1
|
Zhao X, Xiao Y, Jiang M, Cao Y. Pharmacological and toxicological roles of Kruppel-like factors (KLFs) in the cardiovascular system: a review. Mol Biol Rep 2025; 52:506. [PMID: 40418318 DOI: 10.1007/s11033-025-10613-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
Kruppel-like factors (KLFs) are transcription factors (TFs) increasingly implicated in cardiovascular pharmacology and toxicology through molecular mechanisms regulating endothelial function, macrophage polarization, and lipid metabolism. For example, KLF2/4 maintains endothelial homeostasis by modulating endothelial nitric oxide synthase (eNOS) activity and oxidative stress, and KLF4 additionally regulates smooth muscle cell phenotypic switch. KLF6 governs macrophage polarization and pyroptosis, while KLF15 modulates cardiomyocyte lipid metabolism, with dysregulation linked to cardiomyopathy. Not surprisingly, drugs such as statins and phytochemicals, as well as toxicants like ox-LDL, nanomaterials, and radiation, alter KLF expression via non-coding RNA (such as microRNA) or TFs, influencing endothelial cell activation, vascular smooth muscle cell phenotypic switch, macrophage inflammation, and cardiomyocyte apoptosis. KLF-dependent pathways intersect with key toxicological processes, such as autophagy, ferroptosis, and lipid dysregulation, culminating in atherosclerosis and heart failure. Despite preclinical advances demonstrating KLFs as therapeutic targets, clinical translation remains limited, with no KLF-targeted agents in active trials. Future studies should delineate tissue-specific KLF interactions, resolve KLFs' conflicting roles, and explore CRISPR-based KLF-targeting modulation. Bridging molecular mechanisms, such as KLF's regulation of phenotypic transformation pathways in smooth muscle cells, to drug discovery could yield novel therapies for cardiovascular diseases. The present review underscores the need for mechanistic and translational research to harness KLFs in cardiovascular pharmacotherapy and toxicant risk assessment.
Collapse
Affiliation(s)
- Xiaomei Zhao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yangfan Xiao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Miao Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, 421001, China
| | - Yi Cao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
2
|
Han GM, Liu JQ, Dai ZQ, Jin WL, Cai QL, Kong DM, Zhu LN. Multifunctional cerium-based nanozymes as moonlighting protein mimics for atherosclerosis diagnosis and therapy. Chem Sci 2025; 16:8772-8782. [PMID: 40271043 PMCID: PMC12013363 DOI: 10.1039/d5sc01014d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
Moonlighting proteins are multifunctional proteins widely present in organisms, playing crucial roles in various physiological activities. Drawing inspiration from the moonlight proteins, we developed a cerium (Ce)-based nanozyme CF, featuring multiple enzymatic activities along with robust cargo-loading and transport capabilities. The CF was synthesized through a one-step assembly between Ce3+ and a phosphorylated amino acid derivative, achieving high biostability through a simple heat treatment. The nanozyme possesses both superoxide dismutase (SOD) and catalase (CAT) activities, enabling scavenging of reactive oxygen species (ROS) and modulation of inflammation by inhibiting NF-κB pathway activation. Besides its enzymatic activities, CF can also serve as a versatile nanocarrier for various cargoes through one-pot co-assembly. Herein, the CF-based nanoassembly loaded with a near infrared fluorescent dye was demonstrated to work well for the diagnosis of atherosclerotic plaques. The nanoassembly co-assembled with probucol exhibited superior ROS-scavenging and anti-inflammatory effects compared to either CF nanozyme or probucol, attributed to the synergy of the nanozyme and the drug, thus facilitating a highly efficient treatment of atherosclerosis. This work introduces a novel Ce-based nanozyme with multifunctional properties, providing a promising approach to endow nanozymes with moonlighting protein-like characteristics, thereby enhancing their functional capabilities and broadening their application potential in various fields.
Collapse
Affiliation(s)
- Gui-Mei Han
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center of Analytical Science, College of Chemistry, Nankai University Tianjin 300071 P. R. China
- College of Chemistry and Chemical Engineering, Qilu Normal University Jinan 250200 P. R. China
| | - Jing-Qi Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center of Analytical Science, College of Chemistry, Nankai University Tianjin 300071 P. R. China
| | - Zhi-Qi Dai
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center of Analytical Science, College of Chemistry, Nankai University Tianjin 300071 P. R. China
| | - Wei-Liang Jin
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center of Analytical Science, College of Chemistry, Nankai University Tianjin 300071 P. R. China
- Department of Chemistry, School of Science, Tianjin University Tianjin 300354 P. R. China
| | - Qi-Liang Cai
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin 300211 P. R. China
| | - De-Ming Kong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center of Analytical Science, College of Chemistry, Nankai University Tianjin 300071 P. R. China
| | - Li-Na Zhu
- Department of Chemistry, School of Science, Tianjin University Tianjin 300354 P. R. China
| |
Collapse
|
3
|
Al-Kuraishy HM, Sulaiman GM, Mohsin MH, Mohammed HA, Dawood RA, Albuhadily AK, Al-Gareeb AI, Albukhaty S, Abomughaid MM. Targeting of AMPK/MTOR signaling in the management of atherosclerosis: Outmost leveraging. Int J Biol Macromol 2025; 309:142933. [PMID: 40203916 DOI: 10.1016/j.ijbiomac.2025.142933] [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: 07/20/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
Atherosclerosis (AS) is a chronic vascular disorder that is characterized by the thickening and narrowing of arteries due to the development of atherosclerotic plaques. The traditional risk factors involved in AS are obesity, type 2 diabetes (T2D), dyslipidemia, hypertension, and smoking. Furthermore, non-traditional risk factors for AS, such as inflammation, sleep disturbances, physical inactivity, air pollution, and alterations of gut microbiota, gained attention in relation to the pathogenesis of AS. Interestingly, the pathogenesis of AS, is complex and related to different abnormalities of cellular and sub-cellular signaling pathways. It has been illustrated that AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (MTOR) pathways are involved in AS pathogenesis. Mounting evidence indicated that AMPK plays a critical role in attenuating the development of AS by activating autophagy, which is impaired during atherogenesis. AMPK has a vasculoprotective effect by reducing lipid accumulation, inflammatory cell proliferation, and the release of pro-inflammatory cytokines, as well as decreasing inflammatory cell adhesion to the vascular endothelium. AMPK activation by metformin inhibits the migration of vascular smooth muscle cells (VSMCs) and AS development. However, the MTOR pathway contributes to AS by inhibiting autophagy, highlighting autophagy as a crucial link between the AMPK and MTOR pathways in AS pathogenesis. The MTOR is a key inducer of endothelial dysfunction and is involved in the development of AS. Therefore, both the AMPK and MTOR pathways play a crucial role in the pathogenesis of AS. However, the exact role of AMPK and MTOR pathways in the pathogenesis of AS is not fully clarified. Therefore, this review aims to discuss the potential role of the AMPK/MTOR signaling pathway in AS, and how AMPK activators and MTOR inhibitors influence the development and progression of AS. In conclusion, AMPK activators and MTOR inhibitors have vasculoprotective effects against the development and progression of AS.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ghassan M Sulaiman
- Department of Applied Sciences, University of Technology, Baghdad, Iraq.
| | - Mayyadah H Mohsin
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Retaj A Dawood
- Department of Biology, College of Science, Al-Mustaqbal University, Hilla 51001, Iraq
| | - Ali K Albuhadily
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir ibn Hayyan Medical University, Al-Ameer Qu, PO.Box13 Kufa, Najaf, Iraq
| | | | - Mosleh M Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 255, Bisha 67714, Saudi Arabia
| |
Collapse
|
4
|
Li Y, Luo M, Ye C, Bao H. Recent advancements in dual-locked optical probe for precise imaging of atherosclerosis. Org Biomol Chem 2025; 23:4024-4035. [PMID: 40196887 DOI: 10.1039/d5ob00312a] [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: 04/09/2025]
Abstract
Atherosclerosis (AS) is the primary pathological basis for cardiovascular and cerebrovascular events, making early diagnosis and dynamic risk assessment crucial for reducing the incidence of thrombotic occurrences. Traditional imaging techniques, such as CT, MRI, and PET, provide morphological information about plaques; however, they are limited in their ability to detect the molecular activity characteristics of these plaques, which hampers the assessment of thrombotic risk. In recent years, molecular optical probes have offered new insights into plaque activity detection by targeting specific biomarkers. However, single-target probes often produce false positive signals due to the cross-expression of biomarkers, which limits their clinical application. To overcome this challenge, dual-lock optical probes have been developed, achieving dual-target synergistic activation, which significantly enhances the specificity and signal-to-noise ratio of imaging. This article reviews the targeted imaging strategies and recent advancements in dual-lock optical probes in the context of AS, with a particular emphasis on their application in lipid droplet enrichment, oxidative stress, and specific enzymes. Although the technology still faces challenges regarding sensitivity, specificity, and multi-target design, its potential for future development is substantial. Through interdisciplinary collaboration and technological innovation, dual-lock optical probes are poised to evolve from 'imaging tools' to 'integrated diagnosis and treatment platforms', thereby advancing the precise diagnosis and treatment of complex diseases.
Collapse
Affiliation(s)
- Yulong Li
- College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350117, P. R. of China
- Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, State Key Laboratory of Structural Chemistry, Center for Excellence in Molecular Synthesis, Chinese Academy of Sciences, 155 Yangqiao Road West, Fuzhou 350002, Fujian, P. R. of China.
- Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, P. R. of China
| | - Mengyao Luo
- College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350117, P. R. of China
- Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, State Key Laboratory of Structural Chemistry, Center for Excellence in Molecular Synthesis, Chinese Academy of Sciences, 155 Yangqiao Road West, Fuzhou 350002, Fujian, P. R. of China.
- Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, P. R. of China
| | - Changqing Ye
- Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, State Key Laboratory of Structural Chemistry, Center for Excellence in Molecular Synthesis, Chinese Academy of Sciences, 155 Yangqiao Road West, Fuzhou 350002, Fujian, P. R. of China.
| | - Hongli Bao
- Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, State Key Laboratory of Structural Chemistry, Center for Excellence in Molecular Synthesis, Chinese Academy of Sciences, 155 Yangqiao Road West, Fuzhou 350002, Fujian, P. R. of China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. of China
- Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, P. R. of China
| |
Collapse
|
5
|
Chen H, Zhang Y, Aikebaier M, Du Y, Liu Y, Zha Q, Zheng L, Shan S, Wang Y, Chen J, Li Y, Yang K, Yang Y, Cui W. Decursin-Loaded Nanovesicles Target Macrophages Driven by the Pathological Process of Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417489. [PMID: 40285666 DOI: 10.1002/advs.202417489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/12/2025] [Indexed: 04/29/2025]
Abstract
Atherosclerosis (AS) is a major pathological factor contributing to the mortality associated with ischemic heart disease and is driven primarily by macrophage-mediated lipid accumulation and inflammatory processes. Conventional cardiovascular pharmacotherapies address these pathological mechanisms but often show limited efficacy, highlighting the need for innovative agents capable of effectively reducing lipid accumulation and inflammation with minimal toxicity. In this study, decursin, a monomer derived from traditional Chinese medicine, is shown to inhibit both lipid accumulation and inflammatory responses in macrophages through direct interaction with protein kinase Cδ (PKCδ), resulting in low cytotoxicity in vitro and negligible toxicity in vivo. To address the short half-life of decursin, a targeted cascade drug delivery system (ALD@EM), which is specifically designed to target AS pathophysiology, is developed. This system employs ICAM-1 and VCAM-1 antibodies for plaque localization and incorporates low-density lipoproteins (LDLs) to facilitate chemotaxis to lesion sites, with an inner layer of apoptotic endothelial cell membranes to increase macrophage internalization and drug release. As a result, ALD@EM nanovesicles significantly increased the accumulation and therapeutic efficacy of decursin within plaques, substantially reducing lipid deposition and plaque inflammation, thereby offering a novel strategy for targeted AS treatment.
Collapse
Affiliation(s)
- Hui Chen
- Department of Endocrinology, The Affiliated Hospital of Yunnan University, Kunming, Yunnan, 650021, China
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yifeng Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mirenuer Aikebaier
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yawei Du
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200002, China
| | - Yan Liu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Qing Zha
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Lan Zheng
- Department of Traditional Chinese Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shuyao Shan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanping Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jiawei Chen
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yiping Li
- Department of Endocrinology, The Affiliated Hospital of Yunnan University, Kunming, Yunnan, 650021, China
| | - Ke Yang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Yang
- Department of Endocrinology, The Affiliated Hospital of Yunnan University, Kunming, Yunnan, 650021, China
| | - Wenguo Cui
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
6
|
Liang J, Xie Y, Li P, Li H, Li P, Huang Z, Liu G, Zhong Y, Li B, Zhang J, Wen J. The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio and its combination with obesity indicators as a predictor of all cause and cardiovascular mortality in non-diabetic individuals. BMC Public Health 2025; 25:1513. [PMID: 40269817 PMCID: PMC12016409 DOI: 10.1186/s12889-025-22789-y] [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: 01/10/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) represents a novel composite lipid marker for atherosclerosis and cardiovascular disease (CVD). Nevertheless, the correlation between NHHR and mortality in the non-diabetic population remains indistinct. METHODS This study included 20,774 non-diabetic individuals from the 1999-2018 National Health and Nutrition Examination Survey (NHANES). We employed a weighted multivariate Cox proportional hazards model and restricted cubic splines to assess the associations between NHHR, its combination with obesity indicators, and all-cause and CVD mortality. RESULTS During a mean follow-up period of 62 months, a total of 897 participant deaths were recorded, of which 155 were attributed to cardiovascular causes. The restricted cubic splines revealed a U-shaped association between NHHR and all-cause mortality, while an L-shaped association was observed for CVD mortality. The analysis of threshold efects revealed that the infection points for NHHR and all-cause and CVD mortality were 2.65 and 2.07, respectively. The cubic spline revealed a nonlinear correlation was observed between NHHR-BMI, NHHR-WC and NHHR-WHtR and all-cause and CVD mortality. CONCLUSION NHHR and its combination with obesity indicators can be a meaningful predictor of all-cause mortality and CVD mortality in non-diabetic individuals.
