1
|
Yao Q, Wang B, Yu J, Pan Q, Yu Y, Feng X, Chen W, Yang J, Gao C, Cao H. ROS-responsive nanoparticle delivery of obeticholic acid mitigate primary sclerosing cholangitis. J Control Release 2024; 374:112-126. [PMID: 39117112 DOI: 10.1016/j.jconrel.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Primary sclerosing cholangitis (PSC) is a challenging cholestatic liver disease marked by progressive bile duct inflammation and fibrosis that has no FDA-approved therapy. Although obeticholic acid (OCA) has been sanctioned for PSC, its clinical utility in PSC is constrained by its potential hepatotoxicity. Here, we introduce a novel therapeutic construct consisting of OCA encapsulated within a reactive oxygen species (ROS)-responsive, biodegradable polymer, further cloaked with human placenta-derived mesenchymal stem cell (hP-MSC) membrane (MPPFTU@OCA). Using PSC patient-derived organoid models, we assessed its cellular uptake and cytotoxicity. Moreover, using a PSC mouse model induced by 3,5-diethoxycarbonyl-1,4-dihydro-collidine (DDC), we demonstrated that intravenous administration of MPPFTU@OCA not only improved cholestasis via the FXR-SHP pathway but also reduced macrophage infiltration and the accumulation of intracellular ROS, and alleviated mitochondria-induced apoptosis. Finally, we verified the ability of MPPFTU@OCA to inhibit mitochondrial ROS thereby alleviating apoptosis by measuring the mitochondrial adenosine triphosphate (ATP) concentration, ROS levels, and membrane potential (ΔΨm) using H2O2-stimulated PSC-derived organoids. These results illuminate the synergistic benefits of integrating an ROS-responsive biomimetic platform with OCA, offering a promising therapeutic avenue for PSC.
Collapse
Affiliation(s)
- Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Beiduo Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou City 310058, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China; Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, 79 Qingchun Rd, Hangzhou City 310003, China; National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China; National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Yingduo Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Xudong Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Wenyi Chen
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Jinfeng Yang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China; National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou City 310058, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China; Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, 79 Qingchun Rd, Hangzhou City 310003, China; National Clinical Research Center for Infectious Diseases, Hangzhou, China.
| |
Collapse
|
2
|
Luo R, Le H, Wu Q, Gong C. Nanoplatform-Based In Vivo Gene Delivery Systems for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312153. [PMID: 38441386 DOI: 10.1002/smll.202312153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Indexed: 07/26/2024]
Abstract
Gene therapy uses modern molecular biology methods to repair disease-causing genes. As a burgeoning therapeutic, it has been widely applied for cancer therapy. Since 1989, there have been numerous clinical gene therapy cases worldwide. However, a few are successful. The main challenge of clinical gene therapy is the lack of efficient and safe vectors. Although viral vectors show high transfection efficiency, their application is still limited by immune rejection and packaging capacity. Therefore, the development of non-viral vectors is overwhelming. Nanoplatform-based non-viral vectors become a hotspot in gene therapy. The reasons are mainly as follows. 1) Non-viral vectors can be engineered to be uptaken by specific types of cells or tissues, providing effective targeting capability. 2) Non-viral vectors can protect goods that need to be delivered from degradation. 3) Nanoparticles can transport large-sized cargo such as CRISPR/Cas9 plasmids and nucleoprotein complexes. 4) Nanoparticles are highly biosafe, and they are not mutagenic in themselves compared to viral vectors. 5) Nanoparticles are easy to scale preparation, which is conducive to clinical conversion and application. Here, an overview of the categories of nanoplatform-based non-viral gene vectors, the limitations on their development, and their applications in cancer therapy.
Collapse
Affiliation(s)
- Rui Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Le
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinjie Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Changyang Gong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
3
|
Petcov TE, Straticiuc M, Iancu D, Mirea DA, Trușcă R, Mereuță PE, Savu DI, Mogoșanu GD, Mogoantă L, Popescu RC, Kopatz V, Jinga SI. Unveiling Nanoparticles: Recent Approaches in Studying the Internalization Pattern of Iron Oxide Nanoparticles in Mono- and Multicellular Biological Structures. J Funct Biomater 2024; 15:169. [PMID: 38921542 PMCID: PMC11204647 DOI: 10.3390/jfb15060169] [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: 03/30/2024] [Revised: 05/15/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Nanoparticle (NP)-based solutions for oncotherapy promise an improved efficiency of the anticancer response, as well as higher comfort for the patient. The current advancements in cancer treatment based on nanotechnology exploit the ability of these systems to pass biological barriers to target the tumor cell, as well as tumor cell organelles. In particular, iron oxide NPs are being clinically employed in oncological management due to this ability. When designing an efficient anti-cancer therapy based on NPs, it is important to know and to modulate the phenomena which take place during the interaction of the NPs with the tumor cells, as well as the normal tissues. In this regard, our review is focused on highlighting different approaches to studying the internalization patterns of iron oxide NPs in simple and complex 2D and 3D in vitro cell models, as well as in living tissues, in order to investigate the functionality of an NP-based treatment.
Collapse
Affiliation(s)
- Teodora Eliana Petcov
- Department of Bioengineering and Biotechnology, Faculty of Medical Engineering, National University for Science and Technology Politehnica of Bucharest, 1–7 Gheorghe Polizu Street, 011061 Bucharest, Romania; (T.E.P.); (S.I.J.)
| | - Mihai Straticiuc
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Decebal Iancu
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Dragoș Alexandru Mirea
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Roxana Trușcă
- National Research Center for Micro and Nanomaterials, National University for Science and Technology Politehnica of Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania;
| | - Paul Emil Mereuță
- Department of Applied Nuclear Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.I.); (D.A.M.); (P.E.M.)
| | - Diana Iulia Savu
- Department of Life and Environmental Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania
| | - George Dan Mogoșanu
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareș Street, 200349 Craiova, Romania;
| | - Laurențiu Mogoantă
- Research Center for Microscopic Morphology and Immunology, University of Medicine and Pharmacy of Craiova, 2 Petru Rareș Street, 200349 Craiova, Romania;
| | - Roxana Cristina Popescu
- Department of Bioengineering and Biotechnology, Faculty of Medical Engineering, National University for Science and Technology Politehnica of Bucharest, 1–7 Gheorghe Polizu Street, 011061 Bucharest, Romania; (T.E.P.); (S.I.J.)
