu J, Zhong Y, Chen W, Zhao Z, Ding J, Shu G, Chen M, Lu C, Ji J. The biological applications of near-infrared optical nanomaterials in atherosclerosis. J Nanobiotechnology 2024; 22:478. [PMID: 39135099 PMCID: PMC11320980 DOI: 10.1186/s12951-024-02703-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 07/05/2024] [Indexed: 08/15/2024] Open
Abstract
PURPOSE OF REVIEW Atherosclerosis, a highly pathogenic and lethal disease, is difficult to locate accurately via conventional imaging because of its scattered and deep lesions. However, second near-infrared (NIR-II) nanomaterials show great application potential in the tracing of atherosclerotic plaques due to their excellent penetration and angiographic capabilities. RECENT FINDINGS With the development of nanotechnology, among many nanomaterials available for the visual diagnosis and treatment of cardiovascular diseases, optical nanomaterials provide strong support for various biomedical applications because of their advantages, such as noninvasive, nondestructive and molecular component imaging. Among optical nanomaterials of different wavelengths, NIR-II-range (900 ~ 1700 nm) nanomaterials have been gradually applied in the visual diagnosis and treatment of atherosclerosis and other vascular diseases because of their deep biological tissue penetration and limited background interference. This review explored in detail the prospects and challenges of the biological imaging and clinical application of NIR-II nanomaterials in treating atherosclerosis.
Collapse
Affiliation(s)
- Lin Shen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Yanran Bi
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Junchao Yu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Yi Zhong
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Weiqian Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Jiayi Ding
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Gaofeng Shu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Chenying Lu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China.
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China.
| |
Collapse
|
10
|
Huang H, Zheng Y, Chang M, Song J, Xia L, Wu C, Jia W, Ren H, Feng W, Chen Y. Ultrasound-Based Micro-/Nanosystems for Biomedical Applications. Chem Rev 2024; 124:8307-8472. [PMID: 38924776 DOI: 10.1021/acs.chemrev.4c00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Due to the intrinsic non-invasive nature, cost-effectiveness, high safety, and real-time capabilities, besides diagnostic imaging, ultrasound as a typical mechanical wave has been extensively developed as a physical tool for versatile biomedical applications. Especially, the prosperity of nanotechnology and nanomedicine invigorates the landscape of ultrasound-based medicine. The unprecedented surge in research enthusiasm and dedicated efforts have led to a mass of multifunctional micro-/nanosystems being applied in ultrasound biomedicine, facilitating precise diagnosis, effective treatment, and personalized theranostics. The effective deployment of versatile ultrasound-based micro-/nanosystems in biomedical applications is rooted in a profound understanding of the relationship among composition, structure, property, bioactivity, application, and performance. In this comprehensive review, we elaborate on the general principles regarding the design, synthesis, functionalization, and optimization of ultrasound-based micro-/nanosystems for abundant biomedical applications. In particular, recent advancements in ultrasound-based micro-/nanosystems for diagnostic imaging are meticulously summarized. Furthermore, we systematically elucidate state-of-the-art studies concerning recent progress in ultrasound-based micro-/nanosystems for therapeutic applications targeting various pathological abnormalities including cancer, bacterial infection, brain diseases, cardiovascular diseases, and metabolic diseases. Finally, we conclude and provide an outlook on this research field with an in-depth discussion of the challenges faced and future developments for further extensive clinical translation and application.
Collapse
Affiliation(s)
- Hui Huang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Jun Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wei Feng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
11
|
Nankivell V, Vidanapathirana AK, Hoogendoorn A, Tan JTM, Verjans J, Psaltis PJ, Hutchinson MR, Gibson BC, Lu Y, Goldys E, Zheng G, Bursill CA. Targeting macrophages with multifunctional nanoparticles to detect and prevent atherosclerotic cardiovascular disease. Cardiovasc Res 2024; 120:819-838. [PMID: 38696700 PMCID: PMC11218693 DOI: 10.1093/cvr/cvae099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/29/2024] [Accepted: 04/02/2024] [Indexed: 05/04/2024] Open
Abstract
Despite the emergence of novel diagnostic, pharmacological, interventional, and prevention strategies, atherosclerotic cardiovascular disease remains a significant cause of morbidity and mortality. Nanoparticle (NP)-based platforms encompass diverse imaging, delivery, and pharmacological properties that provide novel opportunities for refining diagnostic and therapeutic interventions for atherosclerosis at the cellular and molecular levels. Macrophages play a critical role in atherosclerosis and therefore represent an important disease-related diagnostic and therapeutic target, especially given their inherent ability for passive and active NP uptake. In this review, we discuss an array of inorganic, carbon-based, and lipid-based NPs that provide magnetic, radiographic, and fluorescent imaging capabilities for a range of highly promising research and clinical applications in atherosclerosis. We discuss the design of NPs that target a range of macrophage-related functions such as lipoprotein oxidation, cholesterol efflux, vascular inflammation, and defective efferocytosis. We also provide examples of NP systems that were developed for other pathologies such as cancer and highlight their potential for repurposing in cardiovascular disease. Finally, we discuss the current state of play and the future of theranostic NPs. Whilst this is not without its challenges, the array of multifunctional capabilities that are possible in NP design ensures they will be part of the next frontier of exciting new therapies that simultaneously improve the accuracy of plaque diagnosis and more effectively reduce atherosclerosis with limited side effects.
Collapse
Affiliation(s)
- Victoria Nankivell
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Achini K Vidanapathirana
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Ayla Hoogendoorn
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Johan Verjans
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Peter J Psaltis
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Mark R Hutchinson
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Brant C Gibson
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- School of Science, RMIT University, Melbourne, Victoria, Australia
| | - Yiqing Lu
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- School of Engineering, Macquarie University, Sydney, NSW, Australia
| | - Ewa Goldys
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Graduate School of Biomedical Engineering, University of New South Wales, High Street, NSW, 2052, Australia
| | - Gang Zheng
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, M5G 1L7, Canada
| | - Christina A Bursill
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| |
Collapse
|
12
|
Lv F, Fang H, Huang L, Wang Q, Cao S, Zhao W, Zhou Z, Zhou W, Wang X. Curcumin Equipped Nanozyme-Like Metal-Organic Framework Platform for the Targeted Atherosclerosis Treatment with Lipid Regulation and Enhanced Magnetic Resonance Imaging Capability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309062. [PMID: 38696653 PMCID: PMC11234396 DOI: 10.1002/advs.202309062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/21/2024] [Indexed: 05/04/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) has become the leading cause of death worldwide, and early diagnosis and treatment of atherosclerosis (AS) are crucial for reducing the occurrence of acute cardiovascular events. However, early diagnosis of AS is challenging, and oral anti-AS drugs suffer from limitations like imprecise targeting and low bioavailability. To overcome the aforementioned shortcomings, Cur/MOF@DS is developed, a nanoplatform integrating diagnosis and treatment by loading curcumin (Cur) into metal-organic frameworks with nanozymes and magnetic resonance imaging (MRI) properties. In addition, the surface-modification of dextran sulfate (DS) enables PCN-222(Mn) effectively target scavenger receptor class A in macrophages or foam cells within the plaque region. This nanoplatform employs mechanisms that effectively scavenge excessive reactive oxygen species in the plaque microenvironment, promote macrophage autophagy and regulate macrophage polarization to realize lipid regulation. In vivo and in vitro experiments confirm that this nanoplatform has outstanding MRI performance and anti-AS effects, which may provide a new option for early diagnosis and treatment of AS.
Collapse
Affiliation(s)
- Fanzhen Lv
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Huaqiang Fang
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Li Huang
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Qingqing Wang
- School of PharmacyNanchang UniversityNanchangJiangxi330006China
| | - Shuangyuan Cao
- The National Engineering Research Center for Bioengineering Drugs and the TechnologiesInstitute of Translational MedicineNanchang UniversityNanchangJiangxi330006China
| | - Wenpeng Zhao
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Zhibin Zhou
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Weimin Zhou
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Xiaolei Wang
- School of PharmacyNanchang UniversityNanchangJiangxi330006China
- The National Engineering Research Center for Bioengineering Drugs and the TechnologiesInstitute of Translational MedicineNanchang UniversityNanchangJiangxi330006China
| |
Collapse
|
13
|
Deng X, Wang J, Yu S, Tan S, Yu T, Xu Q, Chen N, Zhang S, Zhang M, Hu K, Xiao Z. Advances in the treatment of atherosclerosis with ligand-modified nanocarriers. EXPLORATION (BEIJING, CHINA) 2024; 4:20230090. [PMID: 38939861 PMCID: PMC11189587 DOI: 10.1002/exp.20230090] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/08/2023] [Indexed: 06/29/2024]
Abstract
Atherosclerosis, a chronic disease associated with metabolism, poses a significant risk to human well-being. Currently, existing treatments for atherosclerosis lack sufficient efficiency, while the utilization of surface-modified nanoparticles holds the potential to deliver highly effective therapeutic outcomes. These nanoparticles can target and bind to specific receptors that are abnormally over-expressed in atherosclerotic conditions. This paper reviews recent research (2018-present) advances in various ligand-modified nanoparticle systems targeting atherosclerosis by specifically targeting signature molecules in the hope of precise treatment at the molecular level and concludes with a discussion of the challenges and prospects in this field. The intention of this review is to inspire novel concepts for the design and advancement of targeted nanomedicines tailored specifically for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xiujiao Deng
- Department of PharmacyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
- Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouChina
| | - Jinghao Wang
- Department of PharmacyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
| | - Shanshan Yu
- Department of PharmacyZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Suiyi Tan
- Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouChina
| | - Tingting Yu
- Department of PharmacyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
| | - Qiaxin Xu
- Department of PharmacyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
| | - Nenghua Chen
- Department of PharmacyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
| | - Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ming‐Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical, ScienceNational Institutes for Quantum Science and TechnologyChibaJapan
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical, ScienceNational Institutes for Quantum Science and TechnologyChibaJapan
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical TranslationJinan UniversityGuangzhouChina
| |
Collapse
|
14
|
Wang S, He H, Mao Y, Zhang Y, Gu N. Advances in Atherosclerosis Theranostics Harnessing Iron Oxide-Based Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308298. [PMID: 38368274 PMCID: PMC11077671 DOI: 10.1002/advs.202308298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/06/2024] [Indexed: 02/19/2024]
Abstract
Atherosclerosis, a multifaceted chronic inflammatory disease, has a profound impact on cardiovascular health. However, the critical limitations of atherosclerosis management include the delayed detection of advanced stages, the intricate assessment of plaque stability, and the absence of efficacious therapeutic strategies. Nanotheranostic based on nanotechnology offers a novel paradigm for addressing these challenges by amalgamating advanced imaging capabilities with targeted therapeutic interventions. Meanwhile, iron oxide nanoparticles have emerged as compelling candidates for theranostic applications in atherosclerosis due to their magnetic resonance imaging capability and biosafety. This review delineates the current state and prospects of iron oxide nanoparticle-based nanotheranostics in the realm of atherosclerosis, including pivotal aspects of atherosclerosis development, the pertinent targeting strategies involved in disease pathogenesis, and the diagnostic and therapeutic roles of iron oxide nanoparticles. Furthermore, this review provides a comprehensive overview of theranostic nanomedicine approaches employing iron oxide nanoparticles, encompassing chemical therapy, physical stimulation therapy, and biological therapy. Finally, this review proposes and discusses the challenges and prospects associated with translating these innovative strategies into clinically viable anti-atherosclerosis interventions. In conclusion, this review offers new insights into the future of atherosclerosis theranostic, showcasing the remarkable potential of iron oxide-based nanoparticles as versatile tools in the battle against atherosclerosis.
