1
|
Li X, Chen Y, Cao X, Feng W, Chen Y, Zhang J. Inflammatory Macrophage-Targeted Atherosclerosis Treatment by miRNA-Delivered, MRI-Visible, and Anti-Inflammatory Nanomedicine. ACS NANO 2025; 19:20472-20490. [PMID: 40433973 DOI: 10.1021/acsnano.4c16585] [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: 05/29/2025]
Abstract
Atherosclerosis, a principal cause of fatal cardiovascular diseases, is fundamentally a chronic inflammatory disease. Addressing this, the combined regulation of oxidative stress and inflammation through synergistic modalities offers an efficient therapeutic avenue. In this work, we rationally designed and engineered a highly efficient functional nanosystem, referred to as polydopamine nanoparticles doped with arginine and gadolinium ions (AGPDAR-146a), for the targeted delivery of therapeutic oligonucleotides, specifically microRNA-146a (miR-146a), to inflammatory macrophages within atherosclerotic plaques. AGPDAR-146a nanoparticles effectively load and deliver miR-146a, achieving enhanced accumulation in inflammatory macrophages through the specific interaction between miR-146a and class A scavenger receptors. Functionally, AGPDAR-146a nanoparticles excel in eliminating reactive oxygen species and exert anti-inflammatory effects, principally by modulating the nuclear factor kappa-light-chain-enhancer of activated B cells pathway and the interferon regulatory factor 5 protein, consequently helping to reduce and stabilize atherosclerotic plaques. Additionally, the intrinsic T1 magnetic resonance imaging capability of AGPDAR-146a nanoparticles enables real-time visualization of the progression of plaque inflammation. Therefore, the engineered nanosystem not only underscores the therapeutic potential of miR-146a in atherosclerosis but also illustrates a versatile microRNA delivery strategy applicable to various diseases characterized by oxidative stress and inflammation.
Collapse
Affiliation(s)
- Xiaodan Li
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200040, P. R. China
| | - Yixin Chen
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200040, P. R. China
| | - Xin Cao
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200040, P. R. China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Jun Zhang
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200040, P. R. China
- National Center for Neurological Disorders, Shanghai 200040, P. R. China
| |
Collapse
|
2
|
Dai Y, Yang L, Cao G, Mo L, Yang C, Zhu Y, Guo Y, Hong Y, Xu H, Lu S, Du S, He J. Combination therapy and drug co-delivery systems for atherosclerosis. J Control Release 2025; 381:113543. [PMID: 39986476 DOI: 10.1016/j.jconrel.2025.02.039] [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: 11/13/2024] [Revised: 01/25/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of plaque within the arteries. Despite advances in therapeutic strategies including anti-inflammatory, antioxidant, and lipid metabolism modulation treatments over the past two decades, the treatment of atherosclerosis remains challenging, as arterial damage is the result of interconnected pathological factors. Therefore, current monotherapies often fail to address the complex nature of this disease, leading to insufficient therapeutic outcomes. This review addressed this paucity of effective treatment options by comprehensively exploring the potential for combination therapies and advanced drug co-delivery systems for the treatment of atherosclerosis. We investigated the pathological features of and risk factors for atherosclerosis, underscoring the importance of drug combination therapies for the treatment of atherosclerotic diseases. We discuss herein mathematical models for quantifying the efficacy of the combination therapies and provide a systematic summary of drug combinations for the treatment of atherosclerosis. We also provide a detailed review of the latest advances in nanoparticle-based drug co-delivery systems for the treatment of atherosclerosis, focusing on the design of carriers with high biocompatibility and efficacy. By exploring the possibilities and challenges inherent to this approach, we aim to highlight cutting-edge technologies that can foster the development of innovative strategies, optimize drug co-administration, improve treatment outcomes, and reduce the burden of atherosclerosis-related morbidity and mortality on the healthcare system.
Collapse
Affiliation(s)
- Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Li Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Guosheng Cao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Liqing Mo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Can Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuxi Zhu
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA
| | - Yujie Guo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yi Hong
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Hanlin Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Shan Lu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Shi Du
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH 43210, USA.
| | - Jianhua He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| |
Collapse
|
3
|
Gu X, Du L, Lin R, Ding Z, Guo Z, Wei J, Li Y. How Advanced Is Nanomedicine for Atherosclerosis? Int J Nanomedicine 2025; 20:3445-3470. [PMID: 40125442 PMCID: PMC11928726 DOI: 10.2147/ijn.s508757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in nanotechnology have opened new avenues for precision therapy, personalized medicine, and multifunctional theranostics in atherosclerosis (AS). This review provides a comprehensive overview of the role of nanoparticles (NPs) in precision medicine for AS, discussing their applications, challenges, and future prospects. The review first analyzes the current treatment landscape of AS and outlines potential biological targets for therapy. Various nanocarriers, including organic, inorganic, and hybrid systems, are evaluated for their therapeutic potential, with a focus on targeted drug delivery, anti-inflammatory therapy, vascular repair, plaque stabilization, and lipid clearance. Additionally, the review explores NP preparation methods, emphasizing strategies to enhance drug loading, stability, and controlled release. Finally, the translational challenges of NP-based therapies, including biocompatibility, large-scale production, regulatory hurdles, and clinical implementation, are critically analyzed. Future directions highlight the importance of interdisciplinary collaboration and technological innovation in advancing nanoparticle-based precision medicine for AS.