- Department of Life and Environmental Physics, National Institute for R&D in Physics and Nuclear Engineering “Horia Hulubei”, 30 Reactorului Street, 077125 Magurele, Romania
| | - Verena Kopatz
- Department of Radiation Oncology, Medical University of Vienna, 18–20 Waehringer Guertel Street, 1090 Vienna, Austria;
| | - Sorin Ion Jinga
- Department of Bioengineering and Biotechnology, Faculty of Medical Engineering, National University for Science and Technology Politehnica of Bucharest, 1–7 Gheorghe Polizu Street, 011061 Bucharest, Romania; (T.E.P.); (S.I.J.)
| |
Collapse
|
4
|
Inam H, Sprio S, Tavoni M, Abbas Z, Pupilli F, Tampieri A. Magnetic Hydroxyapatite Nanoparticles in Regenerative Medicine and Nanomedicine. Int J Mol Sci 2024; 25:2809. [PMID: 38474056 DOI: 10.3390/ijms25052809] [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: 01/29/2024] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
This review focuses on the latest advancements in magnetic hydroxyapatite (mHA) nanoparticles and their potential applications in nanomedicine and regenerative medicine. mHA nanoparticles have gained significant interest over the last few years for their great potential, offering advanced multi-therapeutic strategies because of their biocompatibility, bioactivity, and unique physicochemical features, enabling on-demand activation and control. The most relevant synthetic methods to obtain magnetic apatite-based materials, either in the form of iron-doped HA nanoparticles showing intrinsic magnetic properties or composite/hybrid compounds between HA and superparamagnetic metal oxide nanoparticles, are described as highlighting structure-property correlations. Following this, this review discusses the application of various magnetic hydroxyapatite nanomaterials in bone regeneration and nanomedicine. Finally, novel perspectives are investigated with respect to the ability of mHA nanoparticles to improve nanocarriers with homogeneous structures to promote multifunctional biological applications, such as cell stimulation and instruction, antimicrobial activity, and drug release with on-demand triggering.
Collapse
Affiliation(s)
- Hina Inam
- Institute of Science, Technology and Sustainability for Ceramics (ISSMC), National Research Council of Italy (CNR), 48018 Faenza, Italy
- Department of Material Science and Technology, University of Parma, 43121 Parma, Italy
| | - Simone Sprio
- Institute of Science, Technology and Sustainability for Ceramics (ISSMC), National Research Council of Italy (CNR), 48018 Faenza, Italy
| | - Marta Tavoni
- Institute of Science, Technology and Sustainability for Ceramics (ISSMC), National Research Council of Italy (CNR), 48018 Faenza, Italy
- Department of Material Science and Technology, University of Parma, 43121 Parma, Italy
| | - Zahid Abbas
- Institute of Science, Technology and Sustainability for Ceramics (ISSMC), National Research Council of Italy (CNR), 48018 Faenza, Italy
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy
| | - Federico Pupilli
- Institute of Science, Technology and Sustainability for Ceramics (ISSMC), National Research Council of Italy (CNR), 48018 Faenza, Italy
- Department of Chemical Sciences, University of Padova, 35122 Padova, Italy
| | - Anna Tampieri
- Institute of Science, Technology and Sustainability for Ceramics (ISSMC), National Research Council of Italy (CNR), 48018 Faenza, Italy
| |
Collapse
|
5
|
Sembada AA, Lenggoro IW. Transport of Nanoparticles into Plants and Their Detection Methods. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:131. [PMID: 38251096 PMCID: PMC10819755 DOI: 10.3390/nano14020131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024]
Abstract
Nanoparticle transport into plants is an evolving field of research with diverse applications in agriculture and biotechnology. This article provides an overview of the challenges and prospects associated with the transport of nanoparticles in plants, focusing on delivery methods and the detection of nanoparticles within plant tissues. Passive and assisted delivery methods, including the use of roots and leaves as introduction sites, are discussed, along with their respective advantages and limitations. The barriers encountered in nanoparticle delivery to plants are highlighted, emphasizing the need for innovative approaches (e.g., the stem as a new recognition site) to optimize transport efficiency. In recent years, research efforts have intensified, leading to an evendeeper understanding of the intricate mechanisms governing the interaction of nanomaterials with plant tissues and cells. Investigations into the uptake pathways and translocation mechanisms within plants have revealed nuanced responses to different types of nanoparticles. Additionally, this article delves into the importance of detection methods for studying nanoparticle localization and quantification within plant tissues. Various techniques are presented as valuable tools for comprehensively understanding nanoparticle-plant interactions. The reliance on multiple detection methods for data validation is emphasized to enhance the reliability of the research findings. The future outlooks of this field are explored, including the potential use of alternative introduction sites, such as stems, and the continued development of nanoparticle formulations that improve adhesion and penetration. By addressing these challenges and fostering multidisciplinary research, the field of nanoparticle transport in plants is poised to make significant contributions to sustainable agriculture and environmental management.