Collapse
Affiliation(s)
- Jiahua Liang
- Meizhou Hospital of Guangzhou University of Chinese Medicine, Meizhou, Guangdong, China
| | - Yuxin Xie
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Peilin Li
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huamei Li
- Meizhou Hospital of Guangzhou University of Chinese Medicine, Meizhou, Guangdong, China
| | - Ping Li
- Meizhou Hospital of Guangzhou University of Chinese Medicine, Meizhou, Guangdong, China
| | - Zhihua Huang
- Meizhou Hospital of Guangzhou University of Chinese Medicine, Meizhou, Guangdong, China
| | - Guangjiao Liu
- Meizhou Hospital of Guangzhou University of Chinese Medicine, Meizhou, Guangdong, China
| | - Yueqiao Zhong
- Meizhou Hospital of Guangzhou University of Chinese Medicine, Meizhou, Guangdong, China
| | - Bin Li
- Meizhou Hospital of Guangzhou University of Chinese Medicine, Meizhou, Guangdong, China
| | - Jialing Zhang
- Meizhou Hospital of Guangzhou University of Chinese Medicine, Meizhou, Guangdong, China
| | - Junmao Wen
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Jichang Road 16#, District Baiyun, Guangzhou, 510405, Guangdong, China.
| |
Collapse
|
7
|
Wang H, Li Y, Zhang L, Lu M, Li C, Li Y. Anti-Inflammatory Lipid Mediators from Polyunsaturated Fatty Acids: Insights into their Role in Atherosclerosis Microenvironments. Curr Atheroscler Rep 2025; 27:48. [PMID: 40198469 DOI: 10.1007/s11883-025-01285-z] [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] [Accepted: 03/05/2025] [Indexed: 04/10/2025]
Abstract
PURPOSE OF REVIEW Inflammation has become a major residual risk factor for atherosclerotic cardiovascular disease (ASCVD). Certain lipid mediators, known as specialized proresolving mediators (SPMs), are mainly derived from polyunsaturated fatty acids (PUFAs) and can promote inflammation resolution while maintaining host autoimmunity. This review investigates the synthesis and ligand action pathways of these lipid mediators, as well as their regulatory mechanisms in the microenvironment of atherosclerotic plaques. Furthermore, it explores their clinical therapeutic potential, aiming to offer new insights into novel anti-inflammatory drug targets for the treatment of ASCVD. RECENT FINDINGS Reduced levels of SPMs are associated with the progression of atherosclerosis. SPMs inhibit inflammatory responses in the plaque microenvironment by limiting immune cell infiltration, reducing oxidative stress, and promoting the clearance of apoptotic cells, all of which contribute to plaque stabilization. Tyrosine-protein kinase Mer (MerTK), TRIF-related adaptor molecule (TRAM), and high mobility group box 1 (HMGB1) play crucial roles in the modulation of SPM production. Clinical use of ω-3 PUFAs has been shown to reduce the incidence of fatal cardiovascular events. Furthermore, aspirin not only initiates the synthesis of specific SPMs but also extends their activity within the body. The enhanced production of SPMs promotes inflammation resolution in the plaque microenvironment without inducing immunosuppression. This characteristic highlights MerTK, TRAM, and HMGB1 as potential targets for the development of anti-inflammatory drugs. Investigating targets and compounds that enhance the production of SPMs presents a promising strategy for developing future anti-inflammatory agents.
Collapse
Affiliation(s)
- Hongqin Wang
- Post-doctoral Mobile Station of Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- , Daxue Road No. 4655, Changqing District, Jinan, Shandong Province, People's Republic of China
| | - Yuan Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- , Daxue Road No. 4655, Changqing District, Jinan, Shandong Province, People's Republic of China
| | - Lei Zhang
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- , Daxue Road No. 4655, Changqing District, Jinan, Shandong Province, People's Republic of China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- , Daxue Road No. 4655, Changqing District, Jinan, Shandong Province, People's Republic of China.
| | - Yunlun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
8
|
Wu M, Chen M, Zhao Y, Zhang X, Ding X, Yuan J, Shi J, Yu W, Zhu H. Neutrophil Hitchhiking-Mediated Delivery of ROS-Scavenging Biomimetic Nanoparticles for Enhanced Treatment of Atherosclerosis. SMALL METHODS 2025:e2402019. [PMID: 40109147 DOI: 10.1002/smtd.202402019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/01/2025] [Indexed: 03/22/2025]
Abstract
Atherosclerosis (AS), a chronic inflammatory disease and a leading cause of cardiovascular morbidity and mortality worldwide, is a significant contributor to disability. Neutrophil extracellular traps (NETs) have been closely associated with the progression of AS and plaque vulnerability. However, developing a treatment strategy that specifically targets neutrophils and effectively reduces NET release at the lesion site remains a major challenge. In this study, a biomimetic nanosystem with neutrophil-targeting properties is engineered. Coating Prussian blue nanoparticles with bacterial biomimetic membranes (MPB NPs) enables specific recognition and internalization by neutrophils. By hitching onto neutrophils, the MPB NPs scavenge intracellular reactive oxygen species (ROS) and suppress NET formation at the lesion site. Importantly, MPB NPs reduce the size of atherosclerotic plaques by 3.29-fold, from 22.53% to 6.85%, stabilize the plaques, and halt their progression in atherosclerotic mouse models. These findings suggest that MPB NPs offer a promising therapeutic strategy for atherosclerosis, and provide a versatile platform for the treatment of NET-associated diseases.
Collapse
Affiliation(s)
- Ming Wu
- Department of Ultrasonography, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Mengjuan Chen
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuzhen Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xijun Zhang
- Department of Ultrasonography, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Xiao Ding
- Department of Ultrasonography, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Jianjun Yuan
- Department of Ultrasonography, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenyan Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Haohui Zhu
- Department of Ultrasonography, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| |
Collapse
|
9
|
Fan B, Hong J, Wu Q, Shen W, Hu N, Xing Y, Zhang J, Cai W, Zhang R. Matrix metalloproteinase-responsive melanin nanoparticles utilize live neutrophils for targeted high-risk plaque detection and atherosclerosis regression. Acta Biomater 2025; 195:496-508. [PMID: 39956306 DOI: 10.1016/j.actbio.2025.02.033] [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: 11/06/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
Abrupt rupture of atherosclerotic plaque is the predominant contributor to acute cardiovascular events. It is of clinical importance to effectively identify and inhibit high-risk plaque progression. However, this remains a major challenge due to the inadequate targeting of theranostic agents to atherosclerotic lesions. Herein, we utilize live neutrophils to encapsulate melanin-based theranostics (termed MNPpep-Gd) to enhance their plaque targeting, leveraging the inherent inflammatory tropism of neutrophils in atherosclerosis progression. The MNPpep-Gd are fabricated using the water-insoluble gadolinium-chelated melanin nanoparticle modified with a detachable polyethylene glycol (PEG) segment via a matrix metalloproteinase (MMP)-cleavable peptide linker. Our work demonstrated that overexpressed MMP in high-risk plaques can induce an increase in particle size and prolonged retention time of the MNPpep-Gd nanoprobe in lesions, making it a highly efficient contrast agent for magnetic resonance (MR) and photoacoustic (PA) dual-modal imaging atherosclerotic plaque. Concurrently, the melanin nanoparticles function as a therapeutic agent by scavenging multiple toxic reactive oxygen species (ROS), inhibiting the pro-inflammatory cytokines expression, and significantly reducing the foam cell formation. As a result, NE/MNPpep remarkably alleviates atherosclerosis progression by a 24.7 % reduction for plaque area in ApoE-/- mice. Immunohistochemical analysis confirmed that NE/MNPpep treatment significantly reduced the macrophage content by 21.3 % and lipid burden by 15.6 % in plaques. In conclusion, our innovative nanoagent actively targets atherosclerotic sites, offers a noninvasive approach for identifying high-risk atherosclerotic plaques, and significantly contributes to the alleviation of lesion development in ApoE-/- mice. STATEMENT OF SIGNIFICANCE: Effective identification and inhibition of high-risk plaque progression hold clinical importance. However, it remains a major challenge due to the insufficient targeting of theranostic agents to plaques. Herein, a biomimetic nanoplatform is developed to actively target atherosclerosis plaque with the assistance of neutrophils, thereby minimizing off-target effects. Then, overexpressed MMP2 in high-risk plaques trigger the aggregation of hydrophobic Gd3+-labeled melanin nanoparticles, enhancing both MRI/PAI intensities for precise diagnosis. Additionally, the native antioxidant activity of melanin reduces inflammatory level, alleviates oxidative damage, and inhibits plaque progression in ApoE-/- mice. This study offers valuable insights for accurate plaque assessment and provides effective guidance for subsequent management strategies.
Collapse
Affiliation(s)
- Bo Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China; School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, 030001, China.
| | - Jie Hong
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Qian Wu
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Weiguang Shen
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Nan Hu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yang Xing
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Juan Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Wenwen Cai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital, Fifth Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
10
|
Cheng Q, Sun J, Wang B, Ding A, Sun W, Yang F, Zhang J. Detecting mitochondrial hypochlorous acid and viscosity in atherosclerosis models via NIR fluorescent probes. Bioorg Chem 2025; 156:108191. [PMID: 39848167 DOI: 10.1016/j.bioorg.2025.108191] [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: 10/09/2024] [Revised: 12/22/2024] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
The assessment of early atherosclerosis (AS) via fluorescence imaging is crucial for advancing early diagnosis research. Abnormal inflammation biomarkers, including hypochlorous acid (HClO) and viscosity within mitochondria, have been closely linked to the pathogenesis of AS. However, current fluorescent probes predominantly rely on unimodal imaging that targets a single biomarker and lacks mitochondrial specificity, which can result in potential false signal readouts due to the complex intracellular environment. Herein, we report the development of three fluorescence probes (M-1, M-2, and M-3) designed to achieve simultaneous detection and imaging of mitochondrial HClO and viscosity. Spectroscopic analysis revealed these probes exhibit high specificity towards HClO and viscosity, superior sensitivity at sub-nanomolar levels, and rapid response within 2 min. Among them, M-2 demonstrated the best performance in terms of fluorescence emission (extending into near-infrared region) and sensitivity (as low as 0.072 nM), and was selected for biological evaluation. When applied to living cells, the probe M-2 demonstrated high preferential accumulation within mitochondria and a bimodal off-on fluorescence response, allowing the evaluation of mitochondrial HClO and viscosity levels using dual-channel microscopy. With this probe, we effectively monitored abnormal HClO and viscosity levels in early AS mice, providing a highly sensitive and precise tool for early AS diagnosis.
Collapse
Affiliation(s)
- Quanli Cheng
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Jian Sun
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Baoguo Wang
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Aixiang Ding
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China
| | - Wan Sun
- Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou 253023, China
| | - Fan Yang
- Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou 253023, China
| | - Jian Zhang
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
11
|
Ji M, Dong J, Ye Z, Kang J, Han G, Hong X, Wei Y, Chen X, Sun W, Liu D. Endogenous HClO-Gated Cascade MicroRNA Imaging for Precise Diagnosis of Atherosclerosis In Vivo. J Am Chem Soc 2025; 147:7113-7126. [PMID: 39932706 DOI: 10.1021/jacs.5c00031] [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: 02/27/2025]
Abstract
Precise imaging of atherosclerotic plaques using biomarkers could prompt the diagnosis and clinical management of atherosclerosis (AS)-driven cardiovascular diseases. MicroRNA-155 (miR-155) plays a critical role in AS development, with its expression notably upregulated in foam cells within plaques. However, miRNA imaging methods for atherosclerotic plaques face significant challenges, including low specificity, inefficient delivery, and poor cell selectivity. Herein, we develop an endogenous hypochlorous acid (HClO)-gated cascade signal amplification strategy for precise miR-155 imaging in living foam cells, enabling accurate in vivo and ex vivo detection of atherosclerotic plaques. This strategy utilizes a phosphorothioate (PT)-modified hairpin probe that is specifically deprotected by HClO and uncaged by miR-155, triggering a catalytic hairpin assembly (CHA) to amplify fluorescence signals. The PT-CHA probes are encapsulated in lipid nanoparticles (LNs), followed by conjugating with phosphatidylserine (PS)-binding peptide (PBP) for selectively targeting foam cells, enabling in vivo miR-155 imaging in atherosclerotic plaques. The fluorescence intensity of PT-CHA@LN-PBP in the aorta region shows clear differentiation among AS-bearing mice, miR-155-/- mice, and healthy mice. Moreover, the fluorescence intensity strongly correlates with plaque area and AS progression and can discriminate plaque vulnerability risk with an area under the curve (AUC) of 0.94. Imaging of human aortic tissues further validates the probe's capacity to distinguish atherosclerotic plaques from normal endarterium. These findings establish PT-CHA@LN-PBP as a noninvasive, reliable diagnostic tool for precise assessment of AS.
Collapse
Affiliation(s)
- Moxuan Ji
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jiantong Dong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhuo Ye
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Jingjing Kang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Guimei Han
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaoqin Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yongchun Wei
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xin Chen
- Department of Cardiovascular Medicine, Tianjin First Center Hospital, Tianjin 300192, China
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Dingbin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Centers for Cell Responses and New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
12
|
Qin YS, Yi J, Chen YJ, Zhang W, Tang SF. Recent Advances in Micro/Nanomotor for the Therapy and Diagnosis of Atherosclerosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11443-11468. [PMID: 39648908 DOI: 10.1021/acsami.4c15165] [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: 12/10/2024]
Abstract
Atherosclerotic cardiovascular disease poses a significant global public health threat with a high incidence that can result in severe mortality and disability. The lack of targeted effects from traditional therapeutic drugs on atherosclerosis may cause damage to other organs and tissues, necessitating the need for a more focused approach to address this dilemma. Micro/nanomotors are self-propelled micro/nanoscale devices capable of converting external energy into autonomous movement, which offers advantages in enhancing penetration depth and retention while increasing contact area with abnormal sites, such as atherosclerotic plaque, inflammation, and thrombosis, within blood vessel walls. Recent studies have demonstrated the crucial role micro/nanomotors play in treating atherosclerotic cardiovascular disease. Hence, this review highlights the recent progress of micro/nanomotor technology in atherosclerotic cardiovascular disease, including the effective promotion of micro/nanomotors in the circulatory system, overcoming hemorheological barriers, targeting the atherosclerotic plaque microenvironment, and targeting intracellular drug delivery, to facilitate atherosclerotic plaque localization and therapy. Furthermore, we also describe the potential application of micro/nanomotors in the imaging of vulnerable plaque. Finally, we discuss key challenges and prospects for treating atherosclerotic cardiovascular disease while emphasizing the importance of designing individualized management strategies specific to its causes and microenvironmental factors.
