1
|
Li S, Li F, Wang Y, Li W, Wu J, Hu X, Tang T, Liu X. Multiple delivery strategies of nanocarriers for myocardial ischemia-reperfusion injury: current strategies and future prospective. Drug Deliv 2024; 31:2298514. [PMID: 38147501 PMCID: PMC10763895 DOI: 10.1080/10717544.2023.2298514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Acute myocardial infarction, characterized by high morbidity and mortality, has now become a serious health hazard for human beings. Conventional surgical interventions to restore blood flow can rapidly relieve acute myocardial ischemia, but the ensuing myocardial ischemia-reperfusion injury (MI/RI) and subsequent heart failure have become medical challenges that researchers have been trying to overcome. The pathogenesis of MI/RI involves several mechanisms, including overproduction of reactive oxygen species, abnormal mitochondrial function, calcium overload, and other factors that induce cell death and inflammatory responses. These mechanisms have led to the exploration of antioxidant and inflammation-modulating therapies, as well as the development of myocardial protective factors and stem cell therapies. However, the short half-life, low bioavailability, and lack of targeting of these drugs that modulate these pathological mechanisms, combined with liver and spleen sequestration and continuous washout of blood flow from myocardial sites, severely compromise the expected efficacy of clinical drugs. To address these issues, employing conventional nanocarriers and integrating them with contemporary biomimetic nanocarriers, which rely on passive targeting and active targeting through precise modifications, can effectively prolong the duration of therapeutic agents within the body, enhance their bioavailability, and augment their retention at the injured myocardium. Consequently, these approaches significantly enhance therapeutic effectiveness while minimizing toxic side effects. This article reviews current drug delivery systems used for MI/RI, aiming to offer a fresh perspective on treating this disease.
Collapse
Affiliation(s)
- Shengnan Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Fengmei Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Yan Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Xiongbin Hu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Xinyi Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
2
|
Villa-Martínez E, Rios A, Gutiérrez-Vidal R, Escalante B. Potentiation of anti-angiogenic eNOS-siRNA transfection by ultrasound-mediated microbubble destruction in ex vivo rat aortic rings. PLoS One 2024; 19:e0308075. [PMID: 39088581 PMCID: PMC11293687 DOI: 10.1371/journal.pone.0308075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/16/2024] [Indexed: 08/03/2024] Open
Abstract
Nitric oxide (NO) regulates vascular homeostasis and plays a key role in revascularization and angiogenesis. The endothelial nitric oxide synthase (eNOS) enzyme catalyzes NO production in endothelial cells. Overexpression of the eNOS gene has been implicated in pathologies with dysfunctional angiogenic processes, such as cancer. Therefore, modulating eNOS gene expression using small interfering RNAs (siRNAs) represents a viable strategy for antitumor therapy. siRNAs are highly specific to the target gene, thus reducing off-target effects. Given the widespread distribution of endothelium and the crucial physiological role of eNOS, localized delivery of nucleic acid to the affected area is essential. Therefore, the development of an efficient eNOS-siRNA delivery carrier capable of controlled release is imperative for targeting specific vascular regions, particularly those associated with tumor vascular growth. Thus, this study aims to utilize ultrasound-mediated microbubble destruction (UMMD) technology with cationic microbubbles loaded with eNOS-siRNA to enhance transfection efficiency and improve siRNA delivery, thereby preventing sprouting angiogenesis. The efficiency of eNOS-siRNA transfection facilitated by UMMD was assessed using bEnd.3 cells. Synthesis of nitric oxide and eNOS protein expression were also evaluated. The silencing of eNOS gene in a model of angiogenesis was assayed using the rat aortic ring assay. The results showed that from 6 to 24 h, the transfection of fluorescent siRNA with UMMD was twice as high as that of lipofection. Moreover, transfection of eNOS-siRNA with UMMD enhanced the knockdown level (65.40 ± 4.50%) compared to lipofectamine (40 ± 1.70%). Silencing of eNOS gene with UMMD required less amount of eNOS-siRNA (42 ng) to decrease the level of eNOS protein expression (52.30 ± 0.08%) to the same extent as 79 ng of eNOS-siRNA using lipofectamine (56.30 ± 0.10%). NO production assisted by UMMD was reduced by 81% compared to 67% reduction transfecting with lipofectamine. This diminished NO production led to higher attenuation of aortic ring outgrowth. Three-fold reduction compared to lipofectamine transfection. In conclusion, we propose the combination of eNOS-siRNA and UMMD as an efficient, safe, non-viral nucleic acid transfection strategy for inhibition of tumor progression.
Collapse
Affiliation(s)
- Elisa Villa-Martínez
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, México
| | - Amelia Rios
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, México
| | - Roxana Gutiérrez-Vidal
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, México
- Programa de Investigadoras e Investigadores por México, CONAHCyT/Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, México
| | - Bruno Escalante
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, México
| |
Collapse
|
3
|
Alqarni Z, Rezgui Y, Petri I, Ghoroghi A. Viral infection transmission and indoor air quality: A systematic review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 923:171308. [PMID: 38432379 DOI: 10.1016/j.scitotenv.2024.171308] [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: 12/14/2023] [Revised: 02/03/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Respiratory disease transmission in indoor environments presents persistent challenges for health authorities, as exemplified by the recent COVID-19 pandemic. This underscores the urgent necessity to investigate the dynamics of viral infection transmission within indoor environments. This systematic review delves into the methodologies of respiratory infection transmission in indoor settings and explores how the quality of indoor air (IAQ) can be controlled to alleviate this risk while considering the imperative of sustainability. Among the 2722 articles reviewed, 178 were retained based on their focus on respiratory viral infection transmission and IAQ. Fifty eight articles delved into SARS-CoV-2 transmission, 21 papers evaluated IAQ in contexts of other pandemics, 53 papers assessed IAQ during the SARS-CoV-2 pandemic, and 46 papers examined control strategies to mitigate infectious transmission. Furthermore, of the 46 papers investigating control strategies, only nine considered energy consumption. These findings highlight clear gaps in current research, such as analyzing indoor air and surface samples for specific indoor environments, oversight of indoor and outdoor parameters (e.g., temperature, relative humidity (RH), and building orientation), neglect of occupancy schedules, and the absence of considerations for energy consumption while enhancing IAQ. This study distinctly identifies the indoor environmental conditions conducive to the thriving of each respiratory virus, offering IAQ trade-offs to mitigate the risk of dominant viruses at any given time. This study argues that future research should involve digital twins in conjunction with machine learning (ML) techniques. This approach aims to enhance IAQ by analyzing the transmission patterns of various respiratory viruses while considering energy consumption.