Collapse
Affiliation(s)
- Anca Awal Sembada
- Department of Applied Physics and Chemical Engineering, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan;
- School of Life Sciences and Technology, Bandung Institute of Technology, Bandung 40132, Indonesia
| | - I. Wuled Lenggoro
- Department of Applied Physics and Chemical Engineering, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan;
| |
Collapse
|
6
|
Wang J, Wang P, Shao Y, He D. Advancing Treatment Strategies: A Comprehensive Review of Drug Delivery Innovations for Chronic Inflammatory Respiratory Diseases. Pharmaceutics 2023; 15:2151. [PMID: 37631365 PMCID: PMC10458134 DOI: 10.3390/pharmaceutics15082151] [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: 07/20/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Chronic inflammatory respiratory diseases, such as asthma, chronic obstructive pulmonary disease (COPD), and cystic fibrosis, present ongoing challenges in terms of effective treatment and management. These diseases are characterized by persistent inflammation in the airways, leading to structural changes and compromised lung function. There are several treatments available for them, such as bronchodilators, immunomodulators, and oxygen therapy. However, there are still some shortcomings in the effectiveness and side effects of drugs. To achieve optimal therapeutic outcomes while minimizing systemic side effects, targeted therapies and precise drug delivery systems are crucial to the management of these diseases. This comprehensive review focuses on the role of drug delivery systems in chronic inflammatory respiratory diseases, particularly nanoparticle-based drug delivery systems, inhaled corticosteroids (ICSs), novel biologicals, gene therapy, and personalized medicine. By examining the latest advancements and strategies in these areas, we aim to provide a thorough understanding of the current landscape and future prospects for improving treatment outcomes in these challenging conditions.
Collapse
Affiliation(s)
- Junming Wang
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; (J.W.); (P.W.); (Y.S.)
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Pengfei Wang
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; (J.W.); (P.W.); (Y.S.)
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Yiru Shao
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; (J.W.); (P.W.); (Y.S.)
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Daikun He
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; (J.W.); (P.W.); (Y.S.)
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai 201508, China
- Department of General Practice, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
7
|
Niu H, Zhao P, Sun W. Biomaterials for chimeric antigen receptor T cell engineering. Acta Biomater 2023; 166:1-13. [PMID: 37137403 DOI: 10.1016/j.actbio.2023.04.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/23/2023] [Accepted: 04/27/2023] [Indexed: 05/05/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cells have achieved breakthrough efficacies against hematological malignancies, but their unsatisfactory efficacies in solid tumors limit their applications. The prohibitively high prices further restrict their access to broader populations. Novel strategies are urgently needed to address these challenges, and engineering biomaterials can be one promising approach. The established process for manufacturing CAR-T cells involves multiple steps, and biomaterials can help simplify or improve several of them. In this review, we cover recent progress in engineering biomaterials for producing or stimulating CAR-T cells. We focus on the engineering of non-viral gene delivery nanoparticles for transducing CAR into T cells ex vivo/in vitro or in vivo. We also dive into the engineering of nano-/microparticles or implantable scaffolds for local delivery or stimulation of CAR-T cells. These biomaterial-based strategies can potentially change the way CAR-T cells are manufactured, significantly reducing their cost. Modulating the tumor microenvironment with the biomaterials can also considerably enhance the efficacy of CAR-T cells in solid tumors. We pay special attention to progress made in the past five years, and perspectives on future challenges and opportunities are also discussed. STATEMENT OF SIGNIFICANCE: Chimeric antigen receptor T (CAR-T) cell therapies have revolutionized the field of cancer immunotherapy with genetically engineered tumor recognition. They are also promising for treating many other diseases. However, the widespread application of CAR-T cell therapy has been hampered by the high manufacturing cost. Poor penetration of CAR-T cells into solid tissues further restricted their use. While biological strategies have been explored to improve CAR-T cell therapies, such as identifying new cancer targets or integrating smart CARs, biomaterial engineering provides alternative strategies toward better CAR-T cells. In this review, we summarize recent advances in engineering biomaterials for CAR-T cell improvement. Biomaterials ranging from nano-, micro-, and macro-scales have been developed to assist CAR-T cell manufacturing and formulation.
Collapse
Affiliation(s)
- Huanqing Niu
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA; State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, Jiangsu 211816, China
| | - Penghui Zhao
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Wujin Sun
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA; Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA; Center for Emerging, Zoonotic, and Arthropod-Born Pathogens, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
8
|
Zheng J, Jiang X, Li Y, Gao J. Inorganic nanoparticle-integrated mesenchymal stem cells: A potential biological agent for multifaceted applications. MedComm (Beijing) 2023; 4:e313. [PMID: 37533768 PMCID: PMC10390757 DOI: 10.1002/mco2.313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 08/04/2023] Open
Abstract
Mesenchymal stem cell (MSC)-based therapies are flourishing. MSCs could be used as potential therapeutic agents for regenerative medicine due to their own repair function. Meanwhile, the natural predisposition toward inflammation or injury sites makes them promising carriers for targeted drug delivery. Inorganic nanoparticles (INPs) are greatly favored for their unique properties and potential applications in biomedical fields. Current research has integrated INPs with MSCs to enhance their regenerative or antitumor functions. This model also allows the in vivo fate tracking of MSCs in multiple imaging modalities, as many INPs are also excellent contrast agents. Thus, INP-integrated MSCs would be a multifunctional biologic agent with great potential. In this review, the current roles performed by the integration of INPs with MSCs, including (i) enhancing their repair and regeneration capacity via the improvement of migration, survival, paracrine, or differentiation properties, (ii) empowering tumor-killing ability through agent loaded or hyperthermia, and (iii) conferring traceability are summarized. An introduction of INP-integrated MSCs for simultaneous treatment and tracking is also included. The promising applications of INP-integrated MSCs in future treatments are emphasized and the challenges to their clinical translation are discussed.