Collapse
Affiliation(s)
- Xiang Gu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Lixin Du
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ruifang Lin
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zehui Ding
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhihua Guo
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jiaming Wei
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ya Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| |
Collapse
|
4
|
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
|
5
|
Xiao Y, Liang Z, Shyngys M, Baekova A, Cheung S, Muljadi MB, Bai Q, Zeng L, Choi CHJ. In Vivo Interactions of Nucleic Acid Nanostructures With Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2314232. [PMID: 39263835 PMCID: PMC11733725 DOI: 10.1002/adma.202314232] [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: 12/27/2023] [Revised: 07/03/2024] [Indexed: 09/13/2024]
Abstract
Nucleic acid nanostructures, derived from the assembly of nucleic acid building blocks (e.g., plasmids and oligonucleotides), are important intracellular carriers of therapeutic cargoes widely utilized in preclinical nanomedicine applications, yet their clinical translation remains scarce. In the era of "translational nucleic acid nanotechnology", a deeper mechanistic understanding of the interactions of nucleic acid nanostructures with cells in vivo will guide the development of more efficacious nanomedicines. This review showcases the recent progress in dissecting the in vivo interactions of four key types of nucleic acid nanostructures (i.e., tile-based, origami, spherical nucleic acid, and nucleic acid nanogel) with cells in rodents over the past five years. Emphasis lies on the cellular-level distribution of nucleic acid nanostructures in various organs and tissues and the cellular responses induced by their cellular entry. Next, in the spirit of preclinical translation, this review features the latest interactions of nucleic acid nanostructures with cells in large animals and humans. Finally, the review offers directions for studying the interactions of nucleic acid nanostructures with cells from both materials and biology perspectives and concludes with some regulatory updates.
Collapse
Affiliation(s)
- Yu Xiao
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Zhihui Liang
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Moldir Shyngys
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Aiana Baekova
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Suen Cheung
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Mathias Billy Muljadi
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Qianqian Bai
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Lula Zeng
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Chung Hang Jonathan Choi
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkShatinNew TerritoriesHong Kong
| |
Collapse
|
6
|
Peng F, Wang Z, Qiu Z, Zhang W, Zhao Y, Li C, Shi B. Nanomedicine in cardiology: Precision drug delivery for enhanced patient outcomes. Life Sci 2024; 358:123199. [PMID: 39488265 DOI: 10.1016/j.lfs.2024.123199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Cardiovascular diseases as a primary driver of global morbidity and mortality. Despite the array of therapeutic avenues in clinical practice, predominantly pharmaceutical and surgical interventions, they often fall short of fully addressing the clinical exigencies of cardiovascular patients. In recent years, nanocarriers have shown great potential in the treatment and diagnose of cardiovascular diseases. They can enhance drug targeting and bioavailability while reducing side effects. Additionally, by improving imaging and detection technologies, they enhance early diagnosis and disease monitoring capabilities. These advancements in technology offer new solutions for precision medicine in cardiovascular diseases, advancing treatment efficacy and disease management. Crafted from biomaterials, metals, or their amalgamations, these nanocarriers approximate the dimensions of biologically active molecules like proteins and DNA. Cardiovascular nanomedicine, in its infancy, has only recently burgeoned. Yet, with continual refinement in nanocarrier architecture, drug delivery mechanisms, and therapeutic outcomes, the potential of nanomedical technologies in clinical contexts becomes increasingly evident. This review aims to consolidate the strides made in nanocarrier research concerning the treatment and diagnose of cardiovascular diseases.
Collapse
Affiliation(s)
- Fengli Peng
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zimu Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Zhimei Qiu
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wei Zhang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Chaofu Li
- Department of cardiology, Chongqing University Central Hospital (Chongqing Emergency Medical Center), College of Bioengineering, Chongqing University, Chongqing, China.
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
7
|
Ding Y, Sun Y, Wang H, Zhao H, Yin R, Zhang M, Pan X, Zhu X. Atherosis-associated lnc_000048 activates PKR to enhance STAT1-mediated polarization of THP-1 macrophages to M1 phenotype. Neural Regen Res 2024; 19:2488-2498. [PMID: 38526285 PMCID: PMC11090429 DOI: 10.4103/nrr.nrr-d-23-01355] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 12/12/2023] [Accepted: 01/20/2024] [Indexed: 03/26/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202419110-00029/figure1/v/2024-03-08T184507Z/r/image-tiff Our previous study has demonstrated that lnc_000048 is upregulated in large-artery atherosclerotic stroke and promotes atherosclerosis in ApoE-/- mice. However, little is known about the role of lnc_000048 in classically activated macrophage (M1) polarization. In this study, we established THP-1-derived testing state macrophages (M0), M1 macrophages, and alternately activated macrophages (M2). Real-time fluorescence quantitative PCR was used to verify the expression of marker genes and the expression of lnc_000048 in macrophages. Flow cytometry was used to detect phenotypic proteins (CD11b, CD38, CD80). We generated cell lines with lentivirus-mediated upregulation or downregulation of lnc_000048. Flow cytometry, western blot, and real-time fluorescence quantitative PCR results showed that down-regulation of lnc_000048 reduced M1 macrophage polarization and the inflammation response, while over-expression of lnc_000048 led to the opposite effect. Western blot results indicated that lnc_000048 enhanced the activation of the STAT1 pathway and mediated the M1 macrophage polarization. Moreover, catRAPID prediction, RNA-pull down, and mass spectrometry were used to identify and screen the protein kinase RNA-activated (PKR), then catRAPID and RPIseq were used to predict the binding ability of lnc_000048 to PKR. Immunofluorescence (IF)-RNA fluorescence in situ hybridization (FISH) double labeling was performed to verify the subcellular colocalization of lnc_000048 and PKR in the cytoplasm of M1 macrophage. We speculate that lnc_000048 may form stem-loop structure-specific binding and activate PKR by inducing its phosphorylation, leading to activation of STAT1 phosphorylation and thereby enhancing STAT1 pathway-mediated polarization of THP-1 macrophages to M1 and inflammatory factor expression. Taken together, these results reveal that the lnc_000048/PKR/STAT1 axis plays a crucial role in the polarization of M1 macrophages and may be a novel therapeutic target for atherosclerosis alleviation in stroke.