Collapse
Affiliation(s)
- Zahi Alqarni
- School of Engineering, Cardiff University, Cardiff CF24 3AA, UK; School of Computer Science, King Khalid University, Abha 62529, Saudi Arabia.
| | - Yacine Rezgui
- School of Engineering, Cardiff University, Cardiff CF24 3AA, UK
| | - Ioan Petri
- School of Engineering, Cardiff University, Cardiff CF24 3AA, UK
| | - Ali Ghoroghi
- School of Engineering, Cardiff University, Cardiff CF24 3AA, UK
| |
Collapse
|
4
|
Shakya G, Cattaneo M, Guerriero G, Prasanna A, Fiorini S, Supponen O. Ultrasound-responsive microbubbles and nanodroplets: A pathway to targeted drug delivery. Adv Drug Deliv Rev 2024; 206:115178. [PMID: 38199257 DOI: 10.1016/j.addr.2023.115178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/21/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024]
Abstract
Ultrasound-responsive agents have shown great potential as targeted drug delivery agents, effectively augmenting cell permeability and facilitating drug absorption. This review focuses on two specific agents, microbubbles and nanodroplets, and provides a sequential overview of their drug delivery process. Particular emphasis is given to the mechanical response of the agents under ultrasound, and the subsequent physical and biological effects on the cells. Finally, the state-of-the-art in their pre-clinical and clinical implementation are discussed. Throughout the review, major challenges that need to be overcome in order to accelerate their clinical translation are highlighted.
Collapse
Affiliation(s)
- Gazendra Shakya
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Marco Cattaneo
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Giulia Guerriero
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Anunay Prasanna
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Samuele Fiorini
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Outi Supponen
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland.
| |
Collapse
|
5
|
Cattaneo M, Supponen O. Shell viscosity estimation of lipid-coated microbubbles. SOFT MATTER 2023; 19:5925-5941. [PMID: 37490014 DOI: 10.1039/d3sm00871a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Understanding the shell rheology of ultrasound contrast agent microbubbles is vital for anticipating their bioeffects in clinical practice. Past studies using sophisticated acoustic and optical techniques have made enormous progress in this direction, enabling the development of shell models that adequately reproduce the nonlinear behaviour of the coated microbubble under acoustic excitation. However, there have also been puzzling discrepancies and missing physical explanations for the dependency of shell viscosity on the equilibrium bubble radius, which demands further experimental investigations. In this study, we aim to unravel the cause of such behaviour by performing a refined characterisation of the shell viscosity. We use ultra-high-speed microscopy imaging, optical trapping and wide-field fluorescence to accurately record the individual microbubble response upon ultrasound driving across a range of bubble sizes. An advanced model of bubble dynamics is validated and employed to infer the shell viscosity of single bubbles from their radial time evolution. The resulting values reveal a prominent variability of the shell viscosity of about an order of magnitude and no dependency on the bubble size, which is contrary to previous studies. We find that the method called bubble spectroscopy, which has been used extensively in the past to determine the shell viscosity, is highly sensitive to methodology inaccuracies, and we demonstrate through analytical arguments that the previously reported unphysical trends are an artifact of these biases. We also show the importance of correct bubble sizing, as errors in this aspect can also lead to unphysical trends in shell viscosity, when estimated through a nonlinear fitting from the time response of the bubble.
Collapse
Affiliation(s)
- Marco Cattaneo
- Institute of Fluid Dynamics, Department of Mechanical and Process Engineering, ETH Zürich, Sonneggstrasse 3, 8092 Zürich, Switzerland.
| | - Outi Supponen
- Institute of Fluid Dynamics, Department of Mechanical and Process Engineering, ETH Zürich, Sonneggstrasse 3, 8092 Zürich, Switzerland.
| |
Collapse
|
6
|
Navarro-Becerra JA, Borden MA. Targeted Microbubbles for Drug, Gene, and Cell Delivery in Therapy and Immunotherapy. Pharmaceutics 2023; 15:1625. [PMID: 37376072 DOI: 10.3390/pharmaceutics15061625] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Microbubbles are 1-10 μm diameter gas-filled acoustically-active particles, typically stabilized by a phospholipid monolayer shell. Microbubbles can be engineered through bioconjugation of a ligand, drug and/or cell. Since their inception a few decades ago, several targeted microbubble (tMB) formulations have been developed as ultrasound imaging probes and ultrasound-responsive carriers to promote the local delivery and uptake of a wide variety of drugs, genes, and cells in different therapeutic applications. The aim of this review is to summarize the state-of-the-art of current tMB formulations and their ultrasound-targeted delivery applications. We provide an overview of different carriers used to increase drug loading capacity and different targeting strategies that can be used to enhance local delivery, potentiate therapeutic efficacy, and minimize side effects. Additionally, future directions are proposed to improve the tMB performance in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
| | - Mark A Borden
- Mechanical Engineering Department, University of Colorado Boulder, Boulder, CO 80309, USA
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
7
|
Li W, Jin Q, Zhang L, He S, Song Y, Xu L, Deng C, Wang L, Qin X, Xie M. Ultrasonic Microbubble Cavitation Deliver Gal-3 shRNA to Inhibit Myocardial Fibrosis after Myocardial Infarction. Pharmaceutics 2023; 15:pharmaceutics15030729. [PMID: 36986588 PMCID: PMC10051524 DOI: 10.3390/pharmaceutics15030729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Galectin-3 (Gal-3) participates in myocardial fibrosis (MF) in a variety of ways. Inhibiting the expression of Gal-3 can effectively interfere with MF. This study aimed to explore the value of Gal-3 short hairpin RNA (shRNA) transfection mediated by ultrasound-targeted microbubble destruction (UTMD) in anti-myocardial fibrosis and its mechanism. A rat model of myocardial infarction (MI) was established and randomly divided into control and Gal-3 shRNA/cationic microbubbles + ultrasound (Gal-3 shRNA/CMBs + US) groups. Echocardiography measured the left ventricular ejection fraction (LVEF) weekly, and the heart was harvested to analyze fibrosis, Gal-3, and collagen expression. LVEF in the Gal-3 shRNA/CMB + US group was improved compared with the control group. On day 21, the myocardial Gal-3 expression decreased in the Gal-3 shRNA/CMBs + US group. Furthermore, the proportion of the myocardial fibrosis area in the Gal-3 shRNA/CMBs + US group was 6.9 ± 0.41% lower than in the control group. After inhibition of Gal-3, there was a downregulation in collagen production (collagen I and III), and the ratio of Col I/Col III decreased. In conclusion, UTMD-mediated Gal-3 shRNA transfection can effectively silence the expression of Gal-3 in myocardial tissue, reduce myocardial fibrosis, and protect the cardiac ejection function.