Collapse
Affiliation(s)
- Yu-Sheng Qin
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| | - Juan Yi
- Department of Laboratory Medicine, Liuzhou Traditional Chinese Medical Hospital, The Third Affiliated Hospital of Guangxi University of Chinese Medicine, Liuzhou 545006, China
| | - Yan-Jun Chen
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei Zhang
- Department of Radiology, Liuzhou People's Hospital, Liuzhou 545006, China
| | - Shi-Fu Tang
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| |
Collapse
|
13
|
Chen Y, Pan D, Zhu Q, Lu M, Zhang Y, Gao Z, Zhang L, Yi Y, Liu L, Liu Q, Li S, Shen C, Tang Q, Jiang C. Biomimetic metal-phenolic nanocarrier for co-delivery of multiple phytomedical bioactive components for anti-atherosclerotic therapy. Int J Pharm 2025; 671:125228. [PMID: 39832572 DOI: 10.1016/j.ijpharm.2025.125228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Atherosclerosis, a major cause of cardiovascular diseases, involves complex pathophysiological processes. The co-delivery of multiple bioactive components derived from phytomedicine to atherosclerotic plaque is challenging, especially for those with varied solubilities. This study introduces a novel metal-phenolic network-based core-shell recombinant high-density lipoprotein nanocarrier (SSPH-MPN@rHDL) for co-delivering multiple bioactive components from Salvia miltiorrhiza and Carthamus tinctorius, including salvianic acid A (SAA), salvianolic acid B (SAB), protocatechuic aldehyde (PCA), hydroxysafflor yellow A (HSYA), and tanshinone IIA (TS-IIA). These components have varied solubilities, presenting challenges for achieving synergistic therapeutic effects. The SSPH-MPN@rHDL system encapsulates the four hydrophilic components (i.e. SAA, SAB, PCA, HSYA) within a quaternary metal-phenolic network and a hydrophobic component (i.e. TS-IIA) in an outer lipid layer, facilitating targeted plaque delivery. In vitro and in vivo experimental results demonstrated that SSPH-MPN@rHDL enhanced anti-atherosclerotic efficacy through combined antioxidant, anti-inflammatory, and lipid-lowering actions. This approach offers new perspectives on using nanotechnology to optimize the delivery of phytomedicinal compounds for cardiovascular therapy.
Collapse
Affiliation(s)
- Yao Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515 China
| | - Dongmei Pan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515 China
| | - Qinglan Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515 China
| | - Meiting Lu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515 China
| | - Ying Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China
| | - Ziting Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China
| | - Lu Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515 China
| | - Yankui Yi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515 China
| | - Li Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515 China
| | - Qiang Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515 China
| | - Shasha Li
- College of Pharmacy, Xinjiang Medical University, Urumqi, People's Republic of China
| | - Chunyan Shen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515 China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515 China.
| | - Qingfa Tang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515 China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515 China.
| | - Cuiping Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515 China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515 China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515 China.
| |
Collapse
|
14
|
Chen L, Shi Y, Xiao D, Huang Y, Jiang Y, Liang M, Liang F, Xue J, Chen H, Liu Z, Wang X, Zhuang F, Zhou G, Huo H, Cai Z, Shao Q, He B. NR4A1 deficiency promotes carotid plaque vulnerability by activating integrated stress response via targeting Bcat1. Cell Mol Life Sci 2025; 82:91. [PMID: 39985585 PMCID: PMC11846829 DOI: 10.1007/s00018-025-05602-2] [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: 11/14/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/24/2025]
Abstract
Rupture of vulnerable carotid atherosclerotic plaque is one of the leading causes of ischemic stroke. However, the mechanisms driving the transition from stable to vulnerable plaques have not yet been elucidated. NR4A1 is an orphan nuclear receptor that functions in various inflammatory diseases. To explore the role of NR4A1 in vulnerable plaque formation, we generated a vulnerable plaque mouse model by combining partial ligation of the left common carotid artery and left renal artery in ApoE-/- and ApoE-/-;NR4A1-/- mice. Our research revealed that NR4A1 deficiency significantly worsened the pathology of vulnerable plaque, increasing intraplaque hemorrhage, rupture with thrombus, and the occurrence of multilayer with discontinuity. Moreover, NR4A1 deficiency exacerbated macrophage infiltration, inflammation, and oxidative stress. Mechanistically, we identified Bcat1 as the target of NR4A1. NR4A1 modulated the integrated stress response (ISR) in macrophages by transcriptionally inhibiting Bcat1, thus influencing the progression of vulnerable plaque. ISR inhibitor GSK2606414 or Bcat1 inhibitor ERG240 significantly ameliorated atherosclerotic plaque formation and increased plaque stability. Notably, supplementation with Celastrol, an herbal extract, stabilized atherosclerotic plaques in mice. These findings suggest that NR4A1 deficiency exacerbates vulnerable plaque by activating ISR via targeting Bcat1. The NR4A1/Bcat1/ISR axis is therefore an important therapeutic target for stabilizing atherosclerotic plaque.
Collapse
MESH Headings
- Animals
- Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Plaque, Atherosclerotic/pathology
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/genetics
- Mice
- Mice, Inbred C57BL
- Macrophages/metabolism
- Disease Models, Animal
- Mice, Knockout
- Oxidative Stress
- Male
- Humans
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Inflammation/pathology
Collapse
Affiliation(s)
- Long Chen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yiping Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Danrui Xiao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yijie Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yangjing Jiang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Min Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Feng Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Jieyuan Xue
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Haiping Chen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Zhitong Liu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xia Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Fei Zhuang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Guo Zhou
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Huanhuan Huo
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Qin Shao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
15
|
Huang X, Zhou Y, Guo Y, Yan D, Sun P, Cao Y, Chen Y, Peng J. Selenium-Doped Copper Formate Nanozymes with Antisenescence and Oxidative Stress Reduction for Atherosclerosis Treatment. NANO LETTERS 2025; 25:2662-2669. [PMID: 39819038 DOI: 10.1021/acs.nanolett.4c05348] [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: 01/19/2025]
Abstract
Atherosclerosis, resulting from chronic inflammation of the arterial wall, serves as the underlying cause of multiple major cardiovascular diseases. Current anti-inflammatory therapies often exhibit limited and unsatisfactory efficacy. To address this, we have designed a selenium-doped copper formate (Cuf-Se) nanozyme for the treatment of atherosclerosis, which possesses superoxide dismutase (SOD) and glutathione peroxidase (GPx)-like activities. The Cuf-Se can efficiently scavenge reactive oxygen species (ROS), inhibit cellular senescence, and prevent the formation of foam cells. It acts on macrophages to reduce cellular ROS levels and lipid oxidation, thereby significantly inhibiting inflammation-related processes. Notably, while inhibiting foam cell formation, Cuf-Se also alleviates endothelial cells senescence. After intravenous administration, Cuf-Se effectively inhibits the formation of atherosclerosis in mice through the synergistic effects of antisenescence and antioxidant properties, reducing plaque area by approximately 5-fold. This study provides an effective strategy for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xiao Huang
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yue Zhou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Yukang Guo
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Dan Yan
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ping Sun
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Yongbin Cao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Yuanyuan Chen
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Jian Peng
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| |
Collapse
|
16
|
Sebatana R, Kudzai KD, Magura A, Mdlophane A, Zeevaart JR, Sathekge M, Kahts M, Mdanda S, Witika BA. An Insight to Nanoliposomes as Smart Radiopharmaceutical Delivery Tools for Imaging Atherosclerotic Plaques: Positron Emission Tomography Applications. Pharmaceutics 2025; 17:240. [PMID: 40006607 PMCID: PMC11858949 DOI: 10.3390/pharmaceutics17020240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/27/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Atherosclerosis is a chronic progressive disease which is known to cause acute cardiovascular events as well as cerebrovascular events with high mortality. Unlike many other diseases, atherosclerosis is often diagnosed only after an acute or fatal event. At present, the clinical problems of atherosclerosis mainly involve the difficulty in confirming the plaques or identifying the stability of the plaques in the early phase. In recent years, the development of nanotechnology has come with various advantages including non-invasive imaging enhancement, which can be studied for the imaging of atherosclerosis. For targeted imaging and atherosclerosis treatment, nanoliposomes provide enhanced stability, drug administration, extended circulation, and less toxicity. This review discusses the current advances in the development of tailored liposomal nano-radiopharmaceutical-based techniques and their applications to atherosclerotic plaque diagnosis. This review further highlights liposomal nano-radiopharmaceutical localisation and biodistribution-key processes in the pathophysiology of atherosclerosis. Finally, this review discusses the direction and future of liposomal nano-radiopharmaceuticals as a potential clinical tool for the assessment and diagnosis of atherosclerotic plaque.
Collapse
Affiliation(s)
- Reabetswe Sebatana
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
| | - Kahwenga D. Kudzai
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| | - Allan Magura
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| | - Amanda Mdlophane
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
| | - Jan Rijn Zeevaart
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
- Radiochemistry, The South African Nuclear Energy Corporation (Necsa) SOC Ltd., Pelindaba 0240, South Africa
| | - Mike Sathekge
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
| | - Maryke Kahts
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| | - Sipho Mdanda
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
| | - Bwalya Angel Witika
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| |
Collapse
|
17
|
Chen Q, Yang X, Yu Y, Duan X, Ni R, Song G, Zhu L, Zhong Y, Qu K, Qin X, Zhang K, Luo Y, Wu W. Biomimetic Cerium-Assisted Supra-Carbon Dots Assembly for Reactive Oxygen Species-Activated Atherosclerosis Theranostic. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408980. [PMID: 39777854 DOI: 10.1002/smll.202408980] [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: 10/01/2024] [Revised: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Theranostic applications in atherosclerosis plaque microenvironment-triggered nanoplatforms are significantly compromised by the complex synthesis procedure, non-specific distribution, and limited therapeutic function. Therefore, development of a facile and feasible method to construct a pathology-based stimuli-responsive nanoplatform with satisfactory theranostic performance remains a demanding and highly anticipated goal. Herein, a novel class of multifunctional supra-carbon dots (CDs), denoted as MM@Ce-CDs NPs, by a simple nanoassembly and a subsequent coating with macrophage membrane (MM), is developed for the targeted reactive oxygen species-trigged theranostic and positive regulation of the pathological plaque microenvironment in AS. The harvested MM@Ce-CDs NPs exhibit activatable fluorescence properties, photoacoustic characteristics, and cascade enzyme performances, which can be effectively activated under ROS stimulation in the plaque pathological microenvironment, enabling precise control over theranostic functions, while markedly enhancing diagnostic accuracy and therapeutic efficacy for AS management. Besides, MM@Ce-CDs NPs can effectively manipulate the plaque microenvironment by reducing ROS levels and inflammation, alleviating M1 macrophage infiltration, and inhibiting foam cell formation, all together suppressing the pathological plaque development through the synergistic mechanisms. In addition, MM@Ce-CDs NPs inherit the biomimetic biological functions from MM, facilitating a highly specific target delivery to AS.
Collapse
Affiliation(s)
- Qiao Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing University Three Gorges Hospital, Chongqing, 400044, China
| | - Xu Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing University Three Gorges Hospital, Chongqing, 400044, China
| | - Yao Yu
- Thyroid Breast Surgery Department, Dazhou Central Hospital, Dazhou, 635000, China
| | - Xinmei Duan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing University Three Gorges Hospital, Chongqing, 400044, China
| | - Rongrong Ni
- Medical Department, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Guojing Song
- Urology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Li Zhu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing University Three Gorges Hospital, Chongqing, 400044, China
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing University Three Gorges Hospital, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing University Three Gorges Hospital, Chongqing, 400044, China
| | - Xian Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing University Three Gorges Hospital, Chongqing, 400044, China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing University Three Gorges Hospital, Chongqing, 400044, China
| | - Yang Luo
- JinFeng Laboratory, Chongqing, 401329, China
- Department of Laboratory Medicine, Chongqing General Hospital, School of Medicine, Chongqing University, Chongqing, 400044, China
- College of Life Science and Laboratory Medicine, Kunming Medical University, Kunming, Yunnan, 650050, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing University Three Gorges Hospital, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| |
Collapse
|
18
|
Pan W, Shao S, Cao X, Dai X, Zheng Y, Cheng J, Feng W, Wu R, Chen Y. In Situ Conversion of Atherosclerotic Plaques' Iron into Nanotheranostics. J Am Chem Soc 2025; 147:3553-3569. [PMID: 39836090 DOI: 10.1021/jacs.4c15068] [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: 01/22/2025]
Abstract
The presence of a substantial necrotic core in atherosclerotic plaques markedly heightens the risk of rupture, a consequence of elevated iron levels that exacerbate oxidative stress and lipid peroxidation, thereby sustaining a detrimental cycle of ferroptosis and inflammation. Concurrently targeting both ferroptosis and inflammation is crucial for the effective treatment of vulnerable plaques. In this study, we introduce gallium hexacyanoferrate nanoabsorption catalysts (GaHCF NACs) designed to disrupt this pathological cycle. GaHCF NACs function as highly efficient iron chelators with robust antiferroptosis properties. Through in situ capture of iron within atherosclerotic plaques, these catalysts enhance reactive oxygen species scavenging, initiating an amplified therapeutic response. GaHCF NACs significantly advance plaque regression, stabilization, and vascular functional recovery by inhibiting MAPK13 (p38-δ MAPK) signaling, a key mediator of inflammation and cell death. Importantly, the in situ iron capture process generates a detectable photoacoustic signal, offering a notable diagnostic advantage that allows real-time monitoring of plague status. This multifunctional nanocatalytic platform in situ transforms toxic iron within atherosclerotic plaques into both a therapeutic and diagnostic agent, adapting dynamically to the microenvironment and representing a promising strategy for reducing plaque vulnerability and preventing rupture.
