1
|
Zhang M, An Z, Jiang Y, Wei M, Li X, Wang Y, Wang H, Gong Y. Self-assembled redox-responsive BRD4 siRNA nanoparticles: fomulation and its in vitro delivery in gastric cancer cells. J Chemother 2024:1-15. [PMID: 38291982 DOI: 10.1080/1120009x.2024.2308980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024]
Abstract
With the development of newer biomarkers in the diagnosis of gastric cancer (GC), therapeutic targets are emerging and molecular-targeted therapy is in progress RNA interference has emerged as a promising method of gene targeting therapy. However, naked small interfering RNA (siRNA) is unstable and susceptible to degradation, so employing vectors for siRNA delivery is the focus of our research. Therefore, we developed LMWP modified PEG-SS-PEI to deliver siRNA targeting BRD4 (L-NPs/siBRD4) for GC therapy. L-NPs/siBRD4 were prepared by electrostatic interaction and characterized by dynamic light scattering (DLS) and transmission electron microscopy (TEM). The release characteristics, cellular uptake and intracellular localization were also investigated. The in vitro anticancer activity of the prepared nanoparticles was analysed by MTT, Transwell invasion and wound healing assay. Quantitative real time-polymerase chain reaction (qRT-PCR) and Western blot were used to detect the effect of gene silencing. The results showed that the optimal N/P was 30 and the prepared L-NPs/siBRD4 uniformly distributed in the system with a spherical and regular shape. L-NPs/siBRD4 exhibited an accelerated release in GSH-containing media from 12h to 24h. The uptake of L-NPs/siBRD4 was enhanced and mainly co-localized in the lysosomes. After 6h incubation, LMWP modified PEG-SS-PEI helped siRNA escape from the lysosomes and diffused into the cytoplasm. L-NPs/siBRD4 significantly inhibited the proliferation, migration and invasion of cells. This might be related with the silence of BRD4, then inhibition of PI3K/Akt and c-Myc. Our results demonstrate that L-NPs/siBRD4 are a novel delivery system with anticancer, which may provide a more effective strategy for GC treatment.
Collapse
Affiliation(s)
- Mengying Zhang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Zhonghua An
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Yiming Jiang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Meijiao Wei
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Xiangbo Li
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Yifan Wang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Hongbo Wang
- Gastrointestinal Surgery Department, Jimo District People's Hospital, Qingdao, China
| | - Yanling Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| |
Collapse
|
2
|
Jiao L, Sun Z, Sun Z, Liu J, Deng G, Wang X. Nanotechnology-based non-viral vectors for gene delivery in cardiovascular diseases. Front Bioeng Biotechnol 2024; 12:1349077. [PMID: 38303912 PMCID: PMC10830866 DOI: 10.3389/fbioe.2024.1349077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/08/2024] [Indexed: 02/03/2024] Open
Abstract
Gene therapy is a technique that rectifies defective or abnormal genes by introducing exogenous genes into target cells to cure the disease. Although gene therapy has gained some accomplishment for the diagnosis and therapy of inherited or acquired cardiovascular diseases, how to efficiently and specifically deliver targeted genes to the lesion sites without being cleared by the blood system remains challenging. Based on nanotechnology development, the non-viral vectors provide a promising strategy for overcoming the difficulties in gene therapy. At present, according to the physicochemical properties, nanotechnology-based non-viral vectors include polymers, liposomes, lipid nanoparticles, and inorganic nanoparticles. Non-viral vectors have an advantage in safety, efficiency, and easy production, possessing potential clinical application value when compared with viral vectors. Therefore, we summarized recent research progress of gene therapy for cardiovascular diseases based on commonly used non-viral vectors, hopefully providing guidance and orientation for future relevant research.
Collapse
Affiliation(s)
- Liping Jiao
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Zhuokai Sun
- Queen Mary School, Nanchang University, Nanchang, China
| | - Zhihong Sun
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jie Liu
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Guanjun Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Xiaozhong Wang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, China
| |
Collapse
|
3
|
Zhang B, Jiang X. Magnetic Nanoparticles Mediated Thrombolysis-A Review. IEEE OPEN JOURNAL OF NANOTECHNOLOGY 2023; 4:109-132. [PMID: 38111792 PMCID: PMC10727495 DOI: 10.1109/ojnano.2023.3273921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Nanoparticles containing thrombolytic medicines have been developed for thrombolysis applications in response to the increasing demand for effective, targeted treatment of thrombosis disease. In recent years, there has been a great deal of interest in nanoparticles that can be navigated and driven by a magnetic field. However, there are few review publications concerning the application of magnetic nanoparticles in thrombolysis. In this study, we examine the current state of magnetic nanoparticles in the application of in vitro and in vivo thrombolysis under a static or dynamic magnetic field, as well as the combination of magnetic nanoparticles with an acoustic field for dual-mode thrombolysis. We also discuss four primary processes of magnetic nanoparticles mediated thrombolysis, including magnetic nanoparticle targeting, magnetic nanoparticle trapping, magnetic drug release, and magnetic rupture of blood clot fibrin networks. This review will offer unique insights for the future study and clinical development of magnetic nanoparticles mediated thrombolysis approaches.
Collapse
Affiliation(s)
- Bohua Zhang
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695 USA
| | - Xiaoning Jiang
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695 USA
| |
Collapse
|
4
|
Awen A, Hu D, Gao D, Wang Z, Wu Y, Zheng H, Guan L, Mu Y, Sheng Z. Dual-modal molecular imaging and therapeutic evaluation of coronary microvascular dysfunction using indocyanine green-doped targeted microbubbles. Biomater Sci 2023; 11:2359-2371. [PMID: 36883518 DOI: 10.1039/d2bm02155b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Coronary microvascular dysfunction (CMD), which causes a series of cardiovascular diseases, seriously endangers human health. However, precision diagnosis of CMD is still challenging due to the lack of sensitive probes and complementary imaging technologies. Herein, we demonstrate indocyanine green-doped targeted microbubbles (named T-MBs-ICG) as dual-modal probes for highly sensitive near-infrared (NIR) fluorescence imaging and high-resolution ultrasound imaging of CMD in mouse models. In vitro results show that T-MBs-ICG can specifically target fibrin, a specific CMD biomarker, via the cysteine-arginine-glutamate-lysine-alanine (CREKA) peptide modified on the surface of microbubbles. We further employ T-MBs-ICG to achieve NIR fluorescence imaging of injured myocardial tissue in a CMD mouse model, leading to a signal-to-background ratio (SBR) of up to 50, which is 20 fold higher than that of the non-targeted group. Furthermore, ultrasound molecular imaging of T-MBs-ICG is obtained within 60 s after intravenous injection, providing molecular information on ventricular and myocardial structures and fibrin with a resolution of 1.033 mm × 0.466 mm. More importantly, we utilize comprehensive dual-modal imaging of T-MBs-ICG to evaluate the therapeutic efficacy of rosuvastatin, a cardiovascular drug for the clinical treatment of CMD. Overall, the developed T-MBs-ICG probes with good biocompatibility exhibit great potential in the clinical diagnosis of CMD.
Collapse
Affiliation(s)
- Alimina Awen
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, Xinjiang, 830011, P. R. China.
| | - Dehong Hu
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, P. R. China.
| | - Duyang Gao
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, P. R. China.
| | - Zihang Wang
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, Xinjiang, 830011, P. R. China.
| | - Yayun Wu
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, P. R. China.
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, P. R. China.
| | - Lina Guan
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, Xinjiang, 830011, P. R. China.
| | - Yuming Mu
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, Xinjiang, 830011, P. R. China.
| | - Zonghai Sheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, P. R. China.
| |
Collapse
|
5
|
Zhang N, Ru B, Hu J, Xu L, Wan Q, Liu W, Cai W, Zhu T, Ji Z, Guo R, Zhang L, Li S, Tong X. Recent advances of CREKA peptide-based nanoplatforms in biomedical applications. J Nanobiotechnology 2023; 21:77. [PMID: 36869341 PMCID: PMC9985238 DOI: 10.1186/s12951-023-01827-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Nanomedicine technology is a rapidly developing field of research and application that uses nanoparticles as a platform to facilitate the diagnosis and treatment of diseases. Nanoparticles loaded with drugs and imaging contrast agents have already been used in clinically, but they are essentially passive delivery carriers. To make nanoparticles smarter, an important function is the ability to actively locate target tissues. It enables nanoparticles to accumulate in target tissues at higher concentrations, thereby improving therapeutic efficacy and reducing side effects. Among the different ligands, the CREKA peptide (Cys-Arg-Glu-Lys-Ala) is a desirable targeting ligand and has a good targeting ability for overexpressed fibrin in different models, such as cancers, myocardial ischemia-reperfusion, and atherosclerosis. In this review, the characteristic of the CREKA peptide and the latest reports regarding the application of CREKA-based nanoplatforms in different biological tissues are described. In addition, the existing problems and future application perspectives of CREKA-based nanoplatforms are also addressed.
Collapse
Affiliation(s)
- Nannan Zhang
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Bin Ru
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Jiaqi Hu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Langhai Xu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Quan Wan
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Wenlong Liu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - WenJun Cai
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Tingli Zhu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Zhongwei Ji
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Ran Guo
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Lin Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| | - Shun Li
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| | - Xiangmin Tong
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
6
|
Sedighi M, Shrestha N, Mahmoudi Z, Khademi Z, Ghasempour A, Dehghan H, Talebi SF, Toolabi M, Préat V, Chen B, Guo X, Shahbazi MA. Multifunctional Self-Assembled Peptide Hydrogels for Biomedical Applications. Polymers (Basel) 2023; 15:1160. [PMID: 36904404 PMCID: PMC10007692 DOI: 10.3390/polym15051160] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Self-assembly is a growth mechanism in nature to apply local interactions forming a minimum energy structure. Currently, self-assembled materials are considered for biomedical applications due to their pleasant features, including scalability, versatility, simplicity, and inexpensiveness. Self-assembled peptides can be applied to design and fabricate different structures, such as micelles, hydrogels, and vesicles, by diverse physical interactions between specific building blocks. Among them, bioactivity, biocompatibility, and biodegradability of peptide hydrogels have introduced them as versatile platforms in biomedical applications, such as drug delivery, tissue engineering, biosensing, and treating different diseases. Moreover, peptides are capable of mimicking the microenvironment of natural tissues and responding to internal and external stimuli for triggered drug release. In the current review, the unique characteristics of peptide hydrogels and recent advances in their design, fabrication, as well as chemical, physical, and biological properties are presented. Additionally, recent developments of these biomaterials are discussed with a particular focus on their biomedical applications in targeted drug delivery and gene delivery, stem cell therapy, cancer therapy and immune regulation, bioimaging, and regenerative medicine.