Collapse
Affiliation(s)
- Wenqu Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Shukun He
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yishu Song
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Lingling Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Cheng Deng
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Lufang Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaojuan Qin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Correspondence: (X.Q.); (M.X.); Tel.: +86-136-0710-8938 (M.X.); Fax: +86-27-85726386 (M.X.)
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Correspondence: (X.Q.); (M.X.); Tel.: +86-136-0710-8938 (M.X.); Fax: +86-27-85726386 (M.X.)
| |
Collapse
|
8
|
Wu Y, Deng C, Xu J, Wang W, Chen Y, Qin X, Lv Q, Xie M. Enhanced Local Delivery of microRNA-145a-5P into Mouse Aorta via Ultrasound-Targeted Microbubble Destruction Inhibits Atherosclerotic Plaque Formation. Mol Pharm 2023; 20:1086-1095. [PMID: 36656656 DOI: 10.1021/acs.molpharmaceut.2c00799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) play a key role in the formation and rupture of atherosclerotic plaques. Previous studies have confirmed that microRNA-145 (miR-145) is involved in the phenotypic regulation of VSMCs and reduction of atherosclerosis. At present, seeking safe and effective gene delivery remains a key problem restricting the development of gene therapy. In recent years, ultrasound-targeted microbubble destruction (UTMD) has become a safe and effective transfection method that is widely used in the basic research of gene therapy for heart and tumor diseases. Here, we synthesized cationic microbubbles to encapsulate miR-145 and targeted their release into VSMCs in vitro and in vivo using ultrasound. The feasibility of this gene therapy was verified by fluorescence microscopy and an in vivo imaging system. The results showed that treatment with miR-145 delivered via UTMD considerably improved the gene transfection efficiency and promoted the contraction phenotype of VSMCs in vitro. In vivo, this treatment reduced the atherosclerotic plaque area by 48.04% compared with treatment with free miR-145. Therefore, UTMD-mediated miRNA therapy may provide a new targeted therapeutic approach for atherosclerotic plaques.
Collapse
Affiliation(s)
- Yu Wu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Cheng Deng
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Jia Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Wei Wang
- Department of Ultrasound, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaojuan Qin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
9
|
Sridharan B, Lim HG. Exosomes and ultrasound: The future of theranostic applications. Mater Today Bio 2023; 19:100556. [PMID: 36756211 PMCID: PMC9900624 DOI: 10.1016/j.mtbio.2023.100556] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Biomaterials and pertaining formulations have been very successful in various diagnostic and therapeutic applications because of its ability to overcome pharmacological limitations. Some of them have gained significant focus in the recent decade for their theranostic properties. Exosomes can be grouped as biomaterials, since they consist of various biological micro/macromolecules and possess all the properties of a stable biomaterial with size in nano range. Significant research has gone into isolation and exploitation of exosomes as potential theranostic agent. However, the limitations in terms of yield, efficacy, and target specificity are continuously being addressed. On the other hand, several nano/microformulations are responsive to physical or chemical alterations and were successfully stimulated by tweaking the physical characteristics of the surrounding environment they are in. Some of them are termed as photodynamic, sonodynamic or thermodynamic therapeutic systems. In this regard, ultrasound and acoustic systems were extensively studied for its ability towards altering the properties of the systems to which they were applied on. In this review, we have detailed about the diagnostic and therapeutic applications of exosomes and ultrasound separately, consisting of their conventional applications, drawbacks, and developments for addressing the challenges. The information were categorized into various sections that provide complete overview of the isolation strategies and theranostic applications of exosomes in various diseases. Then the ultrasound-based disease diagnosis and therapy were elaborated, with special interest towards the use of ultrasound in enhancing the efficacy of nanomedicines and nanodrug delivery systems, Finally, we discussed about the ability of ultrasound in enhancing the diagnostic and therapeutic properties of exosomes, which could be the future of theranostics.