Collapse
Affiliation(s)
- Juan‐Juan Zheng
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Xin‐Chi Jiang
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Yao‐Sheng Li
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Jian‐Qing Gao
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Hangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
9
|
Cai G, Yang Z, Chen YC, Huang Y, Liang L, Feng S, Zhao J. Magnetic Bead Manipulation in Microfluidic Chips for Biological Application. CYBORG AND BIONIC SYSTEMS 2023; 4:0023. [PMID: 37287460 PMCID: PMC10243203 DOI: 10.34133/cbsystems.0023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 03/20/2023] [Indexed: 10/21/2023] Open
Abstract
Magnetic beads manipulation in microfluidic chips is a promising research field for biological application, especially in the detection of biological targets. In this review, we intend to present a thorough and in-depth overview of recent magnetic beads manipulation in microfluidic chips and its biological application. First, we introduce the mechanism of magnetic manipulation in microfluidic chip, including force analysis, particle properties, and surface modification. Then, we compare some existing methods of magnetic manipulation in microfluidic chip and list their biological application. Besides, the suggestions and outlook for future developments in the magnetic manipulation system are also discussed and summarized.
Collapse
Affiliation(s)
- Gaozhe Cai
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology,
Chinese Academy of Sciences, Shanghai 200050, China
| | - Zixin Yang
- School of Communication and Information Engineering,
Shanghai University, Shanghai 200444, China
| | - Yu-Cheng Chen
- School of Electrical and Electronics Engineering,
Nanyang Technological University, 50 Nanyang Ave., Singapore 639798, Singapore
| | - Yaru Huang
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology,
Chinese Academy of Sciences, Shanghai 200050, China
- School of Life Sciences,
Shanghai Normal University, Shanghai, 200235, China
| | - Lijuan Liang
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology,
Chinese Academy of Sciences, Shanghai 200050, China
| | - Shilun Feng
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology,
Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering,
University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianlong Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology,
Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering,
University of Chinese Academy of Sciences, Beijing 100049, China
- Xiangfu Laboratory, Jiaxing, Zhejiang 314102, China
| |
Collapse
|
10
|
Bioinspired Pd-Cu Alloy Nanoparticles as Accept Agent for Dye Degradation Performances. Int J Mol Sci 2022; 23:ijms232214072. [PMID: 36430550 PMCID: PMC9698934 DOI: 10.3390/ijms232214072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/24/2022] [Accepted: 11/04/2022] [Indexed: 11/17/2022] Open
Abstract
Dye degradation is a key reaction in organic decomposition production through electron donor transferring. Palladium (Pd) is the best-known element for synthesis Pd-based catalyst, the surface status determines the scope of relative applications. Here we first prepare Pd-Cu alloy nanoparticles (NPs) by co-reduction of Cu(acac)2 (acac = acetylacetonate) and Pd(C5HF6O2)2 in the presence of sodium borohydride (NaBH4) and glutathione (GSH). The obtained Pd-Cu is about ~10 nm with super-hydrophilicity in aqueous mediums. The structural analysis clearly demonstrated the uniform distribution of Pd and Cu element. The colloidal solution keeps stability even during 30 days. Bimetallic Pd-Cu NPs shows biocompatibility in form of cell lines (IMEF, HACAT, and 239 T) exposed to colloidal solution (50 µg mL-1) for 2 days. It shows the catalytic multi-performance for dye degradation such as methyl orange (MO), rhodamine B (RhB), and methylene blue (MB), respectively. The as-synthesized nanoparticles showed one of the best multiple catalytic activities in the industrially important (electro)-catalytic reduction of 4-nitrophenol (4-NP) to corresponding amines with noticeable reduced reaction time and increased rate constant without the use of any large area support. In addition, it exhibits peroxidase-like activity in the 3, 3', 5, 5'-Tetramethylbenzidine (TMB) color test and exhibit obvious difference with previous individual metal materials. By treated with high intensity focused ultrasound filed (HIFU), Pd-Cu NPs might be recrystallized and decreased the diameters than before. The enhancement in catalytic performance is observed obviously. This work expedites rational design and synthesis of the high-hierarchy alloy catalyst for biological and environment-friendly agents.
Collapse
|
11
|
Zhang X, Kong F, Wang T, Huang X, Li W, Zhang M, Wen T, Liu J, Zhang Y, Meng J, Xu H. Iron oxide nanoparticles cause surface coating- and core chemistry-dependent endothelial cell ferroptosis. Nanotoxicology 2022; 16:829-843. [PMID: 36660964 DOI: 10.1080/17435390.2022.2154176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Iron oxide nanoparticles (IONPs) are mostly intended to be administrated intravenously, understanding the interaction of IONPs with vascular endothelial cells is extremely crucial for developing safe application regimes of IONPs. In this work, interactions of three kinds of IONPs to endothelial cells were investigated both in human umbilical vein endothelial cells (HUVECs) and in healthy mice. Both meso-2,3-dimercaptosuccinic acid (DMSA) coated Fe3O4 NPs (DMSA-Fe3O4 NPs) and DMSA-Fe2O3 NPs induced cell growth inhibition, while polyglucose sorbitol carboxymethyether coated Fe2O3 NPs(PSC-Fe2O3 NPs) did not. The PSC coating inhibited the cellular uptake of the IONPs. Both DMSA-Fe3O4 and DMSA-Fe2O3 NPs induced ferroptosis of HUVEC through upregulating phospholipid peroxides, which could be inhibited by typical ferroptosis inhibitors ferrostatin-1, Trolox and deferoxamine. Moreover, transforming growth factor beta 1 (TGFβ1) was upregulated by DMSA-Fe3O4 NPs at protein and gene level. The inhibitor of TGFβ1 receptor LY210 could reduce the effect. When being intravenously injected in mice, DMSA-Fe3O4 NPs were observed locating in the liver, increased the levels of lipid peroxidation (4-hydroxynonenal), acyl-CoA synthetase long-chain family member 4(ACSL4) and TGFβ1, indicating ferroptosis occurrence in vivo. The ferroptosis of vascular endothelial cells in exposure with IONPs depended on the surface coating and core chemistry of the NPs. Both DMSA-Fe3O4 NPs and DMSA-Fe2O3 NPs could induce the ferroptosis of endothelial cells, while PSC-Fe2O3 NPs did not induce ferroptosis and apoptosis possibly due to the very low cellular uptake. DMSA-Fe3O4 NPs and TGFβ1 formed feedforward loop to induce ferroptosis.