Collapse
Affiliation(s)
- Yuanyuan Ding
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hongyan Wang
- Qingdao Cadre Health Care Service Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hongqin Zhao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Ruihua Yin
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
8
|
Lin Y, Liu J, Chong SY, Ting HJ, Tang X, Yang L, Zhang S, Qi X, Pei P, Yi Z, Huang C, Hou X, Gao L, Torta F, Liu X, Liu B, Kah JCY, Wang J. Dual-Function Nanoscale Coordination Polymer Nanoparticles for Targeted Diagnosis and Therapeutic Delivery in Atherosclerosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401659. [PMID: 39185808 PMCID: PMC11579969 DOI: 10.1002/smll.202401659] [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: 03/03/2024] [Revised: 08/08/2024] [Indexed: 08/27/2024]
Abstract
Atherosclerosis is the primary cause of cardiovascular events such as heart attacks and strokes. However, current medical practice lacks non-invasive, reliable approaches for both imaging atherosclerotic plaques and delivering therapeutic agents directly therein. Here, a biocompatible and biodegradable pH-responsive nanoscale coordination polymers (NCPs) based theranostic system is reported for managing atherosclerosis. NCPs are synthesized with a pH-responsive benzoic-imine (BI) linker and Gd3+. Simvastatin (ST), a statin not used for lowering blood cholesterol but known for its anti-inflammatory and antioxidant effects in mice, is chosen as the model drug. By incorporating ST into the hydrophobic domain of a lipid bilayer shell on NCPs surfaces, ST/NCP-PEG nanoparticles are created that are designed for dual purposes: they diagnose and treat atherosclerosis. When administered intravenously, they target atherosclerotic plaques, breaking down in the mild acidic microenvironment of the plaque to release ST, which reduces inflammation and oxidative stress, and Gd-complexes for MR imaging of the plaques. ST/NCP-PEG nanoparticles show efficacy in slowing the progression of atherosclerosis in live models and allow for simultaneous in vivo monitoring without observed toxicity in major organs. This positions ST/NCP-PEG nanoparticles as a promising strategy for the spontaneous diagnosis and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yuanzhe Lin
- Department of SurgeryYong Loo Lin School of MedicineNational University of Singapore1E Kent Ridge RdSingapore119228Singapore
- Department of Biomedical EngineeringNational University of Singapore4 Engineering Drive 3, Block E4, #04‐08Singapore117583Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117609Singapore
| | - Jingjing Liu
- Institute of Translational MedicineMedical CollegeYangzhou UniversityYangzhouJiangsu225001China
- Department of Chemical and Biomolecular EngineeringNational University of SingaporeSingapore117585Singapore
| | - Suet Yen Chong
- Department of SurgeryYong Loo Lin School of MedicineNational University of Singapore1E Kent Ridge RdSingapore119228Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117609Singapore
- Cardiovascular Research InstituteNational University Heart Centre Singapore (NUHCS)14 Medical DriveSingapore117599Singapore
| | - Hui Jun Ting
- Department of SurgeryYong Loo Lin School of MedicineNational University of Singapore1E Kent Ridge RdSingapore119228Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117609Singapore
| | - Xichuan Tang
- Department of SurgeryYong Loo Lin School of MedicineNational University of Singapore1E Kent Ridge RdSingapore119228Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117609Singapore
| | - Liqiang Yang
- Department of SurgeryYong Loo Lin School of MedicineNational University of Singapore1E Kent Ridge RdSingapore119228Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117609Singapore
| | - Sitong Zhang
- Department of SurgeryYong Loo Lin School of MedicineNational University of Singapore1E Kent Ridge RdSingapore119228Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117609Singapore
| | - Xinyi Qi
- Department of SurgeryYong Loo Lin School of MedicineNational University of Singapore1E Kent Ridge RdSingapore119228Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117609Singapore
| | - Peng Pei
- Department of SurgeryYong Loo Lin School of MedicineNational University of Singapore1E Kent Ridge RdSingapore119228Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117609Singapore
- Department of ChemistryNational University of Singapore3 Science Drive 3Singapore117543Singapore
| | - Zhigao Yi
- Department of ChemistryNational University of Singapore3 Science Drive 3Singapore117543Singapore
| | - Chenyuan Huang
- Department of SurgeryYong Loo Lin School of MedicineNational University of Singapore1E Kent Ridge RdSingapore119228Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117609Singapore
| | - Xiao Hou
- Department of SurgeryYong Loo Lin School of MedicineNational University of Singapore1E Kent Ridge RdSingapore119228Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117609Singapore
| | - Liang Gao
- Department of BiochemistryYong Loo Lin School of MedicineNational University of SingaporeSingapore117596Singapore
- Singapore Lipidomics Incubator (SLING)Life Sciences InstituteNational University of SingaporeSingapore117456Singapore
| | - Federico Torta
- Department of BiochemistryYong Loo Lin School of MedicineNational University of SingaporeSingapore117596Singapore
- Singapore Lipidomics Incubator (SLING)Life Sciences InstituteNational University of SingaporeSingapore117456Singapore
| | - Xiaogang Liu
- Department of ChemistryNational University of Singapore3 Science Drive 3Singapore117543Singapore
| | - Bin Liu
- Department of Chemical and Biomolecular EngineeringNational University of SingaporeSingapore117585Singapore
| | - James Chen Yong Kah