Collapse
Affiliation(s)
- Wenqi Pan
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080 Shanghai, P. R. China
| | - Sihui Shao
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080 Shanghai, P. R. China
| | - Xinyue Cao
- Materdicine Lab, School of Life Sciences, Shanghai University, 200444 Shanghai, P. R. China
| | - Xinyue Dai
- Materdicine Lab, School of Life Sciences, Shanghai University, 200444 Shanghai, P. R. China
| | - Yi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080 Shanghai, P. R. China
| | - Jingyun Cheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080 Shanghai, P. R. China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, 200444 Shanghai, P. R. China
| | - Rong Wu
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080 Shanghai, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, 200444 Shanghai, P. R. China
| |
Collapse
|
19
|
Tang C, Wang H, Guo L, Cui Y, Zou C, Hu J, Zhang H, Yang G, Zhou W. Multifunctional Nanomedicine for Targeted Atherosclerosis Therapy: Activating Plaque Clearance Cascade and Suppressing Inflammation. ACS NANO 2025; 19:3339-3361. [PMID: 39812806 DOI: 10.1021/acsnano.4c12131] [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: 01/16/2025]
Abstract
Atherosclerosis (AS) is a prevalent inflammatory vascular disease characterized by plaque formation, primarily composed of foam cells laden with lipids. Despite lipid-lowering therapies, effective plaque clearance remains challenging due to the overexpression of the CD47 molecule on apoptotic foam cells, inhibiting macrophage-mediated cellular efferocytosis and plaque resolution. Moreover, AS lesions are often associated with severe inflammation and oxidative stress, exacerbating disease progression. Herein, we introduce a multifunctional nanomedicine (CEZP) targeting AS pathogenesis via a "cell efferocytosis-lipid degradation-cholesterol efflux" paradigm, with additional anti-inflammatory properties. CEZP comprises poly(lactic-co-glycolic acid) nanoparticles encapsulated within a metal-organic framework shell coordinated with zinc ions (Zn2+) and epigallocatechin gallate (EGCG), enabling CpG encapsulation. Upon intravenous administration, CEZP accumulates at AS plaque sites, facilitating macrophage uptake and orchestrating AS treatment through synergistic mechanisms. CpG enhances cellular efferocytosis, Zn2+ promotes intracellular lipid degradation, and EGCG upregulates adenosine 5'-triphosphate-binding cassette transporters for cholesterol efflux while also exhibiting antioxidant and anti-inflammatory effects. In vivo validation confirms CEZP's ability to stabilize plaques, reduce lipid burden, and modulate the macrophage phenotype. Moreover, CEZP is excreted from the body without safety concerns, offering a low-toxicity nonsurgical strategy for AS plaque eradication.
Collapse
Affiliation(s)
- Cui Tang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Hui Wang
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Lina Guo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, Beijing 100034, China
| | - Chan Zou
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Jianming Hu
- First Department of Pathology, Affiliated Hospital, Shihezi University, Xinjiang Uygur Autonomous Region, Shihezi City 832002, China
| | - Hanyong Zhang
- Academician Workstation, Changsha Medical University, Changsha 410219, China
| | - Guoping Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- National-Local Joint Engineering Laboratory of Drug Clinical Evaluation Technology, Changsha, Hunan 410000, China
- Hunan Engineering Research Center for Optimization of Drug Formulation and Early Clinical Evaluation, Changsha, Hunan 410013, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
- Academician Workstation, Changsha Medical University, Changsha 410219, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Affiliated Hospital, Shihezi University, Shihezi City, Xinjiang 832002, China
| |
Collapse
|
20
|
Wang J, Su S, Zheng H, Zhang C, Chen W, Zhang S, Xiao Q, Sheng J, Yang L. Rational Development of a Lipid Droplets and Hypochlorous Acid In-Sequence Responsive Fluorescent Probe for Accurate Imaging of Atherosclerotic Plaques. Anal Chem 2025; 97:758-767. [PMID: 39723762 DOI: 10.1021/acs.analchem.4c05265] [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: 12/28/2024]
Abstract
To answer the call for effective and timely intervention in cardiovascular diseases (CVDs), the development of fluorescent probes that can precisely identify atherosclerotic plaques, the root cause of various fatal CVDs, is highly desirable but remains a great challenge. Herein, by integrating bis(trifluoromethyl)benzyl and phenothiazine into the coumarin matrix, a robust fluorescent probe, NOR1, has been developed. NOR1 responds sequentially to lipid droplets (LDs) and HClO via fluorescence turn-on and ratiometric readouts, respectively, with a fast response rate (within 70 s for LDs and 80 s for HClO), excellent sensitivity (detection limit: 0.41 μg/mL for LDs and 23.38 nM for HClO), and high selectivity. Based on these impressive features, NOR1 was successfully applied to discriminate foam cells from others by simultaneously monitoring two hallmark events, lipid accumulation and oxidative stress, in foam cells. Furthermore, the use of NOR1 to monitor in real time the transformation process of A7r5 cells into foam cells under high LDL/glucose conditions was successfully realized for the first time. Importantly, we further demonstrate the ability of NOR1 to precisely identify atherosclerotic plaques with clear margin delineation, highlighting its potential utility in elucidating the pathological mechanism and clinical diagnosis of atherosclerosis.
Collapse
Affiliation(s)
- Jingshuai Wang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, China
| | - Shuxing Su
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, China
| | - Hongyong Zheng
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, China
| | - Chi Zhang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, China
| | - Wenqiang Chen
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, China
| | - Siqi Zhang
- School of Pharmaceutical and Chemical Engineering, Taizhou University, Jiaojiang, Taizhou 318000, Zhejiang, China
| | - Qi Xiao
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, China
| | - Jiarong Sheng
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, China
| | - Lei Yang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, China
| |
Collapse
|
21
|
Yang F, Huangfu N, Shen J, Ying J, Mao H, Yin Y, Su P, Zhao R, Chen X, Zhu H, He W, Cui H, Wang K. Multifunctional Optical Sensor for the Comprehensive Detection of Zinc Ions in Cardiovascular Disease. ACS Sens 2024; 9:6824-6832. [PMID: 39601238 DOI: 10.1021/acssensors.4c02672] [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: 11/29/2024]
Abstract
Cardiovascular diseases (CVDs) are a major global health concern, highlighting the need for effective diagnostic tools. Zinc ions (Zn2+) play a role in CVDs, but their detection is challenging. This study presents a multifunctional optical sensor, HD-Zn, designed to detect Zn2+ in relation to CVDs. We developed a novel fluorescence probe, HD-Zn, by conjugating N,N-di(2-picolyl)ethylenediamine (DPEN) to HD via an amide bond, which results in fluorescence quenching due to photoinduced electron transfer (PeT). Adding Zn2+ significantly increased fluorescence intensity in the near-infrared region (NIR-I). The probe showed a linear response to varying Zn2+ concentrations, with a detection limit of 9.8 nM, appropriate for physiological conditions. Fluorescence imaging in RAW264.7 macrophages indicated lower intracellular Zn2+ levels in foam cells compared to healthy cells, linked to CVDsprogression. In vivo imaging in mouse models showed decreased fluorescence intensity in the aorta with disease progression. Our findings confirm that HD-Zn is a reliable tool for measuring Zn2+ levels in plaques and demonstrate its biosafety for detecting Zn2+ in serum and urine, offering potential for clinical applications in CVDs diagnosis and monitoring.
Collapse
Affiliation(s)
- Fangkun Yang
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
- Key Laboratory of Precision Prevention and Treatment for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315211, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Zhejiang 315211, China
| | - Ning Huangfu
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
- Key Laboratory of Precision Prevention and Treatment for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315211, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Zhejiang 315211, China
| | - Jiaxi Shen
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Jiajun Ying
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Hengyi Mao
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Yongfang Yin
- Department of Gastroenterology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Pengpeng Su
- School of Medicine, Wenzhou Medical University, Zhejiang 325035, China
| | - Ruochi Zhao
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Xiao Chen
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Hailiang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| | - Wenming He
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
- Key Laboratory of Precision Prevention and Treatment for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315211, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Zhejiang 315211, China
| | - Hanbin Cui
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
- Key Laboratory of Precision Prevention and Treatment for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315211, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Zhejiang 315211, China
| | - Kai Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| |
Collapse
|
22
|
Yang F, Cao J, Jia X, Zhu H, Zhou J, Liu Z, Cui H, Wang K. Novel Approach for Biosensor-Based Imaging of Cysteine Levels in Ischemeic Heart Disease: Insights from Preclinical Models and Human Samples. ACS Sens 2024; 9:6815-6823. [PMID: 39591537 DOI: 10.1021/acssensors.4c02643] [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: 11/28/2024]
Abstract
Cardiovascular diseases (CVDs) pose a serious threat to human health, with atherosclerosis being a leading cause of heart disease and stroke. Elevated cysteine (Cys) levels have been closely linked to an increased risk of cardiovascular diseases, underscoring its significance in cardiovascular health. However, current detection methods for cysteine in serum and atherosclerotic plaques present challenges in sensitivity, specificity, dynamic monitoring, and invasiveness. The development of more sensitive, specific, and noninvasive assays is needed to enable accurate monitoring of cysteine levels. This study introduces the development and characterization of Cys-NPs, a sensitive and selective tool for imaging cysteine in foam cells and atherosclerotic mice. Encapsulation of the HD-probe using DSPE-PEG to obtain Cys-NPs effectively reduced interference from glutathione (GSH), leading to successful preparation and validation of Cys-NPs's nanoscale structure. At the same time, Cys-NPs was able to use the differences in Hcy and Cys concentrations in vivo to better assess Cys levels in vivo. In vitro and in vivo studies demonstrated Cys-NPs's effective imaging of cysteine in foam cells and atherosclerotic mice, highlighting its potential for noninvasive assessment of cysteine levels in ischemic heart disease research and clinical practice.
Collapse
Affiliation(s)
- Fangkun Yang
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
- Key Laboratory of Precision Prevention and Treatment for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315211, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Zhejiang 315211, China
| | - Jie Cao
- The Fifth Dental Center, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China
| | - Xiaoxue Jia
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, Maryland 20740, United States
| | - Hailiang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jieli Zhou
- UM-SJTU Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhaocheng Liu
- Department of Laboratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi 214023, China
| | - Hanbin Cui
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
- Key Laboratory of Precision Prevention and Treatment for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315211, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Zhejiang 315211, China
| | - Kai Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
23
|
Sun X, Tian T, Lian Y, Cui Z. Current Advances in Viral Nanoparticles for Biomedicine. ACS NANO 2024; 18:33827-33863. [PMID: 39648920 DOI: 10.1021/acsnano.4c13146] [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: 12/10/2024]
Abstract
Viral nanoparticles (VNPs) have emerged as crucial tools in the field of biomedicine. Leveraging their biological and physicochemical properties, VNPs exhibit significant advantages in the prevention, diagnosis, and treatment of human diseases. Through techniques such as chemical bioconjugation, infusion, genetic engineering, and encapsulation, these VNPs have been endowed with multifunctional capabilities, including the display of functional peptides or proteins, encapsulation of therapeutic drugs or inorganic particles, integration with imaging agents, and conjugation with bioactive molecules. This review provides an in-depth analysis of VNPs in biomedicine, elucidating their diverse types, distinctive features, production methods, and complex design principles behind multifunctional VNPs. It highlights recent innovative research and various applications, covering their roles in imaging, drug delivery, therapeutics, gene delivery, vaccines, immunotherapy, and tissue regeneration. Additionally, the review provides an assessment of their safety and biocompatibility and discusses challenges and future opportunities in the field, underscoring the vast potential and evolving nature of VNP research.
Collapse
Affiliation(s)
- Xianxun Sun
- School of Life Sciences, Jianghan University, Wuhan 430056, China
| | - Tao Tian
- School of Life Sciences, Jianghan University, Wuhan 430056, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yindong Lian
- School of Life Sciences, Jianghan University, Wuhan 430056, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
24
|
Shen L, Chen M, Su Y, Bi Y, Shu G, Chen W, Lu C, Zhao Z, Lv L, Zou J, Chen X, Ji J. NIR-II Imaging for Tracking the Spatiotemporal Immune Microenvironment in Atherosclerotic Plaques. ACS NANO 2024; 18:34171-34185. [PMID: 39630481 DOI: 10.1021/acsnano.4c10739] [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: 12/18/2024]
Abstract
The inflammatory immune microenvironment is responsible for atherosclerotic plaque erosion and rupture. Near-infrared-II (NIR-II) fluorescence imaging has the potential to continuously monitor the spatiotemporal changes in the plaque immune microenvironment. Herein, we constructed three different NIR-II probes based on benzo[1,2-c;4,5-c']bis[1,2,5]thiadiazole-4,7-bis(9,9-dioctyl-9H-fluoren-2-yl)thiophene (denoted as BBT-2FT): VHPK/BBT-2FT NPs, where VHPK is a specific peptide targeting vascular cell adhesion molecule-1; iNOS/BBT-2FT NPs for modulating the polarization of M1 macrophages by inducible NO synthase (iNOS) antibodies; and Arg-1/BBT-2FT for counterbalancing the inflammatory responses of M1 macrophages. These tracers enable precise tracking of atherosclerotic plaques and M1 and M2 macrophages through NIR-II imaging. VHPK/BBT-2FT NPs can accurately trace atherosclerotic plaques at various stages. Arg-1/BBT-2FT NPs precisely located M2 macrophages in the early plaque microenvironment with upregulation of peroxisome proliferator-activated receptor γ (PPAR-γ), signal transducer and activator of transcription (STAT) 6, and ATP-binding cassette transporter A1 (ABCA1), indicating that M2 macrophage polarization is crucial for early plaque lipid clearance. Meanwhile, iNOS/BBT-2FT NPs accurately tracked M1 macrophages in the advanced plaque microenvironment. The results showed that M1 macrophage polarization induces the formation of an inflammatory microenvironment through anaerobic glycolytic metabolism and pyroptosis in the advanced hypoxic plaque microenvironment, as indicated by the upregulation of hypoxia-inducible factor 1 alpha (HIF-1α), STAT1, NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3), pyruvate dehydrogenase kinase 1 (PDK1), and glucose transporter 1 (GLUT-1). Combining immunological approaches with NIR-II imaging has revealed that hypoxia-induced metabolic reprogramming of macrophages is a key factor in dynamic changes in the immune microenvironment of atherosclerotic plaques. Furthermore, our strategy shows the potential for real-time diagnosis and clinical prevention of unstable plaque rupture in atherosclerosis.
Collapse
Affiliation(s)
- Lin Shen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, 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, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| | - Yanping Su
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, 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, Imaging Diagnostic and Interventional Minimally Invasive Institute, 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, Imaging Diagnostic and Interventional Minimally Invasive Institute, 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, Imaging Diagnostic and Interventional Minimally Invasive Institute, 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, Imaging Diagnostic and Interventional Minimally Invasive Institute, 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, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| | - Lingchun Lv
- Department of Cardiovascular Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Jianhua Zou
- Departments of Diagnostic Radiology Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios 138667, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Lower Kent Ridge Road, 4 Science Drive 2, Singapore 117544, Singapore
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| |
Collapse
|
25
|
Li Y, Feng Q, Wang L, Gao X, Xi Y, Ye L, Ji J, Yang X, Zhai G. Current targeting strategies and advanced nanoplatforms for atherosclerosis therapy. J Drug Target 2024; 32:128-147. [PMID: 38217526 DOI: 10.1080/1061186x.2023.2300694] [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: 09/16/2023] [Accepted: 12/24/2023] [Indexed: 01/15/2024]
Abstract
Atherosclerosis is one of the major causes of death worldwide, and it is closely related to many cardiovascular diseases, such as stroke, myocardial infraction and angina. Although traditional surgical and pharmacological interventions can effectively retard or slow down the progression of atherosclerosis, it is very difficult to prevent or even reverse this disease. In recent years, with the rapid development of nanotechnology, various nanoagents have been designed and applied to different diseases including atherosclerosis. The unique atherosclerotic microenvironment with signature biological components allows nanoplatforms to distinguish atherosclerotic lesions from normal tissue and to approach plaques specifically. Based on the process of atherosclerotic plaque formation, this review summarises the nanodrug delivery strategies for atherosclerotic therapy, trying to provide help for researchers to understand the existing atherosclerosis management approaches as well as challenges and to reasonably design anti-atherosclerotic nanoplatforms.