Collapse
Affiliation(s)
- Mahsa Sedighi
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Birjand University of Medical Sciences, Birjand 9717853076, Iran
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853076, Iran
| | - Neha Shrestha
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
- Department of Biomedicine and Translational Research, Research Institute for Bioscience and Biotechnology, Kathmandu P.O. Box 7731, Nepal
| | - Zahra Mahmoudi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan 6517838636, Iran
| | - Zahra Khademi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Alireza Ghasempour
- Student Research Committee, Birjand University of Medical Sciences, Birjand 9717853076, Iran
| | - Hamideh Dehghan
- Student Research Committee, Birjand University of Medical Sciences, Birjand 9717853076, Iran
| | - Seyedeh Fahimeh Talebi
- Student Research Committee, Birjand University of Medical Sciences, Birjand 9717853076, Iran
| | - Maryam Toolabi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Bozhi Chen
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xindong Guo
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| |
Collapse
|
7
|
Zhang W, Li M, Wang X, Zhang W, Wang H, Li P, Tang B. Precision Navigation of Venous Thrombosis Guided by Viscosity-Activatable Near-Infrared Fluorescence. Anal Chem 2023; 95:2382-2389. [PMID: 36653196 DOI: 10.1021/acs.analchem.2c04395] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Thrombus are blood clots formed by abnormal hemostasis in blood vessels and are closely associated with various diseases such as pulmonary embolism, myocardial infarction and stroke. Early diagnosis and treatment of thrombus is the key to reducing the high risk of thrombotic disease. Given that early thrombus is small in early size, free instability, wide regional distribution and fast formation, it is urgent to develop all-inclusive detection methods that combine high signal-to-noise ratio, in situ dynamic and rapid in-depth tissue imaging. Near-infrared (NIR) fluorescence imaging, with its excellent high spatiotemporal resolution and tissue penetration depth, is a powerful technique for direct visualization of thrombotic events in situ. Considering the fibrin highly expressed in the thrombus is a typical thrombotic target. Moreover, the viscosity of the thrombus is markedly higher than its surroundings. Therefore, we developed a fibrin-targeting and viscosity-activating thrombus NIR fluorescent probe (TIR-V) for high-resolution and high-sensitivity in situ lighten-up thrombus. TIR-V has the advantages of good thrombus targeting, significant "off-on" fluorescence specific response to viscosity, bright NIR fluorescence and good biocompatibility. The thrombus is clearly delineated by a high signal-to-noise ratio NIR fluorescence imaging, enabling imaging detection and precise navigation of thrombotic regions. This work demonstrates the potential of TIR-V as a bifunctional probe for definitive diagnostic imaging and direct navigation of thrombotic lesions in clinical applications.
Collapse
Affiliation(s)
- Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Mengmei Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Xin Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
8
|
Luo Z, Wu S, Zhou J, Xu W, Xu Q, Lu L, Xie C, Liu Y, Lu W. All-stage targeted therapy for the brain metastasis from triple-negative breast cancer. Acta Pharm Sin B 2023; 13:359-371. [PMID: 36815053 PMCID: PMC9939358 DOI: 10.1016/j.apsb.2022.03.026] [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: 02/21/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/27/2022] Open
Abstract
Brain metastasis is a common and serious complication of breast cancer, which is commonly associated with poor survival and prognosis. In particular, the treatment of brain metastasis from triple-negative breast cancer (BM-TNBC) has to face the distinct therapeutic challenges from tumor heterogeneity, circulating tumor cells (CTCs), blood-brain barrier (BBB) and blood-tumor barrier (BTB), which is in unmet clinical needs. Herein, combining with the advantages of synthetic and natural targeting moieties, we develop a "Y-shaped" peptide pVAP-decorated platelet-hybrid liposome drug delivery system to address the all-stage targeted drug delivery for the whole progression of BM-TNBC. Inherited from the activated platelet, the hybrid liposomes still retain the native affinity toward CTCs. Further, the peptide-mediated targeting to breast cancer cells and transport across BBB/BTB are demonstrated in vitro and in vivo. The resultant delivery platform significantly improves the drug accumulation both in orthotopic breast tumors and brain metastatic lesions, and eventually exhibits an outperformance in the inhibition of BM-TNBC compared with the free drug. Overall, this work provides a promising prospect for the comprehensive treatment of BM-TNBC, which could be generalized to other cell types or used in imaging platforms in the future.
Collapse
Affiliation(s)
- Zimiao Luo
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Jianfen Zhou
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Weixia Xu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yu Liu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China,Minhang Branch, Zhongshan Hospital and Institute of Fudan-Minghang Academic Health System, Minghang Hospital, Fudan University, Shanghai 201199, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China,Corresponding author.
| |
Collapse
|
9
|
Recent advances in nanomedicines for imaging and therapy of myocardial ischemia-reperfusion injury. J Control Release 2023; 353:563-590. [PMID: 36496052 DOI: 10.1016/j.jconrel.2022.11.057] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
Myocardial ischemia-reperfusion injury (IRI) is becoming a typical cardiovascular disease with increasing worldwide incidence. It is usually induced by the restoration of normal blood flow to the ischemic myocardium after a period of recanalization and directly leads to myocardial damage. Notably, the pathological mechanism of myocardial IRI is closely related to inflammation, oxidative stress, Ca2+ overload, and the opening of mitochondrial permeability transition pore channels. Therefore, monitoring of these changes and imaging lesions is a key to timely clinical diagnosis. Nanomedicines have shown great value in the diagnosis and treatment of myocardial IRI, with advantages including passive/active targeting, prolonged circulation, improved bioavailability, versatile carrier selection, and synergistic integration of different imaging and therapeutic agents in single particles with the same pharmaceutics. Because theranostic nanomedicines for myocardial IRI have advanced rapidly, we conduct an updated review on this topic. The special focus is on how to rationally design the nanomedicines to achieve optimal imaging and therapy. We hope this review would stimulate the interest of researchers with different backgrounds and expedite the development of nanomedicines for myocardial IRI.
Collapse
|
10
|
Fatima M, Abourehab MAS, Aggarwal G, Jain GK, Sahebkar A, Kesharwani P. Advancement of cell-penetrating peptides in combating triple-negative breast cancer. Drug Discov Today 2022; 27:103353. [PMID: 36099963 DOI: 10.1016/j.drudis.2022.103353] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/23/2022] [Accepted: 09/06/2022] [Indexed: 11/03/2022]
Abstract
Extensive research efforts have been made and are still ongoing in the search for an ideal anti-cancer therapy. Almost all chemotherapeutics require a carrier or vehicle, a drug delivery system that can transport the drug specifically to the targeted cancer cells, sparing normal cells. Cell-penetrating peptides (CPPs) provide an effective and efficient pathway for the intra-cellular transportation of various bioactive molecules in several biomedical therapies. They are now well-recognized as facilitators of intracellular cargo delivery and have excellent potential for targeted anti-cancer therapy. In this review, we explain CPPs, recent progress in the development of new CPPs, and their utilization to transport cargoes such as imaging agents, chemotherapeutics, and short-interfering RNAs (siRNA) into tumor cells, contributing to the advancement of novel tumor-specific delivery systems.
Collapse
Affiliation(s)
- Mahak Fatima
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110 062, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia 61519, Egypt
| | - Geeta Aggarwal
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi 110 017, India
| | - Gaurav K Jain
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi 110 017, India
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110 062, India.
| |
Collapse
|
11
|
El Hauadi K, Resina L, Zanuy D, Esteves T, Ferreira FC, Pérez-Madrigal MM, Alemán C. Dendritic Self-assembled Structures from Therapeutic Charged Pentapeptides. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:12905-12914. [PMID: 36229043 PMCID: PMC9988208 DOI: 10.1021/acs.langmuir.2c02010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/04/2022] [Indexed: 06/16/2023]
Abstract
CRENKA [Cys-Arg-(NMe)Glu-Lys-Ala, where (NMe)Glu refers to N-methyl-Glu], an anti-cancer pentapeptide that induces prostate tumor necrosis and significant reduction in tumor growth, was engineered to increase the resistance to endogenous proteases of its parent peptide, CREKA (Cys-Arg-Glu-Lys-Ala). Considering their high tendency to aggregate, the self-assembly of CRENKA and CREKA into well-defined and ordered structures has been examined as a function of peptide concentration and pH. Spectroscopic studies and atomistic molecular dynamics simulations reveal significant differences between the secondary structures of CREKA and CRENKA. Thus, the restrictions imposed by the (NMe)Glu residue reduce the conformational variability of CRENKA with respect to CREKA, which significantly affects the formation of well-defined and ordered self-assembly morphologies. Aggregates with poorly defined morphology are obtained from solutions with low and moderate CREKA concentrations at pH 4, whereas well-defined dendritic microstructures with fractal geometry are obtained from CRENKA solutions with similar peptide concentrations at pH 4 and 7. The formation of dendritic structures is proposed to follow a two-step mechanism: (1) pseudo-spherical particles are pre-nucleated through a diffusion-limited aggregation process, pre-defining the dendritic geometry, and (2) such pre-nucleated structures coalesce by incorporating conformationally restrained CRENKA molecules from the solution to their surfaces, forming a continuous dendritic structure. Instead, no regular assembly is obtained from solutions with high peptide concentrations, as their dynamics is dominated by strong repulsive peptide-peptide electrostatic interactions, and from solutions at pH 10, in which the total peptide charge is zero. Overall, results demonstrate that dendritic structures are only obtained when the molecular charge of CRENKA, which is controlled through the pH, favors kinetics over thermodynamics during the self-assembly process.
Collapse
Affiliation(s)
- Karima El Hauadi
- Departament
d’Enginyeria Química and Barcelona Research Center for
Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany 10-14, Barcelona 08019, Spain
| | - Leonor Resina
- Departament
d’Enginyeria Química and Barcelona Research Center for
Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany 10-14, Barcelona 08019, Spain
- Department
of Bioengineering, iBB − Institute for Bioengineering and Biosciences,
Instituto Superior Técnico, Universidade
de Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
- Associate
Laboratory i4HB—Institute for Health and Bioeconomy at Instituto
Superior Técnico, Universidade de
Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
| | - David Zanuy
- Departament
d’Enginyeria Química and Barcelona Research Center for
Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany 10-14, Barcelona 08019, Spain
| | - Teresa Esteves
- Department
of Bioengineering, iBB − Institute for Bioengineering and Biosciences,
Instituto Superior Técnico, Universidade
de Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
- Associate
Laboratory i4HB—Institute for Health and Bioeconomy at Instituto
Superior Técnico, Universidade de
Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
| | - Frederico Castelo Ferreira
- Department
of Bioengineering, iBB − Institute for Bioengineering and Biosciences,
Instituto Superior Técnico, Universidade
de Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
- Associate
Laboratory i4HB—Institute for Health and Bioeconomy at Instituto
Superior Técnico, Universidade de
Lisboa, Avenida Rovisco Pais 1, Lisboa 1049-001, Portugal
| | - Maria M. Pérez-Madrigal
- Departament
d’Enginyeria Química and Barcelona Research Center for
Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany 10-14, Barcelona 08019, Spain
| | - Carlos Alemán
- Departament
d’Enginyeria Química and Barcelona Research Center for
Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany 10-14, Barcelona 08019, Spain
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, Barcelona 08028, Spain
| |
Collapse
|
12
|
Fibrin-Targeted Nanoparticles for Finding, Visualizing and Characterizing Blood Clots in Acute Ischemic Stroke. Pharmaceutics 2022; 14:pharmaceutics14102156. [PMID: 36297588 PMCID: PMC9606925 DOI: 10.3390/pharmaceutics14102156] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/29/2022] Open
Abstract
Recanalization of the occluded artery is the gold standard treatment for acute ischemic stroke, which includes enzymatic fibrinolytic treatment with the use of recombinant tissue plasminogen activators (rtPAs) to disrupt the occluding clot, the use of mechanical thrombectomy to physically remove the clot, or a combination of both. Fibrin is one of the main components of blood clots causing ischemic stroke and is the target of rtPA upon activation of plasminogen in the clot. In addition, fibrin content also influences the efficacy of mechanical thrombectomy. Current imaging methods can successfully identify occlusions in large vessels; however, there is still a need for contrast agents capable of visualizing small thrombi in ischemic stroke patients. In this work, we describe the synthesis and the in vitro characterization of a new diagnostic nanoparticle, as well as the in vivo evaluation in an animal model of thromboembolic stroke. Gd-labeled KCREKA peptides were synthesized and attached onto the surface of PEGylated superparamagnetic nanoparticles. Magnetic resonance imaging (MRI) of blood clots was performed in vitro and in vivo in animal models of thromboembolic stroke. KCREKA-NPs were synthesized by attaching the peptide to the amino (N) termini of the PEG-NPs. The sizes of the nanoparticles, measured via DLS, were similar for both KCREKA-NPs and PEG-NPs (23 ± 4 nm, PDI = 0.11 and 25 ± 8 nm, PDI = 0.24, respectively). In the same line, r2 relaxivities were also similar for the nanoparticles (149 ± 2 mM Fe s−1 and 151 ± 5 mM Fe s−1), whereas the r1 relaxivity was higher for KCREKA-NPs (1.68 ± 0.29 mM Fe s−1 vs. 0.69 ± 0.3 mM Fe s−1). In vitro studies showed that blood clots with low coagulation times were disrupted by rtPA, whereas aged clots were almost insensitive to the presence of rtPA. MRI in vitro studies showed a sharp decrease in the T1 × T2 signals measured for aged clots incubated with KCREKA-NPs compared with fresh clots (47% [22, 80] to 26% [15, 51]). Furthermore, the control blood showed a higher value of the T1 × T2 signal (39% [20, 61]), being the blood clots with low coagulation times the samples with the lowest values measured by MRI. In vivo studies showed a significant T1 × T2 signal loss in the clot region of 24% after i.v. injection of KCREKA-NPs. The thrombus age (2.5% ± 6.1% vs. 81.3% ± 19.8%, p < 0.01) confirmed our ability to identify in vivo fresh blood clots. In this study, we developed and tested a dual MRI nanoparticle, acting as T1 and T2 contrast agents in MRI analyses. The developed KCREKA-NPs showed affinity for the fibrin content of blood clots, and the MRI signals provided by the nanoparticles showed significant differences depending on the clot age. The developed KCREKA-NPs could be used as a tool to predict the efficacy of a recanalization treatment and improve the triage of ischemic stroke patients.