Collapse
Affiliation(s)
- Shi Wang
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Hongliang He
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Yu Mao
- School of MedicineNanjing UniversityNanjing210093P. R. China
| | - Yu Zhang
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Ning Gu
- School of MedicineNanjing UniversityNanjing210093P. R. China
| |
Collapse
|
15
|
Chen Y, Wang H, Pan J, Guo Y, Hu Y, Huang X, Zhou Y, Deng Q, Zhou Q. Macrophage-targeted ultrasound nanobubbles for highly efficient sonodynamic therapy of atherosclerotic plaques by modulating M1-to-M2 polarization. Atherosclerosis 2024; 389:117423. [PMID: 38159431 DOI: 10.1016/j.atherosclerosis.2023.117423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND AND AIMS Sonodynamic therapy (SDT) is a new approach for the treatment of atherosclerosis (AS), yet the poor targeting ability of sonosensitizers limits its therapeutic efficacy. Herein, we reported a plaque-targeted nanoplatform modified with macrophage type A scavenger receptor (SR-A)-targeted peptide (designated as SR-A-Ce6NB) to augment the efficacy of low-intensity pulsed ultrasound (LIPUS)-mediated SDT of atherosclerotic plaque. METHODS SR-A-Ce6NB was fabricated by thin hydration method and biotin-avidin system, and its physicochemical properties, biocompatibility and plaque-targeting ability were investigated. RAW 264.7 cells were used for in vitro experimental studies. Male 6-week-old apolipoprotein E-deficient mice were fed a high-fat diet for 16 weeks to induce aortic atherosclerotic plaques. Plaque-bearing mice were randomly allocated into five groups (n = 6): control group, Ce6 + LIPUS group, Ce6NB + LIPUS group, SR-A-Ce6NB + LIPUS group and atorvastatin group. After treatment in each group, the aortic artery was harvested for Oil red O, H&E, Masson's trichrome staining, immunohistochemical and immunofluorescent staining. RESULTS SR-A-Ce6NB with high stability and excellent biocompatibility was successfully fabricated. SR-A-Ce6NB could actively target activated macrophages and selectively accumulate in the plaque. SR-A-Ce6NB could be triggered by LIPUS and had a more potent sonodynamic effect than free Ce6 to potentiate SDT. SR-A-Ce6NB-mediated SDT enhanced the anti-atherogenic effect via modulating M1-to-M2 macrophage polarization and had an earlier onset of action on plaque than the statin-mediated effect. No apparent side effect was observed after intravenous SR-A-Ce6NB injection and LIPUS exposure. CONCLUSIONS Macrophage-targeted nanoplatform SR-A-Ce6NB-mediated SDT provides a safe, effective and preferable anti-atherogenic therapy by mediating M1-to-M2 macrophage polarization.
Collapse
Affiliation(s)
- Yueying Chen
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Hao Wang
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Juhong Pan
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Yuxin Guo
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Yugang Hu
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Xin Huang
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Yanxiang Zhou
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Qing Deng
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Qing Zhou
- Department of Ultrasonography, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| |
Collapse
|
16
|
Zou L, Zhang Y, Cheraga N, Abodunrin OD, Qu KY, Qiao L, Ma YQ, Hang Y, Huang NP, Chen LJ. M2 Macrophage Membrane-Camouflaged Fe 3 O 4 -Cy7 Nanoparticles with Reduced Immunogenicity for Targeted NIR/MR Imaging of Atherosclerosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304110. [PMID: 37806756 DOI: 10.1002/smll.202304110] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/27/2023] [Indexed: 10/10/2023]
Abstract
Atherosclerosis (AS) is the primary reason behind cardiovascular diseases, leading to approximately one-third of global deaths. Developing a novel multi-model probe to detect AS is urgently required. Macrophages are the primary cells from which AS genesis occurs. Utilizing natural macrophage membranes coated on the surface of nanoparticles is an efficient delivery method to target plaque sites. Herein, Fe3 O4 -Cy7 nanoparticles (Fe3 O4 -Cy7 NPs), functionalized using an M2 macrophage membrane and a liposome extruder for Near-infrared fluorescence and Magnetic resonance imaging, are synthesized. These macrophage membrane-coated nanoparticles (Fe3 O4 @M2 NPs) enhance the recognition and uptake using active macrophages. Moreover, they inhibit uptake using inactive macrophages and human coronary artery endothelial cells. The macrophage membrane-coated nanoparticles (Fe3 O4 @M0 NPs, Fe3 O4 @M1 NPs, Fe3 O4 @M2 NPs) can target specific sites depending on the macrophage membrane type and are related to C-C chemofactor receptor type 2 protein content. Moreover, Fe3 O4 @M2 NPs demonstrate excellent biosafety in vivo after injection, showing a significantly higher Fe concentration in the blood than Fe3 O4 -Cy7 NPs. Therefore, Fe3 O4 @M2 NPs effectively retain the physicochemical properties of nanoparticles and depict reduced immunological response in blood circulation. These NPs mainly reveal enhanced targeting imaging capability for atherosclerotic plaque lesions.
Collapse
Affiliation(s)
- Lin Zou
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Yao Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 210009, Nanjing, China
| | - Nihad Cheraga
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Oluwatosin David Abodunrin
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Kai-Yun Qu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Li Qiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Yu-Qing Ma
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Yue Hang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Ning-Ping Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Li-Juan Chen
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, 211200, Nanjing, China
| |
Collapse
|
17
|
Shao J, Lang Y, Ding M, Yin X, Cui L. Transcription Factor EB: A Promising Therapeutic Target for Ischemic Stroke. Curr Neuropharmacol 2024; 22:170-190. [PMID: 37491856 PMCID: PMC10788889 DOI: 10.2174/1570159x21666230724095558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 07/27/2023] Open
Abstract
Transcription factor EB (TFEB) is an important endogenous defensive protein that responds to ischemic stimuli. Acute ischemic stroke is a growing concern due to its high morbidity and mortality. Most survivors suffer from disabilities such as numbness or weakness in an arm or leg, facial droop, difficulty speaking or understanding speech, confusion, impaired balance or coordination, or loss of vision. Although TFEB plays a neuroprotective role, its potential effect on ischemic stroke remains unclear. This article describes the basic structure, regulation of transcriptional activity, and biological roles of TFEB relevant to ischemic stroke. Additionally, we explore the effects of TFEB on the various pathological processes underlying ischemic stroke and current therapeutic approaches. The information compiled here may inform clinical and basic studies on TFEB, which may be an effective therapeutic drug target for ischemic stroke.
Collapse
Affiliation(s)
- Jie Shao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yue Lang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Manqiu Ding
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xiang Yin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Li Cui
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
18
|
Liao M, Du J, Chen L, Huang J, Yang R, Bao W, Zeng K, Wang W, Aphan BC, Wu Z, Ma L, Lu Q. Sono-activated materials for enhancing focused ultrasound ablation: Design and application in biomedicine. Acta Biomater 2024; 173:36-50. [PMID: 37939816 DOI: 10.1016/j.actbio.2023.11.004] [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: 06/26/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/10/2023]
Abstract
The ablation effect of focused ultrasound (FUS) has played an increasingly important role in the biomedical field over the past decades, and its non-invasive features have great advantages, especially for clinical diseases where surgical treatment is not available or appropriate. Recently, rapid advances in the adjustable morphology, enzyme-mimetic activity, and biostability of sono-activated materials have significantly promoted the medical application of FUS ablation. However, a systematic review of sono-activated materials based on FUS ablation is not yet available. This progress review focuses on the recent design, fundamental principles, and applications of sono-activated materials in the FUS ablation biomedical field. First, the different ablation mechanisms and the key factors affecting ablation are carefully determined. Then, the design of sono-activated materials with high FUS ablation efficiencies is comprehensively discussed. Subsequently, the representative biological applications are summarized in detail. Finally, the primary challenges and future perspectives are also outlined. We believe this timely review will provide key information and insights for further exploration of focused ultrasound ablation and new inspiration for designing future sono-activated materials. STATEMENT OF SIGNIFICANCE: The ablation effect of focused ultrasound (FUS) has played an increasingly important role in the biomedical field over the past decades. However, there are also some challenges of FUS ablation, such as skin burns, tumour recurrence after thermal ablation, and difficulty in controlling cavitation ablation. The rapid advance in adjustable morphology, enzyme-mimetic activity, and biostability of sono-activated materials has significantly promoted the medical application of FUS ablation. However, the systematic review of sono-activated materials based on FUS ablation is not yet available. This progress review focuses on the recent design, fundamental principles, and applications in the FUS ablation biomedical field of sono-activated materials. We believe this timely review will provide key information and insights for further exploration of FUS ablation.