Collapse
Affiliation(s)
- Yingchao Li
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Qixiang Feng
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Luyue Wang
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Xi Gao
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Yanwei Xi
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Lei Ye
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Jianbo Ji
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Xiaoye Yang
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Guangxi Zhai
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
26
|
Li S, Luo S, Yin X, Zhao X, Wang X, Gao S, Xu S, Lu J, Zhou J. Screening of ent-copalyl diphosphate synthase and metabolic engineering to achieve de novo biosynthesis of ent-copalol in Saccharomyces cerevisiae. Synth Syst Biotechnol 2024; 9:784-792. [PMID: 39021361 PMCID: PMC11253141 DOI: 10.1016/j.synbio.2024.06.005] [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: 04/05/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
The diterpene ent-copalol is an important precursor to the synthesis of andrographolide and is found only in green chiretta (Andrographis paniculata). De novo biosynthesis of ent-copalol has not been reported, because the catalytic activity of ent-copalyl diphosphate synthase (CPS) is very low in microorganisms. In order to achieve the biosynthesis of ent-copalol, Saccharomyces cerevisiae was selected as the chassis strain, because its endogenous mevalonate pathway and dephosphorylases could provide natural promotion for the synthesis of ent-copalol. The strain capable of synthesizing diterpene geranylgeranyl pyrophosphate was constructed by strengthening the mevalonate pathway genes and weakening the competing pathway. Five full-length ApCPSs were screened by transcriptome sequencing of A. paniculata and ApCPS2 had the best activity and produced ent-CPP exclusively. The peak area of ent-copalol was increased after the ApCPS2 saturation mutation and its configuration was determined by NMR and ESI-MS detection. By appropriately optimizing acetyl-CoA supply and fusion-expressing key enzymes, 35.6 mg/L ent-copalol was generated. In this study, de novo biosynthesis and identification of ent-copalol were achieved and the highest titer ever reported. It provides a platform strain for the further pathway analysis of andrographolide and derivatives and provides a reference for the synthesis of other pharmaceutical intermediates.
Collapse
Affiliation(s)
- Shan Li
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| | - Shuangshuang Luo
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| | - Xinran Yin
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| | - Xingying Zhao
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| | - Xuyang Wang
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| | - Song Gao
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| | - Sha Xu
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| | - Jian Lu
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
| | - Jingwen Zhou
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, China
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Rd, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
27
|
Cao Y, Yang Y, Guo C, Zong J, Li M, Li X, Yu T. Role of RNA-binding Proteins in Regulating Cell Adhesion and Progression of the Atherosclerotic Plaque and Plaque Erosion. Curr Atheroscler Rep 2024; 27:8. [PMID: 39576410 DOI: 10.1007/s11883-024-01250-2] [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] [Accepted: 10/15/2024] [Indexed: 11/24/2024]
Abstract
PURPOSE OF REVIEW RNA-binding proteins (RBPs) have emerged as crucial regulators of post-transcriptional processes, influencing the fate of RNA. This review delves into the biological functions of RBPs and their role in alternative splicing concerning atherosclerosis (AS), highlighting their participation in essential cellular processes. Our goal is to offer new insights for cardiovascular disease research and treatment. RECENT FINDING Dysregulation of RBPs is associated with various human diseases, including autoimmune and neurological disorders. The role of RBPs in the pathogenesis of AS is progressively being elucidated, as they influence plaque formation and disease progression by regulating cell function and gene expression. RBPs play intricate biological roles in regulating pre-mRNA, including editing, splicing, stability and translation. Alternative splicing has been demonstrated to enhance biological complexity and diversity. Our findings indicate that alternative splicing is extensively involved in the pathogenesis of AS. The dysregulated expression of specific RBPs in AS is linked to the production of adhesion molecules and vascular endothelium damage. Further research on RBPs could pave the way for the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Ying Cao
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, No. 308 Ningxia Road, Qingdao, 266000, People's Republic of China
| | - Chuan Guo
- Industrial Synergy Innovation Center, Linyi Vocational University of Science and Technology, Linyi, 276000, People's Republic of China
| | - Jinbao Zong
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Min Li
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Tao Yu
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China.
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China.
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| |
Collapse
|
28
|
Yu M, Wu H, Hu H, Cheng Y, Qin Y, Yang K, Hu C, Guo W, Kong Y, Zhao W, Cheng X, Jiang H, Wang S. Emerging near-infrared targeting diagnostic and therapeutic strategies for ischemic cardiovascular and cerebrovascular diseases. Acta Biomater 2024:S1742-7061(24)00682-2. [PMID: 39577483 DOI: 10.1016/j.actbio.2024.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 11/10/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Ischemic cardiovascular and cerebrovascular diseases (ICCDs), including thrombosis, ischemic stroke and atherosclerosis, represent a significant threat to human health, and there is an urgent requirement for the implementation of emerging diagnostic and therapeutic approaches to improve symptoms and prognosis. As a promising noninvasive modality offering high spatial and temporal resolution with favorable biocompatible properties, near-infrared (NIR) light has demonstrated a vast and profound potential in the biomedical field in recent years. Meanwhile, nanomedicine carriers are undergoing rapid development due to their high specific surface area, elevated drug loading capacity, and unique physicochemical properties. The combination of NIR light with targeted nanoprobes modified with different functional components not only maintains the high penetration depth of NIR irradiation in biological tissues but also significantly enhances the targeting specificity at the lesion site. This strategy allows for the realization of on-demand drug release and photothermal effects, thus inspiring promising avenues for the diagnosis and treatment of ICCDs. However, the clinical translation of NIR imaging and therapy is still hindered by significant obstacles. The existing literature has provided a comprehensive overview of the advancements in NIR-based nanomedicine research. However, there is a notable absence of reviews that summarize the NIR-mediated targeting strategies against ICCDs in imaging and therapy. Therefore, this review concludes the application of the emerging targeting probes combined with NIR radiation for ICCDs classified by molecular targets, analyzes the current challenges, and provides improvement strategies and prospects for further clinical translation. STATEMENT OF SIGNIFICANCE: Ischemic cardiovascular and cerebrovascular diseases (ICCDs) represent a significant threat to human health. Recently, near-infrared (NIR) light combined with targeting probes have been employed for the diagnosis and treatment of ICCDs, offering exceptional advantages including rapid feedback, high penetration depth, on-demand drug release, and favorable biocompatibility. However, there is a notable absence of reviews that summarize the NIR light-mediated targeting strategies for the imaging and therapy of ICCDs. Therefore, this review summarizes the emerging targeting probes combined with NIR light classified by molecular targets, and the proposes potential improvement strategies for clinical translation. This review elucidates the potential and current status of NIR-based techniques in ICCDs, while also serving as a reference point for additional targeted therapeutic strategies for ICCDs.
Collapse
Affiliation(s)
- Mengran Yu
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Huijun Wu
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Haoyuan Hu
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Ye Cheng
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Youran Qin
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Kaiqing Yang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - ChangHao Hu
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Wei Guo
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Yuxuan Kong
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Weiwen Zhao
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Xueqin Cheng
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Hong Jiang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China.
| | - Songyun Wang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China.
| |
Collapse
|
29
|
Yang L, Xia B, Qian T, Wang J, Wang Y, Dai J, Le C, Yang X, Wu J, Wu W, Xu J, Liu Y, Wang J. hUC-MSCs mitigate atherosclerosis induced by a high-fat diet in ApoE -/- mice by regulating the intestinal microbiota. Heliyon 2024; 10:e38698. [PMID: 39559240 PMCID: PMC11570454 DOI: 10.1016/j.heliyon.2024.e38698] [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: 01/29/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/20/2024] Open
Abstract
Background The mechanism underlying human umbilical cord mesenchymal stem cells (hUC-MSCs) regulating the stability of atherosclerotic plaque was explored by establishing mice models of atherosclerosis induced by a high-fat diet and hUC-MSCs intervention. Methods The ApoE-/- mice atherosclerosis model was constructed using a high-fat diet, and mice were divided into a normal diet group (ND), high-fat diet group (HFD), hUC-MSCs treatment group (HFDM), while the blank control (BC) consisted of C57BL/6J mice. After successful establishment of the model, the feces, hearts, and aorta of mice were collected. Morphological features were detected using HE, oil red O, and Masson staining. Afterward, 16s rRNA gene sequences was used to detect the species and abundance of the intestinal flora in mice, and an atomic force microscope (AFM) was used to detect the Young's modulus of the fibrous cap of atherosclerotic plaques. Lastly, the expression level of the inflammatory factors NLRP3, IL-1β, and IL-18 were detected via immunohistochemistry and immunofluorescence assays. Results In terms of morphological characteristics, the expression level of NLRP3, Young's modulus of the fibrous cap, and plaque stability were significantly reduced in HFD, whereas the ratio of Firmicutes to Bacteroidetes (F/B) was significantly increased. Interestingly, hUC-MSCs treatment reversed the above indices, thus enhancing plaque stability. Conclusion HFD led to dysregulation of intestinal flora homeostasis and induced aberrant expression levels of NLRP3, resulting in a decrease in the Young's modulus of plaques. However, hUC-MSCs treatment improved the biomechanical properties of plaque by modulating the intestinal flora and NLRP3, thereby elevating plaque stability and minimizing the risk of plaque rupture.
Collapse
Affiliation(s)
- Lin Yang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Bing Xia
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Tianbao Qian
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Jie Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Yuanhe Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Jialin Dai
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Cuiyun Le
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Xiaorong Yang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Jun Wu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Wenxin Wu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Jianwei Xu
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Youbin Liu
- Cardiovascular Department of Ganzhou People's Hospital, Ganzhou, 341099, Jiangxi, China
| | - Jiawen Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
- School of Basic Medicine/Institute of Forensic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| |
Collapse
|
30
|
Ma Y, Cao H, Chen B, Xu X, Zhang Q, Chen H, Zhang XB, Song G. Simultaneous In Vivo Imaging of Neutrophil Elastase and Oxidative Stress in Atherosclerotic Plaques Using a Unimolecular Photoacoustic Probe. Angew Chem Int Ed Engl 2024; 63:e202411840. [PMID: 39115358 DOI: 10.1002/anie.202411840] [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: 06/25/2024] [Indexed: 10/04/2024]
Abstract
Atherosclerosis, a major global health concern with high morbidity and mortality rates, involves complex interactions of chronic inflammation, oxidative stress, and proteolytic enzymes. Conventional imaging methods struggle to capture the dynamic biochemical processes in atherosclerotic plaques. Here, we introduce a novel unimolecular photoacoustic probe (UMAPP) designed with specific binding sites for neutrophil elastase (NE) and the redox pair O2⋅-/GSH, enabling real-time monitoring of oxidative stress and activated neutrophils in plaques. UMAPP, comprising a boron-dipyrromethene (BODIPY) core linked to a hydrophilic NE-cleavable tetrapeptide and dual oxidative stress-responsive catechol moieties, facilitates NE-mediated modulation of photoinduced electron transfer impacting photoacoustic intensity at 685 nm (PA685). Furthermore, oxidation and reduction of catechol groups by O2⋅- and GSH induce reversible, ratiometric changes in the photoacoustic spectrum (PA745/PA685 ratio). Initial UMAPP applications successfully distinguished atherosclerotic and healthy mice, evaluated pneumonia's effect on plaque composition and verified the probe's effectiveness in drug-treatment studies by detecting molecular alterations before visible histopathological changes. The integrated molecular imaging capabilities of UMAPP offer promising advancements in atherosclerosis diagnosis and management, enabling early and accurate identification of vulnerable plaques.
Collapse
Affiliation(s)
- Yuan Ma
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, China
| | - Hui Cao
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, China
| | - Baode Chen
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, China
| | - Xinyu Xu
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, China
| | - Qingpeng Zhang
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, China
| | - Haoming Chen
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, China
| | - Xiao-Bing Zhang
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, China
| | - Guosheng Song
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, China
| |
Collapse
|
31
|
Shen JX, Guo R, Wang MY, Zhu HL, Jiang HX, Cui HB, Wang K. Enzyme Activity-Based Optical Probe: Imaging Aminopeptidases N in Vulnerable Plaque and Serum. Anal Chem 2024; 96:17800-17806. [PMID: 39441188 DOI: 10.1021/acs.analchem.4c04159] [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: 10/25/2024]
Abstract
Cardiovascular disease, a chronic and progressive arterial wall disease, is increasingly recognized for its clinical significance. Aminopeptidases N (APN), crucial in the pathophysiological processes of vulnerable plaque, have been linked to endothelial dysfunction, oxidative stress, and plaque formation, thus highlighting their potential as biomarkers for disease progression. However, current detection methods for APN in body fluids and in vivo have limitations, including insufficient sensitivity and specificity, time delays, and the inability to directly reflect enzyme activity in plaques. To address these challenges, we developed an optical probe, HD-APN, for in vivo imaging of aminopeptidases, providing a potential implementation in cardiovascular disease. Our work demonstrated the applicability of HD-APN for specific monitoring of aminopeptidase levels in plaques and serum, shedding light on its potential for further research in cardiovascular disease.