Collapse
Affiliation(s)
- Xue Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Kong
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Tian Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Huang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqing Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meichen Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Wen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Jie Meng
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyan Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Luo M, Lee LKC, Peng B, Choi CHJ, Tong WY, Voelcker NH. Delivering the Promise of Gene Therapy with Nanomedicines in Treating Central Nervous System Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201740. [PMID: 35851766 PMCID: PMC9475540 DOI: 10.1002/advs.202201740] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/19/2022] [Indexed: 06/01/2023]
Abstract
Central Nervous System (CNS) diseases, such as Alzheimer's diseases (AD), Parkinson's Diseases (PD), brain tumors, Huntington's disease (HD), and stroke, still remain difficult to treat by the conventional molecular drugs. In recent years, various gene therapies have come into the spotlight as versatile therapeutics providing the potential to prevent and treat these diseases. Despite the significant progress that has undoubtedly been achieved in terms of the design and modification of genetic modulators with desired potency and minimized unwanted immune responses, the efficient and safe in vivo delivery of gene therapies still poses major translational challenges. Various non-viral nanomedicines have been recently explored to circumvent this limitation. In this review, an overview of gene therapies for CNS diseases is provided and describes recent advances in the development of nanomedicines, including their unique characteristics, chemical modifications, bioconjugations, and the specific applications that those nanomedicines are harnessed to deliver gene therapies.
Collapse
Affiliation(s)
- Meihua Luo
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Australian Institute for Bioengineering and Nanotechnologythe University of QueenslandSt LuciaQLD4072Australia
| | - Leo Kit Cheung Lee
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Bo Peng
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Frontiers Science Center for Flexible ElectronicsXi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical materials & EngineeringNorthwestern Polytechnical UniversityXi'an710072China
| | - Chung Hang Jonathan Choi
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Wing Yin Tong
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO)ClaytonVIC3168Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClaytonVIC3168Australia
- Materials Science and EngineeringMonash University14 Alliance LaneClaytonVIC3800Australia
| |
Collapse
|
13
|
Mao Z, Lin X, Wang P, Yan H. Iron oxide nanoparticles for biomedical applications: an updated patent review (2015-2021). Expert Opin Ther Pat 2022; 32:939-952. [PMID: 35929879 DOI: 10.1080/13543776.2022.2109413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Iron oxide nanoparticles (IONPs) hold the edges of great magnetic properties and fine nanoparticle characteristics, making them an attractive therapeutic agent. In the past seven years, more in-depth investigations were devoted to the intrinsic structure, magnetic properties, and biological effects of IONPs, expanding the range of their therapeutic application scenes. AREAS COVERED This review focuses on the development of IONPs for biomedical applications from the angle of the patent literature reported during the period 2015-2021. EXPERT OPINION While the magnetic properties of IONPs have been extensively explored, the precise control of IONP behavior through external magnetic fields remains a challenge. Further digging into the biological effects of IONPs will facilitate the development of IONP-based immune therapies. Long-term reliable safety evaluations are of necessity and significance to promote the process of clinical translation.
Collapse
Affiliation(s)
- Zeyuan Mao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Xin Lin
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Pengfei Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Hailong Yan
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| |
Collapse
|
14
|
Highly effective rheumatoid arthritis therapy by peptide-promoted nanomodification of mesenchymal stem cells. Biomaterials 2022; 283:121474. [DOI: 10.1016/j.biomaterials.2022.121474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/07/2023]
|
15
|
Liu B, Ji Q, Cheng Y, Liu M, Zhang B, Mei Q, Liu D, Zhou S. Biomimetic GBM-targeted drug delivery system boosting ferroptosis for immunotherapy of orthotopic drug-resistant GBM. J Nanobiotechnology 2022; 20:161. [PMID: 35351131 PMCID: PMC8962245 DOI: 10.1186/s12951-022-01360-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/08/2022] [Indexed: 01/13/2023] Open
Abstract
Background Clinical studies have shown that the efficacy of programmed cell death receptor-1/programmed cell death ligand-1 (PD-1/PD-L1) inhibitors on glioblastoma (GBM) is much lower than what is expected because of the low immunogenicity of GBM. Ferroptosis of cancer cells can induce the maturation of dendritic cells (DC cells) and increase the activity of T cell. The activated T cells release IFN-γ, which subsequently induces the ferroptosis of cancer cells. Thus, the aim of this paper is to set up a new GBM-targeted drug delivery system (Fe3O4-siPD-L1@M-BV2) to boost ferroptosis for immunotherapy of drug-resistant GBM. Results Fe3O4-siPD-L1@M-BV2 significantly increased the accumulation of siPD-L1 and Fe2+ in orthotopic drug-resistant GBM tissue in mice. Fe3O4-siPD-L1@M-BV2 markedly decreased the protein expression of PD-L1 and increased the ratio between effector T cells and regulatory T cells in orthotopic drug-resistant GBM tissue. Moreover, Fe3O4-siPD-L1@M-BV2 induced ferroptosis of GBM cells and maturation of DC cell, and it also increased the ratio between M1-type microglia and M2-type microglia in orthotopic drug-resistant GBM tissue. Finally, the growth of orthotopic drug-resistant GBM in mice was significantly inhibited by Fe3O4-siPD-L1@M-BV2. Conclusion The mutual cascade amplification effect between ferroptosis and immune reactivation induced by Fe3O4-siPD-L1@M-BV2 significantly inhibited the growth of orthotopic drug-resistant GBM and prolonged the survival time of orthotopic drug-resistant GBM mice. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01360-6.