Collapse
Affiliation(s)
| | - Hae Gyun Lim
- Corresponding author. Biomedical Ultrasound Lab, Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
10
|
Cardiovascular Nanotechnology. Nanomedicine (Lond) 2023. [DOI: 10.1007/978-981-16-8984-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
11
|
Tu J, Yu ACH. Ultrasound-Mediated Drug Delivery: Sonoporation Mechanisms, Biophysics, and Critical Factors. BME FRONTIERS 2022; 2022:9807347. [PMID: 37850169 PMCID: PMC10521752 DOI: 10.34133/2022/9807347] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/31/2021] [Indexed: 10/19/2023] Open
Abstract
Sonoporation, or the use of ultrasound in the presence of cavitation nuclei to induce plasma membrane perforation, is well considered as an emerging physical approach to facilitate the delivery of drugs and genes to living cells. Nevertheless, this emerging drug delivery paradigm has not yet reached widespread clinical use, because the efficiency of sonoporation is often deemed to be mediocre due to the lack of detailed understanding of the pertinent scientific mechanisms. Here, we summarize the current observational evidence available on the notion of sonoporation, and we discuss the prevailing understanding of the physical and biological processes related to sonoporation. To facilitate systematic understanding, we also present how the extent of sonoporation is dependent on a multitude of factors related to acoustic excitation parameters (ultrasound frequency, pressure, cavitation dose, exposure time), microbubble parameters (size, concentration, bubble-to-cell distance, shell composition), and cellular properties (cell type, cell cycle, biochemical contents). By adopting a science-backed approach to the realization of sonoporation, ultrasound-mediated drug delivery can be more controllably achieved to viably enhance drug uptake into living cells with high sonoporation efficiency. This drug delivery approach, when coupled with concurrent advances in ultrasound imaging, has potential to become an effective therapeutic paradigm.
Collapse
Affiliation(s)
- Juan Tu
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing, China
| | - Alfred C. H. Yu
- Schlegel Research Institute for Aging, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
12
|
Cardiovascular Nanotechnology. Nanomedicine (Lond) 2022. [DOI: 10.1007/978-981-13-9374-7_12-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
13
|
Peng S, Cai J, Bao S. CMBs carrying PTX and CRISPR/Cas9 targeting C‑erbB‑2 plasmids interfere with endometrial cancer cells. Mol Med Rep 2021; 24:830. [PMID: 34590151 PMCID: PMC8503745 DOI: 10.3892/mmr.2021.12470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/06/2021] [Indexed: 12/27/2022] Open
Abstract
Development of combination therapy to decrease side effects of chemotherapeutic drugs and increase their utilization rate in combination with gene editing is a key research topic in tumor treatment. The present study aimed to investigate the effect of cationic microbubbles (CMBs) carrying paclitaxel (PTX) and C-erbB-2 knockout plasmid on the endometrial cancer cell line HEC-1A and to determine how C-erbB-2 regulates the function of endometrial cancer cells. Cells were treated with CMB, PTX, PTX-CMBs, cationic plasmid-carrying or cationic PTX-carrying plasmid groups. After verifying the most effective combination of PTX-CMBs and plasmids, HEC-1A cells were transfected. Reverse transcription-quantitative (RT-q)PCR and western blotting were used to measure C-erbB-2 and protein expression. After verifying C-erbB-2 knockout, invasion, healing, clone formation and proliferation of HEC-1A cells were assessed. Simultaneously, expression levels of the genes for P21, P27, mammalian target of rapamycin (mTOR), and Bcl-2 associated death promoter (Bad) were measured by RT-qPCR. Compared with the PTX group, CMBs significantly enhanced the absorption efficiency of PTX by HEC-1A cells. C-erbB-2 knockout had an inhibitory effect on the proliferation, migration and invasion of HEC-1A cells; cell proliferation and invasion of the group carrying PTX and plasmids simultaneously were significantly weakened. The C-erbB-2-knockout group exhibited increased expression of P21 and P27. Simultaneously loading PTX and plasmid may be novel combination therapy with great potential. C-erbB-2 may regulate the proliferation of HEC-1A cells by downregulating expression of P21 and P27.
Collapse
Affiliation(s)
- Siyuan Peng
- Department of Gynaecology and Obstetrics, Hainan Hospital Affiliated to University of South China, Haikou, Hainan 570311, P.R. China
| | - Junhong Cai
- Key Laboratory of Cell and Molecular Genetic Translational Medicine in Hainan Province, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Shan Bao
- Department of Gynaecology and Obstetrics, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| |
Collapse
|
14
|
Mei L, Zhang Z. Advances in Biological Application of and Research on Low-Frequency Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:2839-2852. [PMID: 34304908 DOI: 10.1016/j.ultrasmedbio.2021.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 06/13/2023]
Abstract
In recent years, the in-depth study of low-frequency sonophoresis (LFS) has greatly elucidated its biological effects in various therapeutic applications, including drug delivery, enhanced healing, thrombolytic technology, anti-inflammatory effects and tumor treatment. Specifically, numerous studies have reported its use in drug delivery and synergistic antitumor activity, indicating a new treatment direction for cancer. However, there are significant gaps in the understanding of LFS in terms of frequency and sound intensity safety; these issues are becoming increasingly important in understanding the biological effects of LFS ultrasound. This article reviews the treatment mechanism and current applications of LFS technology and discusses and summarizes its safety and application prospects.
Collapse
Affiliation(s)
- Lixia Mei
- Department of Ultrasound, Qiqihar Hospital Affiliated to Southern Medical University, Qiqihar City, Heilongjiang Province, China.
| | - Zhen Zhang
- Department of Ultrasound, First Affiliated Hospital of China Medical University, Shenyang City, Liaoning Province, China.