Collapse
Affiliation(s)
- Min Liao
- Department of Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinpeng Du
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Lin Chen
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Jiayan Huang
- Department of Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Yang
- Department of Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wuyongga Bao
- Department of Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Keyu Zeng
- Department of Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenhui Wang
- Department of Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Benjamín Castañeda Aphan
- Department of Engineering, Medical Imaging Laboratory, Pontificia Universidad Católica del Perú, Lima, Peru
| | - Zhe Wu
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| | - Lang Ma
- Department of Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qiang Lu
- Department of Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
19
|
Cheng J, Huang H, Chen Y, Wu R. Nanomedicine for Diagnosis and Treatment of Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304294. [PMID: 37897322 PMCID: PMC10754137 DOI: 10.1002/advs.202304294] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/11/2023] [Indexed: 10/30/2023]
Abstract
With the changing disease spectrum, atherosclerosis has become increasingly prevalent worldwide and the associated diseases have emerged as the leading cause of death. Due to their fascinating physical, chemical, and biological characteristics, nanomaterials are regarded as a promising tool to tackle enormous challenges in medicine. The emerging discipline of nanomedicine has filled a huge application gap in the atherosclerotic field, ushering a new generation of diagnosis and treatment strategies. Herein, based on the essential pathogenic contributors of atherogenesis, as well as the distinct composition/structural characteristics, synthesis strategies, and surface design of nanoplatforms, the three major application branches (nanodiagnosis, nanotherapy, and nanotheranostic) of nanomedicine in atherosclerosis are elaborated. Then, state-of-art studies containing a sequence of representative and significant achievements are summarized in detail with an emphasis on the intrinsic interaction/relationship between nanomedicines and atherosclerosis. Particularly, attention is paid to the biosafety of nanomedicines, which aims to pave the way for future clinical translation of this burgeoning field. Finally, this comprehensive review is concluded by proposing unresolved key scientific issues and sharing the vision and expectation for the future, fully elucidating the closed loop from atherogenesis to the application paradigm of nanomedicines for advancing the early achievement of clinical applications.
Collapse
Affiliation(s)
- Jingyun Cheng
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Hui Huang
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou Institute of Shanghai UniversityWenzhouZhejiang325088P. R. China
| | - Rong Wu
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| |
Collapse
|
20
|
Lin L, Chen L, Yan J, Chen P, Du J, Zhu J, Yang X, Geng B, Li L, Zeng W. Advances of nanoparticle-mediated diagnostic and theranostic strategies for atherosclerosis. Front Bioeng Biotechnol 2023; 11:1268428. [PMID: 38026849 PMCID: PMC10666776 DOI: 10.3389/fbioe.2023.1268428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Atherosclerotic plaque remains the primary cause of morbidity and mortality worldwide. Accurate assessment of the degree of atherosclerotic plaque is critical for predicting the risk of atherosclerotic plaque and monitoring the results after intervention. Compared with traditional technology, the imaging technologies of nanoparticles have distinct advantages and great development prospects in the identification and characterization of vulnerable atherosclerotic plaque. Here, we systematically summarize the latest advances of targeted nanoparticle approaches in the diagnosis of atherosclerotic plaque, including multimodal imaging, fluorescence imaging, photoacoustic imaging, exosome diagnosis, and highlighted the theranostic progress as a new therapeutic strategy. Finally, we discuss the major challenges that need to be addressed for future development and clinical transformation.
Collapse
Affiliation(s)
- Lin Lin
- School of Medicine, Chongqing University, Chongqing, China
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Lin Chen
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Juan Yan
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Peirong Chen
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Jiahui Du
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Junpeng Zhu
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Xinyu Yang
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Boxin Geng
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Lang Li
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Wen Zeng
- School of Medicine, Chongqing University, Chongqing, China
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| |
Collapse
|
21
|
Guo J, Wang H, Li Y, Zhu S, Hu H, Gu Z. Nanotechnology in coronary heart disease. Acta Biomater 2023; 171:37-67. [PMID: 37714246 DOI: 10.1016/j.actbio.2023.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/17/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
Coronary heart disease (CHD) is one of the major causes of death and disability worldwide, especially in low- and middle-income countries and among older populations. Conventional diagnostic and therapeutic approaches have limitations such as low sensitivity, high cost and side effects. Nanotechnology offers promising alternative strategies for the diagnosis and treatment of CHD by exploiting the unique properties of nanomaterials. In this review, we use bibliometric analysis to identify research hotspots in the application of nanotechnology in CHD and provide a comprehensive overview of the current state of the art. Nanomaterials with enhanced imaging and biosensing capabilities can improve the early detection of CHD through advanced contrast agents and high-resolution imaging techniques. Moreover, nanomaterials can facilitate targeted drug delivery, tissue engineering and modulation of inflammation and oxidative stress, thus addressing multiple aspects of CHD pathophysiology. We discuss the application of nanotechnology in CHD diagnosis (imaging and sensors) and treatment (regulation of macrophages, cardiac repair, anti-oxidative stress), and provide insights into future research directions and clinical translation. This review serves as a valuable resource for researchers and clinicians seeking to harness the potential of nanotechnology in the management of CHD. STATEMENT OF SIGNIFICANCE: Coronary heart disease (CHD) is the one of leading cause of death and disability worldwide. Nanotechnology offers new strategies for diagnosing and treating CHD by exploiting the unique properties of nanomaterials. This review uses bibliometric analysis to uncover research trends in the use of nanotechnology for CHD. We discuss the potential of nanomaterials for early CHD detection through advanced imaging and biosensing, targeted drug delivery, tissue engineering, and modulation of inflammation and oxidative stress. We also offer insights into future research directions and potential clinical applications. This work aims to guide researchers and clinicians in leveraging nanotechnology to improve CHD patient outcomes and quality of life.
Collapse
Affiliation(s)
- Junsong Guo
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hao Wang
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Ying Li
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nano-safety, Institute of High Energy Physics, Beijing 100049, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Houxiang Hu
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China.
| | - Zhanjun Gu
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nano-safety, Institute of High Energy Physics, Beijing 100049, China; Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
22
|
Ju J, Xu D, Mo X, Miao J, Xu L, Ge G, Zhu X, Deng H. Multifunctional polysaccharide nanoprobes for biological imaging. Carbohydr Polym 2023; 317:121048. [PMID: 37364948 DOI: 10.1016/j.carbpol.2023.121048] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/28/2023]
Abstract
Imaging and tracking biological targets or processes play an important role in revealing molecular mechanisms and disease states. Bioimaging via optical, nuclear, or magnetic resonance techniques enables high resolution, high sensitivity, and high depth imaging from the whole animal down to single cells via advanced functional nanoprobes. To overcome the limitations of single-modality imaging, multimodality nanoprobes have been engineered with a variety of imaging modalities and functionalities. Polysaccharides are sugar-containing bioactive polymers with superior biocompatibility, biodegradability, and solubility. The combination of polysaccharides with single or multiple contrast agents facilitates the development of novel nanoprobes with enhanced functions for biological imaging. Nanoprobes constructed with clinically applicable polysaccharides and contrast agents hold great potential for clinical translations. This review briefly introduces the basics of different imaging modalities and polysaccharides, then summarizes the recent progress of polysaccharide-based nanoprobes for biological imaging in various diseases, emphasizing bioimaging with optical, nuclear, and magnetic resonance techniques. The current issues and future directions regarding the development and applications of polysaccharide nanoprobes are further discussed.
Collapse
Affiliation(s)
- Jingxuan Ju
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Danni Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xuan Mo
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiaqian Miao
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Hongping Deng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
23
|
Yang MY, Tu YF, Feng KK, Yin MD, Fang YF, Le JQ, Luo BY, Tan XR, Shao JW. A erythrocyte-platelet hybrid membrane coated biomimetic nanosystem based on ginsenosides and PFH combined with ultrasound for targeted delivery in thrombus therapy. Colloids Surf B Biointerfaces 2023; 229:113468. [PMID: 37515961 DOI: 10.1016/j.colsurfb.2023.113468] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/30/2023] [Accepted: 07/17/2023] [Indexed: 07/31/2023]
Abstract
Thrombus is one of the culprits for global health problems. However, most current antithrombotic drugs are limited by restricted targeting ability and a high risk of systemic bleeding. A hybrid cell membrane-coated biomimetic nanosystem (PM/RM@PLGA@P/R) was constructed in this paper to fulfil the targeted delivery of ginsenoside (Rg1) and perfluorohexane (PFH). Poly lactic-co-glycolic acid (PLGA) is used as carriers to coat Rg1 and PFH. Thanks to the camouflage of erythrocyte membrane (RM) and platelet membrane (PM), the nanosystem in question possesses remarkable features including immune escape and self-targeting. Therefore, a compact nano-core with PLGA@P/R was formed, with a hybrid membrane covering the surface of the core, forming a "core-shell" structure. With its "core-shell" structure, this nanoparticle fancifully combines the advantages of both PFH (the low-intensity focused ultrasound (LIFU)-responsive phase-change thrombolysis) and Rg1(the antioxidant, anti-inflammatory and anticoagulant abilities). Meanwhile, PM/RM@PLGA@P/R nanoparticles exhibits superior in-vitro performance in terms of ROS scavenging, anticoagulant activity and immune escape compared with those without cell membranes (PLGA@P/R). Furthermore, in the animal experiment in which the tail vein thrombosis model was established by injecting k-carrageenan, the combined treatment of LIFU and PM/RM@PLGA@P/R showed a satisfactory antithrombotic efficiency (88.20 %) and a relatively higher biological safety level. This strategy provides new insights into the development of more effective and safer targeted biomimetic nanomedicines for antithrombotic treatments, possessing potential application in synergistic therapy field.