Collapse
Affiliation(s)
- Bao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Qifeng Ji
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Ying Cheng
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Miao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Bangle Zhang
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Qibing Mei
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Daozhou Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China.
| | - Siyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China. .,Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
16
|
Huang T, Zhang T, Gao J. Targeted mitochondrial delivery: A therapeutic new era for disease treatment. J Control Release 2022; 343:89-106. [DOI: 10.1016/j.jconrel.2022.01.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 12/13/2022]
|
17
|
Stimuli-controllable iron oxide nanoparticle assemblies: Design, manipulation and bio-applications. J Control Release 2022; 345:231-274. [DOI: 10.1016/j.jconrel.2022.03.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 02/07/2023]
|
18
|
Yao X, Ma Y, Zhou W, Liao Y, Jiang Z, Lin J, He Q, Wu H, Wei W, Wang X, Björklund M, Ouyang H. In-cytoplasm mitochondrial transplantation for mesenchymal stem cells engineering and tissue regeneration. Bioeng Transl Med 2022; 7:e10250. [PMID: 35111950 PMCID: PMC8780934 DOI: 10.1002/btm2.10250] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 12/15/2022] Open
Abstract
Stem cell therapies are unsatisfactory due to poor cell survival and engraftment. Stem cell used for therapy must be properly "tuned" for a harsh in vivo environment. Herein, we report that transfer of exogenous mitochondria (mito) to adipose-derived mesenchymal stem cells (ADSCs) can effectively boost their energy levels, enabling efficient cell engraftment. Importantly, the entire process of exogeneous mitochondrial endocytosis is captured by high-content live-cell imaging. Mitochondrial transfer leads to acutely enhanced bioenergetics, with nearly 17% of higher adenosine 5'-triphosphate (ATP) levels in ADSCs treated with high mitochondrial dosage and further results in altered secretome profiles of ADSCs. Mitochondrial transfer also induced the expression of 334 mRNAs in ADSCs, which are mainly linked to signaling pathways associated with DNA replication and cell division. We hypothesize that increase in ATP and cyclin-dependent kinase 1 and 2 expression might be responsible for promoting enhanced proliferation, migration, and differentiation of ADSCs in vitro. More importantly, mito-transferred ADSCs display prolonged cell survival, engraftment and horizontal transfer of exogenous mitochondria to surrounding cells in a full-thickness skin defect rat model with improved skin repair compared with nontreated ADSCs. These results demonstrate that intracellular mitochondrial transplantation is a promising strategy to engineer stem cells for tissue regeneration.
Collapse
Affiliation(s)
- Xudong Yao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
- The Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Yuanzhu Ma
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Wenyan Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Youguo Liao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Zongsheng Jiang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Junxin Lin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Qiulin He
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Hongwei Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Wei Wei
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
- The Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Xiaozhao Wang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Mikael Björklund
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
- Department of Sports MedicineZhejiang University School of MedicineHangzhouChina
- China Orthopedic Regenerative Medicine Group (CORMed)HangzhouChina
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of MedicineHangzhouChina
| |
Collapse
|
19
|
Xu D, Su Y, Xu Q, Huang T, Chen Z, Zhang T. Uniform iron oxide nanoparticles reduce the required amount of polyethylenimine in the gene delivery to mesenchymal stem cells. NANOTECHNOLOGY 2021; 33:125101. [PMID: 34874301 DOI: 10.1088/1361-6528/ac4066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/06/2021] [Indexed: 06/13/2023]
Abstract
Cationic polyethylenimine (PEI) is regarded as the 'golden standard' of non-viral gene vectors. However, the superiority of PEI with high positive charge density also induces its major drawback of cytotoxicity, which restricts its application for an effective and safe gene delivery to stem cells. To redress this shortcoming, herein, a magnetic gene complex containing uniform iron oxide nanoparticles (UIONPs), plasmid DNA, and free PEI is prepared through electrostatic interactions for the gene delivery to bone marrow-derived mesenchymal stem cells (BM-MSCs). Results show that UIONPs dramatically promote the gene delivery to BM-MSCs using the assistance of magnetic force. In addition, decreasing the free PEI nitrogen to DNA phosphate (N/P) ratio from 10 to 6 has little adverse impact on the transgene expression levels (over 300 times than that of PEI alone at the N/P ratio of 6) and significantly reduces the cytotoxicity to BM-MSCs. Further investigations confirmed that the decrease of free PEI has little influence on the cellular uptake after applying external magnetic forces, but that the reduced positive charge density decreases the cytotoxicity. The present study demonstrates that magnetic gene delivery not only contributes to the enhanced gene expression but also helps to reduce the required amount of PEI, providing a potential strategy for an efficient and safe gene delivery to stem cells.
Collapse
Affiliation(s)
- Donghang Xu
- Department of Pharmacy, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yuanqin Su
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Qianhao Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Ting Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Zhilan Chen
- Department of Pharmacy, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
20
|
Magnetofection In Vivo by Nanomagnetic Carriers Systemically Administered into the Bloodstream. Pharmaceutics 2021; 13:pharmaceutics13111927. [PMID: 34834342 PMCID: PMC8619128 DOI: 10.3390/pharmaceutics13111927] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/11/2022] Open
Abstract
Nanoparticle-based technologies are rapidly expanding into many areas of biomedicine and molecular science. The unique ability of magnetic nanoparticles to respond to the magnetic field makes them especially attractive for a number of in vivo applications including magnetofection. The magnetofection principle consists of the accumulation and retention of magnetic nanoparticles carrying nucleic acids in the area of magnetic field application. The method is highly promising as a clinically efficient tool for gene delivery in vivo. However, the data on in vivo magnetofection are often only descriptive or poorly studied, insufficiently systematized, and sometimes even contradictory. Therefore, the aim of the review was to systematize and analyze the data that influence the in vivo magnetofection processes after the systemic injection of magnetic nanostructures. The main emphasis is placed on the structure and coating of the nanomagnetic vectors. The present problems and future trends of the method development are also considered.