- Department of Biomedical EngineeringNational University of Singapore4 Engineering Drive 3, Block E4, #04‐08Singapore117583Singapore
| | - Jiong‐Wei Wang
- Department of SurgeryYong Loo Lin School of MedicineNational University of Singapore1E Kent Ridge RdSingapore119228Singapore
- Nanomedicine Translational Research ProgramYong Loo Lin School of MedicineNational University of SingaporeSingapore117609Singapore
- Cardiovascular Research InstituteNational University Heart Centre Singapore (NUHCS)14 Medical DriveSingapore117599Singapore
- Department of PhysiologyYong Loo Lin School of MedicineNational University of Singapore2 Medical DriveSingapore117593Singapore
| |
Collapse
|
9
|
Zhang L, Sun T, Gong M, Zhou C, Zhao Y, Zhang W, Zhang Z, Xiao S, Yang X, Wang M, Liu X, Xie Q, Zhang D. Protein corona dynamicity contributes to biological destiny disparities of nanoparticles. Mater Today Bio 2024; 28:101215. [PMID: 39221215 PMCID: PMC11364921 DOI: 10.1016/j.mtbio.2024.101215] [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: 05/15/2024] [Revised: 08/05/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Extracellular protein coronas (exPCs), which have been identified in various biofluids, are recognized for their pivotal role in mediating the interaction between nanoparticles and the cytomembrane. However, it is still unclear whether various exPCs can induce different levels of intracellular proteostasis, which is of utmost importance in preserving cellular function, and eliciting distinct intracellular biological behaviors. To investigate this, two types of exPC-coated iron oxide nanoparticles (IONPs) are prepared and used to investigate the influence of exPCs on extracellular and intracellular biological outcomes. The results demonstrate that the formation of exPCs promotes the colloidal stability of IONPs, and the discrepancies in the components of the two exPCs, including opsonin, dysopsonin, and lipoprotein, are responsible for the disparities in cellular uptake and endocytic pathways. Moreover, the differential evolution of the two exPCs during cellular internalization leads to distinct autophagy and glycolysis activities, which can be attributed to the altered depletion of angiopoietin 1 during the formation of intracellular protein coronas, which ultimately impacts the PI3K/AKT-mTOR signaling. These findings offer valuable insights into the dynamic characteristics of exPCs during cellular internalization, and their consequential implications for cellular internalization and intracellular metabolism activity, which may facilitate the comprehension of PCs on biological effects of NPs and expedite the design and application of biomedical nanoparticles.
Collapse
Affiliation(s)
| | | | | | - Chunyu Zhou
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yue Zhao
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Wansu Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Zhipeng Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Shilin Xiao
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Xiaofeng Yang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Miaomiao Wang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Xu Liu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Qian Xie
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Dong Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| |
Collapse
|
10
|
Tang C, Zhou K, Wu D, Zhu H. Nanoparticles as a Novel Platform for Cardiovascular Disease Diagnosis and Therapy. Int J Nanomedicine 2024; 19:8831-8846. [PMID: 39220195 PMCID: PMC11365508 DOI: 10.2147/ijn.s474888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular disease (CVD) is a major global health issue with high mortality and morbidity rates. With the advances in nanotechnology, nanoparticles are receiving increasing attention in diagnosing and treating CVD. Previous studies have explored the use of nanoparticles in noninvasive diagnostic technologies, such as magnetic resonance imaging and computed tomography. Nanoparticles have been extensively studied as drug carriers and prognostic factors, demonstrating synergistic efficacy. This review summarized the current applications of nanoparticles in CVD and discussed their opportunities and challenges for further exploration.
Collapse
Affiliation(s)
- Chuanyun Tang
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Di Wu
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
11
|
Fan G, Hou S, Zhang W, Jiang H, Xiao F, Yu J, Tian L. Polymer-DNA Carriers Co-Deliver Photosensitizer and siRNA for Light-Promoted Gene Transfection and Hypoxia-Relieved Photodynamic Therapy. Angew Chem Int Ed Engl 2024; 63:e202405600. [PMID: 38757208 DOI: 10.1002/anie.202405600] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 05/18/2024]
Abstract
Photochemical internalization is an efficient strategy relying on photodynamic reactions to promote siRNA endosomal escape for the success of RNA-interference gene regulation, which makes gene-photodynamic combined therapy highly synergistic and efficient. However, it is still desired to explore capable carriers to improve the delivery efficiency of the immiscible siRNA and organic photosensitizers simultaneously. Herein, we employ a micellar nanostructure (PSNA) self-assembled from polymer-DNA molecular chimeras to fulfill this task. PSNA can plentifully load photosensitizers in its hydrophobic core simply by the nanoprecipitation method. Moreover, it can organize siRNA self-assembly by the densely packed DNA shell, which leads to a higher loading capacity than the typical electrostatic condensation method. The experimental results prove that this PSNA carrier can greatly facilitate siRNA escape from the endosome/lysosome and enhance transfection. Accordingly, the PSNA-administrated therapy exhibits a significantly improved anti-tumor efficacy owing to the highly efficient co-delivery capability.
