1
|
Wang Y, Gao L, Cao Y, Yan D, Lukman R, Zhang J, Li Q, Liu J, Du F, Zhang L. Research progress on the synthesis, performance regulation, and applications of Prussian blue nanozymes. Int J Biol Macromol 2025; 295:139535. [PMID: 39761892 DOI: 10.1016/j.ijbiomac.2025.139535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/14/2025]
Abstract
Nanocatalytic medicine offers a novel solution to address the issues of low efficacy, potential side effects, and the development of drug resistance associated with traditional therapies. Therefore, developing highly efficient and durable nanozymes is of great significance for treating diseases related to oxidative stress. In recent years, prussian blue nanoparticles (PBNPs) have been demonstrated to possess multiple enzyme-like catalytic activities and are thus referred to as prussian blue nanozymes (PBNZs). Their excellent biocompatibility and biodegradability make PBNZs promising candidates as biomedical materials. Due to their remarkable catalytic activities, PBNZs have shown great potential in various biomedical applications, such as heavy metal detoxification, antioxidative damage, and anticancer therapies. This paper systematically summarizes the Synthetic strategies of PBNZs, analyzes the regulatory factors of their catalytic performance, and highlights the corresponding modulation methods. Furthermore, the biomedical applications of PBNZs are also reviewed. This study aims to provide researchers with insights and inspirations for the design and preparation of high-performance PBNZs.
Collapse
Affiliation(s)
- Yiyang Wang
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Lei Gao
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Yue Cao
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Duanfeng Yan
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Rilwanu Lukman
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Jingxi Zhang
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Quan Li
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Jiaying Liu
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Fengyi Du
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China; Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, PR China
| | - Li Zhang
- Department of Critical Care Medicine Unit, Shanghai Baoshan District Wusong Central Hospital (Zhongshan Hospital Wusong Branch, Fudan University), Shanghai 201900, PR China.
| |
Collapse
|
2
|
Zhang D, Zhang J, Bian X, Zhang P, Wu W, Zuo X. Iron Oxide Nanoparticle-Based T 1 Contrast Agents for Magnetic Resonance Imaging: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 15:33. [PMID: 39791792 PMCID: PMC11722098 DOI: 10.3390/nano15010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
This review highlights recent progress in utilizing iron oxide nanoparticles (IONPs) as a safer alternative to gadolinium-based contrast agents (GBCAs) for magnetic resonance imaging (MRI). It consolidates findings from multiple studies, discussing current T1 contrast agents (CAs), the synthesis techniques for IONPs, the theoretical principles for designing IONP-based MRI CAs, and the key factors that impact their T1 contrast efficacy, such as nanoparticle size, morphology, surface modifications, valence states, and oxygen vacancies. Furthermore, we summarize current strategies to achieve IONP-based responsive CAs, including self-assembly/disassembly and distance adjustment. This review also evaluates the biocompatibility, organ accumulation, and clearance pathways of IONPs for clinical applications. Finally, the challenges associated with the clinical translation of IONP-based T1 CAs are included.
Collapse
Affiliation(s)
- Dongmei Zhang
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
| | - Jing Zhang
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
| | - Xianglin Bian
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
| | - Pei Zhang
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
| | - Weihua Wu
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
| | - Xudong Zuo
- School of Mathematics and Physics, Jiangsu University of Technology, Changzhou 213100, China; (D.Z.)
- The Jiangsu Key Laboratory of Clean Energy Storage and Conversion, Jiangsu University of Technology, Changzhou 213100, China
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
3
|
Thamizhchelvan AM, Ma H, Wu T, Nguyen D, Padelford J, Whitworth TJ, Li Y, Yang L, Mao H. Shape-dependent cellular uptake of iron oxide nanorods: mechanisms of endocytosis and implications on cell labeling and cellular delivery. NANOSCALE 2024; 16:21398-21415. [PMID: 39329423 PMCID: PMC11429166 DOI: 10.1039/d4nr02408g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
The effects of nanoparticle morphology, especially size and shape, on their interactions with cells are of great interest in understanding the fate of nanoparticles in biological systems and designing them for biomedical applications. While size and shape-dependent cell behavior, endocytosis mechanism, and subcellular distribution of nanoparticles have been investigated extensively with gold and other nanoparticles, studies on iron oxide nanoparticles (IONP), one of the most promising and well-thought-of nanomaterials in biomedical applications, were limited. In this study, we synthesized oligosaccharide-coated water-soluble iron oxide nanorods (IONR) with different core sizes (nm) and different aspect ratios (i.e., length/width), such as IONR(L) at 140/6 nm and IONR(S) at 50/7 nm as well as spherical IONP (20 nm). We investigated how their sizes and shapes affect uptake mechanisms, localization, and cell viability in different cell lines. The results of transmission electron microscopy (TEM) and confocal fluorescence microscopic imaging confirmed the internalization of these nanoparticles in different types of cells and subsequent accumulation in the subcellular compartments, such as the endosomes, and into the cytosol. Specifically, IONR(L) exhibited the highest cellular uptake compared to IONR(S) and spherical IONP, 1.36-fold and 1.17-fold higher than that of spherical IONP in macrophages and pediatric brain tumor medulloblastoma cells, respectively. To examine the cellular uptake mechanisms preferred by the different IONR and IONP, we used different endocytosis inhibitors to block specific cellular internalization pathways when cells were treated with different nanoparticles. The results from these blocking experiments showed that IONR(L) enter macrophages and normal kidney cells through clathrin-mediated, dynamin-dependent, and macropinocytosis/phagocytosis pathways, while they are internalized in cancer cells primarily via clathrin/caveolae-mediated and phagocytosis mechanisms. Overall, our findings provide new insights into further development of magnetic IONR-based imaging probes and drug delivery systems for biomedical applications.
Collapse
Affiliation(s)
- Anbu Mozhi Thamizhchelvan
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | - Hedi Ma
- 5M Biomed, LLC, Atlanta, Georgia 30303, USA
| | - Tianhe Wu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | - Darlene Nguyen
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | | - Ted J Whitworth
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Yuancheng Li
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
- 5M Biomed, LLC, Atlanta, Georgia 30303, USA
| | - Lily Yang
- Department of Surgery Emory University School of Medicine, Atlanta, Georgia 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
4
|
Shang K, Xu C, Cao Z, Cui M, Sun J, Xiao H, Zhang L, Wang Y, Han H. Polymer-based delivery systems with metal complexes as contrast agents for medical imaging. Coord Chem Rev 2024; 518:216071. [DOI: 10.1016/j.ccr.2024.216071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Wu T, Zhu W, Duan R, Sun J, Bao S, Chen K, Han B, Chen Y, Lu Y. Magnetic vagus nerve stimulation ameliorates contrast-induced acute kidney injury by circulating plasma exosomal miR-365-3p. J Nanobiotechnology 2024; 22:666. [PMID: 39468562 PMCID: PMC11520859 DOI: 10.1186/s12951-024-02928-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Contrast-induced acute kidney injury (CI-AKI) is manifested by a rapid decline in renal function occurring within 48-72 h in patients exposed to iodinated contrast media (CM). Although intravenous hydration is currently the effective method confirmed to prevent CI-AKI, it has several drawbacks. Some investigations have demonstrated the nephroprotective effects of vagus nerve stimulation (VNS) against kidney ischemia-reperfusion injury, but no direct research has investigated the use of VNS for treating CI-AKI. Additionally, most current VNS treatment applies invasive electrical stimulator implantation, which is largely limited by the complications. Our recent publications introduce the magnetic vagus nerve stimulation (mVNS) system pioneered and successfully used for the treatment of myocardial infarction. However, it remains uncertain whether mVNS can mitigate CI-AKI and its specific underlying mechanisms. Therefore, we herein evaluate the potential therapeutic effects of mVNS on CM-induced nephropathy in rats and explore the underlying mechanisms. RESULTS mVNS treatment was found to significantly improve the damaged renal function, including the reduction of elevated serum creatinine (Scr), blood urea nitrogen (BUN), and urinary N-acetyl-β-D-glucosaminidase (NAG) with increased urine output. Pathologically, mVNS treatment alleviated the renal tissue structure injury, and suppressed kidney injury molecule-1 (KIM-1) expression and apoptosis in renal tubular epithelial cells. Mechanistically, increased circulating plasma exosomal miR-365-3p after mVNS treatment enhanced the autophagy and reduced CM-induced apoptosis in renal tubular epithelial cells by targeting Ras homolog enriched in brain (Rheb). CONCLUSIONS In summary, we demonstrated that mVNS can improve CI-AKI through enhanced autophagy and apoptosis inhibition, which depended on plasma exosomal miR-365-3p. Our findings highlight the therapeutic potential of mVNS for CI-AKI in clinical practice. However, further research is needed to determine the optimal stimulation parameters to achieve the best therapeutic effects.
Collapse
Affiliation(s)
- Tianyu Wu
- XuZhou Clinical School of Xuzhou Medical University, Department of Central Laboratory, Xuzhou Central Hospital, No.199 Jiefang South Road, Xuzhou, 221009, P.R. China
| | - Wenwu Zhu
- XuZhou Clinical School of Xuzhou Medical University, Department of Cardiology, Xuzhou Central Hospital, XuZhou Institute of Cardiovascular disease, No.199 Jiefang South Road, Xuzhou, 221009, P.R. China
| | - Rui Duan
- XuZhou Clinical School of Xuzhou Medical University, Department of Cardiology, Xuzhou Central Hospital, XuZhou Institute of Cardiovascular disease, No.199 Jiefang South Road, Xuzhou, 221009, P.R. China
| | - Jianfei Sun
- The State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, P.R. China
| | - Siyuan Bao
- The State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, P.R. China
| | - Kaiyan Chen
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, P.R. China
| | - Bing Han
- XuZhou Clinical School of Xuzhou Medical University, Department of Cardiology, Xuzhou Central Hospital, XuZhou Institute of Cardiovascular disease, No.199 Jiefang South Road, Xuzhou, 221009, P.R. China.
| | - Yuqiong Chen
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, PR China.
| | - Yao Lu
- XuZhou Clinical School of Xuzhou Medical University, Department of Cardiology, Xuzhou Central Hospital, XuZhou Institute of Cardiovascular disease, No.199 Jiefang South Road, Xuzhou, 221009, P.R. China.
| |
Collapse
|
6
|
Guo Y, Shi Z, Han L, Qin X, You J, Zhang Q, Chen X, Zhao Y, Sun J, Xia Y. Infection-Sensitive SPION/PLGA Scaffolds Promote Periodontal Regeneration via Antibacterial Activity and Macrophage-Phenotype Modulation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:41855-41868. [PMID: 39093305 DOI: 10.1021/acsami.4c06430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Inflammation caused by a bacterial infection and the subsequent dysregulation of the host immune-inflammatory response are detrimental to periodontal regeneration. Herein, we present an infection-sensitive scaffold prepared by layer-by-layer assembly of Feraheme-like superparamagnetic iron oxide nanoparticles (SPIONs) on the surface of a three-dimensional-printed polylactic-co-glycolic acid (PLGA) scaffold. The SPION/PLGA scaffold is magnetic, hydrophilic, and bacterial-adhesion resistant. As indicated by gene expression profiling and confirmed by quantitative real-time reverse transcription polymerase chain reaction and flow cytometry analysis, the SPION/PLGA scaffold facilitates macrophage polarization toward the regenerative M2 phenotype by upregulating IL-10, which is the molecular target of repair promotion, and inhibits macrophage polarization toward the proinflammatory M1 phenotype by downregulating NLRP3, which is the molecular target of anti-inflammation. As a result, macrophages modulated by the SPS promote osteogenic differentiation of bone marrow mesenchymal stromal cells (BMSCs) in vitro. In a rat periodontal defect model, the SPION/PLGA scaffold increased IL-10 secretion and decreased NLRP3 and IL-1β secretion with Porphyromonas gingivalis infection, achieving superior periodontal regeneration than the PLGA scaffold alone. Therefore, this antibacterial SPION/PLGA scaffold has anti-inflammatory and bacterial antiadhesion properties to fight infection and promote periodontal regeneration by immunomodulation. These findings provide an important strategy for developing engineered scaffolds to treat periodontal defects.