Collapse
|
21
|
Huang T, Zhang T, Jiang X, Li A, Su Y, Bian Q, Wu H, Lin R, Li N, Cao H, Ling D, Wang J, Tabata Y, Gu Z, Gao J. Iron oxide nanoparticles augment the intercellular mitochondrial transfer-mediated therapy. SCIENCE ADVANCES 2021; 7:eabj0534. [PMID: 34586849 PMCID: PMC8480934 DOI: 10.1126/sciadv.abj0534] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/30/2021] [Indexed: 05/24/2023]
Abstract
The transfer of mitochondria between cells has recently been revealed as a spontaneous way to protect the injured cells. However, the utilization of this natural transfer process for disease treatment is so far limited by its unsatisfactory transfer efficiency and selectivity. Here, we demonstrate that iron oxide nanoparticles (IONPs) can augment the intercellular mitochondrial transfer from human mesenchymal stem cells (hMSCs) selectively to diseased cells, owing to the enhanced formation of connexin 43–containing gap junctional channels triggered by ionized IONPs. In a mouse model of pulmonary fibrosis, the IONP-engineered hMSCs achieve a remarkable mitigation of fibrotic progression because of the promoted intercellular mitochondrial transfer, with no serious safety issues identified. The present study reports a potential method of using IONPs to enable hMSCs for efficient and safe transfer of mitochondria to diseased cells to restore mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Ting Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinchi Jiang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ai Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuanqin Su
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiong Bian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honghui Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ruyi Lin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ni Li
- Department of Cardiothoracic Surgery, Ningbo Medical Center, Lihuili Hospital Affiliated to Ningbo University, Ningbo, Zhejiang 315041, China
| | - Hongcui Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Daishun Ling
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinqiang Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yasuhiko Tabata
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Zhen Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
- Westlake Laboratory of Life Sciences and Biomedicine, Zhejiang, China
| |
Collapse
|
22
|
Lien J, Bull T, Michelmore RW, Guo T. Fast Fluorescence Titration Quantification of Plasmid DNA with DNA Attractive Magnetic Nanoparticles. Anal Chem 2021; 93:12854-12861. [PMID: 34516097 DOI: 10.1021/acs.analchem.0c04892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Fluorescence titration using magnetic nanoparticles (FTMN) was performed as a rapid, inexpensive, and simple method for quantifying the amount of fluorophore-intercalated plasmid DNA on these DNA attractive nanoparticles. Binding of the propidium iodide (PI)-intercalated DNA (PI/DNA) to polyethylenimine (PEI)-coated monodisperse iron oxide magnetic nanoparticles (PEI-MNs) was confirmed with transmission electron microscopy after the two species were mixed in water for less than a minute. The amount of DNA on PEI-MNs in aqueous solution, however, could not be easily determined using direct fluorescence measurements due to strong scattering by aggregated MNs, especially at high nanoparticle concentrations. Instead, fluorescence measurements were taken immediately after the solution of PI/DNA and PEI-MN mixtures was treated with a magnet to pull the PEI-MNs out of the solution. The detected fluorescence signal of the remaining free PI/DNA in the solution decreased as the concentration of PEI-MNs in the pre-treated solutions increased, resulting in a titration curve, which was used to determine the amount of DNA on MNs, the dissociation constant, and binding energy after the concentration of PEI-MNs was calibrated with microwave-plasma atomic emission spectroscopy. Quantitative polymerase chain reaction was used to understand the binding of DNA to MNs and to measure the amount of free PI/DNA in solution, and the results were similar to those obtained with the FTMN method.
Collapse
Affiliation(s)
- Jennifer Lien
- Department of Chemistry, University of California, Davis, California 95616, United States.,Innovative Genomics Institute, 2151 Berkeley Way, Berkeley, California 94704, United States
| | - Tawni Bull
- The Genome Center, Department of Plant Sciences, University of California, Davis, California 95616, United States
| | - Richard W Michelmore
- The Genome Center, Department of Plant Sciences, University of California, Davis, California 95616, United States.,Departments of Molecular and Cellular Biology, Medical Microbiology and Immunology, University of California, Davis, California 95616, United States.,Innovative Genomics Institute, 2151 Berkeley Way, Berkeley, California 94704, United States
| | - Ting Guo
- Department of Chemistry, University of California, Davis, California 95616, United States
| |
Collapse
|
23
|
Kralj S, Marchesan S. Bioinspired Magnetic Nanochains for Medicine. Pharmaceutics 2021; 13:1262. [PMID: 34452223 PMCID: PMC8398308 DOI: 10.3390/pharmaceutics13081262] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been widely used for medicine, both in therapy and diagnosis. Their guided assembly into anisotropic structures, such as nanochains, has recently opened new research avenues; for instance, targeted drug delivery. Interestingly, magnetic nanochains do occur in nature, and they are thought to be involved in the navigation and geographic orientation of a variety of animals and bacteria, although many open questions on their formation and functioning remain. In this review, we will analyze what is known about the natural formation of magnetic nanochains, as well as the synthetic protocols to produce them in the laboratory, to conclude with an overview of medical applications and an outlook on future opportunities in this exciting research field.
Collapse
Affiliation(s)
- Slavko Kralj
- Department for Materials Synthesis, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Silvia Marchesan
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy;
| |
Collapse
|
24
|
Takayama Y, Kusamori K, Nishikawa M. Mesenchymal stem/stromal cells as next-generation drug delivery vehicles for cancer therapeutics. Expert Opin Drug Deliv 2021; 18:1627-1642. [PMID: 34311638 DOI: 10.1080/17425247.2021.1960309] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Drug delivery to solid tumors remains a significant therapeutic challenge. Mesenchymal stem/stromal cells (MSCs) home to tumor tissues and can be employed as tumor targeted drug/gene delivery vehicles. Reportedly, therapeutic gene- or anti-cancer drug-loaded MSCs have shown remarkable anti-tumor effects in preclinical studies, and some clinical trials for assessing therapeutic MSCs in patients with cancer have been registered. AREAS COVERED In the present review, we first discuss the source and interdonor heterogeneity of MSCs, their tumor-homing mechanism, and the route of MSC administration in MSC-based cancer therapy. We then summarize the therapeutic applications of MSCs as a drug delivery vehicle for therapeutic genes or anti-cancer drugs and the drug delivery mechanism from drug-loaded MSCs to cancer cells. EXPERT OPINION Although numerous preclinical studies have revealed significant anti-tumor effects, several clinical trials assessing MSC-based cancer gene therapy have failed to demonstrate corroborative results, documenting limited therapeutic effects. Notably, a successful clinical outcome with MSC-based cancer therapy would require the interdonor heterogeneity of administered MSCs to be resolved, along with improved tumor-homing efficiency and optimized drug delivery efficiency from MSCs to cancer cells.