Collapse
Affiliation(s)
- Guiling Fan
- Department of Materials Science and Engineering, Southern University of Science and Technology, 1088 Xueyuan Blvd., Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| | - Shengxin Hou
- Department of Materials Science and Engineering, Southern University of Science and Technology, 1088 Xueyuan Blvd., Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| | - Wenkang Zhang
- Department of Materials Science and Engineering, Southern University of Science and Technology, 1088 Xueyuan Blvd., Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| | - Hengfeng Jiang
- Department of Materials Science and Engineering, Southern University of Science and Technology, 1088 Xueyuan Blvd., Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| | - Fan Xiao
- Department of Materials Science and Engineering, Southern University of Science and Technology, 1088 Xueyuan Blvd., Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| | - Jiantao Yu
- Department of Materials Science and Engineering, Southern University of Science and Technology, 1088 Xueyuan Blvd., Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| | - Leilei Tian
- Department of Materials Science and Engineering, Southern University of Science and Technology, 1088 Xueyuan Blvd., Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| |
Collapse
|
12
|
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
|
13
|
Wei Q, Xiao Y, Du L, Li Y. Advances in Nanoparticles in the Prevention and Treatment of Myocardial Infarction. Molecules 2024; 29:2415. [PMID: 38893291 PMCID: PMC11173599 DOI: 10.3390/molecules29112415] [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: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Myocardial infarction (MI) is one of the most prevalent types of cardiovascular disease. During MI, myocardial cells become ischemic and necrotic due to inadequate blood perfusion, leading to irreversible damage to the heart. Despite the development of therapeutic strategies for the prevention and treatment of MI, their effects are still unsatisfactory. Nanoparticles represent a new strategy for the pre-treatment and treatment of MI, and novel multifunctional nanoparticles with preventive and therapeutic capabilities hold promise for the prevention and treatment of this disease. This review summarizes the common types and properties of nanoparticles, and focuses on the research progress of nanoparticles for the prevention and treatment of MI.
Collapse
Affiliation(s)
| | | | | | - Ya Li
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.W.); (Y.X.); (L.D.)
| |
Collapse
|
14
|
Liu Y, Lin Z, Wang Y, Chen L, Wang Y, Luo C. Nanotechnology in inflammation: cutting-edge advances in diagnostics, therapeutics and theranostics. Theranostics 2024; 14:2490-2525. [PMID: 38646646 PMCID: PMC11024862 DOI: 10.7150/thno.91394] [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: 10/22/2023] [Accepted: 02/14/2024] [Indexed: 04/23/2024] Open
Abstract
Inflammatory dysregulation is intimately associated with the occurrence and progression of many life-threatening diseases. Accurate detection and timely therapeutic intervention on inflammatory dysregulation are crucial for the effective therapy of inflammation-associated diseases. However, the clinical outcomes of inflammation-involved disorders are still unsatisfactory. Therefore, there is an urgent need to develop innovative anti-inflammatory strategies by integrating emerging technological innovations with traditional therapeutics. Biomedical nanotechnology is one of the promising fields that can potentially transform the diagnosis and treatment of inflammation. In this review, we outline recent advances in biomedical nanotechnology for the diagnosis and treatment of inflammation, with special attention paid to nanosensors and nanoprobes for precise diagnosis of inflammation-related diseases, emerging anti-inflammatory nanotherapeutics, as well as nanotheranostics and combined anti-inflammatory applications. Moreover, the prospects and challenges for clinical translation of nanoprobes and anti-inflammatory nanomedicines are highlighted.
Collapse
Affiliation(s)
- Yuting Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Ziqi Lin
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Yuting Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Liuhui Chen
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Yuequan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| |
Collapse
|
15
|
Sekita A, Unterweger H, Berg S, Ohlmeyer S, Bäuerle T, Zheng KH, Coolen BF, Nederveen AJ, Cabella C, Rossi S, Stroes ESG, Alexiou C, Lyer S, Cicha I. Accumulation of Iron Oxide-Based Contrast Agents in Rabbit Atherosclerotic Plaques in Relation to Plaque Age and Vulnerability Features. Int J Nanomedicine 2024; 19:1645-1666. [PMID: 38406599 PMCID: PMC10893894 DOI: 10.2147/ijn.s430693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/14/2023] [Indexed: 02/27/2024] Open
Abstract
Purpose In this study, a detailed characterization of a rabbit model of atherosclerosis was performed to assess the optimal time frame for evaluating plaque vulnerability using superparamagnetic iron oxide nanoparticle (SPION)-enhanced magnetic resonance imaging (MRI). Methods The progression of atherosclerosis induced by ballooning and a high-cholesterol diet was monitored using angiography, and the resulting plaques were characterized using immunohistochemistry and histology. Morphometric analyses were performed to evaluate plaque size and vulnerability features. The accumulation of SPIONs (novel dextran-coated SPIONDex and ferumoxytol) in atherosclerotic plaques was investigated by histology and MRI and correlated with plaque age and vulnerability. Toxicity of SPIONDex was evaluated in rats. Results Weak positive correlations were detected between plaque age and intima thickness, and total macrophage load. A strong negative correlation was observed between the minimum fibrous cap thickness and plaque age as well as the mean macrophage load. The accumulation of SPION in the atherosclerotic plaques was detected by MRI 24 h after administration and was subsequently confirmed by Prussian blue staining of histological specimens. Positive correlations between Prussian blue signal in atherosclerotic plaques, plaque age, and macrophage load were detected. Very little iron was observed in the histological sections of the heart and kidney, whereas strong staining of SPIONDex and ferumoxytol was detected in the spleen and liver. In contrast to ferumoxytol, SPIONDex administration in rabbits was well tolerated without inducing hypersensitivity. The maximum tolerated dose in rat model was higher than 100 mg Fe/kg. Conclusion Older atherosclerotic plaques with vulnerable features in rabbits are a useful tool for investigating iron oxide-based contrast agents for MRI. Based on the experimental data, SPIONDex particles constitute a promising candidate for further clinical translation as a safe formulation that offers the possibility of repeated administration free from the risks associated with other types of magnetic contrast agents.