Collapse
Affiliation(s)
- Yu Guo
- The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Zihan Shi
- The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Liping Han
- The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| | - Xuan Qin
- The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jiayi You
- The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Qian Zhang
- The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Suzhou Stomatological Hospital, Suzhou, Jiangsu 215000, China
| | - Xichen Chen
- Analytical and Testing Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yantao Zhao
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China
- State Key Laboratory of Military Stomatology, Shanxi, Xi'an 710032, China
| | - Jianfei Sun
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yang Xia
- The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
7
|
Zhao Y, Pan J, Han B, Hou W, Li B, Wang J, Wang G, He Y, Ma M, Zhou J, Yu C, Sun SK. Ultrahigh-Resolution Visualization of Vascular Heterogeneity in Brain Tumors via Magnetic Nanoparticles-Enhanced Susceptibility-Weighted Imaging. ACS NANO 2024; 18:21112-21124. [PMID: 39094075 DOI: 10.1021/acsnano.4c02611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The precise assessment of vascular heterogeneity in brain tumors is vital for diagnosing, grading, predicting progression, and guiding treatment decisions. However, currently, there is a significant shortage of high-resolution imaging approaches. Herein, we propose a contrast-enhanced susceptibility-weighted imaging (CE-SWI) utilizing the minimalist dextran-modified Fe3O4 nanoparticles (Dextran@Fe3O4 NPs) for ultrahigh-resolution mapping of vasculature in brain tumors. The Dextran@Fe3O4 NPs are prepared via a facile coprecipitation method under room temperature, and exhibit small hydrodynamic size (28 nm), good solubility, excellent biocompatibility, and high transverse relaxivity (r2*, 159.7 mM-1 s-1) under 9.4 T magnetic field. The Dextran@Fe3O4 NPs-enhanced SWI can increase the contrast-to-noise ratio (CNR) of cerebral vessels to 2.5 times that before injection and achieves ultrahigh-spatial-resolution visualization of microvessels as small as 0.1 mm in diameter. This advanced imaging capability not only allows for the detailed mapping of both enlarged peritumoral drainage vessels and the intratumoral microvessels, but also facilitates the sensitive imaging detection of vascular permeability deterioration in a C6 cells-bearing rat glioblastoma model. Our proposed Dextran@Fe3O4 NPs-enhanced SWI provides a powerful imaging technique with great clinical translation potential for the precise theranostics of brain tumors.
Collapse
Affiliation(s)
- Yujie Zhao
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jinbin Pan
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Bing Han
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wenjing Hou
- Department of Diagnostic and Therapeutic Ultrasonography, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Bingjie Li
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jiaojiao Wang
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Guohe Wang
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300204, China
| | - Yujing He
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Min Ma
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Junzi Zhou
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300204, China
| |
Collapse
|
8
|
Liu S, Sun J. Magnetic nanomaterials mediate precise magnetic therapy. Biomed Phys Eng Express 2024; 10:052001. [PMID: 38981447 DOI: 10.1088/2057-1976/ad60cb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 07/09/2024] [Indexed: 07/11/2024]
Abstract
Magnetic nanoparticle (MNP)-mediated precision magnet therapy plays a crucial role in treating various diseases. This therapeutic strategy compensates for the limitations of low spatial resolution and low focusing of magnetic stimulation, and realizes the goal of wireless teletherapy with precise targeting of focal areas. This paper summarizes the preparation methods of magnetic nanomaterials, the properties of magnetic nanoparticles, the biological effects, and the measurement methods for detecting magnetism; discusses the research progress of precision magnetotherapy in the treatment of psychiatric disorders, neurological injuries, metabolic disorders, and bone-related disorders, and looks forward to the future development trend of precision magnet therapy.
Collapse
Affiliation(s)
- Sha Liu
- Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, People's Republic of China
| | - Jianfei Sun
- Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, People's Republic of China
| |
Collapse
|
9
|
Wu L, Zhang Y, Zhao F, Sheng J, Gu N. Magnetic Characterization of Human Intrinsically Magnetic Monocytes Through a Novel Optical Tracking-Based Magnetic Sensor. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307306. [PMID: 38312110 DOI: 10.1002/smll.202307306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/17/2024] [Indexed: 02/06/2024]
Abstract
Intrinsically magnetic cells naturally occur within organisms and are believed to be linked to iron metabolism and certain cellular functions while the functional significance of this magnetism is largely unexplored. To better understand this property, an approach named Optical Tracking-based Magnetic Sensor (OTMS) has been developed. This multi-target tracking system is designed to measure the magnetic moment of individual cells. The OTMS generates a tunable magnetic field and induces movement in magnetic cells that are subsequently analyzed through a learning-based tracking-by-detection system. The magnetic moment of numerous cells can be calculated simultaneously, thereby providing a quantitative tool to assess cellular magnetic properties within populations. Upon deploying the OTMS, a stable population of magnetic cells in human peripheral monocytes is discovered. Further application in the analysis of clinical blood samples reveals an intriguing pattern: the proportion of magnetic monocytes differs significantly between systemic lupus erythematosus (SLE) patients and healthy volunteers. This variation is positively correlated with disease activity, a trend not observed in patients with rheumatoid arthritis (RA). The study, therefore, presents a new frontier in the investigation of the magnetic characteristics of naturally occurring magnetic cells, opening the door to potential diagnostic and therapeutic applications that leverage cellular magnetism.
Collapse
Affiliation(s)
- Linyuan Wu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
| | - Yuxin Zhang
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
| | - Fengfeng Zhao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jingyi Sheng
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
- Medical School, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
10
|
Wang K, Zhang M, Geng Z, Zhang S, Deng Z, Tan J, Zhang Q, Jiao Z, Gu N. One-Step Preparation of Magnetic Lipid Bubbles: Magnetothermal Effect Induces the Simultaneous Formation of Gas Nuclei and Self-Assembly of Phospholipids. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30755-30765. [PMID: 38847111 DOI: 10.1021/acsami.4c03788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
In recent years, enveloped micro-nanobubbles have garnered significant attention in research due to their commendable stability, biocompatibility, and other notable properties. Currently, the preparation methods of enveloped micro-nanobubbles have limitations such as complicated preparation process, large bubble size, wide distribution range, low yield, etc. There exists an urgent demand to devise a simple and efficient method for the preparation of enveloped micro-nanobubbles, ensuring both high concentration and a uniform particle size distribution. Magnetic lipid bubbles (MLBs) are a multifunctional type of enveloped micro-nanobubble combining magnetic nanoparticles with lipid-coated bubbles. In this study, MLBs are prepared simply and efficiently by a magneto internal heat bubble generation process based on the interfacial self-assembly of iron oxide nanoparticles induced by the thermogenic effect in an alternating magnetic field. The mean hydrodynamic diameter of the MLBs obtained was 384.9 ± 8.5 nm, with a polydispersity index (PDI) of 0.248 ± 0.021, a zeta potential of -30.5 ± 1.0 mV, and a concentration of (7.92 ± 0.46) × 109 bubbles/mL. Electron microscopy results show that the MLBs have a regular spherical stable core-shell structure. The superparamagnetic iron oxide nanoparticles (SPIONs) and phospholipid layers adsorbed around the spherical gas nuclei of the MLBs, leading the particles to demonstrate commendable superparamagnetic and magnetic properties. In addition, the effects of process parameters on the morphology of MLBs, including phospholipid concentration, phospholipid proportiona, current intensity, magnetothermal time, and SPION concentration, were investigated and discussed to achieve controlled preparation of MLBs. In vitro imaging results reveal that the higher the concentration of MLBs loaded with iron oxide nanoparticles, the better the in vitro ultrasound (US) imaging and magnetic resonance imaging (MRI) results. This study proves that the magneto internal heat bubble generation process is a simple and efficient technique for preparing MLBs with high concentration, regular structure, and commendable properties. These findings lay a robust foundation for the mass production and application of enveloped micro-nanobubbles, particularly in biomedical fields and other related domains.
Collapse
Affiliation(s)
- Kailin Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Mengnan Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Zejin Geng
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
- Joint Research Institute of Southeast University and Monash University, Suzhou, Jiangsu 215123, PR China
| | - Shuo Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Zhuang Deng
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Jin Tan
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Zhen Jiao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
- Joint Research Institute of Southeast University and Monash University, Suzhou, Jiangsu 215123, PR China
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
- Medical School, Nanjing University, Nanjing, Jiangsu 210093, PR China
| |
Collapse
|
11
|
Bian X, Guo T, Chen G, Nie D, Yue M, Zhu Y, Lin M. The therapeutic effect and MR molecular imaging of FA-PEG-FePt/DDP nanoliposomes in AMF on ovarian cancer. Int J Nanomedicine 2024; 19:5227-5243. [PMID: 38855734 PMCID: PMC11162642 DOI: 10.2147/ijn.s453601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/17/2024] [Indexed: 06/11/2024] Open
Abstract
Purpose This study aimed to construct targeting drug-loading nanocomposites (FA-FePt/DDP nanoliposomes) to explore their potential in ovarian cancer therapy and molecular magnetic resonance imaging (MMRI). Methods FA-FePt-NPs were prepared by coupling folate (FA) with polyethylene-glycol (PEG)-coated ferroplatinum nanoparticles and characterized. Then cisplatin (DDP) was encapsulated in FA-FePt-NPs to synthesize FA-PEG-FePt/DDP nanoliposomes by thin film-ultrasonic method and high-speed stirring, of which MMRI potential, magnetothermal effect, and the other involved performance were analyzed. The therapeutic effect of FA-FePt/DDP nanoliposomes combined with magnetic fluid hyperthermia (MFH) on ovarian cancer in vitro and in vivo was evaluated. The expression levels of Bax and epithelial-mesenchymal transition related proteins were detected. The biosafety was also preliminarily observed. Results The average diameter of FA-FePt-NPs was about 30 nm, FA-FePt/DDP nanoliposomes were about 70 nm in hydrated particle size, with drug slow-release and good cell-specific targeted uptake. In an alternating magnetic field (AMF), FA-FePt/DDP nanoliposomes could rapidly reach the ideal tumor hyperthermia temperature (42~44 °C). MRI scan showed that FA-FePt-NPs and FA-FePt/DDP nanoliposomes both could suppress the T2 signal, indicating a good potential for MMRI. The in vitro and in vivo experiments showed that FA-FePt/DDP-NPs in AMF could effectively inhibit the growth of ovarian cancer by inhibiting cancer cell proliferation, invasion, and migration, and inducing cancer cell apoptosis, much better than that of the other individual therapies; molecularly, E-cadherin and Bax proteins in ovarian cancer cells and tissues were significantly increased, while N-cadherin, Vimentin, and Bcl-2 proteins were inhibited, effectively inhibiting the malignant progression of ovarian cancer. In addition, no significant pathological injury and dysfunction was observed in major visceras. Conclusion We successfully synthesized FA-FePt/DDP nanoliposomes and confirmed their good thermochemotherapeutic effect in AMF and MMRI potential on ovarian cancer, with no obvious side effects, providing a favorable strategy of integrated targeting therapy and diagnosis for ovarian cancer.