Collapse
Affiliation(s)
- Jia-Xi Shen
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Rui Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Miao-Yan Wang
- Affiliated Children's Hospital of Jiangnan University, Wuxi 214023, China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Hao-Xiang Jiang
- Affiliated Children's Hospital of Jiangnan University, Wuxi 214023, China
| | - Han-Bin Cui
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Kai Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
- Affiliated Children's Hospital of Jiangnan University, Wuxi 214023, China
| |
Collapse
|
32
|
Li J, Miao Y, Wang K, Pan W, Li N, Tang B. Synergistically Activated Aggregation-Induced Emission Probe for Precise In Situ Staining of Lipids in Atherosclerotic Plaques. Anal Chem 2024; 96:17868-17878. [PMID: 39446031 DOI: 10.1021/acs.analchem.4c04559] [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: 10/25/2024]
Abstract
Visualizing the localization and distribution of lipids within arteries is crucial for studying atherosclerosis. However, existing lipid-specific probes face challenges such as strong hydrophobicity and nonspecific staining of lipophilic organelles or tissues, making them impractical for the precise identification of atherosclerotic plaques. To address this issue, we design a synergistically activated probe, Cbz-Lys-Lys-TPEB, which responds to cathepsin B (CTB) and H2O2 for the in situ generation of aggregation-induced emission luminogens (AIEgens). This enables specific staining of lipids within arteries and precise imaging of atherosclerotic plaques. The probe combines a tetraphenylethene building block with a hydrophilic peptide sequence (Cbz-Lys-Lys) and phenylboric acid module, providing excellent water solubility and fluorescence quenching in a molecular dispersion state. Upon interaction with H2O2 and CTB within plaques, the hydrophilic Cbz-Lys-Lys-TPEB probe is specifically cleaved and converted into hydrophobic AIEgens, leading to rapid aggregation and significant fluorescence enhancement. Interestingly, the in situ-liberated AIEgens display distinct lipid binding ability, effectively tracking the location and distribution of lipids in plaques. This synergistic target-activated AIEgen liberation strategy demonstrates significant feasibility for the reliable and accurate identification of atherosclerotic plaques, holding tremendous potential for clinical diagnosis and risk stratification of atherosclerosis.
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, P. R. China
| | - Yihui Miao
- 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, P. R. 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, P. R. 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, P. R. 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, P. R. 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, P. R. China
- Laoshan Laboratory, Qingdao 266237, P. R. China
| |
Collapse
|
33
|
Yang FK, Cao J, Zhang T, Jiang HX, Cui HB, Wang K. Dual-Activated Photoacoustic Probe for Reliably Detecting Hydroxyl Radical in Ischemic Cardiovascular Disease in Mouse and Human Samples. ACS Sens 2024; 9:5445-5453. [PMID: 39364916 DOI: 10.1021/acssensors.4c01665] [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: 10/05/2024]
Abstract
Cardiovascular disease (CVD) is a chronic disease characterized by the accumulation of lipids and fibrous tissue within the arterial walls, potentially leading to vascular obstruction and an increased risk of heart disease and stroke. Hydroxyl radicals play a significant role in the formation and progression of CVD as they can instigate lipid peroxidation, resulting in cellular damage and inflammatory responses. However, precisely detecting hydroxyl radicals in CVD lesions presents significant challenges due to their high reactivity and short lifespan. Herein, we present the development and application of a novel activatable optical probe, Cy-OH-LP, designed to detect hydroxyl radicals in lipid-rich environments specifically. Built on the Cy7 molecular skeleton, Cy-OH-LP exhibits near-infrared absorption and fluorescence characteristics, and its specific response to hydroxyl radicals enables a turn-on signal in both photoacoustic and fluorescence spectra. The probe demonstrated excellent selectivity and stability in various tests. Furthermore, Cy-OH-LP was successfully applied in an in vivo model to detect hydroxyl radicals in mouse models, providing a potential tool for diagnosing and monitoring AS. The biosafety of Cy-OH-LP was also verified, showing low cytotoxicity and no significant organ damage in mice. The findings suggest that Cy-OH-LP is a promising tool for the specific detection of hydroxyl radicals in lipid-rich environments, providing new possibilities for research and clinical applications in the field of oxidative stress-related diseases.
Collapse
Affiliation(s)
- Fang-Kun Yang
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Jie Cao
- The Fifth Dental Center, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Ting Zhang
- Wuxi Maternity and Chield Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi 214002, China
| | - Hao-Xiang Jiang
- Affiliated Children's Hospital of Jiangnan University, Wuxi 214023, China
| | - Han-Bin Cui
- Department of Cardiology, First Affiliated Hospital of Ningbo University (Ningbo First Hospital), School of Medicine, Ningbo University, Ningbo 315211, China
| | - Kai Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
- Affiliated Children's Hospital of Jiangnan University, Wuxi 214023, China
| |
Collapse
|
34
|
You Y, Tang C, Lin S, Li W, Li Y, Yan D, Wang D, Chen X. Activatable fluorescent probes for atherosclerosis theranostics. iScience 2024; 27:111009. [PMID: 39429791 PMCID: PMC11490739 DOI: 10.1016/j.isci.2024.111009] [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] [Indexed: 10/22/2024] Open
Abstract
The onset of atherosclerosis (AS) is insidious, and early stage patients have atypical clinical symptoms. After being diagnosed in late stage, it is often prone to sudden and fatal cardiovascular events. Therefore, it is highly desirable to develop precise and efficient diagnosis and therapy strategies of AS. Benefiting from high signal-to-noise ratio, low detection limit, high specificity and sensitivity, a series of activatable fluorescent probes based on atherosclerotic microenvironment have emerged for identification and treatment of AS. In this review, we focus on the atherosclerotic microenvironment and briefly summarize the correlation between the structural transformation and fluorescence signal changes of mono-/double-activatable fluorescent probes upon biomarkers stimulation. Moreover, their cutting-edge progress for AS theranostics is described. Finally, the outlook for activatable theranostic probes based on atherosclerotic microenvironment is discussed to aim at promoting innovative research in imaging-guided precise AS therapy.
Collapse
Affiliation(s)
- Yuanyuan You
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Chengwei Tang
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Songling Lin
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Wenman Li
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yuchao Li
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Dingyuan Yan
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Dong Wang
- Center for AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xiaohui Chen
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
35
|
Wang L, Zhang X, Zhang H, Wang X, Ren X, Bian W, Shi C, Wang J, Li L, Zhang R, Zhang H. Novel Metal-Free Nanozyme for Targeted Imaging and Inhibition of Atherosclerosis via Macrophage Autophagy Activation to Prevent Vulnerable Plaque Formation and Rupture. ACS APPLIED MATERIALS & INTERFACES 2024; 16:51944-51956. [PMID: 39287614 PMCID: PMC11450685 DOI: 10.1021/acsami.4c08671] [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: 05/26/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024]
Abstract
Atherosclerosis is a primary cause of cardiovascular and cerebrovascular diseases, with the unpredictable rupture of vulnerable atherosclerotic plaques enriched with lipid-laden macrophages being able to lead to heart attacks and strokes. Activating macrophage autophagy presents itself as a promising strategy for preventing vulnerable plaque formation and reducing the risk of rupture. In this study, we have developed a novel metal-free nanozyme (HCN@DS) that integrates the functions of multimodal imaging-guided therapy for atherosclerosis. HCN@DS has demonstrated high macrophage-targeting abilities due to its affinity toward scavenger receptor A (SR-A), along with excellent photoacoustic and photothermal imaging capabilities for guiding the precise treatment. It combines mild photothermal effects with moderate reactive oxygen species (ROS) generation to treat atherosclerosis. This controlled approach activates autophagy in atherosclerotic macrophages, inhibiting foam cell formation by reducing the uptake of oxidized low-density lipoproteins (oxLDL) and promoting efferocytosis and cholesterol efflux in macrophages. Additionally, it prevents plaque rupture by inhibiting apoptosis and inflammation within the plaque. Therefore, this metal-free nanozyme holds great potential for reducing the risk of atherosclerosis due to its high biosafety, excellent targeting ability, dual-modality imaging capability, and appropriate modulation of autophagy.
Collapse
Affiliation(s)
- Lingjie Wang
- Department
of Medical Imaging, First Hospital of Shanxi
Medical University, Taiyuan 030001, China
| | - Xiaoqian Zhang
- Department
of Medical Imaging, First Hospital of Shanxi
Medical University, Taiyuan 030001, China
- Department
of Biochemistry and Molecular Biology, Basic Medical College, Shanxi Medical University, Taiyuan 030001, China
| | - Hongrong Zhang
- Department
of Medical Imaging, First Hospital of Shanxi
Medical University, Taiyuan 030001, China
| | - Xiaozhe Wang
- Department
of Medical Imaging, First Hospital of Shanxi
Medical University, Taiyuan 030001, China
| | - Xiaofeng Ren
- Department
of Biochemistry and Molecular Biology, Basic Medical College, Shanxi Medical University, Taiyuan 030001, China
| | - Wei Bian
- Department
of Biochemistry and Molecular Biology, Basic Medical College, Shanxi Medical University, Taiyuan 030001, China
| | - Caiyun Shi
- Department
of Medical Imaging, First Hospital of Shanxi
Medical University, Taiyuan 030001, China
| | - Jingying Wang
- Shanxi
Provincial Center for Disease Control and Prevention, Taiyuan 030001, China
| | - Liping Li
- Department
of Medical Imaging, First Hospital of Shanxi
Medical University, Taiyuan 030001, China
- Department
of Biochemistry and Molecular Biology, Basic Medical College, Shanxi Medical University, Taiyuan 030001, China
| | - Ruiping Zhang
- Department
of Medical Imaging, First Hospital of Shanxi
Medical University, Taiyuan 030001, China
- Department
of Medical Imaging, Shanxi Provincial Peoples
Hospital, Taiyuan 030001, China
| | - Hua Zhang
- Department
of Medical Imaging, First Hospital of Shanxi
Medical University, Taiyuan 030001, China
| |
Collapse
|
36
|
Wang K, Gu KF, Cao J, Yang YS, Zhu HL, Shang JH, Zhou JL. Activatable Photoacoustic/Near-Infrared Probes for the Detection of Copper Ions of Cardiovascular Disease In Vivo and in Urine. ACS Sens 2024; 9:4898-4905. [PMID: 39236153 DOI: 10.1021/acssensors.4c01490] [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: 09/07/2024]
Abstract
Copper ions, implicated in processes such as oxidative stress and inflammation, are believed to play a crucial role in cardiovascular disease, a prevalent and deadly disease. Despite this, current diagnostic methods fail to detect early stage cardiovascular disease or track copper ion accumulation, limiting our understanding of the disease's progression. Therefore, the development of noninvasive techniques to image copper ions in cardiovascular disease is urgently needed to enhance diagnostic precision and therapeutic strategies. In this study, we report the successful synthesis and application of a copper ion-activated photoacoustic probe, CS-Cu, which exhibits high sensitivity and selectivity toward copper ions both in vitro and in vivo. CS-Cu was able to noninvasively monitor the changes in copper ion levels and differentiate between different mice based on copper ions in urine. Furthermore, the probe demonstrated good photoacoustic stability and exhibited no significant toxicity in the mice. These findings suggest that CS-Cu could be a promising tool for early detection and monitoring of Cu2+ levels in vivo and urine, providing a new perspective on the role of copper ions in cardiovascular disease.
Collapse
Affiliation(s)
- Kai Wang
- Affiliated Children's Hospital of Jiangnan University, Wuxi 214023, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Ke-Feng Gu
- Affiliated Children's Hospital of Jiangnan University, Wuxi 214023, China
| | - Jie Cao
- The Fifth Dental Center, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Yu-Shun Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jin-Hui Shang
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jie-Li Zhou
- UM-SJTU Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
37
|
Chen X, Zhang Z, Qiao G, Sun Z, Lu W. Immune and inflammatory insights in atherosclerosis: development of a risk prediction model through single-cell and bulk transcriptomic analyses. Front Immunol 2024; 15:1448662. [PMID: 39364414 PMCID: PMC11446800 DOI: 10.3389/fimmu.2024.1448662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/29/2024] [Indexed: 10/05/2024] Open
Abstract
Background Investigation into the immune heterogeneity linked with atherosclerosis remains understudied. This knowledge gap hinders the creation of a robust theoretical framework essential for devising personalized immunotherapies aimed at combating this disease. Methods Single-cell RNA sequencing (scRNA-seq) analysis was employed to delineate the immune cell-type landscape within atherosclerotic plaques, followed by assessments of cell-cell interactions and phenotype characteristics using scRNA-seq datasets. Subsequently, pseudotime trajectory analysis was utilized to elucidate the heterogeneity in cell fate and differentiation among macrophages. Through integrated approaches, including single-cell sequencing, Weighted Gene Co-expression Network Analysis (WGCNA), and machine learning techniques, we identified hallmark genes. A risk score model and a corresponding nomogram were developed and validated using these genes, confirmed through Receiver Operating Characteristic (ROC) curve analysis. Additionally, enrichment and immune characteristic analyses were conducted based on the risk score model. The model's applicability was further corroborated by in vitro and in vivo validation of specific genes implicated in atherosclerosis. Result This comprehensive scRNA-seq analysis has shed new light on the intricate immune landscape and the role of macrophages in atherosclerotic plaques. The presence of diverse immune cell populations, with a particularly enriched macrophage population, was highlighted by the results. Macrophage heterogeneity was intricately characterized, revealing four distinct subtypes with varying functional attributes that underscore their complex roles in atherosclerotic pathology. Intercellular communication analysis revealed robust macrophage interactions with multiple cell types and detailed pathways differing between proximal adjacent and atherosclerotic core groups. Furthermore, pseudotime trajectories charted the developmental course of macrophage subpopulations, offering insights into their differentiation fates within the plaque microenvironment. The use of machine learning identified potential diagnostic markers, culminating in the identification of RNASE1 and CD14. The risk score model based on these biomarkers exhibited high accuracy in diagnosing atherosclerosis. Immune characteristic analysis validated the risk score model's efficacy in defining patient profiles, distinguishing high-risk individuals with pronounced immune cell activities. Finally, experimental validation affirmed RNASE1's involvement in atherosclerotic progression, suggesting its potential as a therapeutic target. Conclusion Our findings have advanced our understanding of atherosclerosis immunopathology and paved the way for novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Xiaosan Chen
- Heart Center of Henan Provincial People’s Hospital, Central China Fuwai
Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | | | | | | | | |
Collapse
|
38
|
Xie Q, Zhang X, Liu F, Luo J, Liu C, Zhang Z, Yang Y, Li X. Identification and verification of immune-related genes for diagnosing the progression of atherosclerosis and metabolic syndrome. BMC Cardiovasc Disord 2024; 24:405. [PMID: 39095691 PMCID: PMC11295872 DOI: 10.1186/s12872-024-04026-3] [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: 02/23/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Atherosclerosis and metabolic syndrome are the main causes of cardiovascular events, but their underlying mechanisms are not clear. In this study, we focused on identifying genes associated with diagnostic biomarkers and effective therapeutic targets associated with these two diseases. METHODS Transcriptional data sets of atherosclerosis and metabolic syndrome were obtained from GEO database. The differentially expressed genes were analyzed by RStudio software, and the function-rich and protein-protein interactions of the common differentially expressed genes were analyzed.Furthermore, the hub gene was screened by Cytoscape software, and the immune infiltration of hub gens was analyzed. Finally, relevant clinical blood samples were collected for qRT-PCR verification of the three most important hub genes. RESULTS A total of 1242 differential genes (778 up-regulated genes and 464 down-regulated genes) were screened from GSE28829 data set. A total of 1021 differential genes (492 up-regulated genes and 529 down-regulated genes) were screened from the data set GSE98895. Then 23 up-regulated genes and 11 down-regulated genes were screened by venn diagram. Functional enrichment analysis showed that cytokines and immune activation were involved in the occurrence and development of these two diseases. Through the construction of the Protein-Protein Interaction(PPI) network and Cytoscape software analysis, we finally screened 10 hub genes. The immune infiltration analysis was further improved. The results showed that the infiltration scores of 7 kinds of immune cells in GSE28829 were significantly different among groups (Wilcoxon Test < 0.05), while in GSE98895, the infiltration scores of 4 kinds of immune cells were significantly different between groups (Wilcoxon Test < 0.05). Spearman method was used to analyze the correlation between the expression of 10 key genes and 22 kinds of immune cell infiltration scores in two data sets. The results showed that there were 42 pairs of significant correlations between 10 genes and 22 kinds of immune cells in GSE28829 (|Cor| > 0.3 & P < 0.05). There were 41 pairs of significant correlations between 10 genes and 22 kinds of immune cells in GSE98895 (|Cor| > 0.3 & P < 0.05). Finally, our results identified 10 small molecules with the highest absolute enrichment value, and the three most significant key genes (CX3CR1, TLR5, IL32) were further verified in the data expression matrix and clinical blood samples. CONCLUSION We have established a co-expression network between atherosclerotic progression and metabolic syndrome, and identified key genes between the two diseases. Through the method of bioinformatics, we finally obtained 10 hub genes in As and MS, and selected 3 of the most significant genes (CX3CR1, IL32, TLR5) for blood PCR verification. This may be helpful to provide new research ideas for the diagnosis and treatment of AS complicated with MS.