Collapse
Affiliation(s)
- Alexander Sekita
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Harald Unterweger
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sonja Berg
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sabine Ohlmeyer
- Institute of Radiology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tobias Bäuerle
- Preclinical Imaging Platform Erlangen (PIPE), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Kang H Zheng
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Bram F Coolen
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Aart J Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Claudia Cabella
- Bracco Imaging SpA, Centro Ricerche Bracco, Colleretto Giacosa, Turin, Italy
| | - Silvia Rossi
- Bracco Imaging SpA, Centro Ricerche Bracco, Colleretto Giacosa, Turin, Italy
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Christoph Alexiou
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Lyer
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Iwona Cicha
- ENT-Department, Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
16
|
Shi Y, Zhen X, Zhang Y, Li Y, Koo S, Saiding Q, Kong N, Liu G, Chen W, Tao W. Chemically Modified Platforms for Better RNA Therapeutics. Chem Rev 2024; 124:929-1033. [PMID: 38284616 DOI: 10.1021/acs.chemrev.3c00611] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
RNA-based therapies have catalyzed a revolutionary transformation in the biomedical landscape, offering unprecedented potential in disease prevention and treatment. However, despite their remarkable achievements, these therapies encounter substantial challenges including low stability, susceptibility to degradation by nucleases, and a prominent negative charge, thereby hindering further development. Chemically modified platforms have emerged as a strategic innovation, focusing on precise alterations either on the RNA moieties or their associated delivery vectors. This comprehensive review delves into these platforms, underscoring their significance in augmenting the performance and translational prospects of RNA-based therapeutics. It encompasses an in-depth analysis of various chemically modified delivery platforms that have been instrumental in propelling RNA therapeutics toward clinical utility. Moreover, the review scrutinizes the rationale behind diverse chemical modification techniques aiming at optimizing the therapeutic efficacy of RNA molecules, thereby facilitating robust disease management. Recent empirical studies corroborating the efficacy enhancement of RNA therapeutics through chemical modifications are highlighted. Conclusively, we offer profound insights into the transformative impact of chemical modifications on RNA drugs and delineates prospective trajectories for their future development and clinical integration.
Collapse
Affiliation(s)
- Yesi Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xueyan Zhen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yiming Zhang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 310058, China
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
17
|
Ma Y, Wang Q, Du S, Luo J, Sun X, Jia B, Ge J, Dong J, Jiang S, Li Z. Multipathway Regulation for Targeted Atherosclerosis Therapy Using Anti-miR-33-Loaded DNA Origami. ACS NANO 2024. [PMID: 38321605 DOI: 10.1021/acsnano.3c10213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Given the multifactorial pathogenesis of atherosclerosis (AS), a chronic inflammatory disease, combination therapy arises as a compelling approach to effectively address the complex interplay of pathogenic mechanisms for a more desired treatment outcome. Here, we present cRGD/ASOtDON, a nanoformulation based on a self-assembled DNA origami nanostructure for the targeted combination therapy of AS. cRGD/ASOtDON targets αvβ3 integrin receptors overexpressed on pro-inflammatory macrophages and activated endothelial cells in atherosclerotic lesions, alleviates the oxidative stress induced by extracellular and endogenous reactive oxygen species, facilitates the polarization of pro-inflammatory macrophages toward the anti-inflammatory M2 phenotype, and inhibits foam cell formation by promoting cholesterol efflux from macrophages by downregulating miR-33. The antiatherosclerotic efficacy and safety profile of cRGD/ASOtDON, as well as its mechanism of action, were validated in an AS mouse model. cRGD/ASOtDON treatment reversed AS progression and restored normal morphology and tissue homeostasis of the diseased artery. Compared to probucol, a clinical antiatherosclerotic drug with a similar mechanism of action, cRGD/ASOtDON enabled the desired therapeutic outcome at a notably lower dosage. This study demonstrates the benefits of targeted combination therapy in AS management and the potential of self-assembled DNA nanoformulations in addressing multifactorial inflammatory conditions.
Collapse
Affiliation(s)
- Yuxuan Ma
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Qi Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Shiyu Du
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Jingwei Luo
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Xiaolei Sun
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Bin Jia
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Jingru Ge
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Jun Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P. R. China
| | - Shuoxing Jiang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Zhe Li
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| |
Collapse
|
18
|
Sansonetti M, Al Soodi B, Thum T, Jung M. Macrophage-based therapeutic approaches for cardiovascular diseases. Basic Res Cardiol 2024; 119:1-33. [PMID: 38170281 PMCID: PMC10837257 DOI: 10.1007/s00395-023-01027-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024]
Abstract
Despite the advances in treatment options, cardiovascular disease (CVDs) remains the leading cause of death over the world. Chronic inflammatory response and irreversible fibrosis are the main underlying pathophysiological causes of progression of CVDs. In recent decades, cardiac macrophages have been recognized as main regulatory players in the development of these complex pathophysiological conditions. Numerous approaches aimed at macrophages have been devised, leading to novel prospects for therapeutic interventions. Our review covers the advancements in macrophage-centric treatment plans for various pathologic conditions and examines the potential consequences and obstacles of employing macrophage-targeted techniques in cardiac diseases.
Collapse
Affiliation(s)
- Marida Sansonetti
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625, Hannover, Germany
| | - Bashar Al Soodi
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625, Hannover, Germany.
- REBIRTH-Center for Translational Regenerative Medicine, Hannover Medical School, 30625, Hannover, Germany.