Collapse
|
13
|
Ai Y, He M, Wan C, Luo H, Xin H, Wang Y, Liang Q. Nanoplatform‐Based Reactive Oxygen Species Scavengers for Therapy of Ischemia‐Reperfusion Injury. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yongjian Ai
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology Tsinghua University‐Peking University Joint Centre for Life Sciences Beijing Key Lab of Microanalytical Methods & Instrumentation Department of Chemistry Center for Synthetic and Systems Biology Tsinghua University Beijing 100084 P. R. China
| | - Meng‐Qi He
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology Tsinghua University‐Peking University Joint Centre for Life Sciences Beijing Key Lab of Microanalytical Methods & Instrumentation Department of Chemistry Center for Synthetic and Systems Biology Tsinghua University Beijing 100084 P. R. China
| | - Chengxian Wan
- Jiangxi Provincial People's Hospital The First Affiliated Hospital of Nanchang Medical College The Affiliated People's Hospital of Nanchang University Nanchang Jiangxi 330006 P. R. China
| | - Hua Luo
- State Key Laboratory of Quality Research in Chinese Medicine Institute of Chinese Medical Sciences University of Macau Macau SAR 999078 China
| | - Hongbo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies Institute of Translational Medicine Nanchang University Nanchang Jiangxi 330088 P. R. China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine Institute of Chinese Medical Sciences University of Macau Macau SAR 999078 China
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology Tsinghua University‐Peking University Joint Centre for Life Sciences Beijing Key Lab of Microanalytical Methods & Instrumentation Department of Chemistry Center for Synthetic and Systems Biology Tsinghua University Beijing 100084 P. R. China
| |
Collapse
|
14
|
Zhang W, Wang J, Xie Z, Zou H, Chen Q, Xu L, Hu L, Fang N, Xu J, Zhou J, Liu J, Ran H, Wang Z, Zhang Y, Guo D. Antithrombotic Therapy by Regulating the ROS-Mediated Thrombosis Microenvironment and Specific Nonpharmaceutical Thrombolysis Using Prussian Blue Nanodroplets. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106252. [PMID: 35246943 DOI: 10.1002/smll.202106252] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/30/2022] [Indexed: 06/14/2023]
Abstract
In thrombotic diseases, the effects of reactive oxygen species (ROS)-mediated oxidative stress as a "perpetrator" in thrombosis must be resolved. Accordingly, an insufficient understanding of thrombus therapy prompted the authors to pursue a more comprehensive and efficient antithrombotic treatment strategy. A Prussian blue (PB)-based nanodroplet system (PB-PFP@PC) is designed using PB and perfluorinated pentane (PFP) in the core, and a targeting peptide (CREKA, Cys-Arg-Glu-Lys-Ala) is attached to poly(lactic-coglycolic acid) (PLGA) as the delivery carrier shell. Upon near-infrared (NIR) laser irradiation, PB and PFP jointly achieve an unprecedented dual strategy for drug-free thrombolysis: photothermal therapy (PTT) combined with optical droplet vaporization (ODV). PB, a nanoenzyme, also regulates the vascular microenvironment via its antioxidant activity to continuously scavenge abnormally elevated ROS and correspondingly reduce inflammatory factors in the thrombus site. This study provides a demonstration of not only the potential of ODV in thrombus therapy but also the mechanism underlying PTT thrombolysis due to thermal ablation-induced fibrin network structural damage. Moreover, PB catalyzes ROS to generate oxygen (O2 ), which combines with the ODV effect, enhancing the ultrasound signal. Thus, regulation of the thrombosis microenvironment combined with specific nonpharmaceutical thrombolysis by PB nanodroplets provides a more comprehensive and efficient antithrombotic therapeutic strategy.
Collapse
Affiliation(s)
- Wenli Zhang
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Junrui Wang
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Zhuoyan Xie
- Department of Ultrasound, Chongqing General Hospital of Chinese Academy of Sciences, Chongqing, 401121, China
| | - Hongmi Zou
- Department of Ophthalmology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Qiaoqi Chen
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Lian Xu
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Liu Hu
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Ni Fang
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Jie Xu
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Jun Zhou
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Jia Liu
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Yu Zhang
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Dajing Guo
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| |
Collapse
|
15
|
Zhong Y, Ye M, Huang L, Hu L, Li F, Ni Q, Zhong J, Wu H, Xu F, Xu J, He X, Wang Z, Ran H, Wu Y, Guo D, Liang XJ. A Fibrin Site-Specific Nanoprobe for Imaging Fibrin-Rich Thrombi and Preventing Thrombus Formation in Venous Vessels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109955. [PMID: 35194836 DOI: 10.1002/adma.202109955] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/11/2022] [Indexed: 06/14/2023]
Abstract
Venous thromboembolism (VTE) is a prevalent public health issue worldwide. Before treatment, spatiotemporally accurate thrombus detection is essential. However, with the currently available imaging technologies, this is challenging. Herein, the development of a novel fibrin-specific nanoprobe (NP) based on the conjugation of poly(lactic-co-glycolic acid) with the pentapeptide Cys-Arg-Glu-Lys-Ala (CREKA) for selective and semiquantitative imaging in vivo is presented. By integrating Fe3 O4 and NIR fluorochrome (IR780), the NP can function as a highly sensitive sensor for the direct analysis of thrombi in vivo. The fibrin-specific NP distinguishes fibrin-rich thrombi from collagen-rich or erythrocyte-rich thrombi, which can be beneficial for future individually tailored therapeutic strategy. Furthermore, loading NPs with the ketotifen fumarate results in mast cell degranulation inhibition, and hence, NPs can prevent thrombosis without the risk of excessive bleeding. Thus, the use of fibrin-specific NPs may serve as a safe alternative approach for the detection and prevention of VTEs in susceptible populations in the future.
Collapse
Affiliation(s)
- Yixin Zhong
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Key Laboratory of Ultrasound Molecular Imaging & Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, P. R. China
| | - Man Ye
- Department of Radiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Rd, Wuchang District, Wuhan, Hubei, 430060, P. R. China
| | - Liandi Huang
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Key Laboratory of Ultrasound Molecular Imaging & Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, P. R. China
| | - Liu Hu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, P. R. China
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Qiankun Ni
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jie Zhong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hongyun Wu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, P. R. China
| | - Fengfei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jie Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, P. R. China
| | - Xiaojing He
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, P. R. China
| | - Zhigang Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Key Laboratory of Ultrasound Molecular Imaging & Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, P. R. China
| | - Haitao Ran
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Key Laboratory of Ultrasound Molecular Imaging & Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, P. R. China
| | - Yunzhu Wu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, P. R. China
| | - Dajing Guo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Rd, Yuzhong District, Chongqing, 400010, P. R. China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
16
|
Hu Q, Fang Z, Ge J, Li H. Nanotechnology for cardiovascular diseases. Innovation (N Y) 2022; 3:100214. [PMID: 35243468 PMCID: PMC8866095 DOI: 10.1016/j.xinn.2022.100214] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/30/2022] [Accepted: 01/30/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiovascular diseases have become the major killers in today's world, among which coronary artery diseases (CADs) make the greatest contributions to morbidity and mortality. Although state-of-the-art technologies have increased our knowledge of the cardiovascular system, the current diagnosis and treatment modalities for CADs still have limitations. As an emerging cross-disciplinary approach, nanotechnology has shown great potential for clinical use. In this review, recent advances in nanotechnology in the diagnosis of CADs will first be elucidated. Both the sensitivity and specificity of biosensors for biomarker detection and molecular imaging strategies, such as magnetic resonance imaging, optical imaging, nuclear scintigraphy, and multimodal imaging strategies, have been greatly increased with the assistance of nanomaterials. Second, various nanomaterials, such as liposomes, polymers (PLGA), inorganic nanoparticles (AuNPs, MnO2, etc.), natural nanoparticles (HDL, HA), and biomimetic nanoparticles (cell-membrane coating) will be discussed as engineered as drug (chemicals, proteins, peptides, and nucleic acids) carriers targeting pathological sites based on their optimal physicochemical properties and surface modification potential. Finally, some of these nanomaterials themselves are regarded as pharmaceuticals for the treatment of atherosclerosis because of their intrinsic antioxidative/anti-inflammatory and photoelectric/photothermal characteristics in a complex plaque microenvironment. In summary, novel nanotechnology-based research in the process of clinical transformation could continue to expand the horizon of nanoscale technologies in the diagnosis and therapy of CADs in the foreseeable future.