Collapse
Affiliation(s)
- Ming-Yue Yang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Yi-Fan Tu
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Ke-Ke Feng
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Meng-Die Yin
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Yi-Fan Fang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jing-Qing Le
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Bang-Yue Luo
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Xia-Rong Tan
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jing-Wei Shao
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China.
| |
Collapse
|
24
|
Liu H, Ji M, Bi Y, Xiao P, Zhao J, Gou J, Yin T, He H, Ding H, Tang X, Zhang Y. Integration of MyD88 inhibitor into mesoporous cerium oxide nanozymes-based targeted delivery platform for enhancing treatment of ulcerative colitis. J Control Release 2023; 361:493-509. [PMID: 37572964 DOI: 10.1016/j.jconrel.2023.08.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
Excessive reactive oxygen species (ROS) and stressed inflammatory response are major characteristics of ulcerative colitis, which cause disease progression and aggravation. Herein, a novel mesoporous cerium oxide nanozymes (MCN) was designed and then loaded with Myeloid differentiation factor-88 (MyD88) inhibitor for synergistic treatment of colitis by scavenging ROS and regulating inflammation. This innovative MCN with average particle size of 200.7 nm, specific surface area of 119.78 m2/g and mesopores of 4.47 nm not only exhibited excellent SOD-like and CAT-like activities to scavenge ROS but also could act as a carrier to load MyD88 inhibitor, TJ-M2010-5, (abbreviated as TJ-5) into their mesopores, achieving the effect of 'two birds with one stone'. Besides, the modification of dextran sulfate sodium (TJ-5/MCN/DSS) increased the internalization of nanozymes into activated macrophages and enhanced in vitro anti-inflammatory ability. To enhance colon targeting, we coated TJ-5/MCN/DSS with the enteric material Eudragit S100, preventing premature release or absorption of the drug in the gastrointestinal tract after oral administration. The results demonstrated that TJ-5/MCN/DSS/Eudragit not only achieved delayed drug release and improved colon targeting but also exhibited optimal therapeutic efficacy in colitis mice. Mechanistically, the MCN-mediated ROS scavenging and TJ-5-mediated MyD88 blockade synergistically inhibited the NF-κB signaling pathway, thereby reducing the inflammatory response. Importantly, TJ-5/MCN/DSS/Eudragit did not induce systemic toxicity. In conclusion, our work not only presents a novel carrier capable of scavenging ROS but also provides proof of concept for the synergistic treatment of colitis using this carrier in combination with MyD88 inhibitors. This study proposes a safe and efficient strategy for targeting ROS-associated inflammation.
Collapse
Affiliation(s)
- Hongbing Liu
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Muse Ji
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuti Bi
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Peifu Xiao
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiansong Zhao
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingxin Gou
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haibing He
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Huaiwei Ding
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Xing Tang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Zhang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
25
|
Wu C, Mao J, Wang X, Yang R, Wang C, Li C, Zhou X. Advances in treatment strategies based on scavenging reactive oxygen species of nanoparticles for atherosclerosis. J Nanobiotechnology 2023; 21:271. [PMID: 37592345 PMCID: PMC10433664 DOI: 10.1186/s12951-023-02058-z] [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: 05/25/2023] [Accepted: 08/09/2023] [Indexed: 08/19/2023] Open
Abstract
The development of atherosclerosis (AS) is closely linked to changes in the plaque microenvironment, which consists primarily of the cells that form plaque and the associated factors they secrete. The onset of inflammation, lipid deposition, and various pathological changes in cellular metabolism that accompany the plaque microenvironment will promote the development of AS. Numerous studies have shown that oxidative stress is an important condition that promotes AS. The accumulation of reactive oxygen species (ROS) is oxidative stress's most important pathological change. In turn, the effects of ROS on the plaque microenvironment are complex and varied, and these effects are ultimately reflected in the promotion or inhibition of AS. This article reviews the effects of ROS on the microenvironment of atherosclerotic plaques and their impact on disease progression over the past five years and focuses on the progress of treatment strategies based on scavenging ROS of nanoparticles for AS. Finally, we also discuss the prospects and challenges of AS treatment.
Collapse
Affiliation(s)
- Chengxi Wu
- Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, No. 25, Taiping Street, Luzhou, Sichuan, 646000, China
| | - Jingying Mao
- Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, No. 25, Taiping Street, Luzhou, Sichuan, 646000, China
| | - Xueqin Wang
- Department of Thyroid Surgery, people's Hospital of Deyang, Deyang, Sichuan, 618000, China
| | - Ronghao Yang
- Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, No. 25, Taiping Street, Luzhou, Sichuan, 646000, China
| | - Chenglong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, 1-1 Xianglin Road, Luzhou, Sichuan, 646000, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, 1-1 Xianglin Road, Luzhou, Sichuan, 646000, China.
| | - Xiangyu Zhou
- Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, No. 25, Taiping Street, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
26
|
Ghamkhari A, Tafti HA, Rabbani S, Ghorbani M, Ghiass MA, Akbarzadeh F, Abbasi F. Ultrasound-Triggered Microbubbles: Novel Targeted Core-Shell for the Treatment of Myocardial Infarction Disease. ACS OMEGA 2023; 8:11335-11350. [PMID: 37008126 PMCID: PMC10061684 DOI: 10.1021/acsomega.3c00067] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/07/2023] [Indexed: 06/19/2023]
Abstract
Myocardial infarction (MI) is known as a main cardiovascular disease that leads to extensive cell death by destroying vasculature in the affected cardiac muscle. The development of ultrasound-mediated microbubble destruction has inspired extensive interest in myocardial infarction therapeutics, targeted delivery of drugs, and biomedical imaging. In this work, we describe a novel therapeutic ultrasound system for the targeted delivery of biocompatible microstructures containing basic fibroblast growth factor (bFGF) to the MI region. The microspheres were fabricated using poly(lactic-co-glycolic acid)-heparin-polyethylene glycol- cyclic arginine-glycine-aspartate-platelet (PLGA-HP-PEG-cRGD-platelet). The micrometer-sized core-shell particles consisting of a perfluorohexane (PFH)-core and a PLGA-HP-PEG-cRGD-platelet-shell were prepared using microfluidics. These particles responded adequately to ultrasound irradiation by triggering the vaporization and phase transition of PFH from liquid to gas in order to achieve microbubbles. Ultrasound imaging, encapsulation efficiency cytotoxicity, and cellular uptake of bFGF-MSs were evaluated using human umbilical vein endothelial cells (HUVECs) in vitro. In vivo imaging demonstrated effective accumulation of platelet- microspheres injected into the ischemic myocardium region. The results revealed the potential use of bFGF-loaded microbubbles as a noninvasive and effective carrier for MI therapy.
Collapse
Affiliation(s)
- Aliyeh Ghamkhari
- Institute
of Polymeric Materials and Faculty of Polymer Engineering, Sahand University of Technology, Tabriz 5331817634, Iran
| | - Hossein Ahmadi Tafti
- Research
Center for Advanced Technologies in Cardiovascular Medicine, Tehran
Heart Center, Tehran University of Medical
Sciences, Tehran 1416753955, Iran
| | - Shahram Rabbani
- Research
Center for Advanced Technologies in Cardiovascular Medicine, Tehran
Heart Center, Tehran University of Medical
Sciences, Tehran 1416753955, Iran
| | - Marjan Ghorbani
- Nutrition
Research Center, Tabriz University of Medical Sciences, Tabriz IR 51656-65811, Iran
| | - Mohammad Adel Ghiass
- Tissue
Engineering Department, Tarbiat Modares
University, Tehran 1411713116, Iran
| | - Fariborz Akbarzadeh
- Cardiovascular
Research Center, Tabriz University of Medical
Sciences, Tabriz 5166/15731, Iran
| | - Farhang Abbasi
- Institute
of Polymeric Materials and Faculty of Polymer Engineering, Sahand University of Technology, Tabriz 5331817634, Iran
| |
Collapse
|
27
|
Sha X, Dai Y, Chong L, Wei M, Xing M, Zhang C, Li J. Pro-efferocytic macrophage membrane biomimetic nanoparticles for the synergistic treatment of atherosclerosis via competition effect. J Nanobiotechnology 2022; 20:506. [PMID: 36456996 PMCID: PMC9714205 DOI: 10.1186/s12951-022-01720-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Macrophages participate in many links in the pathological process of atherosclerosis (AS) and the regulation of influence of macrophages at the molecular level might be a new avenue for AS treatment. For this aim, the macrophage membrane biomimetic nanoparticles, derived from macrophage membrane coated SHP1i-loaded liposome NPs (MM@Lips-SHP1i) was designed. Due to the reservation of intrinsic membrane proteins and function from macrophages, the biomimic nanoparticles could effectively evade clearance by the immune system, prolong blood circulation time and actively tend and aggregate to atherosclerotic plaques. More importantly, in the plaque area, MM@Lips-SHP1i nanoparticles could compete with macrophages in vivo to bind with oxidized low-density lipoprotein (oxLDL) and lipopolysaccharide (LPS), reduce uptake of new lipids by macrophages, reduce foam cell formation, and inhibit the expression of pro-inflammatory cytokines. In addition, small molecule inhibitor of SHP-1, the downstream effector molecule of CD47 loaded in macrophage membrane biomimetic nanoparticles could interrupt CD47-SIRPα signal transduction in monocytes and macrophages, thereby enhancing the efferocytosis of macrophages, inhibiting the progression of plaque, achieving synergistic treatment of atherosclerosis. This work focuses on the key process in the formation of AS, macrophage foaming and chronic inflammation, and is based on the fact that macrophage membrane biomimetic nanoparticles can preserve the key surface proteins of macrophages closely related to the formation of AS, providing a new avenue to inhibit the progression of AS by utilizing the biological characteristics of macrophage membrane in macrophage membrane biomimetic nanoparticles.