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), 30625, Hannover, Germany.
| | - Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
19
|
Cheng J, Huang H, Chen Y, Wu R. Nanomedicine for Diagnosis and Treatment of Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304294. [PMID: 37897322 PMCID: PMC10754137 DOI: 10.1002/advs.202304294] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/11/2023] [Indexed: 10/30/2023]
Abstract
With the changing disease spectrum, atherosclerosis has become increasingly prevalent worldwide and the associated diseases have emerged as the leading cause of death. Due to their fascinating physical, chemical, and biological characteristics, nanomaterials are regarded as a promising tool to tackle enormous challenges in medicine. The emerging discipline of nanomedicine has filled a huge application gap in the atherosclerotic field, ushering a new generation of diagnosis and treatment strategies. Herein, based on the essential pathogenic contributors of atherogenesis, as well as the distinct composition/structural characteristics, synthesis strategies, and surface design of nanoplatforms, the three major application branches (nanodiagnosis, nanotherapy, and nanotheranostic) of nanomedicine in atherosclerosis are elaborated. Then, state-of-art studies containing a sequence of representative and significant achievements are summarized in detail with an emphasis on the intrinsic interaction/relationship between nanomedicines and atherosclerosis. Particularly, attention is paid to the biosafety of nanomedicines, which aims to pave the way for future clinical translation of this burgeoning field. Finally, this comprehensive review is concluded by proposing unresolved key scientific issues and sharing the vision and expectation for the future, fully elucidating the closed loop from atherogenesis to the application paradigm of nanomedicines for advancing the early achievement of clinical applications.
Collapse
Affiliation(s)
- Jingyun Cheng
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Hui Huang
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou Institute of Shanghai UniversityWenzhouZhejiang325088P. R. China
| | - Rong Wu
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| |
Collapse
|
20
|
Perera B, Wu Y, Nguyen NT, Ta HT. Advances in drug delivery to atherosclerosis: Investigating the efficiency of different nanomaterials employed for different type of drugs. Mater Today Bio 2023; 22:100767. [PMID: 37600355 PMCID: PMC10433009 DOI: 10.1016/j.mtbio.2023.100767] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/06/2023] [Accepted: 08/06/2023] [Indexed: 08/22/2023] Open
Abstract
Atherosclerosis is the build-up of fatty deposits in the arteries, which is the main underlying cause of cardiovascular diseases and the leading cause of global morbidity and mortality. Current pharmaceutical treatment options are unable to effectively treat the plaque in the later stages of the disease. Instead, they are aimed at resolving the risk factors. Nanomaterials and nanoparticle-mediated therapies have become increasingly popular for the treatment of atherosclerosis due to their targeted and controlled release of therapeutics. In this review, we discuss different types of therapeutics used to treat this disease and focus on the different nanomaterial strategies employed for the delivery of these drugs, enabling the effective and efficient resolution of the atherosclerotic plaque. The ideal nanomaterial strategy for each drug type (e.g. statins, nucleic acids, small molecule drugs, peptides) will be comprehensively discussed.
Collapse
Affiliation(s)
- Binura Perera
- School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia
- Queensland Micro-Nanotechnology Centre, Griffith University, Nathan, Queensland, 4111, Australia
| | - Yuao Wu
- School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia
| | - Nam-Trung Nguyen
- School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia
| | - Hang Thu Ta
- School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia
- Queensland Micro-Nanotechnology Centre, Griffith University, Nathan, Queensland, 4111, Australia
| |
Collapse
|
21
|
Gao M, Tang M, Ho W, Teng Y, Chen Q, Bu L, Xu X, Zhang XQ. Modulating Plaque Inflammation via Targeted mRNA Nanoparticles for the Treatment of Atherosclerosis. ACS NANO 2023; 17:17721-17739. [PMID: 37669404 DOI: 10.1021/acsnano.3c00958] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Atherosclerosis is a common pathology present in many cardiovascular diseases. Although the current therapies (including statins and inhibitors of the serine protease PCSK9) can effectively reduce low-density lipoprotein (LDL) cholesterol levels to guideline-recommended levels, major adverse cardiovascular events still occur frequently. Indeed, the subendothelial retention of lipoproteins in the artery wall triggers multiple events of inflammation in macrophages and is a major contributor to the pathological progression of atherosclerosis. It has been gradually recognized that modulating inflammation is, therefore, an attractive avenue to forestall and treat atherosclerosis and its complications. Unfortunately, challenges with specificity and efficacy in managing plaque inflammation have hindered progress in atherosclerosis treatment. Herein, we report an NP-mediated mRNA therapeutic approach to target atherosclerotic lesional macrophages, modulating inflammation in advanced atherosclerotic lesions for the treatment of atherosclerosis. We demonstrated that the targeted NPs containing IL-10 mRNA colocalized with M2-like macrophages and induced IL-10 production in atherosclerotic plaques following intravenous administration to Western diet (WD)-fed Ldlr-/- mice. Additionally, the lesions showed a significantly alleviated inflammatory response, as evidenced by reduced oxidative stress and macrophage apoptosis, resulting in decreased lipid deposition, diminished necrotic areas, and increased fiber cap thickness. These results demonstrate the successful delivery of mRNA therapeutics to macrophage-enriched plaques in a preclinical model of advanced atherosclerosis, showing that this targeted NP inflammation management approach has great potential for translation into a wide range of clinical applications.