Collapse
Affiliation(s)
- Qinqin Hu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zheyan Fang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Hua Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Zhang X, Zhang J, Zhang Y, Zhang Y, Hou T, Wang S. Homing peptide combined with DNAzyme-based ELISA-like assay for highly specific and sensitive detection of fibrin. Talanta 2022; 238:122995. [PMID: 34857328 DOI: 10.1016/j.talanta.2021.122995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/17/2022]
Abstract
A highly sensitive and specific ELISA-like chemiluminescence method for detection of fibrin has been developed. In the sensing platform, the homing peptide (CREKA), as recognition molecule, which can specially recognize the fibrin on microtiter plate, combined with G-quadruplex-based DNAzyme to form the probe of G-quadruplex-hemin DNAzyme-CREKA. After the sample solution was coated on the plates, the probe was crosslinked with fibrin through the interaction of CREKA and fibrin. Finally, luminol-H2O2 chemiluminesecence (CL) reaction was exploited for quantitative analysis of fibrin. The liner range for fibrin detection was from 0.112 pmol L-1 to 5.6 pmol L-1 with the detection limit of fibrin as low as 0.04 pmol L-1, based on a signal-to-noise ratio (S/N) of 3. Furthermore, on the basis of the high amplification efficiency of the rolling circle amplification (RCA) reaction, the method enabled to analyze fibrin with a detection limit corresponding to 0.06 fmol L-1, whose sensitivity increased 3 orders of magnitude than that of above method in the absence of RCA reaction. In particular, combined with the separation and washing steps of ELISA, the proposed method possessed higher selectivity, high-throughput and low cost, which shows promise for applications in clinical diagnosis.
Collapse
Affiliation(s)
- Xifang Zhang
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252000, China
| | - Jinrong Zhang
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252000, China
| | - Yuanfu Zhang
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252000, China.
| | - Yinghong Zhang
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252000, China
| | - Tingting Hou
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252000, China
| | - Shuhao Wang
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252000, China.
| |
Collapse
|
18
|
Tiwari A, Elgrably B, Saar G, Vandoorne K. Multi-Scale Imaging of Vascular Pathologies in Cardiovascular Disease. Front Med (Lausanne) 2022; 8:754369. [PMID: 35071257 PMCID: PMC8766766 DOI: 10.3389/fmed.2021.754369] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Cardiovascular disease entails systemic changes in the vasculature. The endothelial cells lining the blood vessels are crucial in the pathogenesis of cardiovascular disease. Healthy endothelial cells direct the blood flow to tissues as vasodilators and act as the systemic interface between the blood and tissues, supplying nutrients for vital organs, and regulating the smooth traffic of leukocytes into tissues. In cardiovascular diseases, when inflammation is sensed, endothelial cells adjust to the local or systemic inflammatory state. As the inflamed vasculature adjusts, changes in the endothelial cells lead to endothelial dysfunction, altered blood flow and permeability, expression of adhesion molecules, vessel wall inflammation, thrombosis, angiogenic processes, and extracellular matrix production at the endothelial cell level. Preclinical multi-scale imaging of these endothelial changes using optical, acoustic, nuclear, MRI, and multimodal techniques has progressed, due to technical advances and enhanced biological understanding on the interaction between immune and endothelial cells. While this review highlights biological processes that are related to changes in the cardiac vasculature during cardiovascular diseases, it also summarizes state-of-the-art vascular imaging techniques. The advantages and disadvantages of the different imaging techniques are highlighted, as well as their principles, methodologies, and preclinical and clinical applications with potential future directions. These multi-scale approaches of vascular imaging carry great potential to further expand our understanding of basic vascular biology, to enable early diagnosis of vascular changes and to provide sensitive diagnostic imaging techniques in the management of cardiovascular disease.
Collapse
Affiliation(s)
- Ashish Tiwari
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Betsalel Elgrably
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Galit Saar
- Biomedical Core Facility, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Katrien Vandoorne
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
19
|
Gazaille C, Sicot M, Saulnier P, Eyer J, Bastiat G. Local Delivery and Glioblastoma: Why Not Combining Sustained Release and Targeting? FRONTIERS IN MEDICAL TECHNOLOGY 2022; 3:791596. [PMID: 35047971 PMCID: PMC8757870 DOI: 10.3389/fmedt.2021.791596] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma is one of the most aggressive brain tumors and is associated with a very low overall median survival despite the current treatment. The standard of care used in clinic is the Stupp's protocol which consists of a maximal resection of the tumor when possible, followed by radio and chemotherapy using temozolomide. However, in most cases, glioblastoma cells infiltrate healthy tissues and lead to fatal recurrences. There are a lot of hurdles to overcome in the development of new therapeutic strategies such as tumor heterogeneity, cell infiltration, alkylating agent resistance, physiological barriers, etc., and few treatments are on the market today. One of them is particularly appealing because it is a local therapy, which does not bring additional invasiveness since tumor resection is included in the gold standard treatment. They are implants: the Gliadel® wafers, which are deposited post-surgery. Nevertheless, in addition to presenting important undesirable effects, it does not bring any major benefit in the therapy despite the strategy being particularly attractive. The purpose of this review is to provide an overview of recent advances in the development of innovative therapeutic strategies for glioblastoma using an implant-type approach. The combination of this local strategy with effective targeting of the tumor microenvironment as a whole, also developed in this review, may be of interest to alleviate some of the obstacles encountered in the treatment of glioblastoma.
Collapse
Affiliation(s)
| | - Marion Sicot
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, Angers, France
| | | | - Joël Eyer
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, Angers, France
| | | |
Collapse
|
20
|
Qin H, Teng R, Liu Y, Li J, Yu M. Drug Release from Gelsolin-Targeted Phase-Transition Nanoparticles Triggered by Low-Intensity Focused Ultrasound. Int J Nanomedicine 2022; 17:61-71. [PMID: 35023919 PMCID: PMC8747719 DOI: 10.2147/ijn.s341421] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/26/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose Current strategies for tumour-induced sentinel lymph node detection and metastasis therapy have limitations. It is essential to identify and provide warnings earlier for tumour metastasis to carry out effective clinical interventions. In addition, traditional cancer chemotherapy encounters drastic limitations due to the nonspecific delivery of antitumour drugs and severe side effects. We aimed to exploit the potential of gelsolin (GSN) monoclonal antibody as a targeting agent and perfluorohexane (PFH) as a phase-transition agent to maximize the cytotoxic effect of poly(lactic-co-glycolic acid) (PLGA) nanoparticle-based drug controllable release systems for Hca-F cells. Methods We co-encapsulated PFH and doxorubicin (DOX) into PLGA nanoparticles (NPs) and further conjugated GSN monoclonal antibody onto the surface of NPs to form GSN-targeted phase transition polymer NPs (GSN-PLGA-PFH-DOX) for both imaging and therapy of tumours and metastatic lymph nodes. To promote and trigger drug release on demand, low-intensity focused ultrasound (LIFU) was applied to achieve a controllable release of the encapsulated drug. Results GSN-PLGA-PFH-DOX NPs exhibited characteristics such as a narrow size distribution and smooth surface. GSN-PLGA-PFH-DOX NPs could also specifically bind to Hca-F cells and increase the ultrasound contrast agent (UCA) image contrast intensity. GSN-PLGA-PFH-DOX NPs enable GSN-mediated targeting and biotherapeutic effects as well as LIFU-responsive drug release, resulting in synergistic cytotoxic effects in GSN-overexpressing cells in vitro. Conclusion Our work might provide a strategy for the imaging and chemotherapy of primary tumours and their metastases.
Collapse
Affiliation(s)
- Haocheng Qin
- Department of Ultrasound, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, People's Republic of China
| | - Rong Teng
- Department of Ultrasound, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, People's Republic of China
| | - Yan Liu
- Department of Ultrasound, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, People's Republic of China
| | - Juan Li
- Department of Oncology, The Second People's Hospital of Lianyungang, Lianyungang, 222023, People's Republic of China
| | - Ming Yu
- Department of Ultrasound, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, People's Republic of China
| |
Collapse
|
21
|
Wang Z, Huang H, Chen Y, Zheng Y. Current Strategies for Microbubble-Based Thrombus Targeting: Activation-Specific Epitopes and Small Molecular Ligands. Front Bioeng Biotechnol 2021; 9:699450. [PMID: 34336810 PMCID: PMC8322734 DOI: 10.3389/fbioe.2021.699450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/22/2021] [Indexed: 11/26/2022] Open
Abstract
Microbubbles with enhanced ultrasound represent a potentially potent evolution to the administration of a free drug in the treatment of thrombotic diseases. Conformational and expressional changes of several thrombotic biological components during active coagulation provide epitopes that allow site-specific delivery of microbubble-based agents to the thrombus for theranostic purpose. Through the interaction with these epitopes, emerging high-affinity small molecular ligands are able to selectively target the thrombi with tremendous advantages over traditional antibody-based strategy. In this mini-review, we summarize recent novel strategies for microbubble-based targeting of thrombus through epitopes located at activated platelets and fibrin. We also discuss the challenges of current targeting modalities and supramolecular carrier systems for their translational use in thrombotic pathologies.
Collapse
Affiliation(s)
- Zhaojian Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Huaigu Huang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Yuexin Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Yuehong Zheng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
22
|
Liu M, Wu C, Ke L, Li Z, Wu YL. Emerging Biomaterials-Based Strategies for Inhibiting Vasculature Function in Cancer Therapy. SMALL METHODS 2021; 5:e2100347. [PMID: 34927997 DOI: 10.1002/smtd.202100347] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Indexed: 06/14/2023]
Abstract
The constant feeding of oxygen and nutrients through the blood vasculature has a vital role in maintaining tumor growth. Interestingly, recent endeavors have shown that nanotherapeutics with the strategy to block tumor blood vessels feeding nutrients and oxygen for starvation therapy can be helpful in cancer treatment. However, this field has not been detailed. Hence, this review will present an exhaustive summary of the existing biomaterial based strategies to disrupt tumor vascular function for effective cancer treatment, including hydrogel or nanogel-mediated local arterial embolism, thrombosis activator loaded nano-material-mediated vascular occlusion and anti-vascular drugs that block tumor vascular function, which may be beneficial to the design of anti-cancer nanomedicine by targeting the tumor vascular system.
Collapse
Affiliation(s)
- Minting Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Lingjie Ke
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Zhiguo Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
23
|
Meng B, Li Y, Ding Y, Xu X, Wang L, Guo B, Zhu B, Zhang J, Xiang L, Dong J, Liu M, Xiang L, Xiang G. Myeloid-derived growth factor inhibits inflammation and alleviates endothelial injury and atherosclerosis in mice. SCIENCE ADVANCES 2021; 7:7/21/eabe6903. [PMID: 34020949 PMCID: PMC8139583 DOI: 10.1126/sciadv.abe6903] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 03/31/2021] [Indexed: 05/26/2023]
Abstract
Whether bone marrow modulates systemic metabolism remains unknown. Here, we found that (i) myeloid cell-specific myeloid-derived growth factor (MYDGF) deficiency exacerbated vascular inflammation, adhesion responses, endothelial injury, and atherosclerosis in vivo. (ii) Myeloid cell-specific MYDGF restoration attenuated vascular inflammation, adhesion responses and leukocyte homing and alleviated endothelial injury and atherosclerosis in vivo. (iii) MYDGF attenuated endothelial inflammation, apoptosis, permeability, and adhesion responses induced by palmitic acid in vitro. (iv) MYDGF alleviated endothelial injury and atherosclerosis through mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4)/nuclear factor κB (NF-κB) signaling. Therefore, we concluded that MYDGF inhibits endothelial inflammation and adhesion responses, blunts leukocyte homing, protects against endothelial injury and atherosclerosis in a manner involving MAP4K4/NF-κB signaling, and serves as a cross-talk factor between bone marrow and arteries to regulate the pathophysiology of arteries. Bone marrow functions as an endocrine organ and serves as a potential therapeutic target for metabolic disorders.