Collapse
Affiliation(s)
- Xuan Sha
- grid.417303.20000 0000 9927 0537School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004 China
| | - Yue Dai
- grid.417303.20000 0000 9927 0537School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004 China ,grid.413389.40000 0004 1758 1622Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006 China
| | - Lijuan Chong
- grid.417303.20000 0000 9927 0537School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004 China ,grid.413389.40000 0004 1758 1622Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006 China
| | - Min Wei
- grid.417303.20000 0000 9927 0537School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004 China ,grid.413389.40000 0004 1758 1622Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006 China
| | - Mengyuan Xing
- grid.417303.20000 0000 9927 0537School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004 China
| | - Chun Zhang
- grid.417303.20000 0000 9927 0537School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004 China ,grid.413389.40000 0004 1758 1622Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006 China
| | - Jingjing Li
- grid.417303.20000 0000 9927 0537School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004 China ,grid.413389.40000 0004 1758 1622Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006 China
| |
Collapse
|
28
|
Scavenger receptor-targeted plaque delivery of microRNA-coated nanoparticles for alleviating atherosclerosis. Proc Natl Acad Sci U S A 2022; 119:e2201443119. [PMID: 36122215 PMCID: PMC9522431 DOI: 10.1073/pnas.2201443119] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Atherosclerosis treatments by gene regulation are garnering attention, yet delivery of gene cargoes to atherosclerotic plaques remains inefficient. Here, we demonstrate that assembly of therapeutic oligonucleotides into a three-dimensional spherical nucleic acid nanostructure improves their systemic delivery to the plaque and the treatment of atherosclerosis. This noncationic nanoparticle contains a shell of microRNA-146a oligonucleotides, which regulate the NF-κB pathway, for achieving transfection-free cellular entry. Upon an intravenous injection into apolipoprotein E knockout mice fed with a high-cholesterol diet, this nanoparticle naturally targets class A scavenger receptor on plaque macrophages and endothelial cells, contributing to elevated delivery to the plaques (∼1.2% of the injected dose). Repeated injections of the nanoparticle modulate genes related to immune response and vascular inflammation, leading to reduced and stabilized plaques but without inducing severe toxicity. Our nanoparticle offers a safe and effective treatment of atherosclerosis and reveals the promise of nucleic acid nanotechnology for cardiovascular disease.
Collapse
|
29
|
Yang F, Xue J, Wang G, Diao Q. Nanoparticle-based drug delivery systems for the treatment of cardiovascular diseases. Front Pharmacol 2022; 13:999404. [PMID: 36172197 PMCID: PMC9512262 DOI: 10.3389/fphar.2022.999404] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular disease is the most common health problem worldwide and remains the leading cause of morbidity and mortality. Despite recent advances in the management of cardiovascular diseases, pharmaceutical treatment remains suboptimal because of poor pharmacokinetics and high toxicity. However, since being harnessed in the cancer field for the delivery of safer and more effective chemotherapeutics, nanoparticle-based drug delivery systems have offered multiple significant therapeutic effects in treating cardiovascular diseases. Nanoparticle-based drug delivery systems alter the biodistribution of therapeutic agents through site-specific, target-oriented delivery and controlled drug release of precise medicines. Metal-, lipid-, and polymer-based nanoparticles represent ideal materials for use in cardiovascular therapeutics. New developments in the therapeutic potential of drug delivery using nanoparticles and the application of nanomedicine to cardiovascular diseases are described in this review. Furthermore, this review discusses our current understanding of the potential role of nanoparticles in metabolism and toxicity after therapeutic action, with a view to providing a safer and more effective strategy for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Fangyu Yang
- Department of Clinical Laboratory Medicine, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Jianjiang Xue
- Department of Clinical Laboratory Medicine, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Guixue Wang
- Key Laboratory for Bio-Rheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Qizhi Diao
- Department of Clinical Laboratory Medicine, Sanya Women and Children’s Hospital Managed by Shanghai Children’s Medical Center, Hainan, China
- *Correspondence: Qizhi Diao,
| |
Collapse
|
30
|
Abstract
Atherosclerosis is a chronic inflammatory disease involved in plaque rupture, stroke, thrombosis, and heart attack (myocardial infarction), which is a leading cause of sudden cardiovascular events. In the past decades, various imaging strategies have been designed and employed for the diagnosis of atherosclerosis. Targeted imaging can accurately distinguish pathological tissues from normal tissues and reliably reveal biological information in the occurrence and development of atherosclerosis. By taking advantage of versatile imaging techniques, rationally designed imaging probes targeting biomarkers overexpressed in plaque microenvironments and targeting activated cells by modifying specific ligands accumulated in lesion regions have attracted increasing attention. This Perspective elucidates comprehensively the targeted imaging strategies, current challenges, and future development directions for precise identification and diagnosis of atherosclerosis, which is beneficial to better understand the physiological and pathological progression and exploit novel imaging strategies.
Collapse
Affiliation(s)
- Jingjing Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Kaixian Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People's Republic of China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People's Republic of China
| |
Collapse
|
31
|
Zhang S, Liu Y, Cao Y, Zhang S, Sun J, Wang Y, Song S, Zhang H. Targeting the Microenvironment of Vulnerable Atherosclerotic Plaques: An Emerging Diagnosis and Therapy Strategy for Atherosclerosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110660. [PMID: 35238081 DOI: 10.1002/adma.202110660] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/10/2022] [Indexed: 06/14/2023]
Abstract
Atherosclerosis is considered one of the primary causes of cardiovascular diseases (CVDs). Unpredictable rupture of the vulnerable atherosclerotic plaques triggers adverse cardiovascular events such as acute myocardial syndrome and even sudden cardiac death. Therefore, assessing the vulnerability of atherosclerotic plaques and early intervention are of significance in reducing CVD mortality. Nanomedicine possesses tremendous advantages in achieving the integration of the diagnosis and therapy of atherosclerotic plaques because of its magnetic, optical, thermal, and catalytic properties. Based on the pathological characteristics of vulnerable plaques, stimuli-responsive nanoplatforms and surface-functionalized nanoagents are designed and have drawn great attention for accomplishing the precise imaging and treatment of vulnerable atherosclerotic plaques due to their superior properties, such as high bioavailability, lesion-targeting specificity, on-demand cargo release, and low off-target damage. Here, the characteristics of vulnerable plaques are generalized, and some targeted strategies for boosting the accuracy of plaque vulnerability evaluation by imaging and the efficacy of plaque stabilization therapy (including antioxidant therapy, macrophage depletion therapy, regulation of lipid metabolism therapy, anti-inflammation therapy, etc.) are systematically summarized. In addition, existing challenges and prospects in this field are discussed, and it is believed to provide new thinking for the diagnosis and treatment of CVDs in the near future.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Cardiovascular Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, 130021, China
| | - Yang Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yue Cao
- Department of Neurosurgery, The First Hospital of Jilin University, 71 Ximin Street, Changchun, Jilin, 130021, China
| | - Songtao Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Jian Sun
- Department of Cardiovascular Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, 130021, China
| | - Yinghui Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Shuyan Song
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
- Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
32
|
Fang N, Liu J, Hou J, Zhong Y, Luo Y, Hu L, Zhang W, Wang J, Xu J, Zhou J, Zhang Y, Ran H, Guo D. Magnet-Guided Bionic System with LIFU Responsiveness and Natural Thrombus Tropism for Enhanced Thrombus-Targeting Ability. Int J Nanomedicine 2022; 17:2019-2039. [PMID: 35558339 PMCID: PMC9087377 DOI: 10.2147/ijn.s357050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/21/2022] [Indexed: 11/23/2022] Open
Abstract
Background Arterial thrombosis is a serious threat to human health. Recently, many thrombus-targeted nanoparticles (NPs) have been constructed for detecting thrombi or monitoring thrombolysis, but their thrombus-targeting performance is limited. Considering this drawback, we designed a specific bionic system with enhanced thrombus-targeting ability. Materials and Methods In the bionic system, gelatin was chosen as a carrier, and Fe3O4 served as a magnetic navigation medium and a magnetic resonance (MR) imaging agent. The CREKA peptide, which targets fibrin, was conjugated to the surface of gelatin to prepare targeted NPs (TNPs), which were then engulfed by macrophages to construct the bionic system. At the targeted site, the bionic system released its interior TNPs under low-intensity focused ultrasound (LIFU) irradiation. Moreover, the targeting performance was further improved by the conjugated CREKA peptide. Results In this study, we successfully constructed a bionic system and demonstrated its targeting ability in vitro and in vivo. The results indicated that most TNPs were released from macrophages under LIFU irradiation at 2 W/cm2 for 10 min in vitro. Additionally, the enhanced thrombus-targeting ability, based on the natural tropism of macrophages toward inflammatory thrombi, magnetic navigation and the CREKA peptide, was verified ex vivo and in vivo. Moreover, compared with the bionic system group, the group treated with TNPs had significantly decreased liver and spleen signals in MR images and significantly enhanced liver and spleen signals in fluorescence images, indicating that the bionic system is less likely to be cleared by the reticuloendothelial system (RES) than TNPs, which may promote the accumulation of the bionic system at the site of the thrombus. Conclusion These results suggest that the magnet-guided bionic system with LIFU responsiveness is an excellent candidate for targeting thrombi and holds promise as an innovative drug delivery system for thrombolytic therapy.