| |
Collapse
|
15
|
Gao J, Liu J, Meng Z, Li Y, Hong Y, Wang L, He L, Hu B, Zheng Y, Li T, Cui D, Shen E. Ultrasound-assisted C 3F 8-filled PLGA nanobubbles for enhanced FGF21 delivery and improved prophylactic treatment of diabetic cardiomyopathy. Acta Biomater 2021; 130:395-408. [PMID: 34129954 DOI: 10.1016/j.actbio.2021.06.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/26/2022]
Abstract
Diabetic cardiomyopathy (DCM) is a serious cardiac complication of diabetes that currently lacks specific treatment. Fibroblast growth factor 21 (FGF21) has been proved to have cardioprotective effect in DCM. However, the insufficient cardiac delivery effect of FGF21 limits its application in DCM. Therefore, to improve the therapeutic efficacy of FGF21 in DCM, an effective drug delivery system is urgently required. In this study, perfluoropropane (C3F8) and polyethylenimine (PEI)-doped poly (lactic-co-glycolic acid) (PLGA) nanobubbles (CPPNBs) were synthesized via double-emulsion evaporation and FGF21 was efficiently absorbed (CPPNBs@FGF21) via the electrostatic incorporation effect. CPPNBs@FGF21 could effectively deliver FGF21 to the myocardial tissue through the cavitation effect under low-frequency ultrasound (LFUS). The as-prepared CPPNBs@FGF21 could efficiently load FGF21 after doping with the cationic polymer PEI, and displayed uniform dispersion and favorable biosafety. After filling with C3F8, CPPNBs@FGF21 could be used for distribution monitoring through ultrasound imaging. Moreover, CPPNBs@FGF21 significantly downregulated the expression of ANP, CTGF, and caspase-3 mRNA via the action of LFUS owing to increased FGF21 release, therefore exhibiting enhanced inhibition of myocardial hypertrophy, apoptosis, and interstitial fibrosis in DCM mice. In conclusion, we established an effective protein delivery nanocarrier for the diagnosis and prophylactic treatment of DCM. STATEMENT OF SIGNIFICANCE: Diabetic cardiomyopathy (DCM) is a serious cardiac complication of diabetes that currently lacks effective clinical treatments. Fibroblast growth factor 21 (FGF21) can protect cardiomyocytes from diabetic damage, but insufficient cardiac drug delivery limits the application of FGF21 in DCM. In this study, perfluoropropane (C3F8) and polyethylenimine (PEI)-doped poly (lactic-co-glycolic acid) (PLGA) nanobubbles loaded with FGF21 (CPPNBs@FGF21) were developed for the prophylactic treatment of DCM. CPPNBs@FGF21 could effectively deliver the FGF21 to the myocardial tissue through the cavitation effect of low-frequency ultrasound (LFUS). Our results indicated that CPPNBs@FGF21 combined with LFUS could significantly down-regulate the expressions of ANP, CTGF, and caspase-3 mRNA, and as a result, it prevented the myocardial hypertrophy, apoptosis, and interstitial fibrosis of DCM mice. Overall, we established an effective protein delivery nanocarrier for the diagnosis and prophylactic treatment of DCM.
Collapse
Affiliation(s)
- Jiameng Gao
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China.; Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Jingjing Liu
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China; Department of Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Zheying Meng
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Yanming Li
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Yuping Hong
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Lirui Wang
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Lan He
- Department of Ultrasound in Medicine, Shanghai Eighth People's Hospital, 8 Caobao Road, Shanghai 200235, PR China
| | - Bing Hu
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China.; Department of Ultrasound in Medicine, Shanghai Eighth People's Hospital, 8 Caobao Road, Shanghai 200235, PR China
| | - Yuanyi Zheng
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Tianliang Li
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China.
| | - Daxiang Cui
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China.
| | - E Shen
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China.; Department of Ultrasound in Medicine, Shanghai Eighth People's Hospital, 8 Caobao Road, Shanghai 200235, PR China.
| |
Collapse
|
16
|
Wei T, Li L, He Z. Ultrasound-Mediated Microbubble Destruction Inhibits Skin Melanoma Growth by Affecting YAP1 Translation Using Ribosome Imprinting Sequencing. Front Oncol 2021; 11:619167. [PMID: 33996543 PMCID: PMC8117937 DOI: 10.3389/fonc.2021.619167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/16/2021] [Indexed: 11/13/2022] Open
Abstract
Cutaneous melanoma (CMM) is a skin tumor with a high degree of malignancy. BRAF resistance imposes great difficulty to the treatment of CMM, and partially contributes to the poor prognosis of CMM. YAP is involved in the growth and drug resistance of a variety of tumors, and mechanical signals may affect the activation of YAP1. As a novel ultrasound treatment technology, ultrasound-mediated microbubble destruction (UMMD) has been reported to have a killing effect on isolated CMM cells. In this study, the tumor tissue samples were collected from 64 CMM patients. We found that YAP1 mRNA expression was irrelevant to the clinicopathological characteristics and prognostic survival of the CMM patients. The drug-resistant cell line was constructed and subcutaneously implanted into nude mice, which were further separately treated with UMMD, ultrasound (US), and microbubbles (MB). The result showed that UMMD significantly inhibited the growth of tumor tissues. Ribosome imprinting sequencing (Ribo-seq) is a genetic technology for studying protein translation at genetic level. Ribo-seq, RNA-seq, and RT-qPCR were applied to detect YAP1 expression in CMM mouse tumor tissues. Ribo-seq data revealed that UMMD greatly up-regulated the expression of YAP1, interestingly, the up-regulated YAP1 was found to be negatively correlated with the weight of tumor tissues, while no significant change in YAP1 expression was detected by RNA-seq or RT-qPCR assay. These results indicated that UMMD could inhibit the tumor growth of drug-resistant CMM by affecting the translation efficiency of YAP1, providing a strong basis for the clinical treatment of UMMD in CMM.
Collapse
Affiliation(s)
- Tianhong Wei
- Department of Ultrasonography, Xiangya Hospital, Central South University, Changsha, China
| | - Lan Li
- Department of Ultrasonography, Xiangya Hospital, Central South University, Changsha, China
| | - Zhiyou He
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
17
|
Liu Y, Zhou Y, Xu J, Luo H, Zhu Y, Zeng X, Dong F, Wei Z, Yan F, Zheng H. Ultrasound molecular imaging-guided tumor gene therapy through dual-targeted cationic microbubbles. Biomater Sci 2021; 9:2454-2466. [PMID: 33594996 DOI: 10.1039/d0bm01857k] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The success of gene therapy depends largely on the development of gene vectors and effective gene delivery systems. It has been demonstrated that cationic microbubbles can be loaded with negatively charged plasmid DNA and thus improve gene transfection efficiency. In this study, we developed dual-targeting cationic microbubbles conjugated with iRGD peptides(Cyclo(Cys-Arg-Gly-Asp-Lys-Gly-Pro-Asp-Cys)) and CCR2 (chemokine (C-C motif) receptor 2) antibodies (MBiRGD/CCR2) for ultrasound molecular imaging and targeted tumor gene therapy. The ultrasound molecular imaging experiments showed that there were significantly enhanced ultrasound molecular imaging signals in the tumor that received MBiRGD/CCR2, compared with those that received MBiRGD, MBCCR2, or MBcontrol. As a therapy plasmid, pGPU6/GFP/Neo-shAKT2, carrying an expression cassette for the human AKT2 RNA interference sequence, was used. Our results demonstrated that MBiRGD/CCR2 had a significantly higher gene transfection efficiency than MBiRGD, MBCCR2, or MBcontrol under ultrasound irradiation, resulting in much lower AKT2 protein expression and stronger tumor growth inhibition effects in vivo and in vitro. In conclusion, our study demonstrated a novel gene delivery system via MBiRGD/CCR2 for ultrasound molecular-imaging-guided gene therapy of breast cancer.