Collapse
Affiliation(s)
- Biying Meng
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
- The First School of Clinical Medicine, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Yixiang Li
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Yan Ding
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
- The First School of Clinical Medicine, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Xiaoli Xu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Li Wang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
- The First School of Clinical Medicine, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Bei Guo
- The First School of Clinical Medicine, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Biao Zhu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Jiajia Zhang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Lin Xiang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Jing Dong
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Min Liu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Lingwei Xiang
- ICF, 2635 Century Pkwy NE Unit 1000, Atlanta, GA 30345, USA.
| | - Guangda Xiang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China.
- The First School of Clinical Medicine, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| |
Collapse
|
24
|
Zhang Y, Chen X, Liu L, Tian J, Hao L, Ran HT. Photoacoustic Imaging of Myocardial Infarction Region Using Non-Invasive Fibrin-Targeted Nanoparticles in a Rat Myocardial Ischemia-Reperfusion Model. Int J Nanomedicine 2021; 16:1331-1344. [PMID: 33628023 PMCID: PMC7898240 DOI: 10.2147/ijn.s293736] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/30/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Myocardial infarction (MI) is a serious threat to public health. The early identification of MI is important to promote appropriate treatment strategies for patients. Recently, strategies targeting extracellular matrix (ECM) components have gained attention. Fibrin is an ECM protein involved after MI. In this work, we constructed fibrin-targeted nanoparticles (NPs) by co-assembling a fibrin-targeted peptide (CREKA) and indocyanine green (ICG) and used them to enhance photoacoustic (PA) imaging for noninvasive detection of the infarct region to help diagnose MI. METHODS ICG NPs modified with CREKA were prepared (CREKA-ICG-LIP NPs). Then, the fundamental characteristics, stability, safety, and targeting ability of the NPs were detected. Finally, in an ischemia-reperfusion (IR) injury model, the performance of the NPs in detecting the infarct region in the model on PA imaging was evaluated. RESULTS CREKA-ICG-LIP NPs were successfully constructed and showed excellent basic characteristics, a high safety level, and an excellent targeting ability. After intravenous injection, the CREKA-ICG-LIP NPs accumulated in the injured region in the IR model. Then, the PA signal in the infarct region could be detected by the ultrasound transducer of the Vevo LAZR Photoacoustic Imaging System. CONCLUSION This work provides new insights for non-invasive, real-time imaging techniques to detect the region of myocardial injury and help diagnose MI based on a PA imaging system with high sensitivity in optical imaging and deep penetration in ultrasound imaging.
Collapse
Affiliation(s)
- Yanan Zhang
- Department of Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014, People’s Republic of China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, 400014, People’s Republic of China
| | - Xiajing Chen
- Department of Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014, People’s Republic of China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, 400014, People’s Republic of China
| | - Lingjuan Liu
- Department of Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014, People’s Republic of China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, 400014, People’s Republic of China
| | - Jie Tian
- Department of Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014, People’s Republic of China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, 400014, People’s Republic of China
| | - Lan Hao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Hai-tao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| |
Collapse
|
25
|
Wiwatchaitawee K, Quarterman JC, Geary SM, Salem AK. Enhancement of Therapies for Glioblastoma (GBM) Using Nanoparticle-based Delivery Systems. AAPS PharmSciTech 2021; 22:71. [PMID: 33575970 DOI: 10.1208/s12249-021-01928-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/10/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of malignant brain tumor. Current FDA-approved treatments include surgical resection, radiation, and chemotherapy, while hyperthermia, immunotherapy, and most relevantly, nanoparticle (NP)-mediated delivery systems or combinations thereof have shown promise in preclinical studies. Drug-carrying NPs are a promising approach to brain delivery as a result of their potential to facilitate the crossing of the blood-brain barrier (BBB) via two main types of transcytosis mechanisms: adsorptive-mediated transcytosis (AMT) and receptor-mediated transcytosis (RMT). Their ability to accumulate in the brain can thus provide local sustained release of tumoricidal drugs at or near the site of GBM tumors. NP-based drug delivery has the potential to significantly reduce drug-related toxicity, increase specificity, and consequently improve the lifespan and quality of life of patients with GBM. Due to significant advances in the understanding of the molecular etiology and pathology of GBM, the efficacy of drugs loaded into vectors targeting this disease has increased in both preclinical and clinical settings. Multitargeting NPs, such as those incorporating multiple specific targeting ligands, are an innovative technology that can lead to decreased off-target effects while simultaneously having increased accumulation and action specifically at the tumor site. Targeting ligands can include antibodies, or fragments thereof, and peptides or small molecules, which can result in a more controlled drug delivery system compared to conventional drug treatments. This review focuses on GBM treatment strategies, summarizing current options and providing a detailed account of preclinical findings with prospective NP-based approaches aimed at improving tumor targeting and enhancing therapeutic outcomes for GBM patients.
Collapse
|
26
|
Ye S, Liu Y, Lu Y, Ji Y, Mei L, Yang M, Gong X, Gu Q, Li D, Yang F, Li CJ. Cyclic RGD functionalized liposomes targeted to activated platelets for thrombosis dual-mode magnetic resonance imaging. J Mater Chem B 2021; 8:447-453. [PMID: 31833530 DOI: 10.1039/c9tb01834d] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Thrombotic disease is a serious threat to human health. The rapid and accurate detection of thrombosis is still a clinical challenge. To achieve the accurate diagnosis of thrombosis with magnetic resonance imaging (MRI), nanomaterials-based contrast agents have been developed in recent years. In this study, cyclic RGD functionalized liposomes targeted to the activated platelets are developed for thrombosis dual-mode MRI. The cyclic RGD functionalized liposomes (cRGD@MLP-Gd) encapsulated with gadolinium diethylenetriamine penta-acetic acid (Gd-DTPA) and superparamagnetic iron oxide (SPIO) are prepared, and their thrombus-targeted T1 and T2 MRI potential is evaluated in vitro and in vivo. Results show that cRGD@MLP-Gd could actively bind to the activated platelets and gradually accumulate at the thrombus site with a T1 - T2 contrast enhancement imaging effect in vitro. In in vivo MRI experiments, cRGD@MLP-Gd exhibits a T2 contrast enhancement at 1 h after intravenous administration, followed by a visibly larger T1 contrast enhancement at the thrombus site. This dynamic property showed that cRGD@MLP-Gd could actively bind to thrombus and possessed an enhanced T1 and T2 dual-mode MRI effect in vivo. Our results establish the characterization, feasibility and superiority of cRGD@MLP-Gd for the rapid identification of thrombosis, showing great potential to improve diagnostic accuracy and sensitivity to thrombosis of the MRI technique.
Collapse
Affiliation(s)
- Sen Ye
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Tumor-homing peptides are widely used for improving tumor selectivity of anticancer drugs and imaging agents. The goal is to increase tumor uptake and reduce accumulation at nontarget sites. Here, we describe current approaches for tumor-homing peptide identification and validation, and provide comprehensive overview of classes of tumor-homing peptides undergoing preclinical and clinical development. We focus on unique mechanistic features and applications of a recently discovered class of tumor-homing peptides, tumor-penetrating C-end Rule (CendR) peptides, that can be used for tissue penetrative targeting of extravascular tumor tissue. Finally, we discuss unanswered questions and future directions in the field of development of peptide-guided smart drugs and imaging agents.
Collapse
|
28
|
Co-delivery of EGFR and BRD4 siRNA by cell-penetrating peptides-modified redox-responsive complex in triple negative breast cancer cells. Life Sci 2020; 266:118886. [PMID: 33310044 DOI: 10.1016/j.lfs.2020.118886] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/01/2020] [Accepted: 12/05/2020] [Indexed: 01/07/2023]
Abstract
AIMS Triple negative breast cancer (TNBC) has drawn more and more attention due to its high mitotic indices, high metastatic rate and poor prognosis. Gene therapy, especially RNA interference (RNAi), has become a promising targeted therapy. However, improvement of transfection efficiency and discovery of target genes are major problems for the delivery of small interfering RNAs (siRNA). MATERIALS AND METHODS In the present study, we developed GALA- and CREKA-modified PEG-SS-PEI to deliver siRNAs targeting on EGFR and BRD4 for TNBC therapy. The PEG-SS-PEI/siRNA complexes were prepared by electrostatic interaction and characterized by dynamic light scattering (DLS) and transmission electron microscope (TEM). The release characteristic, stability, cellular uptake and intracellular localization of the complexes were also studied. The effect of the complexes on cell viability was measured in MDA-MB-231 and HUVEC cells. The in vitro anti-tumor activities of the complexes were analyzed by Transwell invasion assay and wound healing assay. The gene silencing effect was evaluated by quantitative real time-polymerase chain reaction (qRT-PCR) and western blot. KEY FINDINGS The results revealed that the GALA- and CREKA-modified PEG-SS-PEI/siRNA complexes showed excellent transfection efficiency with redox-sensitive release profile and good biological compatibility. The complexes protected siRNA from the degradation of RNA enzymes. The complexes significantly inhibited the proliferation, invasion and migration of MDA-MB-231 cells via the synergistic inhibition of EGFR/PI3K/Akt and BRD4/c-Myc pathways. SIGNIFICANCE Taken together, co-delivery of siEGFR and siBRD4 by GALA-PEG-SS-PEI and CREKA-PEG-SS-PEI may provide a more effective strategy for the treatment of TNBC.
Collapse
|
29
|
Vorobiev V, Adriouach S, Crowe LA, Lenglet S, Thomas A, Chauvin AS, Allémann E. Vascular-targeted micelles as a specific MRI contrast agent for molecular imaging of fibrin clots and cancer cells. Eur J Pharm Biopharm 2020; 158:347-358. [PMID: 33271302 DOI: 10.1016/j.ejpb.2020.11.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022]
Abstract
Molecular medical imaging is intended to increase the accuracy of diagnosis, particularly in cardiovascular and cancer-related diseases, where early detection could significantly increase the treatment success rate. In this study, we present mixed micelles formed from four building blocks as a magnetic resonance imaging targeted contrast agent for the detection of atheroma and cancer cells. The building blocks are a gadolinium-loaded DOTA ring responsible for contrast enhancement, a fibrin-specific CREKA pentapeptide responsible for targeting, a fluorescent dye and DSPE-PEG2000. The micelles were fully characterized in terms of their size, zeta potential, stability, relaxivity and toxicity. Target binding assays performed on fibrin clots were quantified by fluorescence and image signal intensities and proved the binding power. An additional internalization assay showed that the micelles were also designed to specifically enter into cancer cells. Overall, these multimodal mixed micelles represent a potential formulation for MRI molecular imaging of atheroma and cancer cells.