Collapse
Affiliation(s)
- Ni Fang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Jia Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Jingxin Hou
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Yixin Zhong
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Ying Luo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Liu Hu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Wenli Zhang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Junrui Wang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Jie Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Jun Zhou
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Yu Zhang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Dajing Guo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
- Correspondence: Dajing Guo, Email
| |
Collapse
|
33
|
Sriamornsak P, Dass CR. Chitosan Nanoparticles in Atherosclerosis-Development to Preclinical Testing. Pharmaceutics 2022; 14:935. [PMID: 35631521 PMCID: PMC9145436 DOI: 10.3390/pharmaceutics14050935] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/04/2022] [Accepted: 04/22/2022] [Indexed: 02/01/2023] Open
Abstract
Chitosan is a natural biopolymer that is present in an abundant supply in sources such as crustacean shells, mushrooms, and insect exoskeletons. It can be used to make a variety of types of drug formulations and is generally safe to use in vivo; plus, it has inherent cholesterol-reducing properties. While an abundance of papers has tested this biopolymer in nanoparticles in cancer and diabetes research, there is a lag of usage, and hence the paucity of information, in the area of cardiovascular research, specifically in atherosclerosis, the topic of this review. This review highlights some of the deficiencies in this niche area of research, examines the range of chitosan nanoparticles that have been researched to date, and proposes several ways forward to advance this field. Nanoparticles used for both diagnostic and therapeutic purposes are reviewed, with a discussion on how these nanoparticles could be better researched in future and what lays ahead as the field potentially moves towards clinical trials in future.
Collapse
Affiliation(s)
- Pornsak Sriamornsak
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand;
- Pharmaceutical Biopolymer Group (PBiG), Silpakorn University, Nakhon Pathom 73000, Thailand
- Academy of Science, The Royal Society of Thailand, Bangkok 10300, Thailand
| | - Crispin R. Dass
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Bentley 6102, Australia
| |
Collapse
|
34
|
Hou J, Zhou J, Chang M, Bao G, Xu J, Ye M, Zhong Y, Liu S, Wang J, Zhang W, Ran H, Wang Z, Chen Y, Guo D. LIFU-responsive nanomedicine enables acoustic droplet vaporization-induced apoptosis of macrophages for stabilizing vulnerable atherosclerotic plaques. Bioact Mater 2022; 16:120-133. [PMID: 35386311 PMCID: PMC8958425 DOI: 10.1016/j.bioactmat.2022.02.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 02/07/2023] Open
Abstract
Due to the high risk of tearing and rupture, vulnerable atherosclerotic plaques would induce serious cardiovascular and cerebrovascular diseases. Despite the available clinical methods can evaluate the vulnerability of plaques and specifically treat vulnerable plaques before a cardiovascular event, but the efficiency is still low and undesirable. Herein, we rationally design and engineer the low-intensity focused ultrasound (LIFU)-responsive FPD@CD nanomedicine for the highly efficient treatment of vulnerable plaques by facilely loading phase transition agent perfluorohexane (PFH) into biocompatible PLGA-PEG-PLGA nanoparticles (PPP NPs) and then attaching dextran sulphate (DS) onto the surface of PPP NPs for targeting delivery. DS, as a typical macrophages-targeted molecule, can achieve the precise vaporization of NPs and subsequently controllable apoptosis of RAW 264.7 macrophages as induced by acoustic droplet vaporization (ADV) effect. In addition, the introduction of DiR and Fe3O4 endows nanomedicine with near-infrared fluorescence (NIRF) and magnetic resonance (MR) imaging capabilities. The engineered FPD@CD nanomedicine that uses macrophages as therapeutic targets achieve the conspicuous therapeutic effect of shrinking vulnerable plaques based on in vivo and in vitro evaluation outcomes. A reduction of 49.4% of vascular stenosis degree in gross pathology specimens were achieved throughout the treatment period. This specific, efficient and biosafe treatment modality potentiates the biomedical application in patients with cardiovascular and cerebrovascular diseases based on the relief of the plaque rupture concerns. A new nanomedicine-enabled treatment strategy has been developed for treating vulnerable plaques by employing ADV. The optimal treatment conditions for ADV have been explored, including LIFU irradiation power intensity and plaque stability. The underlying mechanism of nanomedicine-enabled ADV in the treatment of vulnerable plaques has been studied systematically.
Collapse
Affiliation(s)
- Jingxin Hou
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Jun Zhou
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Meiqi Chang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Guangcheng Bao
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| | - Jie Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Man Ye
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yixin Zhong
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Shuling Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Junrui Wang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Wei Zhang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Dajing Guo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| |
Collapse
|
35
|
Zhang M, Xie Z, Long H, Ren K, Hou L, Wang Y, Xu X, Lei W, Yang Z, Ahmed S, Zhang H, Zhao G. Current advances in the imaging of atherosclerotic vulnerable plaque using nanoparticles. Mater Today Bio 2022; 14:100236. [PMID: 35341094 PMCID: PMC8943324 DOI: 10.1016/j.mtbio.2022.100236] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/13/2022] [Accepted: 03/05/2022] [Indexed: 01/29/2023]
Abstract
Vulnerable atherosclerotic plaques of the artery wall that pose a significant risk of cardio-cerebral vascular accidents remain the global leading cause of morbidity and mortality. Thus, early delineation of vulnerable atherosclerotic plaques is of clinical importance for prevention and treatment. The currently available imaging technologies mainly focus on the structural assessment of the vascular wall. Unfortunately, several disadvantages in these strategies limit the improvement in imaging effect. Nanoparticle technology is a novel diagnostic strategy for targeting and imaging pathological biomarkers. New functionalized nanoparticles that detect hallmarks of vulnerable plaques are promising for advance further control of this critical illness. The review aims to address the current opportunities and challenges for the use of nanoparticle technology in imagining vulnerable plaques.
Collapse
|
36
|
Simultaneous Noninvasive Detection and Therapy of Atherosclerosis Using HDL Coated Gold Nanorods. Diagnostics (Basel) 2022; 12:diagnostics12030577. [PMID: 35328130 PMCID: PMC8947645 DOI: 10.3390/diagnostics12030577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 11/20/2022] Open
Abstract
Cardiovascular disease (CVD) is a major cause of death and disability worldwide. A real need exists in the development of new, improved therapeutic methods for treating CVD, while major advances in nanotechnology have opened new avenues in this field. In this paper, we report the use of gold nanoparticles (GNPs) coated with high-density lipoprotein (HDL) (GNP-HDL) for the simultaneous detection and therapy of unstable plaques. Based on the well-known HDL cardiovascular protection, by promoting the reverse cholesterol transport (RCT), injured rat carotids, as a model for unstable plaques, were injected with the GNP-HDL. Noninvasive detection of the plaques 24 h post the GNP injection was enabled using the diffusion reflection (DR) method, indicating that the GNP-HDL particles had accumulated in the injured site. Pathology and noninvasive CT measurements proved the recovery of the injured artery treated with the GNP-HDL. The DR of the GNP-HDL presented a simple and highly sensitive method at a low cost, resulting in simultaneous specific unstable plaque diagnosis and recovery.
Collapse
|
37
|
Kamegawa R, Naito M, Uchida S, Kim HJ, Kim BS, Miyata K. Bioinspired Silicification of mRNA-Loaded Polyion Complexes for Macrophage-Targeted mRNA Delivery. ACS APPLIED BIO MATERIALS 2021; 4:7790-7799. [PMID: 35006762 DOI: 10.1021/acsabm.1c00704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In vitro transcribed messenger RNA (mRNA) delivery to macrophages is a promising therapeutic modality for inflammatory diseases because it can modulate the immunological activity of macrophages. However, efficient macrophage-targeted mRNA delivery remains challenging. Herein, we fabricated silica-coated polyion complexes (PICs), termed SilPICs, via bioinspired silicification for stable encapsulation of mRNA and scavenger receptor (SR)-mediated macrophage targeting. Silica coating was readily performed by simply mixing mRNA-loaded PICs with tetramethyl orthosilicate in aqueous media at 25 °C. The silica shell formation was verified by a slight increase in size (∼18 nm), a conversion of ζ-potential from positive (+22 mV) to negative (-23 mV), the peak appearance derived from silanol groups and siloxane bonds in the IR spectra, and elemental analyses by scanning transmission electron microscopy-energy-dispersive X-ray spectrometry (STEM-EDS). The silica shell efficiently protected the mRNA payload from enzymatic degradation in a fetal bovine serum-containing medium. Meanwhile, the reversibility of the silica shell allowed mRNA release from SilPICs after silica dissolution into silicic acids under diluted conditions. Furthermore, SilPICs elicited 20-fold higher mRNA transfection efficiency in the macrophage cell line RAW264.7 compared to noncoated PICs, presumably due to the facilitated cellular internalization by the silica shell. These enhancements were compromised in the RAW264.7 cells incubated with dextran sulfate and poly(inosinic acid) as inhibitors of SR type A1 and were not observed in cultured CT26 colon cancer cells, which are SR-negative cells. Collectively, SilPIC is a promising mRNA delivery vehicle with both mRNA protectability and macrophage targetability.
Collapse
Affiliation(s)
- Rimpei Kamegawa
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Mitsuru Naito
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Satoshi Uchida
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| | - Hyun Jin Kim
- Department of Biological Engineering, College of Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Korea
| | - Beob Soo Kim
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
38
|
Abstract
Low-intensity ultrasound-triggered sonodynamic therapy (SDT) is a promising noninvasive therapeutic modality due to its strong tissue penetration ability. In recent years, with the development of nanotechnology, nanoparticle-based sonosensitizer-mediated SDT has been widely investigated. With the increasing demand for precise and personalized treatment, abundant novel sonosensitizers with imaging capabilities have been developed for determining the optimal therapeutic window, thus significantly enhancing treatment efficacy. In this review, we focus on the molecular imaging-guided SDT. The prevalent mechanisms of SDT are discussed, including ultrasonic cavitation, sonoluminescence, reactive oxygen species, and mechanical damage. In addition, we introduce the major molecular imaging techniques and the design principles based on nanoparticles to achieve efficient imaging. Furthermore, the imaging-guided SDT for the treatment of cancer, bacterial infections, and vascular diseases is summarized. The ultimate goal is to design more effective imaging-guided SDT modalities and provide novel ideas for clinical translation of SDT.