Collapse
Affiliation(s)
- Yingying Liu
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen 518020, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Yang L, Chen L, Fang Y, Ma S. Downregulation of GSK-3β Expression via Ultrasound-Targeted Microbubble Destruction Enhances Atherosclerotic Plaque Stability in New Zealand Rabbits. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:710-722. [PMID: 33261913 DOI: 10.1016/j.ultrasmedbio.2020.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/14/2020] [Accepted: 11/01/2020] [Indexed: 06/12/2023]
Abstract
Accumulating evidence suggests that atherosclerosis (AS) is the underlying cause of vascular diseases, including heart disease and stroke. Ultrasound-targeted microbubble destruction (UTMD) technology provides a tolerable, efficient and effective system for drug delivery and gene transfection, which has broad application prospects in the treatment of AS. In addition, glycogen synthase kinase (GSK)-3β has been implicated as a potentially valuable therapeutic agent for AS treatment; however, the specific molecular mechanisms remain unknown. Therefore, this study was conducted to explore the effect of downregulation of GSK-3β expression via UTMD on atherosclerotic plaque stability. We established a THP-1 macrophage-derived foam cell model in vitro and an atherosclerotic plaque model in the right common carotid artery of New Zealand rabbits. We determined levels of the relevant vulnerable plaque stability elements. The results indicate that GSK-3β was upregulated in the foam cells and in atherosclerotic rabbits. Downregulation of GSK-3β expression by UTMD suppressed vulnerable plaque factors and inflammation in vitro and in vivo, changed the cytoskeleton of the foam cells in vitro, increased Young's modulus and decreased the peak intensity of atherosclerotic plaque in vivo. Moreover, GSK-3β inhibition by UTMD did not influence the viability of the foam cells. Collectively, our results indicate that GSK-3β could be a potential target for anti-atherogenic interventions and, in particular, can improve the stability of AS plaques in combination with UTMD.
Collapse
Affiliation(s)
- Lifei Yang
- Department of Ultrasound, Ningbo Urology and Nephrology Hospital, Ningbo, China
| | - Lingzi Chen
- Ningbo University School of Medicine, Ningbo, China
| | - Ye Fang
- Department of Ultrasound, Ningbo Urology and Nephrology Hospital, Ningbo, China
| | - Suya Ma
- Department of Ultrasound, Ningbo Urology and Nephrology Hospital, Ningbo, China.
| |
Collapse
|
19
|
Kumar SU, Telichko AV, Wang H, Hyun D, Johnson EG, Kent MS, Rebhun RB, Dahl JJ, Culp WTN, Paulmurugan R. Acoustically Driven Microbubbles Enable Targeted Delivery of microRNA-Loaded Nanoparticles to Spontaneous Hepatocellular Neoplasia in Canines. ADVANCED THERAPEUTICS 2020; 3:2000120. [PMID: 33415184 PMCID: PMC7784952 DOI: 10.1002/adtp.202000120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Indexed: 01/16/2023]
Abstract
Spatially localized microbubble cavitation by ultrasound offers an effective means of altering permeability of natural barriers (i.e. blood vessel and cell membrane) in favor of nanomaterials accumulation in the target site. In this study, a clinically relevant, minimally invasive ultrasound guided therapeutic approach is investigated for targeted delivery of anticancer microRNA loaded PLGA-b-PEG nanoparticles to spontaneous hepatocellular neoplasia in a canine model. Quantitative assessment of the delivered microRNAs revealed prominent and consistent increase in miRNAs levels (1.5-to 2.3-fold increase (p<0.001)) in ultrasound treated tumor regions compared to untreated control regions. Immunohistology of ultrasound treated tumor tissue presented a clear evidence for higher amount of nanoparticles extravasation from the blood vessels. A distinct pattern of cytokine expression supporting CD8+ T cells mediated "cold-to-hot" tumor transition was evident in all patients. On the outset, proposed platform can enhance delivery of miRNA-loaded nanoparticles to deep seated tumors in large animals to enhance chemotherapy.
Collapse
Affiliation(s)
- Sukumar Uday Kumar
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Arsenii V Telichko
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Huaijun Wang
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Dongwoon Hyun
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Eric G Johnson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Michael S Kent
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Robert B Rebhun
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Jeremy J Dahl
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - William T N Culp
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| |
Collapse
|
20
|
Yang L, Ma J, Guan L, Mu Y. General Characteristics of Microbubble-Adenovirus Vectors Carrying Genes. Cell Mol Bioeng 2020; 14:201-208. [PMID: 33868500 DOI: 10.1007/s12195-020-00663-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/18/2020] [Indexed: 01/12/2023] Open
Abstract
Introduction Transferring genes safely, targeting cells and achieving efficient transfection are urgent problems in gene therapy that need to be solved. Combining microbubbles (MBs) and viruses to construct double vectors has become a promising approach for gene delivery. Understanding the characteristic performance of MBs that carry genes is key to promoting effective gene transfer. Therefore, in this study, we constructed MB-adenovirus vectors and discussed their general characteristics. Methods We constructed MB-adenovirus vectors carrying the chemokine (C-X-C motif) ligand 12 (Cxcl12) and bone morphogenetic protein-2 (Bmp2) genes (pAd-Cxcl12 and pAd-Bmp2, respectively) to explore the general characteristics of double vectors carrying genes. Results The MB-adenovirus vectors had stable physical properties, and no significant differences in diameter, concentration, or pH were noted compared with naked MBs (p > 0.05). Flow cytometry and RT-PCR were used to detect the gene-loading capacity of MBs. The gene-loading efficiency of MBs increased with increasing virus amounts and was highest (91%) when 10.0 µL of virus was added. Beyond 10.0 µL of added virus, the gene-loading efficiency of MBs decreased with the continuous addition of virus. The maximum amounts of pAd-Cxcl12 and pAd-Bmp2 in 100 µL of MBs were approximately 14 and 10 µL, respectively. Conclusions This study indicates that addition of an inappropriate viral load will result in low MB loading efficiency, and the maximum amount of genes loaded by MBs may differ based on the genes carried by the virus.