Collapse
Affiliation(s)
- Vassily Vorobiev
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Souad Adriouach
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Lindsey A Crowe
- Department of Radiology and Medical Informatics, University of Geneva, 1211 Geneva, Switzerland
| | - Sébastien Lenglet
- Forensic Toxicology and Chemistry Unit, University Center for Legal Medicine, Geneva University Hospital, 1211 Geneva, Switzerland
| | - Aurélien Thomas
- Unit of Toxicology, CURML, Lausanne University Hospital, Geneva University Hospitals, Switzerland; Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Anne-Sophie Chauvin
- Institut of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne, Route Cantonale, 1015 Lausanne, Switzerland
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
30
|
Zhao Y, Xie R, Yodsanit N, Ye M, Wang Y, Gong S. Biomimetic fibrin-targeted and H 2O 2-responsive nanocarriers for thrombus therapy. NANO TODAY 2020; 35:100986. [PMID: 33072177 PMCID: PMC7561002 DOI: 10.1016/j.nantod.2020.100986] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Thrombosis is a principle cause of various life-threatening cardiovascular diseases. However, current antithrombotic treatments using drugs only offer limited efficacy due to short half-life, low targeting ability to the thrombus site, and unexpected bleeding complications. Taking into account of the biological characteristics of thrombus including upregulation of hydrogen peroxide (H2O2) and abundance of fibrin, we engineered a H2O2-responsive nanocarrier for thrombus-targeting delivery of an antithrombotic agent (i.e., tirofiban). The nanocarrier was composed of a drug-conjugated dextran nanocore and a red blood cell (RBC) membrane shell, and its surface was functionalized with a fibrin-targeting peptide, CREKA. Tirofiban was conjugated to dextran through a H2O2-cleavable phenylboronic ester linkage. The fibrin-targeting RBC membrane-cloaked dextran-tirofiban conjugate nanoparticles (i.e., T-RBC-DTC NPs) can scavenge H2O2 and provide controlled release of tirofiban to achieve site-specific antithrombotic effects. In RAW 264.7 cells and HUVECs, the T-RBC-DTC NPs effectively scavenged H2O2 and protected cells from H2O2-induced cytotoxicity. In the ferric chloride-induced carotid thrombosis mouse model, the T-RBC-DTC NPs efficiently accumulated at the injured carotid artery and exhibited significantly enhanced antithrombotic activity compared to free drug. The T-RBC-DTC NPs also exhibited good biocompatibility according to histology analysis. Overall, our results indicated that this bioengineered nanocarrier offers a promising therapeutic strategy for thrombotic disorders.
Collapse
Affiliation(s)
- Yi Zhao
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Ruosen Xie
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Material Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Nisakorn Yodsanit
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Mingzhou Ye
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Yuyuan Wang
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Shaoqin Gong
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Material Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Corresponding author. Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA. (S. Gong)
| |
Collapse
|
31
|
Atkinson SP. A preview of selected articles. Stem Cells Transl Med 2020. [PMCID: PMC7519771 DOI: 10.1002/sctm.20-0395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
32
|
Bheri S, Hoffman JR, Park HJ, Davis ME. Biomimetic nanovesicle design for cardiac tissue repair. Nanomedicine (Lond) 2020; 15:1873-1896. [PMID: 32752925 DOI: 10.2217/nnm-2020-0097] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is a major cause of mortality and morbidity worldwide. Exosome therapies are promising for cardiac repair. Exosomes transfer cargo between cells, have high uptake by native cells and are ideal natural carriers for proteins and nucleic acids. Despite their proreparative potential, exosome production is dependent on parent cell state with typically low yields and cargo variability. Therefore, there is potential value in engineering exosomes to maximize their benefits by delivering customized, potent cargo for cardiovascular disease. Here, we outline several methods of exosome engineering focusing on three important aspects: optimizing cargo, homing to target tissue and minimizing clearance. Finally, we put these methods in context of the cardiac field and discuss the future potential of vesicle design.
Collapse
Affiliation(s)
- Sruti Bheri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Jessica R Hoffman
- Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Hyun-Ji Park
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30332, USA.,Department of Pediatrics, Division of Pediatric Cardiology, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
33
|
Xu J, Zhou J, Zhong Y, Zhang Y, Ye M, Hou J, Wang Z, Ran H, Liu J, Guo D. EWVDV-Mediated Platelet-Targeting Nanoparticles for the Multimodal Imaging of Thrombi at Different Blood Flow Velocities. Int J Nanomedicine 2020; 15:1759-1770. [PMID: 32214809 PMCID: PMC7083630 DOI: 10.2147/ijn.s233968] [Citation(s) in RCA: 7] [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: 02/17/2020] [Indexed: 11/23/2022] Open
Abstract
Background There have been many recent reports of molecular probes for thrombi but with unsatisfactory in vivo targeting effects, which could be related to the blood flow velocity in vivo. Therefore, it is worth explaining the relationship between the targeting effect and the blood flow velocity. Methods and Materials In this study, we constructed a platelet-targeting nanoparticle (NP) based on EWVDV for targeting P-selectin combined with the phase transition material perfluorohexane and India ink to achieve the multimodal imaging of thrombi. We studied the targeting effect of the NPs for rabbit blood thrombi under different flow velocities simulating blood flow velocities in vivo. Results The results show the successful fabrication of NPs with the ability to undergo a phase transition via low-intensity focused ultrasound irradiation to achieve ultrasound imaging and with a high binding affinity for activated platelets. In vitro, low flow velocities (20 cm/s) hardly affected the targeting effect of the NPs, while moderate flow velocities (40 cm/s) reduced the number of NPs that target thrombi by 52.6% comparing to static fluid (0 cm/s). High flow velocities (60 cm/s) greatly reduced the targeting effect of the NPs by 83.5%. Conclusion These results can serve as a reference for the design of NPs targeting thrombi at different sites and in different blood vessel types according to the blood flow velocity, thereby establishing a foundation for in vivo experiments.
Collapse
Affiliation(s)
- Jie Xu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jun Zhou
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yixin Zhong
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yu Zhang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Man Ye
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jingxin Hou
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhigang Wang
- Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Haitao Ran
- Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jia Liu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dajing Guo
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
34
|
Puiggalı́-Jou A, del Valle LJ, Alemán C. Encapsulation and Storage of Therapeutic Fibrin-Homing Peptides using Conducting Polymer Nanoparticles for Programmed Release by Electrical Stimulation. ACS Biomater Sci Eng 2020; 6:2135-2145. [DOI: 10.1021/acsbiomaterials.9b01794] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Anna Puiggalı́-Jou
- Departament d’Enginyeria Quı́mica and Barcelona Research Center for Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Luis J. del Valle
- Departament d’Enginyeria Quı́mica and Barcelona Research Center for Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Carlos Alemán
- Departament d’Enginyeria Quı́mica and Barcelona Research Center for Multiscale Science and Engineering, EEBE, Universitat Politècnica de Catalunya, C/Eduard Maristany 10-14, 08019 Barcelona, Spain
- Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| |
Collapse
|
35
|
Bai S, Liao J, Zhang B, Zhao M, You B, Li P, Ran H, Wang Z, Shi R, Zhang G. Multimodal and multifunctional nanoparticles with platelet targeting ability and phase transition efficiency for the molecular imaging and thrombolysis of coronary microthrombi. Biomater Sci 2020; 8:5047-5060. [PMID: 32830839 DOI: 10.1039/d0bm00818d] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this article, we constructed PLGA-cRGD-PFH-ICG NPs through emulsification process and then the bi-modal imaging of coronary microthrombi in ischemia/reperfusion rat model and thrombolysis of clots in vitro were both successfully completed by these NPs.
Collapse
Affiliation(s)
- Sheng Bai
- Department of Ultrasound
- Xiangya Hospital Central South University
- Changsha
- China
| | - Jintang Liao
- Department of Ultrasound
- Xiangya Hospital Central South University
- Changsha
- China
| | - Bo Zhang
- Department of Ultrasound
- Xiangya Hospital Central South University
- Changsha
- China
| | - Min Zhao
- Department of Nuclear Medicine
- Xiangya Hospital Central South University
- Changsha
- China
| | - Baiyang You
- Cardiac Rehabilitation Center
- Department of Rehabilitation
- Xiangya Hospital Central South University
- Changsha
- China
| | - Pan Li
- Institute of Ultrasound imaging of Chongqing Medical University
- Chongqing 400010
- P. R. China
| | - Haitao Ran
- Institute of Ultrasound imaging of Chongqing Medical University
- Chongqing 400010
- P. R. China
| | - Zhigang Wang
- Institute of Ultrasound imaging of Chongqing Medical University
- Chongqing 400010
- P. R. China
| | - Ruizheng Shi
- Department of Cardiovascular Medicine
- Xiangya Hospital Central South University
- Changsha
- China
| | - Guogang Zhang
- Department of Cardiovascular Medicine
- Xiangya Hospital Central South University
- Changsha
- China
| |
Collapse
|
36
|
Ferreira TH, de Oliveira Freitas LB, Fernandes RS, dos Santos VM, Resende JM, Cardoso VN, de Barros ALB, de Sousa EMB. Boron nitride nanotube-CREKA peptide as an effective target system to metastatic breast cancer. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2019. [DOI: 10.1007/s40005-019-00467-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
37
|
Nuzhina JV, Shtil AA, Prilepskii AY, Vinogradov VV. Preclinical Evaluation and Clinical Translation of Magnetite-Based Nanomedicines. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101282] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
38
|
Wan X, Liu C, Lin Y, Fu J, Lu G, Lu Z. pH sensitive peptide functionalized nanoparticles for co-delivery of erlotinib and DAPT to restrict the progress of triple negative breast cancer. Drug Deliv 2019; 26:470-480. [PMID: 30957572 PMCID: PMC6462792 DOI: 10.1080/10717544.2019.1576801] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although a variety of drug delivery strategies have been designed for enhancing the treatment of Triple negative breast cancer (TNBC), combating with TNBCs is still dramatically challenged by the selection of appropriate therapeutic targets and insufficient tumor accumulation or inner penetration of chemotherapeutics. To address these issues, the classical EGFR-inhibitor, erlotinib (EB), was selected as the model drug here and PLA-based nano-platform (NP-EB) was prepared for tumor site drug delivery. Given the significant role of Notch-EGFR interplay in raising severe resistance to EGFR inhibition of EB, gamma secretase inhibitor (GSI)-DAPT was further entrapped into the core of nanoparticles to inhibit the activation of Notch signaling (NP-EB/DART). For achieving the goal of tumor targeting drug delivery, we developed a new peptide CF and decorating it on the surface of EB/DART-dual loaded nanoparticles (CF-NP-EB/DART). Such CF peptide was designed by conjugating two separated peptide CREKA, tumor-homing peptide, and F3, cell penetrating peptide, to together via a pH-sensitive hydrazone bond. By this way, the tumor unspecific property of F3 was sealed and significantly reduced the site effects. However, after the nanoparticles reach the tumor site, the pH-sensitive linkage can be broken down by the unique acidic environment of tumor, and subsequently discovered the F3 peptide to penetrate into tumor cells.