Collapse
Affiliation(s)
- Juan Guo
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Xueting Pan
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Chaohui Wang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| |
Collapse
|
39
|
Guo J, Yang Z, Wang X, Xu Y, Lu Y, Qin Z, Zhang L, Xu J, Wang W, Zhang J, Tang J. Advances in Nanomaterials for Injured Heart Repair. Front Bioeng Biotechnol 2021; 9:686684. [PMID: 34513807 PMCID: PMC8424111 DOI: 10.3389/fbioe.2021.686684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 08/09/2021] [Indexed: 11/30/2022] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is one of the leading causes of mortality worldwide. Because of the limited regenerative capacity of adult myocardium to compensate for the loss of heart tissue after ischemic infarction, scientists have been exploring the possible mechanisms involved in the pathological process of ASCVD and searching for alternative means to regenerate infarcted cardiac tissue. Although numerous studies have pursued innovative solutions for reversing the pathological process of ASCVD and improving the effectiveness of delivering therapeutics, the translation of those advances into downstream clinical applications remains unsatisfactory because of poor safety and low efficacy. Recently, nanomaterials (NMs) have emerged as a promising new strategy to strengthen both the efficacy and safety of ASCVD therapy. Thus, a comprehensive review of NMs used in ASCVD treatment will be useful. This paper presents an overview of the pathophysiological mechanisms of ASCVD and the multifunctional mechanisms of NM-based therapy, including antioxidative, anti-inflammation and antiapoptosis mechanisms. The technological improvements of NM delivery are summarized and the clinical transformations concerning the use of NMs to treat ASCVD are examined. Finally, this paper discusses the challenges and future perspectives of NMs in cardiac regeneration to provide insightful information for health professionals on the latest advancements in nanotechnologies for ASCVD treatment.
Collapse
Affiliation(s)
- Jiacheng Guo
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Zhenzhen Yang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xu Wang
- Department of Medical Record Management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanyan Xu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Yongzheng Lu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Zhen Qin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Li Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Jing Xu
- Department of Cardiac Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Wang
- Henan Medical Association, Zhengzhou, China
| | - Jinying Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Junnan Tang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| |
Collapse
|
40
|
Wang J, Song W, Wang X, Xie Z, Zhang W, Jiang W, Liu S, Hou J, Zhong Y, Xu J, Ran H, Guo D. Tumor-self-targeted "thermoferroptosis-sensitization" magnetic nanodroplets for multimodal imaging-guided tumor-specific therapy. Biomaterials 2021; 277:121100. [PMID: 34492584 DOI: 10.1016/j.biomaterials.2021.121100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/25/2021] [Accepted: 08/25/2021] [Indexed: 01/08/2023]
Abstract
Ferroptosis-based nanomedicine has drawn increasing attention in antitumor therapy because of the advantages of this unconventional mode of apoptosis, but the difficulties of delivery to the tumor site and surface-to-core penetration after arrival seriously hinder further clinical transformation and application. Herein, we propose an unprecedented strategy of injecting magnetic nanodroplets (MNDs) to solve these two longstanding problems. MNDs are nanocarriers that can carry multifunctional drugs and imaging materials. MNDs can effectively accumulate in the tumor site by active tumor targeting (multifunctional drugs) and passive tumor targeting (enhanced permeability and retention effect), allowing diffusion of the MNDs from the surface to the core through mild-temperature magnetic fluid hyperthermia (MHT) under multimodal imaging guidance. Finally, the ferroptosis pathway is activated deep within the tumor site through the drug release. This approach was inspired by the ability of mild-temperature MHT to allow MNDs to quickly pass through the blood vessel-tumor barrier and deeply penetrate the tumor tissue from the surface to the core to amplify the antitumor efficacy of ferroptosis. This strategy is termed as "thermoferroptosis sensitization". Importantly, this behavior can be performed under the guidance of multimodal imaging, making the design of MNDs for cancer therapy safer and more reasonable.
Collapse
Affiliation(s)
- Junrui Wang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China
| | - Weixiang Song
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China
| | - Xingyue Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China
| | - Zhuoyan Xie
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China
| | - Wenli Zhang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China
| | - Weixi Jiang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China
| | - Shuling Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China
| | - Jingxin Hou
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China
| | - Yixin Zhong
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China
| | - Jie Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China
| | - Dajing Guo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, PR China.
| |
Collapse
|
41
|
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
|
42
|
Li J, Qiu H, Gong H, Tong W. Broad-Spectrum Reactive Oxygen Species Scavenging and Activated Macrophage-Targeting Microparticles Ameliorate Inflammatory Bowel Disease. Biomacromolecules 2021; 22:3107-3118. [PMID: 34160209 DOI: 10.1021/acs.biomac.1c00551] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inflammatory bowel disease (IBD) is a refractory chronic inflammatory disease. An excessively high level of reactive oxygen species (ROS) in the colon is one of the characteristics and pathogenic factors of IBD. Therefore, scavenging excessive ROS is a feasible method to treat IBD. Because ROS include many types of species, scavenging a single kind of ROS is not enough to reduce the ROS level and cure IBD effectively. Herein, broad-spectrum ROS scavenging and activated macrophage-targeting microparticles (MPs) are successfully fabricated by coprecipitation of catalase (CAT) and bovine serum albumin into a MnCO3 template followed by deposition of polydopamine (PDA), assembly of targeting molecules on the surface, and finally removal of MnCO3. The CAT content of MPs is about 34.1%. The obtained MPs can effectively scavenge the broad spectrum of ROS and retain 88% of the radical scavenging activity even after the treatment of simulated gastric fluid. The surface-modified dextran sulfate endows MPs with the targeting ability toward activated macrophages, achieving a better therapeutic effect. The MPs with components mostly derived from natural substances exhibit good biocompatibility and can show excellent ROS scavenging ability in cell experiments. In animal experiments, oral administration of a proper dosage of MPs can substantially mitigate colonic inflammation, as evidenced by disease activity index scores reduced by ∼40%, reduced body weight loss, and the production of typical proinflammatory cytokines in the inflammatory colon. This kind of MP can also be utilized for the treatment of other inflammatory diseases.
Collapse
Affiliation(s)
- Jiawei Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Huiqiang Qiu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Hengtai Gong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Weijun Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
43
|
Liu J, Zhou B, Guo Y, Zhang A, Yang K, He Y, Wang J, Cheng Y, Cui D. SR-A-Targeted Nanoplatform for Sequential Photothermal/Photodynamic Ablation of Activated Macrophages to Alleviate Atherosclerosis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:29349-29362. [PMID: 34133141 DOI: 10.1021/acsami.1c06380] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cardiovascular and cerebrovascular diseases induced by atherosclerosis (AS) have become the dominant cause of disability and mortality throughout the world. The typical early pathological process of AS involves the activation of inflammatory macrophages in the vulnerable plaque. In this work, we first employed chitosan-coated carbon nanocages (CS-CNCs) as nanocarriers to load Chlorin e6 (Ce6) and then linked dextran sulfate (DS) to the outermost layer by electrostatic adsorption to create a multifunctional therapeutic nanoplatform, CS-CNCs@Ce6/DS. The DS of the nanoplatform can recognize and bind to the type A scavenger receptor (SR-A), which is expressed only on the activated macrophages of the arterial plaque, so the proposed nanoplatform selectively targets these macrophages and accumulates there. Furthermore, DS can competitively inhibit cellular endocytosis of oxidized low-density lipoproteins via blocking of SR-A. The rapid photothermal conversion capability of CS-CNCs enables efficient therapeutic delivery during photothermal therapy (PTT). Interestingly, near-infrared-accelerated drug release induced by initial 808-nm laser irradiation was observed, thus enhancing the Ce6 concentration in the atherosclerotic plaque area and the efficiency of photodynamic therapy (PDT). Sequential photothermal/photodynamic ablation of the activated macrophages reduced pro-inflammatory cytokine secretion and alleviated the proliferation and migration of smooth muscle cells. These finally resulted in the stabilization and shrinkage of atherosclerotic plaques, further inhibiting the development and exacerbation of AS. Therefore, this work achieved a "1 + 1 greater than 2" effect by providing a novel approach to the treatment of atherosclerotic plaques, which is promising for the prevention of AS-related diseases.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, P. R. China
| | - Bi Zhou
- Department of Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, P. R. China
| | - Yuliang Guo
- Rehabilitation Department at Shanghai Putuo District People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. China
| | - Amin Zhang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Kai Yang
- Department of Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, P. R. China
| | - Yu He
- Department of Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, P. R. China
| | - Jianbo Wang
- Department of Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, P. R. China
| | - Yingsheng Cheng
- Department of Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, P. R. China
| | - Daxiang Cui
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| |
Collapse
|
44
|
Crețu BEB, Dodi G, Shavandi A, Gardikiotis I, Șerban IL, Balan V. Imaging Constructs: The Rise of Iron Oxide Nanoparticles. Molecules 2021; 26:3437. [PMID: 34198906 PMCID: PMC8201099 DOI: 10.3390/molecules26113437] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, an important challenge in nanomedicine imaging has been the work to design multifunctional agents that can be detected by single and/or multimodal techniques. Among the broad spectrum of nanoscale materials being investigated for imaging use, iron oxide nanoparticles have gained significant attention due to their intrinsic magnetic properties, low toxicity, large magnetic moments, superparamagnetic behaviour and large surface area-the latter being a particular advantage in its conjunction with specific moieties, dye molecules, and imaging probes. Tracers-based nanoparticles are promising candidates, since they combine synergistic advantages for non-invasive, highly sensitive, high-resolution, and quantitative imaging on different modalities. This study represents an overview of current advancements in magnetic materials with clinical potential that will hopefully provide an effective system for diagnosis in the near future. Further exploration is still needed to reveal their potential as promising candidates from simple functionalization of metal oxide nanomaterials up to medical imaging.