Collapse
Affiliation(s)
- Lingjie Yang
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, 830011 China.,Xinjiang Key Laboratory of Medical Animal Model Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Juan Ma
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, 830011 China.,Xinjiang Key Laboratory of Medical Animal Model Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Lina Guan
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, 830011 China.,Xinjiang Key Laboratory of Medical Animal Model Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yuming Mu
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, 830011 China.,Xinjiang Key Laboratory of Medical Animal Model Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
21
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|
22
|
Yang Y, Li Q, Guo X, Tu J, Zhang D. Mechanisms underlying sonoporation: Interaction between microbubbles and cells. ULTRASONICS SONOCHEMISTRY 2020; 67:105096. [PMID: 32278246 DOI: 10.1016/j.ultsonch.2020.105096] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 05/04/2023]
Abstract
The past several decades have witnessed great progress in "smart drug delivery", an advance technology that can deliver genes or drugs into specific locations of patients' body with enhanced delivery efficiency. Ultrasound-activated mechanical force induced by the interactions between microbubbles and cells, which can stimulate so-called "sonoporation" process, has been regarded as one of the most promising candidates to realize spatiotemporal-controllable drug delivery to selected regions. Both experimental and numerical studies were performed to get in-depth understanding on how the microbubbles interact with cells during sonoporation processes, under different impact parameters. The current work gives an overview of the general mechanism underlying microbubble-mediated sonoporation, and the possible impact factors (e.g., the properties of cavitation agents and cells, acoustical driving parameters and bubble/cell micro-environment) that could affect sonoporation outcomes. Finally, current progress and considerations of sonoporation in clinical applications are reviewed also.
Collapse
Affiliation(s)
- Yanye Yang
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Qunying Li
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiasheng Guo
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Juan Tu
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China.
| | - Dong Zhang
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China; The State Key Laboratory of Acoustics, Chinese Academy of Science, Beijing 10080, China
| |
Collapse
|
23
|
Kooiman K, Roovers S, Langeveld SAG, Kleven RT, Dewitte H, O'Reilly MA, Escoffre JM, Bouakaz A, Verweij MD, Hynynen K, Lentacker I, Stride E, Holland CK. Ultrasound-Responsive Cavitation Nuclei for Therapy and Drug Delivery. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1296-1325. [PMID: 32165014 PMCID: PMC7189181 DOI: 10.1016/j.ultrasmedbio.2020.01.002] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 05/03/2023]
Abstract
Therapeutic ultrasound strategies that harness the mechanical activity of cavitation nuclei for beneficial tissue bio-effects are actively under development. The mechanical oscillations of circulating microbubbles, the most widely investigated cavitation nuclei, which may also encapsulate or shield a therapeutic agent in the bloodstream, trigger and promote localized uptake. Oscillating microbubbles can create stresses either on nearby tissue or in surrounding fluid to enhance drug penetration and efficacy in the brain, spinal cord, vasculature, immune system, biofilm or tumors. This review summarizes recent investigations that have elucidated interactions of ultrasound and cavitation nuclei with cells, the treatment of tumors, immunotherapy, the blood-brain and blood-spinal cord barriers, sonothrombolysis, cardiovascular drug delivery and sonobactericide. In particular, an overview of salient ultrasound features, drug delivery vehicles, therapeutic transport routes and pre-clinical and clinical studies is provided. Successful implementation of ultrasound and cavitation nuclei-mediated drug delivery has the potential to change the way drugs are administered systemically, resulting in more effective therapeutics and less-invasive treatments.
Collapse
Affiliation(s)
- Klazina Kooiman
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Silke Roovers
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Simone A G Langeveld
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robert T Kleven
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Laboratory for Molecular and Cellular Therapy, Medical School of the Vrije Universiteit Brussel, Jette, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Meaghan A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Martin D Verweij
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands; Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Christy K Holland
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA; Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
24
|
Zheng L, Shen CL, Li JM, Ma YL, Yan N, Tian XQ, Zhao YZ. Assessment of the Preventive Effect Against Diabetic Cardiomyopathy of FGF1-Loaded Nanoliposomes Combined With Microbubble Cavitation by Ultrasound. Front Pharmacol 2020; 10:1535. [PMID: 31998132 PMCID: PMC6967235 DOI: 10.3389/fphar.2019.01535] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 11/27/2019] [Indexed: 12/27/2022] Open
Abstract
Acidic fibroblast growth factor (FGF1) has great potential in preventing diabetic cardiomyopathy. This study aimed to evaluate the preventive effect of FGF1-loaded nanoliposomes (FGF1-nlip) combined with ultrasound-targeted microbubble destruction (UTMD) on diabetic cardiomyopathy (DCM) using ultrasound examination. Nanoliposomes encapsulating FGF1 were prepared by reverse phase evaporation. DM model rats were established by intraperitoneal injection of streptozotocin (STZ), and different forms of FGF1 (FGF1 solution, FGF1-nlip, and FGF1-nlip+UTMD) were used for a 12-week intervention. According to the transthoracic echocardiography and velocity vector imaging (VVI) indexes, the LVEF, LVFS, and VVI indexes (Vs, Sr, SRr) in the FGF1-nlip+UTMD group were significantly higher than those in the DM model group and other FGF1 intervention groups. From the real-time myocardial contrast echocardiography (RT-MCE) indexes, the FGF1-nlip+UTMD group A and A×β showed significant differences from the DM model group and other FGF1 intervention groups. Cardiac catheter hemodynamic testing, CD31 immunohistochemical staining, and electron microscopy also confirmed the same conclusion. These results confirmed that the abnormalities, including myocardial dysfunction and perfusion impairment, could be suppressed to different extents by the twice weekly FGF1 treatments for 12 consecutive weeks (free FGF1, FGF1-nlip, and FGF1-nlip+UTMD), with the strongest improvements observed in the FGF1-nlip+UTMD group. In conclusion, the VVI and RT-MCE techniques can detect left ventricular systolic function and perfusion changes in DM rats, providing a more effective experimental basis for the early detection and treatment evaluation of DCM, which is of great significance for the prevention of DCM.