Collapse
Affiliation(s)
- Xu Wan
- a Department of Pharmacy, South Campus, Renji Hospital, School of Medicine , Shanghai Jiaotong University , Shanghai , People's Republic of China
| | - Chaoqian Liu
- b Department of General Surgery , Changhai Hospital The Second Military Medical University , Shanghai , People's Republic of China
| | - Yinan Lin
- a Department of Pharmacy, South Campus, Renji Hospital, School of Medicine , Shanghai Jiaotong University , Shanghai , People's Republic of China
| | - Jie Fu
- a Department of Pharmacy, South Campus, Renji Hospital, School of Medicine , Shanghai Jiaotong University , Shanghai , People's Republic of China
| | - Guohong Lu
- a Department of Pharmacy, South Campus, Renji Hospital, School of Medicine , Shanghai Jiaotong University , Shanghai , People's Republic of China
| | - Zhengmao Lu
- b Department of General Surgery , Changhai Hospital The Second Military Medical University , Shanghai , People's Republic of China
| |
Collapse
|
39
|
Zhang Y, Zhong Y, Ye M, Xu J, Liu J, Zhou J, Wang S, Guo D, Wang Z, Ran H. Polydopamine-modified dual-ligand nanoparticles as highly effective and targeted magnetic resonance/photoacoustic dual-modality thrombus imaging agents. Int J Nanomedicine 2019; 14:7155-7171. [PMID: 31564871 PMCID: PMC6731970 DOI: 10.2147/ijn.s216603] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022] Open
Abstract
Background Platelet activation and subsequent aggregation are the initial stages of thrombosis. A molecular probe that specifically targets activated platelets and remains retained under high shear stress in vivo can enhance the imaging effect to achieve early and accurate diagnosis. Methods and materials In this study, we constructed nanoparticles (NPs) using polydopamine to carry two peptides that simultaneously bind integrin αIIbβ3 and P-selectin on activated platelets to enhance the targeting of NPs to thrombus. Results The targeting specificity and binding stability of the NPs on red and white thrombi were demonstrated in vitro using a simulated circulatory device and the targeting effect of the NPs on mixed thrombus was studied by magnetic resonance (MR)/photoacoustic (PA) dual-modality imaging in vivo. NPs that were surface modified with both peptides have higher selectivity and retention to red and white thrombi in vitro than NPs with a single or no peptide, and the targeting effect was closely related to the number and distribution of activated platelets as well as the structure and type of thrombus. The NPs also have MR/PA dual-modality imaging functionality, significantly enhancing the imaging of mixed thrombus in vivo. Conclusion These dual-targeted NPs have improved targeting specificity and binding stability to different thrombi under high shear stress and are beneficial for the early diagnosis of thrombosis.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Ultrasound Molecular Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yixin Zhong
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Ultrasound Molecular Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Man Ye
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jie Xu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jia Liu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jun Zhou
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Shike Wang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dajing Guo
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
40
|
Zhong Y, Zhang Y, Xu J, Zhou J, Liu J, Ye M, Zhang L, Qiao B, Wang ZG, Ran HT, Guo D. Low-Intensity Focused Ultrasound-Responsive Phase-Transitional Nanoparticles for Thrombolysis without Vascular Damage: A Synergistic Nonpharmaceutical Strategy. ACS NANO 2019; 13:3387-3403. [PMID: 30855938 DOI: 10.1021/acsnano.8b09277] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Multimodal molecular imaging has shown promise as a complementary approach to thrombus detection. However, the simultaneous noninvasive detection and lysis of thrombi for cardiovascular diseases remain challenging. Herein, a perfluorohexane (PFH)-based biocompatible nanostructure was fabricated, namely, as-prepared Fe3O4-poly(lactic- co-glycolic acid)-PFH-CREKA nanoparticles (NPs), which combine phase transition (PT) thrombolysis capabilities with properties conducive to multimodal imaging. This well-developed PT agent responded effectively to low-intensity focused ultrasound (LIFU) by triggering the vaporization of liquid PFH to achieve thrombolysis. The presence of the CREKA peptide, which binds to the fibrin of the thrombus, allows targeted imaging and efficacious thrombolysis. Then, we found that, compared with thrombolysis using a non-phase-transition agent, PT thrombolysis can produce a robust decrease in the thrombus burden regardless of the acoustic power density of LIFU. In particular, the reduced energy for LIFU-responsive PT during the lysis process guarantees the superior safety of PT thrombolysis. After injecting the NPs intravenously, we demonstrated that this lysis process can be monitored with ultrasound and photoacoustic imaging in vivo to evaluate its efficacy. Therefore, this nonpharmaceutical strategy departs from routine methods and reveals the potential use of PT thrombolysis as an effective and noninvasive alternative to current thrombolytic therapy.
Collapse
Affiliation(s)
- Yixin Zhong
- Department of Radiology , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
| | - Yu Zhang
- Department of Radiology , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
| | - Jie Xu
- Department of Radiology , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
| | - Jun Zhou
- Department of Radiology , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
| | - Jia Liu
- Department of Radiology , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
| | - Man Ye
- Department of Radiology , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
| | - Liang Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
| | - Bin Qiao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
| | - Zhi-Gang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
| | - Hai-Tao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
| | - Dajing Guo
- Department of Radiology , Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Road , Yuzhong District, Chongqing , 400010 , People's Republic of China
| |
Collapse
|
41
|
Chen J, Song Y, Huang Z, Zhang N, Xie X, Liu X, Yang H, Wang Q, Li M, Li Q, Gong H, Qian J, Pang Z, Ge J. Modification with CREKA Improves Cell Retention in a Rat Model of Myocardial Ischemia Reperfusion. Stem Cells 2019; 37:663-676. [PMID: 30779865 DOI: 10.1002/stem.2983] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 01/06/2019] [Accepted: 01/21/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Jing Chen
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
| | - Yanan Song
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
| | - Zheyong Huang
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
| | - Ning Zhang
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
| | - Xinxing Xie
- Department of Cardiology; Rizhao Heart Hospital; Rizhao Shandong People's Republic of China
| | - Xin Liu
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
| | - Hongbo Yang
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
| | - Qiaozi Wang
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
| | - Minghui Li
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
| | - Qiyu Li
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
| | - Hui Gong
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
| | - Juying Qian
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
| | - Zhiqing Pang
- School of Pharmacy, Fudan University; Key Laboratory of Smart Drug Delivery, Ministry of Education; Shanghai People's Republic of China
| | - Junbo Ge
- Department of Cardiology; Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases; Shanghai People's Republic of China
- Institute of Biomedical Science; Fudan University; Shanghai People's Republic of China
| |
Collapse
|
42
|
Pagoto A, Tripepi M, Stefania R, Lanzardo S, Livio Longo D, Garello F, Porpiglia F, Manfredi M, Aime S, Terreno E. An efficient MRI agent targeting extracellular markers in prostate adenocarcinoma. Magn Reson Med 2018; 81:1935-1946. [DOI: 10.1002/mrm.27494] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/23/2018] [Accepted: 07/23/2018] [Indexed: 01/10/2023]
Affiliation(s)
- Amerigo Pagoto
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences University of Torino Torino Italy
| | - Martina Tripepi
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences University of Torino Torino Italy
| | - Rachele Stefania
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences University of Torino Torino Italy
| | - Stefania Lanzardo
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences University of Torino Torino Italy
| | - Dario Livio Longo
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences University of Torino Torino Italy
| | - Francesca Garello
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences University of Torino Torino Italy
| | - Francesco Porpiglia
- Division of Urology University of Torino, San Luigi Gonzaga Hospital Orbassano, Torino Italy
| | - Matteo Manfredi
- Division of Urology University of Torino, San Luigi Gonzaga Hospital Orbassano, Torino Italy
| | - Silvio Aime
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences University of Torino Torino Italy
- IBB‐CNR Sede Secondaria c/o MBC Torino Italy
| | - Enzo Terreno
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences University of Torino Torino Italy
| |
Collapse
|
43
|
Qi GB, Gao YJ, Wang L, Wang H. Self-Assembled Peptide-Based Nanomaterials for Biomedical Imaging and Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1703444. [PMID: 29460400 DOI: 10.1002/adma.201703444] [Citation(s) in RCA: 291] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/22/2017] [Indexed: 05/22/2023]
Abstract
Peptide-based materials are one of the most important biomaterials, with diverse structures and functionalities. Over the past few decades, a self-assembly strategy is introduced to construct peptide-based nanomaterials, which can form well-controlled superstructures with high stability and multivalent effect. More recently, peptide-based functional biomaterials are widely utilized in clinical applications. However, there is no comprehensive review article that summarizes this growing area, from fundamental research to clinic translation. In this review, the recent progress of peptide-based materials, from molecular building block peptides and self-assembly driving forces, to biomedical and clinical applications is systematically summarized. Ex situ and in situ constructed nanomaterials based on functional peptides are presented. The advantages of intelligent in situ construction of peptide-based nanomaterials in vivo are emphasized, including construction strategy, nanostructure modulation, and biomedical effects. This review highlights the importance of self-assembled peptide nanostructures for nanomedicine and can facilitate further knowledge and understanding of these nanosystems toward clinical translation.
Collapse
Affiliation(s)
- Guo-Bin Qi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Yu-Juan Gao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Lei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| |
Collapse
|
44
|
Fakayode OJ, Tsolekile N, Songca SP, Oluwafemi OS. Applications of functionalized nanomaterials in photodynamic therapy. Biophys Rev 2018; 10:49-67. [PMID: 29294258 PMCID: PMC5803176 DOI: 10.1007/s12551-017-0383-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 12/13/2017] [Indexed: 11/25/2022] Open
Abstract
Specially designed functionalized nanomaterials such as superparamagnetic iron oxide, gold, quantum dots and up- and down-conversion lanthanide series nanoparticles have consistently and completely revolutionized the biomedical environment over the past few years due to their specially inferring properties, such as specific drug delivery, plasmonic effect, optical and imaging properties, therapeutic thermal energy productionand excellent irresistible cellular penetration. These properties have been used to improve many existing disease treatment modalities and have led to the development of better therapeutic approaches for the advancement of the treatment of critical human diseases, such as cancers and related malaise. In photodynamic therapy, for example, where the delivery of therapeutic agents should ideally avoid toxicity on nearby healthy cells, superparamagnetic iron oxide nanoparticles have been shown to be capable of making photodynamic therapy (PDT) prodrugs and their associative targeting moieties tumor-specific via their unique response to an external magnetic fields. In this review, the nanomaterials commonly employed for the enhancement of photodynamic therapy are discussed. The review further describes the various methods of synthesis and characterization of these nanomaterials and highlights challenges for improving the efficacy of PDT in the future.
Collapse
Affiliation(s)
- Olayemi J Fakayode
- Department of Applied Chemistry, University of Johannesburg, P.O. Box 17011, Doornfontein, Johannesburg, 2028, South Africa
- Centre for Nanomaterials Science Research, University of Johannesburg, Johannesburg, South Africa
| | - Ncediwe Tsolekile
- Department of Applied Chemistry, University of Johannesburg, P.O. Box 17011, Doornfontein, Johannesburg, 2028, South Africa
- Centre for Nanomaterials Science Research, University of Johannesburg, Johannesburg, South Africa
| | - Sandile P Songca
- Department of Chemistry, University of Zululand, PB X1001, Kwadlangezwa, 3886, South Africa
| | - Oluwatobi S Oluwafemi
- Department of Applied Chemistry, University of Johannesburg, P.O. Box 17011, Doornfontein, Johannesburg, 2028, South Africa.
- Centre for Nanomaterials Science Research, University of Johannesburg, Johannesburg, South Africa.
| |
Collapse
|
45
|
Wang X, Zhang Q, Lv L, Fu J, Jiang Y, Xin H, Yao Q. Glioma and microenvironment dual targeted nanocarrier for improved antiglioblastoma efficacy. Drug Deliv 2017; 24:1401-1409. [PMID: 28933201 PMCID: PMC8241031 DOI: 10.1080/10717544.2017.1378940] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Drug delivery systems based on nanoparticles (nano-DDS) have aroused attentions for the treatment of glioblastoma (GBM), the most malignant brain cancer with a dismal prognosis. However, there are still numerous unmet challenges for traditional nano-DDS, such as the poor nanoparticle penetration, short retention in the GBM parenchyma and low glioma targeting ability. Herein, we used Pep-1 and CREKA peptides to construct a novel multifunctional GBM targeting nano-DDS (PC-NP). Pep-1 was used to overcome the blood-brain tumor barrier (BBTB) and home to glioma cells via interleukin-13 receptor-α2-mediated endocytosis, and CREKA was used to bind to fibrin-fibronectin complexes abundantly expressed in tumor microenvironment for enhanced retention in the GBM. Biological studies showed that the cellular uptake of PC-NP by U87MG cells was significantly enhanced compared with the non-targeting NP. Furthermore, CREKA modification increased the binding capacity of PC-NP to fibrin-fibronectin complexes as confirmed by the competition experiment. In accordance with the increased cellular uptake, PC-NP remarkably increased the cytotoxicity of its payload paclitaxel (PTX) against U87MG cells with an IC50 of 0.176 μg/mL. In vivo fluorescence imaging and antiglioma efficacy evaluation further confirmed that PC-NP accumulated effectively and penetrated deeply into GBM tissue. PC-NP-PTX exhibited a median survival time as long as 61 days in intracranial GBM-bearing mice. In conclusion, our findings indicated PC-NP as a promising nano-DDS for GBM targeting delivery of anticancer drugs.