Collapse
Affiliation(s)
- Bianca Elena-Beatrice Crețu
- Advanced Centre for Research-Development in Experimental Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.E.-B.C.); (I.G.)
| | - Gianina Dodi
- Advanced Centre for Research-Development in Experimental Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.E.-B.C.); (I.G.)
| | - Amin Shavandi
- BioMatter-Biomass Transformation Lab, École Polytechnique de Bruxelles, Université Libre de Bruxelles, 1050 Brussels, Belgium;
| | - Ioannis Gardikiotis
- Advanced Centre for Research-Development in Experimental Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.E.-B.C.); (I.G.)
| | - Ionela Lăcrămioara Șerban
- Physiology Department, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania;
| | - Vera Balan
- Faculty of Medical Bioengineering, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania;
| |
Collapse
|
45
|
Gao B, Xu J, Zhou J, Zhang H, Yang R, Wang H, Huang J, Yan F, Luo Y. Multifunctional pathology-mapping theranostic nanoplatforms for US/MR imaging and ultrasound therapy of atherosclerosis. NANOSCALE 2021; 13:8623-8638. [PMID: 33929480 DOI: 10.1039/d1nr01096d] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Atherosclerotic thrombosis is the leading cause of most life-threatening cardiovascular diseases (CVDs), particularly as a result of rupture or erosion of vulnerable plaques. Rupture or erosion-prone plaques are quite different in cellular composition and immunopathology, requiring different treatment strategies. The current imaging technology cannot distinguish the types of vulnerable plaques, and thus empirical treatment is still applied to all without a tailored and precise treatment. Herein, we propose a novel strategy called "Multifunctional Pathology-mapping Theranostic Nanoplatform (MPmTN)" for the tailored treatment of plaques based on the pathological classification. MPmTNs are made up of poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs), containing contrast imaging materials Fe3O4 and perfluoropentane (PFP), and coated with specific plaque-targeted peptides PP1 and cyclic RGD. The PFP encapsulated inside the MPmTN can undergo a phase change from nanodroplets to gas microbubbles under therapeutic ultrasound (TUS) exposure. The acoustic and biological effects induced by TUS and disruption of microbubbles may further promote therapeutic effects. Hypothetically, MPmTN NPs can target the rupture-prone plaque via the binding of PP1 to class A scavenger receptors (SR-A) on macrophages, induce the apoptosis due to TUS exposure and thus reduce the chronic soakage of inflammatory cells. The MPmTN NPs can also target the erosion-prone plaque through the binding of cRGD to glycoprotein (GP) IIb/IIIa on activated platelets and promote platelet disaggregation under TUS exposure. Therefore, MPmTNs may work as a multifunctional pathology-mapping therapeutic agent. Our in vitro results show that the MPmTN with PP1 and cRGD peptides had a high binding affinity both for activated macrophages and blood clots. Under TUS exposure, the MPmTN could effectively induce macrophage apoptosis, destroy thrombus and exhibit good imaging properties for ultrasound (US) and MRI. In apoE-/- mice, MPmTNs can selectively accumulate at the plaque site and reduce the T2-weighted signal. The apoptosis of macrophages and disaggregation of activated platelets on the plaques were also confirmed in vivo. In summary, this study provides a potential strategy for a tailored treatment of vulnerable plaques based on their pathological nature and a multimodal imaging tool for the risk stratification and assessment of therapeutic efficacy.
Collapse
Affiliation(s)
- Binyang Gao
- Department of Ultrasound, Laboratory of Ultrasound Imaging and Drug, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Li M, Wang Z, Wang P, Li H, Yang L. TFEB: A Emerging Regulator in Lipid Homeostasis for Atherosclerosis. Front Physiol 2021; 12:639920. [PMID: 33679452 PMCID: PMC7925399 DOI: 10.3389/fphys.2021.639920] [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: 12/10/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis, predominantly characterized by the disturbance of lipid homeostasis, has become the main causation of various cardiovascular diseases. Therefore, there is an urgent requirement to explore efficacious targets that act as lipid modulators for atherosclerosis. Transcription factor EB (TFEB), whose activity depends on post-translational modifications, such as phosphorylation, acetylation, SUMOylation, ubiquitination, etc., is significant for normal cell physiology. Recently, increasing evidence implicates a role of TFEB in lipid homeostasis, via its functionality of promoting lipid degradation and efflux through mediating lipophagy, lipolysis, and lipid metabolism-related genes. Furthermore, a regulatory effect on lipid transporters and lipid mediators by TFEB is emerging. Notably, TFEB makes a possible therapeutic target of atherosclerosis by regulating lipid metabolism. This review recapitulates the update and current advances on TFEB mediating lipid metabolism to focus on two intracellular activities: a) how cells perceive external stimuli and initiate transcription programs to modulate TFEB function, and b) how TFEB restores lipid homeostasis in the atherosclerotic process. In-depth research is warranted to develop potent agents against TFEB to alleviate or reverse the progression of atherosclerosis.
Collapse
Affiliation(s)
- Manman Li
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Zitong Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Pengyu Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Hong Li
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University-Daqing, Daqing, China
| |
Collapse
|
47
|
Chen J, Zhang X, Millican R, Sherwood J, Martin S, Jo H, Yoon YS, Brott BC, Jun HW. Recent advances in nanomaterials for therapy and diagnosis for atherosclerosis. Adv Drug Deliv Rev 2021; 170:142-199. [PMID: 33428994 PMCID: PMC7981266 DOI: 10.1016/j.addr.2021.01.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/02/2021] [Accepted: 01/03/2021] [Indexed: 12/18/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease driven by lipid accumulation in arteries, leading to narrowing and thrombosis. It affects the heart, brain, and peripheral vessels and is the leading cause of mortality in the United States. Researchers have strived to design nanomaterials of various functions, ranging from non-invasive imaging contrast agents, targeted therapeutic delivery systems to multifunctional nanoagents able to target, diagnose, and treat atherosclerosis. Therefore, this review aims to summarize recent progress (2017-now) in the development of nanomaterials and their applications to improve atherosclerosis diagnosis and therapy during the preclinical and clinical stages of the disease.
Collapse
Affiliation(s)
- Jun Chen
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Xixi Zhang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | | | | | - Sean Martin
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States; Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Young-Sup Yoon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Brigitta C Brott
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ho-Wook Jun
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
48
|
Modak M, Frey MA, Yi S, Liu Y, Scott EA. Employment of targeted nanoparticles for imaging of cellular processes in cardiovascular disease. Curr Opin Biotechnol 2020; 66:59-68. [PMID: 32682272 PMCID: PMC7744313 DOI: 10.1016/j.copbio.2020.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/13/2020] [Accepted: 06/07/2020] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease (CVD) is a leading cause of global mortality, accounting for pathologies that are primarily of atherosclerotic origin and driven by specific cell populations. A need exists for effective, non-invasive methods to assess the risk of potentially fatal major adverse cardiovascular events (MACE) before occurrence and to monitor post-interventional outcomes such as tissue regeneration. Molecular imaging has widespread applications in CVD diagnostic assessment, through modalities including magnetic resonance imaging (MRI), positron emission tomography (PET), and acoustic imaging methods. However, current gold-standard small molecule contrast agents are not cell-specific, relying on non-specific uptake to facilitate imaging of biologic processes. Nanomaterials can be engineered for targeted delivery to specific cell populations, and several nanomaterial systems have been developed for pre-clinical molecular imaging. Here, we review recent advances in nanoparticle-mediated approaches for imaging of cellular processes in cardiovascular disease, focusing on efforts to detect inflammation, assess lipid accumulation, and monitor tissue regeneration.
Collapse
Affiliation(s)
- Mallika Modak
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Molly A Frey
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| | - Sijia Yi
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Yugang Liu
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA; Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; Department of Microbiology-Immunology, Northwestern University, Chicago, IL 60611, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
49
|
Resistin: Potential biomarker and therapeutic target in atherosclerosis. Clin Chim Acta 2020; 512:84-91. [PMID: 33248946 DOI: 10.1016/j.cca.2020.11.010] [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: 09/25/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022]
Abstract
Resistin, a cysteine-rich secretory protein, has a pleiotropic role in humans. Resistin usually presents as trimer or hexamer in plasma, and targets specific receptors Toll Like Receptor 4 (TLR4) or Adenylyl Cyclase-Associated Protein 1 (CAP1). Upon binding to TLR4 and CAP1, resistin can trigger various intracellular signal transduction pathways to induce vascular inflammation, lipid accumulation, and plaque vulnerability. These pro-atherosclerotic effects of resistin appear in various cell types, including endothelial cells, vessel smooth muscle cells and macrophages, which cause diverse damages to cardiovascular system from dyslipidemia, atherosclerosis rupture and ventricular remodeling. In this review, we gather recent evidence about the pro- atherosclerotic effects of resistin and highlight it as a candidate therapeutic or diagnostic target for cardiovascular disease.
Collapse
|
50
|
Cuthbert GA, Shaik F, Harrison MA, Ponnambalam S, Homer-Vanniasinkam S. Scavenger Receptors as Biomarkers and Therapeutic Targets in Cardiovascular Disease. Cells 2020; 9:cells9112453. [PMID: 33182772 PMCID: PMC7696859 DOI: 10.3390/cells9112453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/23/2022] Open
Abstract
The process of atherosclerosis leads to the formation of plaques in the arterial wall, resulting in a decreased blood supply to tissues and organs and its sequelae: morbidity and mortality. A class of membrane-bound proteins termed scavenger receptors (SRs) are closely linked to the initiation and progression of atherosclerosis. Increasing interest in understanding SR structure and function has led to the idea that these proteins could provide new routes for cardiovascular disease diagnosis, management, and treatment. In this review, we consider the main classes of SRs that are implicated in arterial disease. We consider how our understanding of SR-mediated recognition of diverse ligands, including modified lipid particles, lipids, and carbohydrates, has enabled us to better target SR-linked functionality in disease. We also link clinical studies on vascular disease to our current understanding of SR biology and highlight potential areas that are relevant to cardiovascular disease management and therapy.
Collapse
Affiliation(s)
- Gary A. Cuthbert
- Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK;
- Correspondence: ; Tel.:+44 113 3433007
| | - Faheem Shaik
- School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK; (F.S.); (S.P.)
| | | | - Sreenivasan Ponnambalam
- School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK; (F.S.); (S.P.)
| | | |
Collapse
|