Collapse
Affiliation(s)
- Xuefeng Bian
- Imaging Department, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Ting Guo
- Institute of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Guojie Chen
- Institute of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Dengyun Nie
- Institute of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Miao Yue
- Institute of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Yinxing Zhu
- Institute of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| | - Mei Lin
- Clinical Medical Laboratory, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, People’s Republic of China
| |
Collapse
|
12
|
Wang P, Sun S, Bai G, Zhang R, Liang F, Zhang Y. Nanosized Prussian blue and its analogs for bioimaging and cancer theranostics. Acta Biomater 2024; 176:77-98. [PMID: 38176673 DOI: 10.1016/j.actbio.2023.12.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/29/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
Prussian blue (PB) nanoparticles (NPs) and Prussian blue analogs (PBAs) can form metal-organic frameworks through the programmable coordination of ferrous ions with cyanide. PB and PBAs represent a burgeoning class of hybrid functional nano-systems with a wide-ranging application spectrum encompassing biomedicine, cancer diagnosis, and therapy. A comprehensive overview of recent advancements is crucial for gaining insights for future research. In this context, we reviewed the synthesis techniques and surface modification strategies employed to tailor the dimensions, morphology, and attributes of PB NPs. Subsequently, we explored advanced biomedical utilities of PB NPs, encompassing photoacoustic imaging, magnetic resonance imaging, ultrasound (US) imaging, and multimodal imaging. In particular, the application of PB NPs-mediated photothermal therapy, photodynamic therapy, and chemodynamic therapy to cancer treatment was reviewed. Based on the literature, we envision an evolving trajectory wherein the future of Prussian blue-driven biological applications converge into an integrated theranostic platform, seamlessly amalgamating bioimaging and cancer therapy. STATEMENT OF SIGNIFICANCE: Prussian blue, an FDA-approved coordinative pigment with a centuries-long legacy, has paved the way for Prussian blue nanoparticles (PB NPs), renowned for their remarkable biocompatibility and biosafety. These PB NPs have found their niche in biomedicine, playing crucial roles in both diagnostics and therapeutic applications. The comprehensive review goes beyond PB NP-based cancer therapy. Alongside in-depth coverage of PB NP synthesis and surface modifications, the review delves into their cutting-edge applications in the realm of biomedical imaging, encompassing techniques such as photoacoustic imaging, magnetic resonance imaging, ultrasound imaging, and multimodal imaging.
Collapse
Affiliation(s)
- Pengfei Wang
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Shaohua Sun
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Guosheng Bai
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Ruiqi Zhang
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Fei Liang
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Yuezhou Zhang
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China; Ningbo Institute of Northwestern Polytechnical University, Frontiers Science Center for Flexible Electronics (FSCFE), Key Laboratory of Flexible Electronics of Zhejiang Province, 218 Qingyi Road, Ningbo, 315103, China.
| |
Collapse
|
13
|
Long M, Li Y, He H, Gu N. The Story of Ferumoxytol: Synthesis Production, Current Clinical Applications, and Therapeutic Potential. Adv Healthc Mater 2024; 13:e2302773. [PMID: 37931150 DOI: 10.1002/adhm.202302773] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Ferumoxytol, approved by the U.S. Food and Drug Administration in 2009, is one of the intravenous iron oxide nanoparticles authorized for the treatment of iron deficiency in chronic kidney disease and end-stage renal disease. With its exceptional magnetic properties, catalytic activity, and immune activity, as well as good biocompatibility and safety, ferumoxytol has gained significant recognition in various biomedical diagnoses and treatments. Unlike most existing reviews on this topic, this review primarily focuses on the recent clinical and preclinical advances of ferumoxytol in disease treatment, spanning anemia, cancer, infectious inflammatory diseases, regenerative medicine application, magnetic stimulation for neural modulation, etc. Additionally, the newly discovered mechanisms associated with the biological effects of ferumoxytol are discussed, including its magnetic, catalytic, and immunomodulatory properties. Finally, the summary and future prospects concerning the treatment and application of ferumoxytol-based nanotherapeutics are presented.
Collapse
Affiliation(s)
- Mengmeng Long
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Yan Li
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Hongliang He
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- Medical School, Nanjing University, Nanjing, 210008, P. R. China
| |
Collapse
|
14
|
Yuan Y, Chen B, Song L, An X, Zhang Q, Lu H, Li CM, Guo C. Magnetic two-dimensional nanocomposites for multimodal antitumor therapy: a recent review. J Mater Chem B 2024; 12:1404-1428. [PMID: 38251275 DOI: 10.1039/d3tb02333h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Magnetic two-dimensional nanocomposites (M2D NCs) that synergistically combine magnetic nanomedicine and 2D nanomaterials have emerged in multimodal antitumor therapy, attracting great interest in materials science and biomedical engineering. This review provides a summary of the recent advances of M2D NCs and their multimodal antitumor applications. We first introduce the design and fabrication of M2D NCs, followed by discussing new types of M2D NCs that have been recently reported. Then, a detailed analysis and discussions about the different types of M2D NCs are presented based on the structural categories of 2D NMs, including 2D graphene, transition metal dichalcogenides (TMDs), transition metal carbides/nitrides/carbonitrides (MXenes), black phosphorus (BP), layered double hydroxides (LDHs), metal organic frameworks (MOFs), covalent organic frameworks (COFs) and other 2D nanomaterials. In particular, we focus on the synthesis strategies, magnetic or optical responsive performance, and the versatile antitumor applications, which include magnetic hyperthermia therapy (MHT), photothermal therapy (PTT), photodynamic therapy (PDT), drug delivery, immunotherapy and multimodal imaging. We conclude the review by proposing future developments with an emphasis on the mass production and biodegradation mechanism of the M2D NCs. This work is expected to provide a comprehensive overview to researchers and engineers who are interested in such a research field and promote the clinical translation of M2D NCs in practical applications.
Collapse
Affiliation(s)
- Ying Yuan
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, 99 Xuefu Road, Suzhou, 215009, Jiangsu, P. R. China.
| | - Bo Chen
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, 99 Xuefu Road, Suzhou, 215009, Jiangsu, P. R. China.
| | - Luping Song
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, 99 Xuefu Road, Suzhou, 215009, Jiangsu, P. R. China.
| | - Xingxing An
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, 99 Xuefu Road, Suzhou, 215009, Jiangsu, P. R. China.
| | - Qinrui Zhang
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, 99 Xuefu Road, Suzhou, 215009, Jiangsu, P. R. China.
| | - Hao Lu
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, 99 Xuefu Road, Suzhou, 215009, Jiangsu, P. R. China.
| | - Chang Ming Li
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, 99 Xuefu Road, Suzhou, 215009, Jiangsu, P. R. China.
| | - Chunxian Guo
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, 99 Xuefu Road, Suzhou, 215009, Jiangsu, P. R. China.
| |
Collapse
|
15
|
Wang X, Sun L, Qin X, You J, Zhang J, Xia Y. Enhanced Anti-inflammatory Capacity of the Conditioned Medium Derived from Periodontal Ligament Stem Cells Modified with an Iron-Based Nanodrug. Adv Biol (Weinh) 2023; 7:e2300044. [PMID: 37409394 DOI: 10.1002/adbi.202300044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/28/2023] [Indexed: 07/07/2023]
Abstract
Cell-free therapy using conditioned medium (CM) from mesenchymal stem cells takes full advantage of the bioactive factors secreted by the cells while avoiding disadvantages such as immune rejection and tumor formation due to cell transplantation. In this study, human periodontal ligament stem cells (PDLSCs) are modified with the superparamagnetic iron oxide nanoparticle (SPION)-based nanodrug ferumoxytol (PDLSC-SPION). Compared with PDLSCs, PDLSC-SPION showed good cell viability and better osteogenic differentiation ability. Cell-free CM is collected and the anti-inflammatory capacity of PDLSC CM and PDLSC-SPION CM is assessed by treatment of lipopolysaccharide-stimulated macrophages and IL-17-stimulated human gingival fibroblasts. Both CMs inhibited the expression of proinflammatory cytokines in cells, and the therapeutic effect is more distinct for PDLSC-SPION CM than PDLSC CM, which may be due to their different proteomic compositions. Therefore, modification of PDLSCs with ferumoxytol enhances the anti-inflammatory capacity of its CM, making it more potentially useful for the treatment of inflammatory diseases such as periodontitis.
Collapse
Affiliation(s)
- Xinyue Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Liuxu Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Xuan Qin
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Jiayi You
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Jing Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Yang Xia
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
16
|
Fang G, Zhang Z, Jiang B, Zheng Y, Xiao X, Wang T, Zhang Z, Zhao J. Immunologically active ferumoxytol-poly(I : C) nanomaterials inhibit metastatic melanoma by regulating myeloid-derived suppressor cell differentiation. Biomater Sci 2023. [PMID: 37366334 DOI: 10.1039/d3bm00416c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Nanomaterials have been identified as a potential therapeutic option for targeting myeloid-derived suppressor cells (MDSCs), which are known to play a crucial role in tumor metastasis and treatment resistance. Here, we report a unique immunologically active nanomaterial composed of ferumoxytol and poly(I : C) (FP-NPs) and investigate its immunoregulatory activities on MDSCs in metastatic melanoma. In vivo assays demonstrated that FP-NPs had the ability to significantly impede the progression of metastatic melanoma and decrease the MDSC population in the lungs, spleen, and bone marrow of mice. Both in vivo and in vitro experiments revealed that FP-NPs reduced the number of granulocytic MDSCs and promoted the differentiation of monocytic MDSCs into anti-tumor M1 macrophages. Transcriptome sequencing indicated that FP-NPs significantly altered the expression of several genes involved in immunity. Analysis of Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and quantitative real-time PCR revealed that FP-NPs significantly increased the expression of the myeloid cell differentiation-related gene interferon regulatory factor 7 and activated interferon beta-related signaling pathways, which stimulated the differentiation of MDSCs into M1 macrophages. These findings suggest that FP-NPs, a unique nanomaterial with immunological properties, can induce MDSCs to differentiate into M1 macrophages, potentially offering new treatment prospects for metastatic melanoma in the future.
Collapse
Affiliation(s)
- Gaochuan Fang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| | - Zhonghai Zhang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| | - Bo Jiang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
- Department of Urology, Xuzhou Central Hospital, Xuzhou, 221009, China
| | - Yunuo Zheng
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| | - Xufeng Xiao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| | - Tianlong Wang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| | - Zhengkui Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, China.
| | - Jiaojiao Zhao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| |
Collapse
|
17
|
Shi Z, Jia L, Zhang Q, Sun L, Wang X, Qin X, Xia Y. An altered oral microbiota induced by injections of superparamagnetic iron oxide nanoparticle-labeled periodontal ligament stem cells helps periodontal bone regeneration in rats. Bioeng Transl Med 2023; 8:e10466. [PMID: 37206247 PMCID: PMC10189485 DOI: 10.1002/btm2.10466] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/31/2022] [Accepted: 11/23/2022] [Indexed: 10/20/2023] Open
Abstract
Stem cell injection is good for periodontal regeneration due to the capacity of stem cells to differentiate toward osteogenic direction and to regulate the production of pro- and anti-inflammatory cytokines. However, injected cells are difficult to track in vivo. And there is microbiota in oral cavity, the dysbiosis of which leads to the damage and loss of periodontal tissue. Here, we demonstrated an enhanced periodontal repair was due to an altered oral microbiota. Periodontal defects were surgically prepared in rats, and periodontal ligament stem cells (PDLSCs) labeled by superparamagnetic iron oxide (SPIO) nanoparticles (PC-SPIO) were injected, with PDLSCs and saline treatments as controls. Detected by magnetic resonance imaging (MRI) and histological staining, PC-SPIO was major at limited areas in regenerated periodontal tissues. PC-SPIO-treated rats achieved better periodontal regeneration than the other two groups. Concurrently, the oral microbiota of PC-SPIO-treated rats was changed, presenting SPIO-Lac as a biomarker. SPIO-Lac assisted periodontal repair in vivo, inhibited the inflammation of macrophages induced by lipopolysaccharide (LPS) and antibacterial in vitro. Therefore, our study proved that SPIO-labeled cells can be tracked in periodontal defect and highlighted a potential positive role of an oral microbiota in periodontal regeneration, suggesting the possibility of periodontal repair promotion by manipulating oral microbiota.