Collapse
Affiliation(s)
- Qian Xie
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, Tel, 830054, People's Republic of China
| | - Xuehe Zhang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, Tel, 830054, People's Republic of China
| | - Fen Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Junyi Luo
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, Tel, 830054, People's Republic of China
| | - Chang Liu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, Tel, 830054, People's Republic of China
| | - Zhiyang Zhang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, Tel, 830054, People's Republic of China
| | - Yining Yang
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China.
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, China.
- Key Laboratory of Cardiovascular Disease Research, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| | - Xiaomei Li
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, Tel, 830054, People's Republic of China
| |
Collapse
|
39
|
Chen X, Wu D, Chen Z. Biomedical applications of stimuli-responsive nanomaterials. MedComm (Beijing) 2024; 5:e643. [PMID: 39036340 PMCID: PMC11260173 DOI: 10.1002/mco2.643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/23/2024] Open
Abstract
Nanomaterials have aroused great interests in drug delivery due to their nanoscale structure, facile modifiability, and multifunctional physicochemical properties. Currently, stimuli-responsive nanomaterials that can respond to endogenous or exogenous stimulus display strong potentials in biomedical applications. In comparison with conventional nanomaterials, stimuli-responsive nanomaterials can improve therapeutic efficiency and reduce the toxicity of drugs toward normal tissues through specific targeting and on-demand drug release at pathological sites. In this review, we summarize the responsive mechanism of a variety of stimulus, including pH, redox, and enzymes within pathological microenvironment, as well as exogenous stimulus such as thermal effect, magnetic field, light, and ultrasound. After that, biomedical applications (e.g., drug delivery, imaging, and theranostics) of stimuli-responsive nanomaterials in a diverse array of common diseases, including cardiovascular diseases, cancer, neurological disorders, inflammation, and bacterial infection, are presented and discussed. Finally, the remaining challenges and outlooks of future research directions for the biomedical applications of stimuli-responsive nanomaterials are also discussed. We hope that this review can provide valuable guidance for developing stimuli-responsive nanomaterials and accelerate their biomedical applications in diseases diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| |
Collapse
|
40
|
Li Y, Zhang L, Yang W, Lin L, Pan J, Lu M, Zhang Z, Li Y, Li C. Notoginsenoside R 1 decreases intraplaque neovascularization by governing pericyte-endothelial cell communication via Ang1/Tie2 axis in atherosclerosis. Phytother Res 2024; 38:4036-4052. [PMID: 38886264 DOI: 10.1002/ptr.8257] [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: 08/14/2023] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
Atherosclerosis represents the major cause of mortality worldwide and triggers higher risk of acute cardiovascular events. Pericytes-endothelial cells (ECs) communication is orchestrated by ligand-receptor interaction generating a microenvironment which results in intraplaque neovascularization, that is closely associated with atherosclerotic plaque instability. Notoginsenoside R1 (R1) exhibits anti-atherosclerotic bioactivity, but its effect on angiogenesis in atherosclerotic plaque remains elusive. The aim of our study is to explore the therapeutic effect of R1 on vulnerable plaque and investigate its potential mechanism against intraplaque neovascularization. The impacts of R1 on plaque stability and intraplaque neovascularization were assessed in ApoE-/- mice induced by high-fat diet. Pericytes-ECs direct or non-direct contact co-cultured with VEGF-A stimulation were used as the in vitro angiogenesis models. Overexpressing Ang1 in pericytes was performed to investigate the underlying mechanism. In vivo experiments, R1 treatment reversed atherosclerotic plaque vulnerability and decreased the presence of neovessels in ApoE-/- mice. Additionally, R1 reduced the expression of Ang1 in pericytes. In vitro experiments demonstrated that R1 suppressed pro-angiogenic behavior of ECs induced by pericytes cultured with VEGF-A. Mechanistic studies revealed that the anti-angiogenic effect of R1 was dependent on the inhibition of Ang1 and Tie2 expression, as the effects were partially reversed after Ang1 overexpressing in pericytes. Our study demonstrated that R1 treatment inhibited intraplaque neovascularization by governing pericyte-EC association via suppressing Ang1-Tie2/PI3K-AKT paracrine signaling pathway. R1 represents a novel therapeutic strategy for atherosclerotic vulnerable plaques in clinical application.
Collapse
Affiliation(s)
- Yuan Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenqing Yang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinyuan Pan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunlun Li
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
41
|
Sun X, Jia X, Tan Z, Fan D, Chen M, Cui N, Liu A, Liu D. Oral Nanoformulations in Cardiovascular Medicine: Advances in Atherosclerosis Treatment. Pharmaceuticals (Basel) 2024; 17:919. [PMID: 39065770 PMCID: PMC11279631 DOI: 10.3390/ph17070919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerosis (AS) is the formation of atherosclerotic plaques on the walls of the arteries, causing them to narrow. If this occurs in the coronary arteries, the blood vessels may be completely blocked, resulting in myocardial infarction; if it occurs in the blood vessels of the brain, the blood vessels may be blocked, resulting in cerebral infarction, i.e., stroke. Studies have shown that the pathogenesis of atherosclerosis involves the processes of inflammation, lipid infiltration, oxidative stress, and endothelial damage, etc. SIRT, as a key factor regulating the molecular mechanisms of oxidative stress, inflammation, and aging, has an important impact on the pathogenesis of plaque formation, progression, and vulnerability. Statistics show that AS accounts for about 50 per cent of deaths in Western countries. Currently, oral medication is the mainstay of AS treatment, but its development is limited by side effects, low bioavailability and other unfavourable factors. In recent years, with the rapid development of nano-preparations, researchers have combined statins and natural product drugs within nanopreparations to improve their bioavailability. Based on this, this paper summarises the main pathogenesis of AS and also proposes new oral nanoformulations such as liposomes, nanoparticles, nanoemulsions, and nanocapsules to improve their application in the treatment of AS.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Aidong Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.S.); (X.J.); (Z.T.); (D.F.); (M.C.); (N.C.)
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.S.); (X.J.); (Z.T.); (D.F.); (M.C.); (N.C.)
| |
Collapse
|
42
|
Zong Q, He C, Long B, Huang Q, Chen Y, Li Y, Dang Y, Cai C. Targeted Delivery of Nanoparticles to Blood Vessels for the Treatment of Atherosclerosis. Biomedicines 2024; 12:1504. [PMID: 39062077 PMCID: PMC11275173 DOI: 10.3390/biomedicines12071504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/21/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerosis is a common form of cardiovascular disease, which is one of the most prevalent causes of death worldwide, particularly among older individuals. Surgery is the mainstay of treatment for severe stenotic lesions, though the rate of restenosis remains relatively high. Current medication therapy for atherosclerosis has limited efficacy in reversing the formation of atherosclerotic plaques. The search for new drug treatment options is imminent. Some potent medications have shown surprising therapeutic benefits in inhibiting inflammation and endothelial proliferation in plaques. Unfortunately, their use is restricted due to notable dose-dependent systemic side effects or degradation. Nevertheless, with advances in nanotechnology, an increasing number of nano-related medical applications are emerging, such as nano-drug delivery, nano-imaging, nanorobots, and so forth, which allow for restrictions on the use of novel atherosclerotic drugs to be lifted. This paper reviews new perspectives on the targeted delivery of nanoparticles to blood vessels for the treatment of atherosclerosis in both systemic and local drug delivery. In systemic drug delivery, nanoparticles inhibit drug degradation and reduce systemic toxicity through passive and active pathways. To further enhance the precise release of drugs, the localized delivery of nanoparticles can also be accomplished through blood vessel wall injection or using endovascular interventional devices coated with nanoparticles. Overall, nanotechnology holds boundless potential for the diagnosis and treatment of atherosclerotic diseases in the future.
Collapse
Affiliation(s)
- Qiushuo Zong
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chengyi He
- Department of Vascular Surgery, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| | - Binbin Long
- Department of General Surgery, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan 442099, China;
| | - Qingyun Huang
- Department of Cardiothoracic Surgery, The First Hospital of Putian Affiliated to Fujian Medical University, Putian 351106, China;
| | - Yunfei Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| |
Collapse
|
43
|
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
|
44
|
Popova V, Geneva-Popova M, Popova-Belova S, Doykov M, Valkanov S, Batalov Z, Marinkov A, Karalilova R. Diagnostic and prognostic role of serum interleukin-6 and carotid ultrasonography to detect subclinical atherosclerosis in patients with RA and ANCA-associated vasculitis. Rheumatol Int 2024; 44:1295-1303. [PMID: 38554194 DOI: 10.1007/s00296-024-05568-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 02/23/2024] [Indexed: 04/01/2024]
Abstract
ANCA-associated vasculitis (AAV) can affect multiple organs with severe life-threatening manifestations. Disease monitoring is difficult due to a lack of defined biomarkers. We aimed to assess the diagnostic role of serum interleukin-6 and vascular ultrasonography in AAV and subclinical atherosclerosis. The study included 20 AAV patients and two control groups of 34 patients with rheumatoid arthritis (RA) and 35 healthy controls. The levels of Il-6, carotid intima-media thickness test (CIMT), atherosclerotic plaque, and degree of stenosis were investigated. A GRACE-risk score was calculated for AAV and RA patients. The AAV patients had elevated levels of IL-6 (115 ± 23.96) compared to the RA patients (91.25 ± 42.63) and the healthy controls (15.65 ± 3.30), p < 0.001. IL-6 showed a diagnostic accuracy of 73% in distinguishing AAV from RA patients (AUC = 0.730; 95% CI 0.591 to 0834). In the AAV group, CIMT was 1.09, above the upper reference value of 0.90, p < 0.001. The AAV patients had a higher median GRACE risk score, and 60% of them had a high risk of cardiovascular events as compared to 35% of the RA patients. Sonography of extracranial vessels and serum levels of IL-6 can be used in daily clinical practice to diagnose and monitor patients with AAV.
Collapse
Affiliation(s)
- Velichka Popova
- Department of Rheumatology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria.
| | - Mariela Geneva-Popova
- Department of Rheumatology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | - Mladen Doykov
- Department of Urology and General Medicine, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Stanislav Valkanov
- Department of Urology and General Medicine, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Zguro Batalov
- Department of Rheumatology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Aleksandar Marinkov
- Department of Rheumatology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Rositsa Karalilova
- Department of Rheumatology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
45
|
Hu X, Li S, Wei Z, Wu D, Meng L, Li J, Xu J, Zhang L, Ma Q, Li H, Zeng X, Zhang Q, Li J, Liu D. Prevalence and pattern of multimorbidity in China: a cross-sectional study of 224,142 adults over 60 years old. Front Public Health 2024; 12:1349418. [PMID: 39011332 PMCID: PMC11247643 DOI: 10.3389/fpubh.2024.1349418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 06/18/2024] [Indexed: 07/17/2024] Open
Abstract
Aim To examine the prevalence and potential risk factors of multimorbidity among older adult in China. In addition, we investigated the pattern of multimorbidity. Methods This study is based on data from the fourth Sample Survey of the Aged Population in Urban and Rural China (SSAPUR) in 2015, a comprehensive survey of individuals aged 60 years or older in China. We calculated baseline data and prevalence rates for comorbidities, stratified by household registration, age, sex, education, exercise, and health insurance. Univariate and multivariate logistic regression analyses were conducted to identify potential risk factors for comorbidities. Furthermore, we determined the prevalence rates for the three most frequent disease combinations. Results A total of 215,040 participants were included in our analysis. The prevalence of multimorbidity was 50.5% among the older adult in China. The prevalence rate was slightly higher in rural areas than in urban areas, with rates of 51.5 and 49.6%, respectively (p < 0.001). Moreover, the prevalence rate was higher in females than in males, with rates of 55.2 and 45.3%, respectively (p < 0.001). Multivariate logistic regression analysis revealed that individuals aged 70-79 years (OR:1.40, 95% CI: 1.38-1.43, p < 0.001) and over 80 years (OR:1.41, 95% CI: 1.38-1.45, p < 0.001) had a higher prevalence of multimorbidity than those aged 60-69 years. The most prevalent pair of comorbidities was hypertension and osteoarthropathy, with 19.6% of the participants having these two conditions, accounting for 5.4% of the total participants. Conclusion Our findings indicate a high prevalence of multimorbidity among the older adult in China. Increased expenditure on preventive health care, popularization of general medicine and popular medical education may be adopted by the Government to cope with the high prevalence of multimorbidity.