Collapse
Affiliation(s)
- Xiuzhen Wang
- a Department of Medicinal Chemistry, School of Pharmacy , China Pharmaceutical University , Nanjing , China.,b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Qing Zhang
- b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Lingyan Lv
- b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Junjie Fu
- b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Yan Jiang
- b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Hongliang Xin
- b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Qizheng Yao
- a Department of Medicinal Chemistry, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| |
Collapse
|
46
|
Xu J, Zhou J, Zhong Y, Zhang Y, Liu J, Chen Y, Deng L, Sheng D, Wang Z, Ran H, Guo D. Phase Transition Nanoparticles as Multimodality Contrast Agents for the Detection of Thrombi and for Targeting Thrombolysis: in Vitro and in Vivo Experiments. ACS APPLIED MATERIALS & INTERFACES 2017; 9:42525-42535. [PMID: 29160060 DOI: 10.1021/acsami.7b12689] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Thrombotic disease is extremely harmful to human health, and early detection and treatment can improve the prognosis and reduce mortality. Multimodal molecular imaging can provide abundant information about thrombi, but to date, few studies have used multimodal and multifunctional nanoparticles (NPs) for thrombus detection and for targeting thrombolysis. In this study, phase transition multimodal and multifunctional NPs (EWVDV-Fe-Ink-PFH NPs) were constructed for the first time using a three-step emulsification and carbodiimide method, and the physical and chemical properties of the NPs were investigated. The targeting abilities of the NPs and multimodal imaging, that is, photoacoustic, magnetic resonance, and ultrasound imaging, were successfully achieved in vitro and in vivo. The ability of the EWVDV peptide on the NPs to effectively target the P-selectin of thrombi was confirmed by multimodal imaging and pathology, and the penetration depths of the NPs into the thrombi were far deeper than the previously reported depths. Moreover, a perfluorohexane (PFH) phase transition induced by low-intensity focused ultrasound irradiation enabled the EWVDV-Fe-Ink-PFH NPs to cause thrombolysis in vitro. In summary, EWVDV-Fe-Ink-PFH NPs are a theranostic contrast agent that will provide a simple, effective, and noninvasive approach for the diagnosis and treatment of thrombosis.
Collapse
Affiliation(s)
- Jie Xu
- Department of Radiology and ‡Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Rd, Yuzhong District, Chongqing 400010, P. R. China
| | - Jun Zhou
- Department of Radiology and ‡Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Rd, Yuzhong District, Chongqing 400010, P. R. China
| | - Yixin Zhong
- Department of Radiology and ‡Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Rd, Yuzhong District, Chongqing 400010, P. R. China
| | - Yu Zhang
- Department of Radiology and ‡Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Rd, Yuzhong District, Chongqing 400010, P. R. China
| | - Jia Liu
- Department of Radiology and ‡Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Rd, Yuzhong District, Chongqing 400010, P. R. China
| | - Yuli Chen
- Department of Radiology and ‡Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Rd, Yuzhong District, Chongqing 400010, P. R. China
| | - Liming Deng
- Department of Radiology and ‡Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Rd, Yuzhong District, Chongqing 400010, P. R. China
| | - Danli Sheng
- Department of Radiology and ‡Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Rd, Yuzhong District, Chongqing 400010, P. R. China
| | - Zhigang Wang
- Department of Radiology and ‡Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Rd, Yuzhong District, Chongqing 400010, P. R. China
| | - Haitao Ran
- Department of Radiology and ‡Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Rd, Yuzhong District, Chongqing 400010, P. R. China
| | - Dajing Guo
- Department of Radiology and ‡Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University , No. 74 Linjiang Rd, Yuzhong District, Chongqing 400010, P. R. China
| |
Collapse
|
47
|
Montiel Schneider MG, Lassalle VL. Magnetic iron oxide nanoparticles as novel and efficient tools for atherosclerosis diagnosis. Biomed Pharmacother 2017; 93:1098-1115. [PMID: 28738519 DOI: 10.1016/j.biopha.2017.07.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/14/2017] [Accepted: 07/05/2017] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular complications derivate from atherosclerosis are the main cause of death in western world. An early detection of vulnerable atherosclerotic plaques is primordial for a better care of patients suffering the pathology. In this context nanotechnology has emerged as a promising tool to achieve this goal. Nanoparticles based on magnetic iron oxide (MNPs) have been extensively studied in cardiovascular diseases diagnosis, as well as in the treatment and diagnostic of other pathologies. The present review aims to describe and analyze the most current literature regarding to this topic, offering the level of detail required to reproduce the experimental tasks providing a critical input of the latest available reports. The current diagnostic features are presented and compared, highlighting their advantages and disadvantages. Information on novel technology intended to this purpose is also recompiled and in deep analyzed. Special emphasis is placed in magnetic nanotechnology, remarking the possibility to assess selective and multifunctional systems to the early detection of artherosclerotic pathologies. Finally, in view of the state of the art, the future perspectives about the trends on MNPs in artherosclerorsis diagnostic and treatment have also been addressed.
Collapse
Affiliation(s)
| | - Verónica Leticia Lassalle
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS)-CONICET, Av. Alem 1253, 8000 Bahía Blanca, Argentina.
| |
Collapse
|
48
|
Wang X, Peter K. Molecular Imaging of Atherothrombotic Diseases: Seeing Is Believing. Arterioscler Thromb Vasc Biol 2017; 37:1029-1040. [PMID: 28450298 DOI: 10.1161/atvbaha.116.306483] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/11/2017] [Indexed: 12/13/2022]
Abstract
Molecular imaging, with major advances in the development of both innovative targeted contrast agents/particles and radiotracers, as well as various imaging technologies, is a fascinating, rapidly growing field with many preclinical and clinical applications, particularly for personalized medicine. Thrombosis in either the venous or the arterial system, the latter typically caused by rupture of unstable atherosclerotic plaques, is a major determinant of mortality and morbidity in patients. However, imaging of the various thrombotic complications and the identification of plaques that are prone to rupture are at best indirect, mostly unreliable, or not available at all. The development of molecular imaging toward diagnosis and prevention of thrombotic disease holds promise for major advance in this clinically important field. Here, we review the medical need and clinical importance of direct molecular imaging of thrombi and unstable atherosclerotic plaques that are prone to rupture, thereby causing thrombotic complications such as myocardial infarction and ischemic stroke. We systematically compare the advantages/disadvantages of the various molecular imaging modalities, including X-ray computed tomography, magnetic resonance imaging, positron emission tomography, single-photon emission computed tomography, fluorescence imaging, and ultrasound. We further systematically discuss molecular targets specific for thrombi and those characterizing unstable, potentially thrombogenic atherosclerotic plaques. Finally, we provide examples for first theranostic approaches in thrombosis, combining diagnosis, targeted therapy, and monitoring of therapeutic success or failure. Overall, molecular imaging is a rapidly advancing field that holds promise of major benefits to many patients with atherothrombotic diseases.
Collapse
Affiliation(s)
- Xiaowei Wang
- From the Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute (X.W., K.P.), Departments of Medicine (X.W., K.P.), and Immunology (K.P.), Monash University, Melbourne, Victoria, Australia
| | - Karlheinz Peter
- From the Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute (X.W., K.P.), Departments of Medicine (X.W., K.P.), and Immunology (K.P.), Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
49
|
Huang Z, Song Y, Pang Z, Zhang B, Yang H, Shi H, Chen J, Gong H, Qian J, Ge J. Targeted delivery of thymosin beta 4 to the injured myocardium using CREKA-conjugated nanoparticles. Int J Nanomedicine 2017; 12:3023-3036. [PMID: 28442910 PMCID: PMC5396927 DOI: 10.2147/ijn.s131949] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose Thymosin beta 4 (Tβ4) has multiple beneficial facets for myocardial injury, but its efficiency is limited by the low local concentration within the infarct. Here, we established a Tβ4 delivery system for cardiac repair based on the interaction between the abundant fibrin in the infarct zone and the fibrin-targeting moiety clot-binding peptide cysteine–arginine–glutamic acid–lysine–alanine (CREKA). Methods and results CREKA and Tβ4 were conjugated to nanoparticles (CNP–Tβ4). In vitro binding test revealed that CNP–Tβ4 had a significant binding ability to the surface of fibrin clots when compared to the control clots (NP–Tβ4). Based on the validation of fibrin expression in the early stage of ischemia injury, CNP–Tβ4 was intravenously administered to mice with acute myocardial ischemia–reperfusion injury. CNP–Tβ4 revealed a stronger fibrin-targeting ability than the NP–Tβ4 group and accumulated mainly in the infarcted area and colocalized with fibrin. Subsequently, treatment with CNP–Tβ4 resulted in a better therapeutic effect. Conclusion CRKEA modification favored Tβ4 accumulation and retention in the infarcted region, leading to augmented functional benefits. Fibrin-targeting delivery system represents a generalizable platform technology for regenerative medicine.
Collapse
Affiliation(s)
- Zheyong Huang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Yanan Song
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei
| | - Hongbo Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Hongtao Shi
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Jing Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University.,Institute of Biomedical Science, Fudan University, Shanghai, People's Republic of China
| | - Juying Qian
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University.,Institute of Biomedical Science, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
50
|
Liu J, Xu J, Zhou J, Zhang Y, Guo D, Wang Z. Fe 3O 4-based PLGA nanoparticles as MR contrast agents for the detection of thrombosis. Int J Nanomedicine 2017; 12:1113-1126. [PMID: 28223802 PMCID: PMC5310639 DOI: 10.2147/ijn.s123228] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Thrombotic disease is a great threat to human health, and early detection is particularly important. Magnetic resonance (MR) molecular imaging provides noninvasive imaging with the potential for early disease diagnosis. In this study, we developed Fe3O4-based poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) surface-modified with a cyclic Arg-Gly-Asp (cRGD) peptide as an MR contrast agent for the detection of thrombosis. The physical and chemical characteristics, biological toxicity, ability to target thrombi, and biodistribution of the NPs were studied. The Fe3O4-PLGA-cRGD NPs were constructed successfully, and hematologic and pathologic assays indicated no in vivo toxicity of the NPs. In a rat model of FeCl3-induced abdominal aorta thrombosis, the NPs readily and selectively accumulated on the surface of the thrombosis and under vascular endothelial cells ex vivo and in vivo. In the in vivo experiment, the biodistribution of the NPs suggested that the NPs might be internalized by the macrophages of the reticuloendothelial system in the liver and the spleen. The T2 signal decreased at the mural thrombus 10 min after injection and then gradually increased until 50 min. These results suggest that the NPs are suitable for in vivo molecular imaging of thrombosis under high shear stress conditions and represent a very promising MR contrast agent for sensitive and specific detection of thrombosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhigang Wang
- Department of Ultrasound, Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, People's Republic of China
| |
Collapse
|