Collapse
Affiliation(s)
- Zihan Shi
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingJiangsuPeople's Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsuPeople's Republic of China
| | - Lu Jia
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingJiangsuPeople's Republic of China
- Department of Emergency General Dentistry, Hebei Key Laboratory of StomatologyHebei Medical UniversityShijiazhuangHebeiPeople's Republic of China
| | - Qian Zhang
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingJiangsuPeople's Republic of China
- Suzhou Stomatological HospitalSuzhouJiangsuPeople's Republic of China
| | - Liuxu Sun
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingJiangsuPeople's Republic of China
| | - Xinyue Wang
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingJiangsuPeople's Republic of China
| | - Xuan Qin
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingJiangsuPeople's Republic of China
| | - Yang Xia
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingJiangsuPeople's Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsuPeople's Republic of China
| |
Collapse
|
18
|
Bao S, Lu Y, Zhang J, Xue L, Zhang Y, Wang P, Zhang F, Gu N, Sun J. Rapid improvement of heart repair in rats after myocardial infarction by precise magnetic stimulation on the vagus nerve with an injectable magnetic hydrogel. NANOSCALE 2023; 15:3532-3541. [PMID: 36723151 DOI: 10.1039/d2nr05073k] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The imbalance between the sympathetic and the parasympathetic nervous system is one of the main pathogeneses of myocardial infarction (MI). Vagus nerve stimulation (VNS), which restores autonomic nervous balance by enhancing the parasympathetic drive, is shown to have benefits for patients with MI. As a clinically safe and effective remote neuromodulation method, magnetic stimulation is expected to overcome the problems of infection and nerve injury caused by electrode implantation. However, it is difficult to achieve precise stimulation on a single vagus nerve due to the poor focus of the magnetic field. Here, we described a novel magnetic vagus nerve stimulation (mVNS) system, which consisted of an injectable chitosan/β-glycerophosphate (CS/GP) hydrogel loaded with superparamagnetic iron oxide (SPIO) nanoparticles and a mild magnetic pulse sequence. The injectable hydrogel prepared from clinically safe materials ensured minimally invasive implantation, and the SPIO nanoparticles in the hydrogel mediated the precise magnetic stimulation of a single vagus nerve. Under a mild magnetic field (∼100 mT), a decrease in heart rate and a change in vagus nerve potential were found in rats under in situ injection of a magnetic CS/GP hydrogel. Magnetic stimulation on the vagus nerve for 4 weeks (20 Hz, three times daily, 5 minutes each time) significantly improved the cardiac function and reduced the infarct size of the rats subjected to myocardial infarction, accompanied by suppression of inflammatory cell infiltration and inflammation factor expression. Taken together, these results demonstrated that the mVNS exhibited promising potential for treating myocardial infarction in the clinic.
Collapse
Affiliation(s)
- Siyuan Bao
- The State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, P. R. China.
| | - Yao Lu
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210009, P. R. China.
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated XuZhou Hospital of Nanjing Medical University, No. 199 Jiefang South Road, Xuzhou, 221009, P. R. China
| | - Jian Zhang
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210009, P. R. China.
| | - Le Xue
- The State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, P. R. China.
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, P. R. China
| | - Peng Wang
- The State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, P. R. China.
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Fengxiang Zhang
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210009, P. R. China.
| | - Ning Gu
- School of Medicine, Nanjing University, Nanjing, 210009, P. R. China.
| | - Jianfei Sun
- The State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, P. R. China.
| |
Collapse
|
19
|
Zheng Y, Jiang B, Guo H, Zhang Z, Chen B, Zhang Z, Wu S, Zhao J. The combinational nano-immunotherapy of ferumoxytol and poly(I:C) inhibits melanoma via boosting anti-angiogenic immunity. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 49:102658. [PMID: 36708910 DOI: 10.1016/j.nano.2023.102658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/11/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
Angiogenesis plays a key role in the progression and metastasis of melanoma, and the pro-angiogenic effect of macrophages is one major reason for the failure of current anti-angiogenic therapies. Here, a nano-immunotherapy combining ferumoxytol and poly(I:C) (ferumoxytol/poly(I:C)) has been developed to boost the anti-angiogenic activities of macrophages to inhibit melanoma. Our findings demonstrated that ferumoxytol/poly(I:C) was a highly efficacious anti-tumor therapy with limited toxicity. Both in vivo and in vitro experiments indicated that this combination was successful in impeding angiogenesis. Ferumoxytol/poly(I:C) was demonstrated to reduce the viability of endothelial cells, thus hindering tube formation. Particularly, ferumoxytol/poly(I:C) was able to polarize macrophages to the M1 phenotype and decrease the expression of vascular endothelial growth factor, which in turn amplified the anti-angiogenic properties of ferumoxytol/poly(I:C). This combination of ferumoxytol/poly(I:C) nano-immunotherapy enriches the anti-angiogenic therapeutic nature of ferumoxytol and will shed new light on the treatment of melanoma.
Collapse
Affiliation(s)
- Yunuo Zheng
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China
| | - Bo Jiang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China; Department of Urology, Xuzhou Central Hospital, Xuzhou 221009, China
| | - Hongmei Guo
- Department of Ultrasonography, Weinan Maternal and Child Health Hospital, Weinan 714000, Shaanxi, China
| | - Zhonghai Zhang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China; Department of Physiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Bo Chen
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Zhengkui Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, China.
| | - Shaoyuan Wu
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China.
| | - Jiaojiao Zhao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China.
| |
Collapse
|
20
|
Cardiovascular Nanotechnology. Nanomedicine (Lond) 2023. [DOI: 10.1007/978-981-16-8984-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
21
|
Chen Y, Bai Y, Wang X, Zhang H, Zheng H, Gu N. Plasmonic/magnetic nanoarchitectures: From controllable design to biosensing and bioelectronic interfaces. Biosens Bioelectron 2023; 219:114744. [PMID: 36327555 DOI: 10.1016/j.bios.2022.114744] [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: 07/14/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 02/08/2023]
Abstract
Controllable design of the nanocrystal-assembled plasmonic/magnetic nanoarchitectures (P/MNAs) inspires abundant methodologies to enhance light-matter interactions and control magnetic-induced effects by means of fine-tuning the morphology and ordered packing of noble metallic or magnetic building blocks. The burgeoning development of multifunctional nanoarchitectures has opened up broad range of interdisciplinary applications including biosensing, in vitro diagnostic devices, point-of-care (POC) platforms, and soft bioelectronics. By taking advantage of their customizability and efficient conjugation with capping biomolecules, various nanoarchitectures have been integrated into high-performance biosensors with remarkable sensitivity and versatility, enabling key features that combined multiplexed detection, ease-of-use and miniaturization. In this review, we provide an overview of the representative developments of nanoarchitectures that being built by plasmonic and magnetic nanoparticles over recent decades. The design principles and key mechanisms for signal amplification and quantitative sensitivity have been explored. We highlight the structure-function programmability and prospects of addressing the main limitations for conventional biosensing strategies in terms of accurate selectivity, sensitivity, throughput, and optoelectronic integration. State-of-the-art strategies to achieve affordable and field-deployable POC devices for early multiplexed detection of infectious diseases such as COVID-19 has been covered in this review. Finally, we discuss the urgent yet challenging issues in nanoarchitectures design and related biosensing application, such as large-scale fabrication and integration with portable devices, and provide perspectives and suggestions on developing smart biosensors that connecting the materials science and biomedical engineering for personal health monitoring.
Collapse
Affiliation(s)
- Yi Chen
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Southeast University-Monash University Joint Research Institute, Suzhou, 215123, China.
| | - Yu Bai
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Southeast University-Monash University Joint Research Institute, Suzhou, 215123, China
| | - Xi Wang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Southeast University-Monash University Joint Research Institute, Suzhou, 215123, China
| | - Heng Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Southeast University-Monash University Joint Research Institute, Suzhou, 215123, China
| | - Haoran Zheng
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Southeast University-Monash University Joint Research Institute, Suzhou, 215123, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Southeast University-Monash University Joint Research Institute, Suzhou, 215123, China.
| |
Collapse
|
22
|
Fine-tuned magnetic nanobubbles for magnetic hyperthermia treatment of glioma cells. Biointerphases 2022; 17:061004. [DOI: 10.1116/6.0002110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Magnetic nanoparticle (MNP) induced magnetic hyperthermia has been demonstrated as a promising technique for the treatment of brain tumor. However, lower heating efficiency resulting from low intratumoral accumulation of magnetic nanomaterials is still one of the significant limitations for their thermotherapeutic efficacy. In this study, we have designed a nanobubble structure with MNPs decorated on the shell, which leads to the improvement of magnetocaloric performance under an alternating magnetic field. First, the phospholipid coupled with MNPs as the shell to be self-assembled magnetic nanobubbles (MNBs) was fabricated by a temperature-regulated repeated compression self-assembly approach. Then, the optimal magnetic heating concentration, electric current parameters for producing the magnetic field, and the number of magnetic heating times were investigated for tuning the better magnetoenergy conversion. Finally, the well-defined geometrical orientation of MNPs on the nanobubble structure enhanced hypothermia effect was investigated. The results demonstrate that the MNBs could promote the endocytosis of magnetic nanoparticles by glioma cells, resulting in better therapeutic effect. Therefore, the controlled assembly of MNPs into well-defined bubble structures could serve as a new hyperthermia agent for tumor therapy.
Collapse
|
23
|
Wang J, Guo Y, Jiao Z, Tan J, Zhang M, Zhang Q, Gu N. Generation of micro-nano bubbles by magneto induced internal heat for protecting cells from intermittent hypoxic damage. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.130289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
24
|
The coprecipitation formation study of iron oxide nanoparticles with the assist of a gas/liquid mixed phase fluidic reactor. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
25
|
Liu X, Wang N, Liu X, Deng R, Kang R, Xie L. Vascular Repair by Grafting Based on Magnetic Nanoparticles. Pharmaceutics 2022; 14:pharmaceutics14071433. [PMID: 35890328 PMCID: PMC9320478 DOI: 10.3390/pharmaceutics14071433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 12/11/2022] Open
Abstract
Magnetic nanoparticles (MNPs) have attracted much attention in the past few decades because of their unique magnetic responsiveness. Especially in the diagnosis and treatment of diseases, they are mostly involved in non-invasive ways and have achieved good results. The magnetic responsiveness of MNPs is strictly controlled by the size, crystallinity, uniformity, and surface properties of the synthesized particles. In this review, we summarized the classification of MNPs and their application in vascular repair. MNPs mainly use their unique magnetic properties to participate in vascular repair, including magnetic stimulation, magnetic drive, magnetic resonance imaging, magnetic hyperthermia, magnetic assembly scaffolds, and magnetic targeted drug delivery, which can significantly affect scaffold performance, cell behavior, factor secretion, drug release, etc. Although there are still challenges in the large-scale clinical application of MNPs, its good non-invasive way to participate in vascular repair and the establishment of a continuous detection process is still the future development direction.