Collapse
Affiliation(s)
- Xing Hu
- Health Service Department of the Guard Bureau of the Joint Staff Department, Beijing, China
| | - Shugang Li
- School of Public Health, Capital Medical University, Beijing, China
| | - Zhimin Wei
- Health Service Department of the Guard Bureau of the Joint Staff Department, Beijing, China
| | - Dishan Wu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, National Health Commission Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lingbing Meng
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jianyi Li
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, National Health Commission Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiapei Xu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, National Health Commission Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Luyao Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, National Health Commission Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qinan Ma
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, National Health Commission Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Hui Li
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, National Health Commission Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuezhai Zeng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, National Health Commission Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | | | - Juan Li
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Deping Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, National Health Commission Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
46
|
Bai X, Wang S, Shu L, Cao Q, Hu H, Zhu Y, Chen C. Hawthorn leaf flavonoids alleviate the deterioration of atherosclerosis by inhibiting SCAP-SREBP2-LDLR pathway through sPLA2-ⅡA signaling in macrophages in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:118006. [PMID: 38442806 DOI: 10.1016/j.jep.2024.118006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/23/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hawthorn leaves are a combination of the dried leaves of the Rosaceae plants, i.e., Crataegus pinnatifida Bge. or Crataegus pinnatifida Bge. var. major N. E. Br., is primarily cultivated in East Asia, North America, and Europe. hawthorn leaf flavonoids (HLF) are the main part of extraction. The HLF have demonstrated potential in preventing hypertension, inflammation, hyperlipidemia, and atherosclerosis. However, the potential pharmacological mechanism behind its anti-atherosclerotic effect has yet to be explored. AIM OF THE STUDY The in vivo and in vitro effects of HLF on lipid-mediated foam cell formation were investigated, with a specific focus on the levels of secreted phospholipase A2 type IIA (sPLA2-II A) in macrophage cells. MATERIALS AND METHODS The primary constituents of HLF were analyzed using ultra-high performance liquid chromatography and liquid chromatography-tandem mass spectrometry. In vivo, HLF, at concentrations of 5 mg/kg, 20 mg/kg, and 40 mg/kg, were administered to apolipoprotein E knockout mice (ApoE-/-) fed by high-fat diet (HFD) for 16 weeks. Aorta and serum samples were collected to identify lesion areas and lipids through mass spectrometry analysis to dissect the pathological process. RAW264.7 cells were incubated with oxidized low-density lipoprotein (ox-LDL) alone, or ox-LDL combined with different doses of HLF (100, 50, and 25 μg/ml), or ox-LDL plus 24-h sPLA2-IIA inhibitors, for cell biology analysis. Lipids and inflammatory cytokines were detected using biochemical analyzers and ELISA, while plaque size and collagen content of plaque were assessed by HE and the Masson staining of the aorta. The lipid deposition in macrophages was observed by Oil Red O staining. The expression of sPLA2-IIA and SCAP-SREBP2-LDLR was determined by RT-qPCR and Western blot analysis. RESULTS The chemical profile of HLF was studied using UPLC-Q-TOF-MS/MS, allowing the tentative identification of 20 compounds, comprising 1 phenolic acid, 9 flavonols and 10 flavones, including isovitexin, vitexin-4″-O-glucoside, quercetin-3-O-robibioside, rutin, vitexin-2″-O-rhamnoside, quercetin, etc. HLF decreased total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and non-high-density lipoprotein cholesterol (non-HDL-C) levels in ApoE-/- mice (P < 0.05), reduced ox-LDL uptake, inhibited level of inflammatory factors, such as IL-6, IL-8, TNF-α, and IL-1ꞵ (P < 0.001), and alleviated aortic plaques with a thicker fibrous cap. HLF effectively attenuated foam cell formation in ox-LDL-treated RAW264.7 macrophages, and reduced levels of intracellular TC, free cholesterol (FC), cholesteryl ester (CE), IL-6, TNF-α, and IL-1β (P < 0.001). In both in vivo and in vitro experiments, HLF significantly downregulated the expression of sPLA2-IIA, SCAP, SREBP2, LDLR, HMGCR, and LOX-1 (P < 0.05). Furthermore, sPLA2-IIA inhibitor effectively mitigated inflammatory release in RAW264.7 macrophages and regulated SCAP-SREBP2-LDLR signaling pathway by inhibiting sPLA2-IIA secretion (P < 0.05). CONCLUSION HLF exerted a protective effect against atherosclerosis through inhibiting sPLA2-IIA to diminish SCAP-SREBP2-LDLR signaling pathway, to reduce LDL uptake caused foam cell formation, and to slow down the progression of atherosclerosis in mice.
Collapse
Affiliation(s)
- Xufeng Bai
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Shuwen Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Limei Shu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Qingyu Cao
- College of Pharmacy, Nanchang Medical College, Nanchang, Jiangxi, 330052, China
| | - Huiming Hu
- College of Pharmacy, Nanchang Medical College, Nanchang, Jiangxi, 330052, China; Key Laboratory of Pharmacodynamics and Quality Evaluation on Anti-Inflammatory Chinese Herbs, Jiangxi Administration of Traditional Chinese Medicine, Jiangxi, 330052, China; School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, 4072, Australia.
| | - Yanchen Zhu
- College of Computer Science, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, 4072, Australia.
| |
Collapse
|
47
|
He Z, Chen Q, Duan X, Zhong Y, Zhu L, Mou N, Yang X, Cao Y, Han Z, He H, Wu S, Wang G, Qin X, Qu K, Zhang K, Liu J, Wu W. Reactive oxygen species-responsive nano-platform with dual-targeting and fluorescent lipid-specific imaging capabilities for the management of atherosclerotic plaques. Acta Biomater 2024; 181:375-390. [PMID: 38734284 DOI: 10.1016/j.actbio.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/04/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
Atherosclerosis (AS), a pathological cause of cardiovascular disease, results from endothelial injury, local progressive inflammation, and excessive lipid accumulation. AS plaques rich in foam cells are prone to rupture and form thrombus, which can cause life-threatening complications. Therefore, the assessment of atherosclerotic plaque vulnerability and early intervention are crucial in reducing the mortality rates associated with cardiovascular disease. In this work, A fluorescent probe FC-TPA was synthesized, which switches the fluorescence state between protonated and non-protonated, reducing background fluorescence and enhancing imaging signal-to-noise ratio. On this basis, FC-TPA is loaded into cyclodextrin (CD) modified with phosphatidylserine targeting peptide (PTP) and coated with hyaluronic acid (HA) to construct the intelligent responsive diagnostic nanoplatform (HA@PCFT). HA@PCFT effectively targets atherosclerotic plaques, utilizing dual targeting mechanisms. HA binds strongly to CD44, while PTP binds to phosphatidylserine, enabling nanoparticle aggregation at the lesion site. ROS acts as a smart release switch for probes. Both in vitro and in vivo evaluations confirm impressive lipid-specific fluorescence imaging capabilities of HA@PCFT nanoparticles (NPs). The detection of lipid load in atherosclerotic plaque by fluorescence imaging will aid in assessing the vulnerability of atherosclerotic plaque. STATEMENT OF SIGNIFICANCE: Currently, numerous fluorescent probes have been developed for lipid imaging. However, some challenges including inadequate water solubility, nonspecific distribution patterns, and fluorescence background interference, have greatly limited their further applications in vivo. To overcome these limitations, a fluorescent molecule has been designed and synthesized, thoroughly investigating its photophysical properties through both theoretical and experimental approaches. Interestingly, this fluorescent molecule exhibits the reversible fluorescence switching capabilities, mediated by hydrogen bonds, which effectively mitigate background fluorescence interference. Additionally, the fluorescent molecules has been successfully loaded into nanocarriers functionalized with the active targeting abilities, which has significantly improved the solubility of the fluorescent molecules and reduced their nonspecific distribution in vivo for an efficient target imaging in atherosclerosis. This study provides a valuable reference for evaluating the performance of such fluorescent dyes, and offers a promising perspective on the design of the target delivery systems for atherosclerosis.
Collapse
Affiliation(s)
- Zhigui He
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Qiao Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Xinmei Duan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Li Zhu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Nianlian Mou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Xu Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yu Cao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Zhiqiang Han
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Houhua He
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Shuai Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China; JinFeng Laboratory, Chongqing 401329, China
| | - Xian Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China; Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China; Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China; Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China.
| | - Jie Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China; JinFeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
48
|
Liu Y, Jiang Z, Yang X, Wang Y, Yang B, Fu Q. Engineering Nanoplatforms for Theranostics of Atherosclerotic Plaques. Adv Healthc Mater 2024; 13:e2303612. [PMID: 38564883 DOI: 10.1002/adhm.202303612] [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] [Revised: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Atherosclerotic plaque formation is considered the primary pathological mechanism underlying atherosclerotic cardiovascular diseases, leading to severe cardiovascular events such as stroke, acute coronary syndromes, and even sudden cardiac death. Early detection and timely intervention of plaques are challenging due to the lack of typical symptoms in the initial stages. Therefore, precise early detection and intervention play a crucial role in risk stratification of atherosclerotic plaques and achieving favorable post-interventional outcomes. The continuously advancing nanoplatforms have demonstrated numerous advantages including high signal-to-noise ratio, enhanced bioavailability, and specific targeting capabilities for imaging agents and therapeutic drugs, enabling effective visualization and management of atherosclerotic plaques. Motivated by these superior properties, various noninvasive imaging modalities for early recognition of plaques in the preliminary stage of atherosclerosis are comprehensively summarized. Additionally, several therapeutic strategies are proposed to enhance the efficacy of treating atherosclerotic plaques. Finally, existing challenges and promising prospects for accelerating clinical translation of nanoplatform-based molecular imaging and therapy for atherosclerotic plaques are discussed. In conclusion, this review provides an insightful perspective on the diagnosis and therapy of atherosclerotic plaques.
Collapse
Affiliation(s)
- Yuying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Zeyu Jiang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bin Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
49
|
Li Y, Wang Y, Xia Z, Xie Y, Ke D, Song B, Mu D, Yu R, Xie J. Noninvasive platelet membrane-coated Fe 3O 4 nanoparticles identify vulnerable atherosclerotic plaques. SMART MEDICINE 2024; 3:e20240006. [PMID: 39188703 PMCID: PMC11235982 DOI: 10.1002/smmd.20240006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/27/2024] [Indexed: 08/28/2024]
Abstract
Vulnerable atherosclerotic plaques serve as the primary pathological basis for fatal cardiovascular and cerebrovascular diseases. The precise identification and treatment of these vulnerable plaques hold paramount clinical importance in mitigating the incidence of myocardial infarction and stroke. Nevertheless, the identification of vulnerable plaques within the diffuse atherosclerotic plaques dispersed throughout the systemic circulation continues to pose a substantial challenge in clinical practice. Double emulsion solvent evaporation method, specifically the water-in-oil-in-water (W/O/W) technique, was employed to fabricate Fe3O4-based poly (lactic-co-glycolic acid) (PLGA) nanoparticles (Fe3O4@PLGA). Platelet membranes (PM) were extracted through hypotonic lysis, followed by ultrasound-assisted encapsulation onto the surface of Fe3O4@PLGA, resulting in the formation of PM-coated Fe3O4 nanoparticles (PM/Fe3O4@PLGA). Characterization of PM/Fe3O4@PLGA involved the use of dynamic light scattering, transmission electron microscopy, western blotting, and magnetic resonance imaging (MRI). A model of atherosclerotic vulnerable plaques was constructed by carotid artery coarctation and a high-fat diet fed to ApoE-/- (Apolipoprotein E knockout) mice. Immunofluorescence and MRI techniques were employed to verify the functionality of PM/Fe3O4@PLGA. In this study, we initially synthesized Fe3O4@PLGA as the core material. Subsequently, a platelet membrane was employed as a coating for the Fe3O4@PLGA, aiming to enable the detection of vulnerable atherosclerotic plaques through MRI. In vitro, PM/Fe3O4@PLGA not only exhibited excellent biosafety but also showed targeted collagen characteristics and MR imaging performance. In vivo, the adhesion of PM/Fe3O4@PLGA to atherosclerotic lesions was confirmed in a mouse model of vulnerable atherosclerotic plaques. Simultaneously, PM/Fe3O4@PLGA as a novel contrast agent for MRI has shown effective identification of vulnerable atherosclerotic plaques. In terms of safety profile in vivo, PM/Fe3O4@PLGA has not demonstrated significant organ toxicity or inflammatory response in the bloodstream. In this study, we successfully developed a platelet-membrane-coated nanoparticle system for the targeted delivery of Fe3O4@PLGA to vulnerable atherosclerotic plaques. This innovative system allows for the visualization of vulnerable plaques using MRI, thereby demonstrating its potential for enhancing the clinical diagnosis of vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Yuyu Li
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Beijing Institute of Heart, Lung, and Blood Vessel DiseasesBeijing Anzhen Hospital Affiliated to Capital Medical UniversityBeijingChina
| | - Yujie Wang
- Department of RadiologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Zequn Xia
- Department of CardiologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yangjing Xie
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Department of MedicineCardiovascular CenterMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Daozheng Ke
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Bing Song
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Dan Mu
- Department of RadiologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of RadiologyNanjing Drum Tower Hospital Clinical College of Jiangsu UniversityNanjingChina
| | - Ronghui Yu
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Jun Xie
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Department of CardiologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
50
|
Pan W, Cheng J, Cao X, Zheng Y, Yang Z, Feng W, Chen Y, Wu R. Niobium carbide MXenzyme-Driven comprehensive cholesterol regulation for photoacoustic image-guided and anti-inflammatory photothermal ablation in atherosclerosis. Bioact Mater 2024; 36:565-579. [PMID: 39072287 PMCID: PMC11276926 DOI: 10.1016/j.bioactmat.2024.07.001] [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: 03/28/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Foam cells play a pivotal role in the progression of atherosclerosis progression by triggering inflammation within arterial walls. They release inflammatory molecules that attract additional immune cells, leading to further macrophage recruitment and plaque development. In this study, we develop an osteopontin (OPN) antibody-conjugated niobium carbide (Nb2C-aOPN) MXenzyme designed to selectively target and mildly ablate foam cells while reducing inflammation in the plaque microenvironment. This approach utilizes photonic hyperthermia to decrease plaque size by enhancing cholesterol regulation through both passive cholesterol outflow and positive cholesterol efflux. Nb2C-aOPN MXenzyme exhibits multiple enzyme-mimicking properties, including catalase, superoxide dismutase, peroxidase and glutathione peroxidase, and acts as a scavenger for reactive oxygen and nitrogen species. The inhibition of reactive oxygen and nitrogen species synergizes with photothermal ablation to promote positive cholesterol efflux, leading to reduced macrophage recruitment and a shift in macrophage phenotype from M1 to M2. This integrative strategy on cholesterol regulation and anti-inflammation highlights the potential of multifunctional 2D MXenzyme-based nanomedicine in advancing atherosclerotic regression.
Collapse
Affiliation(s)
- Wenqi Pan
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, PR China
| | - Jingyun Cheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, PR China
| | - Xinyue Cao
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Yi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, PR China
| | - Zhenyu Yang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
- Shanghai Institute of Materdicine, Shanghai, 200051, PR China
| | - Rong Wu
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, PR China
| |
Collapse
|