Collapse
Affiliation(s)
- Lei Zheng
- Department of Ultrasonography, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Department of Ultrasonography of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China.,Department of Ultrasonography, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chuan-Li Shen
- Department of Ultrasonography, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian-Min Li
- Department of Pathology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu-Lei Ma
- Department of Ultrasonography, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Department of Ultrasonography of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Ning Yan
- Department of Ultrasonography, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Department of Ultrasonography of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin-Qiao Tian
- Department of Ultrasonography, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Department of Ultrasonography of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of 6 Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou, China
| |
Collapse
|
25
|
Wang LY, Zheng SS. Advances in low-frequency ultrasound combined with microbubbles in targeted tumor therapy. J Zhejiang Univ Sci B 2019; 20:291-299. [PMID: 30932374 DOI: 10.1631/jzus.b1800508] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The development of low-frequency ultrasound imaging technology and the improvement of ultrasound contrast agent production technology mean that they play an increasingly important role in tumor therapy. The interaction between ultrasound and microbubbles and their biological effects can transfer and release microbubbles carrying genes and drugs to target tissues, mediate the apoptosis of tumor cells, and block the embolization of tumor microvasculature. With the optimization of ultrasound parameters, the development of targeted microbubbles, and the emergence of various composite probes with both diagnostic and therapeutic functions, low-frequency ultrasound combined with microbubble contrast agents will bring new hope for clinical tumor treatment.
Collapse
Affiliation(s)
- Li-Ying Wang
- Department of Ultrasound, Shaoxing Second Hospital, Shaoxing 312000, China
| | - Shu-Sen Zheng
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Health, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
26
|
Wang Y, Li X, Liu L, Liu B, Wang F, Chen C. Tissue Targeting and Ultrasound-Targeted Microbubble Destruction Delivery of Plasmid DNA and Transfection In Vitro. Cell Mol Bioeng 2019; 13:99-112. [PMID: 32030111 DOI: 10.1007/s12195-019-00597-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/27/2019] [Indexed: 02/03/2023] Open
Abstract
Introduction Ultrasound-targeted microbubble destruction (UTMD) has been shown a promising approach for target-specific gene delivery and treatment of many diseases in the past decade. To improve the therapeutic potential of UTMD, the gene carrier of microbubbles should possess adequate DNA condensation capability and (or) specific cell or tissue selectivity. The tissue-targeted and ultrasound-targeted cationic microbubbles were developed to meet gene therapy. Methods A tissue-targeted stearic acid-inserted cationic microbubbles (SCMBs) were prepared for ultrasound-targeted gene delivery. Branched PEI was modified with stearic acid and further mixed with 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) and biot-1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethylene glycol)-2000] (ammonium salt) (Biot-DSPE-PEG2000), intercellular adhesion molecule-1 (ICAM-1) antibody and plasmid DNA to prepare cationic microbubbles through ultrasonic hydration. The ICAM-1 antibody and plasmid DNA were expected to assemble to the surface of SCMBs via biotin-avidin interaction and electrostatic interaction, respectively. Results It was found that the SCMBs had higher zeta potential compared with neutral microbubbles (NMBs) and cationic microbubbles (CMBs). In contrast, DNA incorporated SCMBs4 showed negative potential, exhibiting good DNA-binding capacity. Confocal images showed that the HeLa cells were attached around by the SCMBs4 from the view of green fluorescence of fluorescein isothiocyanate-loaded IgG which conjugated to ICAM-1 antibody on their surface. After ultrasound treatment, HeLa cells treated with SCMBs exhibited slightly stronger red fluorescence under confocal laser scanning microscope, indicating a synergistic promotion for transfection efficiency. Conclusions This tissue- and ultrasound-targeted cationic microbubble demonstrated here showed a promising strategy for improving gene therapy in the future.
Collapse
Affiliation(s)
- Yue Wang
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, 518035 People's Republic of China
| | - Xiaoli Li
- Key Laboratory of Biomedical Materials and Implant Devices, Research Institute of Tsinghua University in Shenzhen, Nanshan Hi-new Technology and Industry Park, Shenzhen, 518057 Guangzhou People's Republic of China
| | - Lanlan Liu
- Key Laboratory of Biomedical Materials and Implant Devices, Research Institute of Tsinghua University in Shenzhen, Nanshan Hi-new Technology and Industry Park, Shenzhen, 518057 Guangzhou People's Republic of China
| | - Bingruo Liu
- Division of Engineering Science, University of Toronto, Toronto, M5S2E8 Canada
| | - Feng Wang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, 603 Jinsui Road, Xinxiang, 453002 Henan People's Republic of China
- Shenzhen Kangning Hospital & Shenzhen Mental Health Center, Shenzhen, 518003 People's Republic of China
| | - Changsheng Chen
- Key Laboratory of Biomedical Materials and Implant Devices, Research Institute of Tsinghua University in Shenzhen, Nanshan Hi-new Technology and Industry Park, Shenzhen, 518057 Guangzhou People's Republic of China
| |
Collapse
|