Collapse
Affiliation(s)
| | | | | | | | | | - Lin Xie
- Correspondence: (R.K.); (L.X.)
| |
Collapse
|
26
|
Liao W, Lu J, Wang Q, Yan S, Li Y, Zhang Y, Wang P, Jiang Q, Gu N. Osteogenesis of Iron Oxide Nanoparticles-Labeled Human Precartilaginous Stem Cells in Interpenetrating Network Printable Hydrogel. Front Bioeng Biotechnol 2022; 10:872149. [PMID: 35573235 PMCID: PMC9099245 DOI: 10.3389/fbioe.2022.872149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/05/2022] [Indexed: 01/22/2023] Open
Abstract
Smart biomaterials combined with stem cell-based therapeutic strategies have brought innovation in the field of bone tissue regeneration. However, little is known about precartilaginous stem cells (PCSCs), which can be used as seed cells and incorporated with bioactive scaffolds for reconstructive tissue therapy of bone defects. Herein, iron oxide nanoparticles (IONPs) were employed to modulate the fate of PCSCs, resulting in the enhanced osteogenic differentiation potential both in vitro and in vivo. PCSCs were isolated from the ring of La-Croix extracted from polydactylism patient and identified through immunohistochemically staining using anti-FGFR-3 antibodies. Potential toxicity of IONPs toward PCSCs was assessed through cell viability, proliferation, and attachment assay, and the results demonstrated that IONPs exhibited excellent biocompatibility. After that, the effects of IONPs on osteogenic differentiation of PCSCs were evaluated and enhanced ALP activity, formation of mineralized nodule, and osteogenic-related genes expressions could be observed upon IONPs treatment. Moreover, in vivo bone regeneration assessment was performed using rabbit femur defects as a model. A novel methacrylated alginate and 4-arm poly (ethylene glycol)-acrylate (4A-PEGAcr)-based interpenetrating polymeric printable network (IPN) hydrogel was prepared for incorporation of IONPs-labeled PCSCs, where 4A-PEGAcr was the common component for three-dimensional (3D) printing. The implantation of IONPs-labeled PCSCs significantly accelerated the bone formation process, indicating that IONPs-labeled PCSCs could endow current scaffolds with excellent osteogenic ability. Together with the fact that the IONPs-labeled PCSCs-incorporated IPN hydrogel (PCSCs-hydrogels) was biosafety and printable, we believed that PCSCs-hydrogels with enhanced osteogenic bioactivity could enrich the stem cell-based therapeutic strategies for bone tissue regeneration.
Collapse
Affiliation(s)
- Wei Liao
- Children’s Hospital of Nanjing Medical University, Nanjing, China
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Jingwei Lu
- Department of Orthopedics, Jinling School of Clinical Medicine, Nanjing Medical University, Jinling Hospital, Nanjing, China
| | - Qianjin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Sen Yan
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| | - Yan Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| | - Yibo Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Peng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- *Correspondence: Qing Jiang, ; Ning Gu,
| | - Ning Gu
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
- *Correspondence: Qing Jiang, ; Ning Gu,
| |
Collapse
|
27
|
Chen Y, Zhang Q, Qin X, Li J, Zhao Y, Xia Y. Superparamagnetic Iron Oxide Nanoparticles Protect Human Gingival Fibroblasts from Porphyromonas gingivalis Invasion and Inflammatory Stimulation. Int J Nanomedicine 2022; 17:45-60. [PMID: 35027826 PMCID: PMC8749050 DOI: 10.2147/ijn.s333496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/23/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Modulating the inflammatory response of human gingival fibroblasts (hGFs) is important for the control of periodontal inflammation because it is a key event in the pathogenesis of periodontitis. Here, we aimed to determine whether polyglucose sorbitol carboxymethyl ether (PSC)-coated superparamagnetic iron oxide nanoparticles (SPIONs) protect hGFs against invasion and inflammatory stimulation by Porphyromonas gingivalis (P. gingivalis). Methods First, we determined the cytotoxicity and antimicrobial activity of PSC-SPIONs. Then, their effects on invasion of hGFs by P. gingivalis were evaluated by counting invading P. gingivalis, fluorescence staining, and transmission electron microscopy. The effect of PSC-SPIONs on inflammation in hGFs induced by P. gingivalis lipopolysaccharide was evaluated by measurement of reactive oxygen species (ROS), and enzyme-linked immunosorbent assays, quantitative reverse transcription-polymerase chain reaction, and Western blotting of key indicator molecules. The effects of dimercaptosuccinic acid (DMSA)-coated SPIONs and the free form of PSC alone were also tested and compared with those of PSC-SPIONs. Results PSC-SPIONs (25 μg/mL) are cytocompatible with hGFs and exhibit no antimicrobial effects on P. gingivalis. However, they inhibit invasion of hGFs by P. gingivalis at 15 μg/mL. They also decrease ROS production and inflammatory cytokine secretion by hGFs at 5, 15, and 25 μg/mL, by downregulating activation of the nuclear factor-kappa B signaling pathway. Furthermore, PSC alone does not inhibit inflammation, while DMSA-SPIONs do. This indicates that the nanosize effects of PSC-SPIONs, rather than their coating material, play the dominant role in their anti-inflammatory activity. Conclusion PSC-SPIONs protect hGFs against P. gingivalis invasion and inflammatory stimulation. Thus, they have potential for clinical application in control of periodontal inflammation.
Collapse
Affiliation(s)
- Yulian Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.,The Affiliated Stomatological Hospital of Soochow University, Suzhou Stomatological Hospital, Suzhou, Jiangsu, People's Republic of China
| | - Qian Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Xuan Qin
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Jin Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Yantao Zhao
- Institute of Orthopedics, Fourth Medical Center of the General Hospital of CPLA, Beijing, People's Republic of China.,Beijing Engineering Research Center of Orthopedics Implants, Beijing, People's Republic of China
| | - Yang Xia
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
28
|
Zhong D, Wang Y, Xie F, Chen S, Yang X, Ma Z, Wang S, Iqbal MZ, Ge J, Zhang Q, Zhao R, Kong X. Biomineralized Prussian Blue Nanotherapeutic for Enhanced Cancer Photothermal Therapy. J Mater Chem B 2022; 10:4889-4896. [DOI: 10.1039/d2tb00775d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Photothermal therapy is a promising tumor ablation technique that converts light into heat energy to kill cancer cells. Prussian blue (PB), a biocompatible photothermal reagent, has been widely explored for...
Collapse
|
29
|
Liu X, Sun Y, Chen B, Li Y, Zhu P, Wang P, Yan S, Li Y, Yang F, Gu N. Novel magnetic silk fibroin scaffolds with delayed degradation for potential long-distance vascular repair. Bioact Mater 2022; 7:126-143. [PMID: 34466722 PMCID: PMC8379427 DOI: 10.1016/j.bioactmat.2021.04.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 01/09/2023] Open
Abstract
Although with the good biological properties, silk fibroin (SF) is immensely restrained in long-distance vascular defect repair due to its relatively fast degradation and inferior mechanical properties. It is necessary to construct a multifunctional composite scaffold based on SF. In this study, a novel magnetic SF scaffold (MSFCs) was prepared by an improved infiltration method. Compared with SF scaffold (SFC), MSFCs were found to have better crystallinity, magnetocaloric properties, and mechanical strength, which was ascribed to the rational introduction of iron-based magnetic nanoparticles (MNPs). Moreover, in vivo and in vitro experiments demonstrated that the degradation of MSFCs was significantly extended. The mechanism of delayed degradation was correlated with the dual effect that was the newly formed hydrogen bonds between SFC and MNPs and the complexing to tyrosine (Try) to inhibit hydrolase by internal iron atoms. Besides, the β-crystallization of protein in MSFCs was increased with the rise of iron concentration, proving the beneficial effect after MNPS doped. Furthermore, although macrophages could phagocytose the released MNPs, it did not affect their function, and even a reasonable level might cause some cytokines to be upregulated. Finally, in vitro and in vivo studies demonstrated that MSFCs showed excellent biocompatibility and the growth promotion effect on CD34-labeled vascular endothelial cells (VECs). In conclusion, we confirm that the doping of MNPs can significantly reduce the degradation of SFC and thus provide an innovative perspective of multifunctional biocomposites for tissue engineering.
Collapse
Affiliation(s)
- Xin Liu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Yuxiang Sun
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Bo Chen
- Materials Science and Devices Institute, Suzhou University of Science and Technology, 1 Kerui Road, Suzhou, 215009, PR China
| | - Yan Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Peng Zhu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, 999078, PR China
| | - Peng Wang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Sen Yan
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Yao Li
- College of Social Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, PR China
| |
Collapse
|
30
|
Cardiovascular Nanotechnology. Nanomedicine (Lond) 2022. [DOI: 10.1007/978-981-13-9374-7_12-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
31
|
Hu S, Chen H, Zhou F, Liu J, Qian Y, Hu K, Yan J, Gu Z, Guo Z, Zhang F, Gu N. Superparamagnetic core-shell electrospun scaffolds with sustained release of IONPs facilitating in vitro and in vivo bone regeneration. J Mater Chem B 2021; 9:8980-8993. [PMID: 34494055 DOI: 10.1039/d1tb01261d] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bone tissue engineering (BTE) is a promising approach to recover insufficient bone in dental implantations. However, the clinical application of BTE scaffolds is limited by their low mechanical strength and lack of osteoinduction. In an attempt to circumvent these limitations and improve osteogenesis, we introduced magnetic iron oxide nanoparticles (IONPs) into a core-shell porous electrospun scaffold and evaluated their impact on the physical, mechanical, and biological properties of the scaffold. We used poly(lactic-co-glycolic acid)/polycaprolactone/beta-tricalcium phosphate (PPT) scaffolds with and without γ-Fe2O3 encapsulation, namely PPT-Fe scaffolds and PPT scaffolds, respectively. The γ-Fe2O3 used in the PPT-Fe scaffolds was coated with polyglucose sorbitol carboxymethylether and was biocompatible. Structurally, PPT-Fe scaffolds showed uniform iron distribution encapsulated within the resorbable PPT scaffolds, and these scaffolds supported sustainable iron release. Furthermore, compared with PPT scaffolds, PPT-Fe scaffolds showed significantly better physical and mechanical properties, including wettability, superparamagnetism, hardness, tensile strength, and elasticity modulus. In vitro tests of rat adipose-derived mesenchymal stem cells (rADSCs) seeded onto the scaffolds showed increased expression of integrin β1, alkaline phosphatase, and osteogenesis-related genes. In addition, enhanced in vivo bone regeneration was observed after implanting PPT-Fe scaffolds in rat calvarial bone defects. Thus, we can conclude that the incorporation of IONPs into porous scaffolds for long-term release can provide a new strategy for BTE scaffold optimization and is a promising approach that can offer enhanced osteogenic capacity in clinical applications.