Collapse
Affiliation(s)
- Mingzhu Gao
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Maoping Tang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | | | - Yilong Teng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Qijing Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Lei Bu
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016, United States
| | | | - Xue-Qing Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| |
Collapse
|
22
|
Benayas E, Espinosa A, Portolés MT, Vila-del Sol V, Morales MP, Serrano MC. Cellular and Molecular Processes Are Differently Influenced in Primary Neural Cells by Slight Changes in the Physicochemical Properties of Multicore Magnetic Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2023; 15:17726-17741. [PMID: 36976318 PMCID: PMC10103129 DOI: 10.1021/acsami.3c02729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/21/2023] [Indexed: 06/18/2023]
Abstract
Herein, we use two exemplary superparamagnetic iron oxide multicore nanoparticles (SPIONs) to illustrate the significant influence of slightly different physicochemical properties on the cellular and molecular processes that define SPION interplay with primary neural cells. Particularly, we have designed two different SPION structures, NFA (i.e., a denser multicore structure accompanied by a slightly less negative surface charge and a higher magnetic response) and NFD (i.e., a larger surface area and more negatively charged), and identified specific biological responses dependent on SPION type, concentration, exposure time, and magnetic actuation. Interestingly, NFA SPIONs display a higher cell uptake, likely driven by their less negative surface and smaller protein corona, more significantly impacting cell viability and complexity. The tight contact of both SPIONs with neural cell membranes results in the significant augmentation of phosphatidylcholine, phosphatidylserine, and sphingomyelin and the reduction of free fatty acids and triacylglycerides for both SPIONs. Nonetheless, NFD induces greater effects on lipids, especially under magnetic actuation, likely indicating a preferential membranal location and/or a tighter interaction with membrane lipids than NFA, in agreement with their lower cell uptake. From a functional perspective, these lipid changes correlate with an increase in plasma membrane fluidity, again larger for more negatively charged nanoparticles (NFD). Finally, the mRNA expression of iron-related genes such as Ireb-2 and Fth-1 remains unaltered, while TfR-1 is only detected in SPION-treated cells. Taken together, these results demonstrate the substantial impact that minor physicochemical differences of nanomaterials may exert in the specific targeting of cellular and molecular processes. A denser multicore structure generated by autoclave-based production is accompanied by a slight difference in surface charge and magnetic properties that become decisive for the biological impact of these SPIONs. Their capacity to markedly modify the lipidic cell content makes them attractive as lipid-targetable nanomedicines.
Collapse
Affiliation(s)
- Esther Benayas
- , Instituto de
Ciencia de Materiales de Madrid, Consejo Superior de
Investigaciones Científicas, calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - Ana Espinosa
- , Instituto de
Ciencia de Materiales de Madrid, Consejo Superior de
Investigaciones Científicas, calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - M. Teresa Portolés
- Departamento
de Bioquímica y Biología Molecular, Facultad de Ciencias
Químicas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico
San Carlos (IdISSC), Madrid 28040, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (IDSCIII), Madrid 28040, Spain
| | - Virginia Vila-del Sol
- Hospital
Nacional de Parapléjicos, Servicio
de Salud de Castilla-La Mancha (SESCAM), Finca de la Peraleda s/n, Toledo 45071, Spain
| | - M. Puerto Morales
- , Instituto de
Ciencia de Materiales de Madrid, Consejo Superior de
Investigaciones Científicas, calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - María C. Serrano
- , Instituto de
Ciencia de Materiales de Madrid, Consejo Superior de
Investigaciones Científicas, calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| |
Collapse
|
23
|
Luo M, Li Y, Peng B, White J, Mäkilä E, Tong WY, Jonathan Choi CH, Day B, Voelcker NH. A Multifunctional Porous Silicon Nanocarrier for Glioblastoma Treatment. Mol Pharm 2023; 20:545-560. [PMID: 36484477 DOI: 10.1021/acs.molpharmaceut.2c00763] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Clinical treatment of glioblastoma (GBM) remains a major challenge because of the blood-brain barrier, chemotherapeutic resistance, and aggressive tumor metastasis. The development of advanced nanoplatforms that can efficiently deliver drugs and gene therapies across the BBB to the brain tumors is urgently needed. The protein "downregulated in renal cell carcinoma" (DRR) is one of the key drivers of GBM invasion. Here, we engineered porous silicon nanoparticles (pSiNPs) with antisense oligonucleotide (AON) for DRR gene knockdown as a targeted gene and drug delivery platform for GBM treatment. These AON-modified pSiNPs (AON@pSiNPs) were selectively internalized by GBM and human cerebral microvascular endothelial cells (hCMEC/D3) cells expressing Class A scavenger receptors (SR-A). AON was released from AON@pSiNPs, knocked down DRR and inhibited GBM cell migration. Additionally, a penetration study in a microfluidic-based BBB model and a biodistribution study in a glioma mice model showed that AON@pSiNPs could specifically cross the BBB and enter the brain. We further demonstrated that AON@pSiNPs could carry a large payload of the chemotherapy drug temozolomide (TMZ, 1.3 mg of TMZ per mg of NPs) and induce a significant cytotoxicity in GBM cells. On the basis of these results, the nanocarrier and its multifunctional strategy provide a strong potential for clinical treatment of GBM and research for targeted drug and gene delivery.
Collapse
Affiliation(s)
- Meihua Luo
- Monash Institute of Pharmaceutics Science, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria3052, Australia.,Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St. Lucia, Queensland4072, Australia.,Cell and Molecular Biology Department, QIMR Berghofer Medical Research Institute, Sid Faithfull Brain Cancer Laboratory, Brisbane, Queensland4006, Australia
| | - Yuchen Li
- Cell and Molecular Biology Department, QIMR Berghofer Medical Research Institute, Sid Faithfull Brain Cancer Laboratory, Brisbane, Queensland4006, Australia
| | - Bo Peng
- Monash Institute of Pharmaceutics Science, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria3052, Australia.,Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical materials & Engineering, Northwestern Polytechnical University, Xi'an710072, China
| | - Jacinta White
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, Victoria3168, Australia
| | - Ermei Mäkilä
- Industrial Physics Laboratory, Department of Physics and Astronomy, University of Turku, Turku20014, Finland
| | - Wing Yin Tong
- Monash Institute of Pharmaceutics Science, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria3052, Australia
| | - Chung Hang Jonathan Choi
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Bryan Day
- Cell and Molecular Biology Department, QIMR Berghofer Medical Research Institute, Sid Faithfull Brain Cancer Laboratory, Brisbane, Queensland4006, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland4072, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland4059, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutics Science, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria3052, Australia.,Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, Victoria3168, Australia.,Materials Science and Engineering, Monash University, 14 Alliance Lane, Clayton, Victoria3800, Australia
| |
Collapse
|