Collapse
Affiliation(s)
- Shuying Hu
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Prosthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Hanbang Chen
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Prosthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Fang Zhou
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Prosthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Jun Liu
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Prosthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Yunzhu Qian
- Center of Stomatology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Ke Hu
- Laboratory of Oral Regenerative Medicine Technology, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing 210000, China
| | - Jia Yan
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Prosthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Zhuxiao Gu
- Jiangsu Key Laboratory for Science and Applications of Molecular Ferroelectrics, Southeast University, Nanjing 211189, China
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, 21218, USA
| | - Feimin Zhang
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Prosthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210009, China
| |
Collapse
|
32
|
Han L, Guo Y, Jia L, Zhang Q, Sun L, Yang Z, Dai Y, Lou Z, Xia Y. 3D magnetic nanocomposite scaffolds enhanced the osteogenic capacities of rat bone mesenchymal stem cells in vitro and in a rat calvarial bone defect model by promoting cell adhesion. J Biomed Mater Res A 2021; 109:1670-1680. [PMID: 33876884 DOI: 10.1002/jbm.a.37162] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 02/04/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022]
Abstract
Magnetic scaffolds incorporated with iron oxide nanoparticles (IONPs) are biocompatible and present excellent osteogenic properties. However, the underlying mechanism is unclear. In this study, 3D-printed poly(lactic-co-glycolic acid) scaffolds were coated with IONPs using layer-by-layer assembly (Fe-scaffold) to prepare magnetic scaffolds. The effects of this modification on osteogenesis were investigated by comparison with untreated scaffolds (Uncoated-scaffold). The results showed that the proliferation of rat bone mesenchymal stem cells (rBMSCs) on the Fe-scaffold was enhanced compared with those on the Uncoated-scaffold (p < 0.05). The alkaline phosphatase activity and expression levels of osteogenic-related genes of cells on the Fe-scaffold were higher than those on the Uncoated-scaffold (p < 0.05). Fe-scaffold was found to promote the cell adhesion compared with Uncoated-scaffold, including increasing the adhered cell number, promoting cell spreading and upregulating the expression levels of adhesion-related genes integrin α1 and β1 and their downstream signaling molecules FAK and ERK1/2 (p < 0.05). Moreover, the amount of new bone formed in rat calvarial defects at 8 weeks decreased in the order: Fe-scaffold > Uncoated-scaffold > Blank-control (samples whose defects were left empty) (p < 0.05). Therefore, 3D magnetic nanocomposite scaffolds enhanced the osteogenic capacities of rBMSCs in vitro and in a rat calvarial bone defect model by promoting cell adhesion. The mechanisms were attributed to the alteration in its hydrophilicity, surface roughness, and chemical composition.
Collapse
Affiliation(s)
- Liping Han
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Guo
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lu Jia
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liuxu Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zukun Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang Dai
- Department of Corona laboratory, Nanjing Suman Plasma Technology Co. Ltd., Nanjing, Jiangsu, China
| | - Zhichao Lou
- College of materials science and engineering, Nanjing Forestry University, Nanjing, Jiangsu, China
| | - Yang Xia
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
33
|
Han X, Tang S, Wang L, Xu X, Yan R, Yan S, Guo Z, Hu K, Yu T, Li M, Li Y, Zhang F, Gu N. Multicellular Spheroids Formation on Hydrogel Enhances Osteogenic/Odontogenic Differentiation of Dental Pulp Stem Cells Under Magnetic Nanoparticles Induction. Int J Nanomedicine 2021; 16:5101-5115. [PMID: 34349510 PMCID: PMC8327189 DOI: 10.2147/ijn.s318991] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/28/2021] [Indexed: 12/11/2022] Open
Abstract
Introduction Promotion odontogenic differentiation of dental pulp stem cells (DPSCs) is essential for dentin regeneration. Physical cellular microenvironment is of critical importance for stem cells differentiation and influences the function of other biological/chemical factors to differentiation. Methods Based on adjusting the mechanical/interfacial properties of hydrogels, multicellular spheroids (MCSs) of DPSCs generated through self-organization. The spheroids were characterized by immunofluorescent staining and flow cytometry. Quantitative real-time polymerase chain reaction, alkaline phosphatase (ALP) activity assay, ALP staining and Alizarin Red S staining were performed to evaluate the osteogenic/odontogenic differentiation of DPSCs with or without magnetic iron oxide nanoparticles (IONPs) induction. Results MCSs of DPSCs exhibited a significant upregulation of E-cadherin and N-cadherin and enriched CD146 positive subpopulation, along with a stronger osteogenic/odontogenic differentiation ability. Moreover, DPSCs spheroids showed more substantial osteogenic differentiation tendency than the classical two-dimensional cultured DPSCs under the stimulation of magnetic IONPs. Conclusion Three-dimensional spheroids culture of DPSCs based on composite viscoelastic materials combined with mechanical/magnetic stimulation may provide a theoretical basis for the subsequent development of dentin or bone regeneration technology.
Collapse
Affiliation(s)
- Xiao Han
- Jiangsu Key Laboratory of Oral Diseases, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.,Laboratory of Oral Regenerative Medicine Technology, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Shijia Tang
- Jiangsu Key Laboratory of Oral Diseases, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Lin Wang
- Jiangsu Key Laboratory of Oral Diseases, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Xueqin Xu
- Laboratory of Oral Regenerative Medicine Technology, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Ruhan Yan
- Laboratory of Oral Regenerative Medicine Technology, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Sen Yan
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Ke Hu
- Jiangsu Key Laboratory of Oral Diseases, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.,Laboratory of Oral Regenerative Medicine Technology, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Tingting Yu
- Department of Medical Genetics, School of Basic Medical Science & Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Mengping Li
- Laboratory of Oral Regenerative Medicine Technology, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Yuqin Li
- Laboratory of Oral Regenerative Medicine Technology, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.,Laboratory of Oral Regenerative Medicine Technology, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Ning Gu
- Laboratory of Oral Regenerative Medicine Technology, School of Biomedical Engineering and Informatics, Department of Biomedical Engineering, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
34
|
Besenhard MO, Panariello L, Kiefer C, LaGrow AP, Storozhuk L, Perton F, Begin S, Mertz D, Thanh NTK, Gavriilidis A. Small iron oxide nanoparticles as MRI T1 contrast agent: scalable inexpensive water-based synthesis using a flow reactor. NANOSCALE 2021; 13:8795-8805. [PMID: 34014243 DOI: 10.1039/d1nr00877c] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Small iron oxide nanoparticles (IONPs) were synthesised in water via co-precipitation by quenching particle growth after the desired magnetic iron oxide phase formed. This was achieved in a millifluidic multistage flow reactor by precisely timed addition of an acidic solution. IONPs (≤5 nm), a suitable size for positive T1 magnetic resonance imaging (MRI) contrast agents, were obtained and stabilised continuously. This novel flow chemistry approach facilitates a reproducible and scalable production, which is a crucial paradigm shift to utilise IONPs as contrast agents and replace currently used Gd complexes. Acid addition had to be timed carefully, as the inverse spinel structure formed within seconds after initiating the co-precipitation. Late quenching allowed IONPs to grow larger than 5 nm, whereas premature acid addition yielded undesired oxide phases. Use of a flow reactor was not only essential for scalability, but also to synthesise monodisperse and non-agglomerated small IONPs as (i) co-precipitation and acid addition occurred at homogenous environment due to accurate temperature control and rapid mixing and (ii) quenching of particle growth was possible at the optimum time, i.e., a few seconds after initiating co-precipitation. In addition to the timing of growth quenching, the effect of temperature and dextran present during co-precipitation on the final particle size was investigated. This approach differs from small IONP syntheses in batch utilising either growth inhibitors (which likely leads to impurities) or high temperature methods in organic solvents. Furthermore, this continuous synthesis enables the low-cost (<£10 per g) and large-scale production of highly stable small IONPs without the use of toxic reagents. The flow-synthesised small IONPs showed high T1 contrast enhancement, with transversal relaxivity (r2) reduced to 20.5 mM-1 s-1 and longitudinal relaxivity (r1) higher than 10 mM-1 s-1, which is among the highest values reported for water-based IONP synthesis.
Collapse
Affiliation(s)
| | - Luca Panariello
- Department of Chemical Engineering, University College London, London, WC1E 7JE, UK.
| | - Céline Kiefer
- Institut de Physique et Chimie des Matériaux de Strasbourg, BP 43, 67034, Strasbourg, France
| | - Alec P LaGrow
- International Iberian Nanotechnology Laboratory, Braga 4715-330, Portugal
| | - Liudmyla Storozhuk
- Biophysics group, Department of Physics and Astronomy, University College London, London, WC1E 6BT, UK.
| | - Francis Perton
- Institut de Physique et Chimie des Matériaux de Strasbourg, BP 43, 67034, Strasbourg, France
| | - Sylvie Begin
- Institut de Physique et Chimie des Matériaux de Strasbourg, BP 43, 67034, Strasbourg, France
| | - Damien Mertz
- Institut de Physique et Chimie des Matériaux de Strasbourg, BP 43, 67034, Strasbourg, France
| | - Nguyen Thi Kim Thanh
- Biophysics group, Department of Physics and Astronomy, University College London, London, WC1E 6BT, UK. and UCL Healthcare Biomagnetic and Nanomaterials Laboratories, 21 Albemarle Street, London, W1S 4BS, UK
| | - Asterios Gavriilidis
- Department of Chemical Engineering, University College London, London, WC1E 7JE, UK.
| |
Collapse
|
35
|
Liu X, Xu Y, Li Y, Pan Y, Zhao S, Hou Y. Ferumoxytol-β-glucan Inhibits Melanoma Growth via Interacting with Dectin-1 to Polarize Macrophages into M1 Phenotype. Int J Med Sci 2021; 18:3125-3139. [PMID: 34400883 PMCID: PMC8364471 DOI: 10.7150/ijms.61525] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/14/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Regulating the polarization of macrophages to antitumor M1 macrophages is a promising strategy for overcoming the immunosuppression of the tumor microenvironment for cancer therapy. Ferumoxytol (FMT) can not only serve as a drug deliver agent but also exerts anti-tumor activity. β-glucan has immuno-modulating properties to prevent tumor growth. Thus, a nanocomposite of FMT surface-coated with β-glucan (FMT-β-glucan) was prepared to explore its effect on tumor suppression. Methods: Male B16F10 melanoma mouse model was established to explore the antitumor effect of FMT-β-glucan. The viability and apoptotic rates of B16F10 cells were detected by cell counting kit-8 and Annexin-V/PI experiments. The levels of M1 markers were quantified by quantitative reverse transcription-polymerase chain reaction and enzyme linked immunosorbent assay. Phagocytic activity and intracellular reactive oxygen species (ROS) in macrophages were evaluated by the neutral red uptake assay and flow cytometry, respectively. Small interfering RNA (siRNA) transfection was applied to knock down the Dectin-1 gene in RAW 264.7 cells. Results: FMT-β-glucan suppressed tumor growth to a greater extent and induced higher infiltration of M1 macrophages than the combination of FMT and β-glucan (FMT+β-glucan) in vivo. In vitro, supernatant from FMT-β-glucan-treated RAW 264.7 cells led to lower cell viability and induced more apoptosis of B16F10 cells than that from the FMT+β-glucan group. Moreover, FMT-β-glucan boosted the expression of M1 type markers, and increased phagocytic activity and ROS in RAW 264.7 cells. Further research indicated that FMT-β-glucan treatment promoted the level of Dectin-1 on the surface of RAW 264.7 cells and that knockdown of Dectin-1 abrogated the phosphorylation levels of several components in MAPK and NF-κB signaling. Conclusion: The nanocomposite FMT-β-glucan suppressed melanoma growth by inducing the M1 macrophage-activated tumor microenvironment.
Collapse
Affiliation(s)
- Xinghan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Yujun Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Yi Li
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Yuchen Pan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Shuli Zhao
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
- ✉ Corresponding authors: Yayi Hou, The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China. Tel.: +86-25-8968-8441; Fax: +86-25-8968-8441. E-mail: ; Shuli Zhao, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China. E-mail:
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
- ✉ Corresponding authors: Yayi Hou, The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China. Tel.: +86-25-8968-8441; Fax: +86-25-8968-8441. E-mail: ; Shuli Zhao, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China. E-mail:
| |
Collapse
|
36
|
Ye D, Li M, Xie Y, Chen B, Han Y, Liu S, Wei QH, Gu N. Optical Imaging and High-Accuracy Quantification of Intracellular Iron Contents. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2005474. [PMID: 33306269 DOI: 10.1002/smll.202005474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/19/2020] [Indexed: 06/12/2023]
Abstract
Precise quantification of intracellular iron contents is important to biomedical applications of magnetic nanoparticles. Current approaches for iron quantification rely on specialized instruments while most only yield iron quantities averaged over plenty of cells. Here, a simple and robust approach, combining digital optical microscopy with the Beer-Lambert's law, that allows for imaging stainable iron distribution in individual cells and the quantification of stainable iron contents with an unprecedented accuracy of femtogram per pixel, is presented. It is further shown that this approach enables studying of the internalization and reduction dynamics of super-paramagnetic iron oxide nanoparticles (SPIONs) by stem cells in single cell level.
Collapse
Affiliation(s)
- Dewen Ye
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Mingxi Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Yuanyuan Xie
- Center for Clinic Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Bo Chen
- Materials Science and Devices Institute, Suzhou University of Science and Technology, 1 Kerui Road, Suzhou, 215009, China
| | - Yuexia Han
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Qi-Huo Wei
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Physics, Nanjing Medical University, Nanjing, 211166, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210009, China
| |
Collapse
|
37
|
Liang Y, Xie J, Yu J, Zheng Z, Liu F, Yang A. Recent advances of high performance magnetic iron oxide nanoparticles: Controlled synthesis, properties tuning and cancer theranostics. NANO SELECT 2020. [DOI: 10.1002/nano.202000169] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Yi‐Jun Liang
- School of Medical Engineering Foshan University Foshan 528000 P.R. China
| | - Jun Xie
- School of Life Science Jiangsu Normal University Xuzhou 221116 P.R. China
| | - Jing Yu
- College of Materials Science and Engineering Zhejiang University of Technology Hangzhou 310014 P.R. China
| | - Zhaoguang Zheng
- School of Medical Engineering Foshan University Foshan 528000 P.R. China
| | - Fang Liu
- School of Medical Engineering Foshan University Foshan 528000 P.R. China
| | - Anping Yang
- School of Medical Engineering Foshan University Foshan 528000 P.R. China
| |
Collapse
|
38
|
Wang H, Ge Y, Sun J, Wang H, Gu N. Magnetic sensor based on image processing for dynamically tracking magnetic moment of single magnetic mesenchymal stem cell. Biosens Bioelectron 2020; 169:112593. [PMID: 32966950 DOI: 10.1016/j.bios.2020.112593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 01/06/2023]
Abstract
Developing an economical and universal method to measure the magnetic moments of magnetic mesenchymal stem cells (MSCs) labelled with superparamagnetic iron oxide (SPIO) nanoparticles is crucial for cell tracking. In this study, we used a gradient magnetic field created by a nickel needle to track the motion of cells. A simple and quantifiable magnetic sensor was employed to evaluate the magnetic properties of single viable MSCs. We measured the magnetic moments of microbeads and MSCs using the proposed method and compared the results with magnetic moments measured using a superconducting quantum interference device and with iron contents measured using an inductively coupled plasma spectrometer, respectively. The correlation coefficients indicated satisfactory agreement in both cases, thus confirming the accuracy of the system. By labelling MSCs with SPIOs, we implemented a miniature magnetic sensor to measure the magnetic moments of single magnetic MSCs quantitatively using an image-processing algorithm. Existing methods for the measurement of magnetic moments at the micro/nanoscale have various limitations. Our system realised the measurement of single viable cells, thereby providing a theoretical foundation for the labelling and tracking of MSCs with SPIO nanoparticles. Additionally, the proposed system is both economical and universal.
Collapse
Affiliation(s)
- Haoyao Wang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Yuqing Ge
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Jianfei Sun
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Hong Wang
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, PR China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China.
| |
Collapse
|
39
|
Yu P, Zheng L, Wang P, Chai S, Zhang Y, Shi T, Zhang L, Peng R, Huang C, Guo B, Jiang Q. Development of a novel polysaccharide-based iron oxide nanoparticle to prevent iron accumulation-related osteoporosis by scavenging reactive oxygen species. Int J Biol Macromol 2020; 165:1634-1645. [PMID: 33049237 DOI: 10.1016/j.ijbiomac.2020.10.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/26/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023]
Abstract
In this work, the biological polysaccharide-based antioxidant polyglucose-sorbitol-carboxymethyl ether (PSC) was used as the precursor to synthesize Fe2O3@PSC nanoparticles, which are expected to scavenge excess reactive oxygen species (ROS) to inhibit osteogenesis and promote osteoclast differentiation in iron accumulation (IA)-related osteoporosis. The Fe2O3@PSC nanoparticles obtained were of a uniform particle size of 7.3 nm with elemental O/Fe/Cl/C at a ratio of 190:7:2:88. In addition, the Fe2O3@PSC nanoparticles showed the ability to supply equivalent amounts of iron as the typical iron agent ferric ammonium citrate (FAC) in vitro and in vivo. Importantly, the Fe2O3@PSC nanoparticles not only induced antioxidative MC3T3-E1 and Raw 264.7 cells to scavenge ROS but also promoted osteogenic differentiation by activating Akt-GSK-3β-β-catenin and inhibiting osteoclast differentiation by inhibiting the MAPK and NF-κB pathways in vitro. In vivo, no IA-related osteoporosis was induced in a mouse model when enough iron was supplied by the Fe2O3@PSC nanoparticles. Overall, the biological polysaccharide-based antioxidant PSC can supply iron and prevent IA-related osteoporosis, indicating that it is a promising novel iron agent for applications to treat iron deficiency diseases.
Collapse
Affiliation(s)
- Pengjun Yu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China; Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing 210093, Jiangsu, PR China
| | - Liming Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China; Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing 210093, Jiangsu, PR China
| | - Peng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China; Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing 210093, Jiangsu, PR China
| | - Senlin Chai
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China; Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing 210093, Jiangsu, PR China
| | - Yibo Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China; Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing 210093, Jiangsu, PR China
| | - Tianshu Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China; Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing 210093, Jiangsu, PR China
| | - Lei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China; Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing 210093, Jiangsu, PR China
| | - Rui Peng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China; Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing 210093, Jiangsu, PR China
| | - Caoxing Huang
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China.
| | - Baosheng Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China; Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing 210093, Jiangsu, PR China.
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, PR China; Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing 210093, Jiangsu, PR China.
| |
Collapse
|
40
|
Wang X, Cheng L. Multifunctional Prussian blue-based nanomaterials: Preparation, modification, and theranostic applications. Coord Chem Rev 2020. [DOI: 10.1016/j.ccr.2020.213393] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
41
|
Zhou Y, Gu N, Yang F. In situ microbubble-assisted, ultrasound-controlled release of superparamagnetic iron oxide nanoparticles from gastro-retentive tablets. Int J Pharm 2020; 586:119615. [PMID: 32650114 DOI: 10.1016/j.ijpharm.2020.119615] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 06/28/2020] [Accepted: 07/01/2020] [Indexed: 11/27/2022]
Abstract
Organic and inorganic nanomaterials have shown great potential in drug delivery applications due to their unique physical and chemical properties. Orally administered nanoparticles have attracted great attention because it is acceptable, convenient, and safe. However, nanoparticles need to overcome numerous hurdles such as acidic gastric environment, the continuous secretion of mucus, and fast gastric emptying after being delivered via an oral route. Here, we used a stimuli-responsive and triggered release strategy for superparamagnetic iron oxide nanoparticles (SPIONPs)-loaded gastro-retentive tablets for in situ bubbles generation. These materials realize SPIOs controlled release and delivery specific to the stomach. The tablet formulation contains a foaming agent (sodium bicarbonate, NaHCO3), adhesive component (HPMC/carbomer 934 P (1:1)), filler (lactose/mannitol (10:1)) and SPIONPs. The in vitro bubble generation and SPIONPs released from the tablets were characterized. The ex vivo gastric adhesive ability, acoustic stimuli performance, and tissue penetration were further evaluated. The results show that when the fabricated tablets interacted with the acidic microenvironment, the carbon dioxide (CO2) could be generated and be captured by ultrasound (US) imaging. Simultaneous with bubble production, SPIONPs are released from the tablets to further control ultrasound-mediated force and deliver SPIONPs entering through the mucus layer. The SPIONPs were loaded in the tablets and could be released in a controllable way; thus, the magnetic resonance imaging (MRI) could also be used to monitor the tablet status and SPIONP delivery process. Therefore, SPIONPs-loaded gastro-retentive effervescent tablets offer effective release and absorption of nanoparticles in the gastric area and be imaged by MRI and US.
Collapse
Affiliation(s)
- Yue Zhou
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
42
|
Chen B, Zhang R, Wu H, Li M, Zhou G, Ji M. Thermoresponsive magnetoliposome encapsulating doxorubicin and high performance Ferumoxytol for effective tumor synergistic therapy in vitro. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
43
|
Chen B, Xing J, Li M, Liu Y, Ji M. DOX@Ferumoxytol-Medical Chitosan as magnetic hydrogel therapeutic system for effective magnetic hyperthermia and chemotherapy in vitro. Colloids Surf B Biointerfaces 2020; 190:110896. [DOI: 10.1016/j.colsurfb.2020.110896] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/26/2020] [Accepted: 02/22/2020] [Indexed: 12/19/2022]
|
44
|
Shi C, Li Y, Gu N. Iron-Based Nanozymes in Disease Diagnosis and Treatment. Chembiochem 2020; 21:2722-2732. [PMID: 32315111 DOI: 10.1002/cbic.202000094] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/21/2020] [Indexed: 12/15/2022]
Abstract
Iron-based nanozymes are currently one of the few clinical inorganic nanoparticles for disease diagnosis and treatment. Overcoming the shortcomings of natural enzymes, such as easy inactivation and low yield, combined with their special nanometer properties and magnetic functions, iron-based nanozymes have broad prospects in biomedicine. This minireview summarizes their preparation, biological activity, catalytic mechanism, and applications in diagnosis and treatment of diseases. Finally, challenges to their future development and the trends of iron-based nanozymes are discussed. The purpose of this minireview is to better understand and reasonably speculate on the rational design of iron-based nanozymes as an increasingly important new paradigm for diagnostics.
Collapse
Affiliation(s)
- Chu Shi
- State Key Laboratory of Bioelectronics Jiangsu Key Laboratory for Biomaterials and Devices School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Yan Li
- State Key Laboratory of Bioelectronics Jiangsu Key Laboratory for Biomaterials and Devices School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics Jiangsu Key Laboratory for Biomaterials and Devices School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210009, China
| |
Collapse
|
45
|
Chen B, Guo Z, Guo C, Mao Y, Qin Z, Ye D, Zang F, Lou Z, Zhang Z, Li M, Liu Y, Ji M, Sun J, Gu N. Moderate cooling coprecipitation for extremely small iron oxide as a pH dependent T 1-MRI contrast agent. NANOSCALE 2020; 12:5521-5532. [PMID: 32091066 DOI: 10.1039/c9nr10397j] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Iron based nanomedicine (IBNM) has been one powerful diagnostic tool as a magnetic resonance imaging (MRI) contrast agent (CA) in the clinic for years. Conventional IBNMs are generally employed as T2-MRI CAs, but most of them are constrained in clinical indication expansion by magnetic susceptibility artifacts. In comparison, extremely small iron oxide (ESIO) with a core size less than 5 nm has demonstrated the T1-MRI effect, which provides prospects for a Gd-based agent alternative. Nevertheless, currently developed ESIOs for T1-MRI CAs always require harsh conditions such as a high temperature and high boiling point reagent. Moreover, very few of the currently developed ESIOs meet the stringent pharmaceutical standard. Herein, on the basis of a crystal nuclear precipitation-dissolution equilibrium mechanism and outer/inner sphere T1-MRI theory, monodisperse ESIOs with an average size of 3.43 nm (polydispersity index of 0.104) are fabricated using a moderate cooling procedure with mild coprecipitation reaction conditions. The as-synthesized ESIOs display around 3-fold higher T1 MRI signal intensity than that of commercial Ferumoxytol (FMT), comparable to that of Gd-based CAs in vitro. Additionally, the T1-MRI performance of the ESIOs is pH dependent and delivers bright signal augmentation. Eventually, the internalization into mesenchymal stem cells of the ESIO is realized in the absence of a transferring agent. Considering the identical structure and composition of the ESIOs as compared to that of FMT, they could meet the pharmaceutical criteria, thus providing great potential as T1-MRI Cas, for instance as stem cell tracers.
Collapse
Affiliation(s)
- Bo Chen
- Materials Science and Devices Institute, Suzhou University of Science and Technology, 1 Kerui Road, Suzhou 215009, Jiangsu, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Li M, Li J, Chen J, Liu Y, Cheng X, Yang F, Gu N. Platelet Membrane Biomimetic Magnetic Nanocarriers for Targeted Delivery and in Situ Generation of Nitric Oxide in Early Ischemic Stroke. ACS NANO 2020; 14:2024-2035. [PMID: 31927980 DOI: 10.1021/acsnano.9b08587] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Early diagnosis and treatment of acute ischemic stroke poses a significant challenge due to its suddenness and short therapeutic time window. Human endogenous cells derived biomimetic drug carriers have provided new options for stroke theranostics since these cells have higher biosafety and targeting abilities than artificial carriers. Inspired by natural platelets (PLTs) and their role in targeting adhesion to the damaged blood vessel during thrombus formation, we fabricated a biomimetic nanocarrier comprising a PLT membrane envelope loaded with l-arginine and γ-Fe2O3 magnetic nanoparticles (PAMNs) for thrombus-targeted delivery of l-arginine and in situ generation of nitric oxide (NO). Results demonstrate that the engineered 200 nm PAMNs inherit the natural properties of the PLT membrane and achieve rapid targeting to ischemic stroke lesions under the guidance of an external magnetic field. Subsequent to the release of l-arginine at the thrombus site, endothelial cells produce NO, which promotes vasodilation to disrupt the local PLT aggregation. Rapid targeting of PAMNs to stroke lesions as well as in situ generation of NO prompts vasodilation, recovery of blood flow, and reperfusion of the stroke microvascular. Thus, these PLT membrane derived nanocarriers are diagnostically beneficial for localizing stroke lesions and a promising modality for executing therapies.
Collapse
Affiliation(s)
- Mingxi Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Jing Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Jinpeng Chen
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Microbiology and Immunology, Medical School , Southeast University , Nanjing 210009 , China
| | - Yang Liu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Xiao Cheng
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| |
Collapse
|
47
|
Wallyn J, Anton N, Vandamme TF. Synthesis, Principles, and Properties of Magnetite Nanoparticles for In Vivo Imaging Applications-A Review. Pharmaceutics 2019; 11:E601. [PMID: 31726769 PMCID: PMC6920893 DOI: 10.3390/pharmaceutics11110601] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/09/2019] [Indexed: 12/16/2022] Open
Abstract
The current nanotechnology era is marked by the emergence of various magnetic inorganic nanometer-sized colloidal particles. These have been extensively applied and hold an immense potential in biomedical applications including, for example, cancer therapy, drug nanocarriers (NCs), or in targeted delivery systems and diagnosis involving two guided-nanoparticles (NPs) as nanoprobes and contrast agents. Considerable efforts have been devoted to designing iron oxide NPs (IONPs) due to their superparamagnetic (SPM) behavior (SPM IONPs or SPIONs) and their large surface-to-volume area allowing more biocompatibility, stealth, and easy bonding to natural biomolecules thanks to grafted ligands, selective-site moieties, and/or organic and inorganic corona shells. Such nanomagnets with adjustable architecture have been the topic of significant progresses since modular designs enable SPIONs to carry out several functions simultaneously such as local drug delivery with real-time monitoring and imaging of the targeted area. Syntheses of SPIONs and adjustments of their physical and chemical properties have been achieved and paved novel routes for a safe use of those tailored magnetic ferrous nanomaterials. Herein we will emphasis a basic notion about NPs magnetism in order to have a better understanding of SPION assets for biomedical applications, then we mainly focus on magnetite iron oxide owing to its outstanding magnetic properties. The general methods of preparation and typical characteristics of magnetite are reviewed, as well as the major biomedical applications of magnetite.
Collapse
Affiliation(s)
| | - Nicolas Anton
- Université de Strasbourg, CNRS, CAMB UMR 7199, F-67000 Strasbourg, France;
| | | |
Collapse
|
48
|
Liu Y, Li J, Chen H, Cai Y, Sheng T, Wang P, Li Z, Yang F, Gu N. Magnet-activatable nanoliposomes as intracellular bubble microreactors to enhance drug delivery efficacy and burst cancer cells. NANOSCALE 2019; 11:18854-18865. [PMID: 31596307 DOI: 10.1039/c9nr07021d] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
To address the thereapeutic challenges in clinical cancer treatment and guarantee efficient and rapid intracellular delivery of drugs while evading efflux and chemotherapy resistance, herein, we designed a liposomal nanostructure equipped with superparamagnetic iron oxide nanoparticles (SPIOs) and anethole trithione (ADT, a hydrogen sulfide (H2S) donor drug). At first, by spatially focused manipulation of the external static magnetic field (SMF), the SPIOs and ADT-loaded liposomes (SPIOs-ADT-LPs) could rapidly overcome the cell membrane barrier to enter the cytoplasm, which could be imaged by magnetic resonance imaging (MRI). Sequentially, the intracellular release of ADT drugs was triggered by enzymatic catalysis to generate acoustic-sensitive H2S gas. At the beginning, during the production of H2S at low concentrations, the cell membrane could be permeabilized to further increase the cellular uptake of SPIOs-ADT-LPs. The continued generation of H2S gas bubbles, imaged by ultrasound (US) imaging, further enhanced the intracellular hydrostatic pressure (above 320 pN per cell) to physically unfold the cytoskeleton, leading to complete cell death. The magneto-acoustic approach based on SPIO-ADT-LPs as intracellular bubble reactors leads to improved anticancer cell efficacy and has potential applications for novel MRI/US dual image-guided bubble bursting of cancer cells.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Jing Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Heming Chen
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yan Cai
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Tianyu Sheng
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Peng Wang
- Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210093, China
| | - Zhiyong Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
49
|
Wang G, Zhao J, Zhang M, Wang Q, Chen B, Hou Y, Lu K. Ferumoxytol and CpG oligodeoxynucleotide 2395 synergistically enhance antitumor activity of macrophages against NSCLC with EGFR L858R/T790M mutation. Int J Nanomedicine 2019; 14:4503-4515. [PMID: 31417255 PMCID: PMC6599896 DOI: 10.2147/ijn.s193583] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 04/22/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose: Drug resistance is a major challenge for epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) treatment of lung cancer. Ferumoxytol (FMT) drives macrophage (MΦ) transformation towards a M1-like phenotype and thereby inhibits tumor growth. CpG oligodeoxynucleotide 2395 (CpG), a toll-like receptor 9 (TLR9) agonist, is an effective therapeutic agent to induce anticancer immune responses. Herein, the effect of co-administered FMT and CpG on MΦ activation for treating non-small cell lung cancer (NSCLC) was explored. Methods: The mRNA expression levels of M1-like genes in RAW 264.7 MΦ cells stimulated by FMT, CpG and FMT and CpG (FMT/CpG) were evaluated by quantitative reverse transcription PCR (qRT-PCR). Then, the effects of FMT/CpG-pretreated MΦ supernatant on apoptosis and proliferation of H1975 cells were detected by flow cytometry, and the expression of EGFR and its downstream signaling pathway in H1975 cells were explored by western blotting. Finally, a H1975 cell xenograft mouse model was used to study the anti-tumor effect of the combination of FMT and CpG in vivo. Results: FMT and CpG synergistically enhanced M1-like gene expression in MΦ, including tumor necrosis factor-α, interleukin (IL)-12, IL-1α, IL-1β, IL-6 and inducible nitric oxide synthase (iNOS). FMT/CpG-pretreated MΦ supernatant inhibited proliferation and induced apoptosis of H1975 cells, accompanied by down-regulation of cell cycle-associated proteins and up-regulation of apoptosis-related proteins. Further studies indicated that the FMT/CpG-pretreated MΦ supernatant suppressed p-EGFR and its downstream AKT/mammalian target of rapamycin signaling pathway in H1975 cells. Furthermore, FMT/CpG suppressed tumor growth in mice accompanied by a decline in the EGFR-positive tumor cell fraction and increased M1 phenotype macrophage infiltration. Conclusion: FMT acted synergistically with CpG to activate MΦ for suppressed proliferation and promoted apoptosis of NSCLC cells via EGFR signaling. Thus, combining FMT and CpG is an effective strategy for the treatment of NSCLC with EGFRL858R/T790M mutation.
Collapse
Affiliation(s)
- Guoqun Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Jiaojiao Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China
| | - Meiling Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Qian Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Bo Chen
- Institute of Materials Science and Devices, Suzhou University of Science and Technology, Suzhou 215009, People's Republic of China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, People's Republic of China
| | - Kaihua Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| |
Collapse
|
50
|
Zhao P, Chen B, Li L, Wu H, Li Y, Shaneen B, Zhan X, Gu N. Missing-in-metastasis protein promotes internalization of magnetic nanoparticles via association with clathrin light chain and Rab7. Biochim Biophys Acta Gen Subj 2019; 1863:502-510. [PMID: 30528490 PMCID: PMC8218922 DOI: 10.1016/j.bbagen.2018.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/05/2018] [Accepted: 12/04/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Magnetic nanoparticles (MNPs) have been widely used in biomedical applications. Proper control of the duration of MNPs in circulation promises to improve further their applications, in particularly drug delivery. It is known that the uptake of tissue-associated MNPs is mainly carried out by macrophages. Yet, the molecular mechanism to control MNPs internalization in macrophages remains to be elusive. Missing-in-metastasis (MIM) is a scaffolding protein that is highly expressed in macrophages and regulates receptor-mediated endocytosis. We hypothesize that uptake of MNPs may also involve the function of MIM. METHODS We investigated the effect of MIM expression on the intracellular trafficking of MNPs by transmission electronic microscopy, flow cytometry, o-phenanthroline photometric analysis, Perl's staining, immunofluorescence microscopy and co-immunoprecipitation. To explore the molecular events in MIM-mediated MNPs uptake, we examined the effect of MNPs on the interaction of MIM with clathrin, Rab5 and Rab7. RESULTS Uptake of MNPs was significantly enhanced in cells overexpressing MIM. Upon exposure to MNPs, MIM was associated with clathrin light chain in endocytic vesicles and Rab7, a protein that regulates late endosomes. However, MNPs caused dissociation of MIM with Rab5, an early endosome-associated protein. CONCLUSIONS MIM regulates internalization of MNPs via promoting their trafficking from plasma membrane to late endosomes. GENERAL SIGNIFICANCE Our data unveiled a novel pathway which MNPs internalization and intracellular trafficking in macrophages. This new pathway may allow us to control the uptake of MNPs within cells by targeting MIM, thereby improving their medical applications.
Collapse
Affiliation(s)
- Peng Zhao
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou 215123, PR China
| | - Bo Chen
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China; Materials Science and Devices Institute, Suzhou University of Science and Technology, 1 Kerui Road, Suzhou, Jiangsu 215009, PR China
| | - Lushen Li
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hao Wu
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 210029, PR China
| | - Yan Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou 215123, PR China
| | - Baxter Shaneen
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xi Zhan
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou 215123, PR China.
| |
Collapse
|