Collapse
Affiliation(s)
- Yukiya Takayama
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba Japan
| | - Kosuke Kusamori
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba Japan
| | - Makiya Nishikawa
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba Japan
| |
Collapse
|
25
|
Development and Characterization of Cellulose/Iron Acetate Nanofibers for Bone Tissue Engineering Applications. Polymers (Basel) 2021; 13:polym13081339. [PMID: 33923866 PMCID: PMC8072972 DOI: 10.3390/polym13081339] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/11/2021] [Accepted: 04/14/2021] [Indexed: 12/31/2022] Open
Abstract
In tissue engineering, design of biomaterial with a micro/nano structure is an essential step to mimic extracellular matrix (ECM) and to enhance biomineralization as well as cell biocompatibility. Composite polymeric nanofiber with iron particles/ions has an important role in biomineralization and collagen synthesis for bone tissue engineering. Herein, we report development of polymeric cellulose acetate (CA) nanofibers (17 wt.%) and traces of iron acetates salt (0.5 wt.%) within a polymeric solution to form electrospinning nanofibers mats with iron nanoparticles for bone tissue engineering applications. The resulting mats were characterized using field emission scanning electron microscopy (FESEM), transmission electron microscope (TEM), Fourier transform infrared (FTIR), thermal gravimetric analysis (TGA), differential scanning calorimetry (DSC), X-ray diffraction (XRD), and X-ray photoelectron spectroscopy (XPS). The resulted morphology indicated that the average diameter of CA decreased after addition of iron from (395 ± 30) to (266 ± 19) nm and had dense fiber distributions that match those of native ECM. Moreover, addition of iron acetate to CA solution resulted in mats that are thermally stable. The initial decomposition temperature was 300 °C of CA/Fe mat > 270 °C of pure CA. Furthermore, a superior apatite formation resulted in a biomineralization test after 3 days of immersion in stimulated environmental condition. In vitro cell culture experiments demonstrated that the CA/Fe mat was biocompatible to human fetal-osteoblast cells (hFOB) with the ability to support the cell attachment and proliferation. These findings suggest that doping traces of iron acetate has a promising role in composite mats designed for bone tissue engineering as simple and economically nanoscale materials. Furthermore, these biomaterials can be used in a potential future application such as drug delivery, cancer treatment, and antibacterial materials.
Collapse
|
26
|
Zou M, Du Y, Liu R, Zheng Z, Xu J. Nanocarrier-delivered small interfering RNA for chemoresistant ovarian cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 12:e1648. [PMID: 33682310 DOI: 10.1002/wrna.1648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the fifth leading cause of cancer-related death in women in the United States. Because success in early screening is limited, and most patients with advanced disease develop resistance to multiple treatment modalities, the overall prognosis of ovarian cancer is poor. Despite the revolutionary role of surgery and chemotherapy in curing ovarian cancer, recurrence remains a major challenge in treatment. Thus, improving our understanding of the pathogenesis of ovarian cancer is essential for developing more effective treatments. In this review, we analyze the underlying molecular mechanisms leading to chemotherapy resistance. We discuss the clinical benefits and potential challenges of using nanocarrier-delivered small interfering RNA to treat chemotherapy-resistant ovarian cancer. We aim to elicit collaborative studies on nanocarrier-delivered small interfering RNA to improve the long-term survival rate and quality of life of patients with ovarian cancer. This article is categorized under: RNA Methods > RNA Nanotechnology Regulatory RNAs/RNAi/Riboswitches > RNAi: Mechanisms of Action.
Collapse
Affiliation(s)
- Mingyuan Zou
- Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruizhen Liu
- The First People's Hospital of Wu'an, Wu'an, Hebei, China
| | - Zeliang Zheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jian Xu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
27
|
Capra M, Lodesani A, Brambilla A, Finazzi M, Duò L, Ciccacci F, Picone A. Reversible metamorphosis from Fe 3O 4 to FeO of epitaxial iron oxide films grown on the Fe-p(1 × 1)O surface. RSC Adv 2021; 11:11513-11518. [PMID: 35423610 PMCID: PMC8698807 DOI: 10.1039/d0ra10650j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/12/2021] [Indexed: 01/08/2023] Open
Abstract
The reduction and oxidation of epitaxial Fe3O4 films grown by reactive deposition on a Fe-p(1 × 1)O surface have been investigated by means of Auger electron spectroscopy (AES), low energy electron diffraction (LEED) and scanning tunneling microcopy (STM). The as-grown iron oxide samples display a square LEED pattern with a lattice constant compatible with a p(1 × 1) bulk terminated Fe3O4(001) surface. STM topographic images of Fe3O4 are characterized by atomically flat terraces separated by highly oriented steps running along the (010) and (100) crystallographic directions of the substrate. Upon annealing at 800 K in an ultra-high vacuum, AES reveals that magnetite transforms to FeO. The sample exposes the (001) surface of the rock salt structure, with a lattice parameter close to that of bulk wüstite. The Fe3O4 phase can be recovered by oxidation at 10−6 mbar of molecular oxygen. The reduction and oxidation of epitaxial Fe3O4 films grown by reactive deposition on a Fe-p(1 × 1)O surface have been investigated by means of surface science techniques.![]()
Collapse
Affiliation(s)
- M. Capra
- Department of Physics
- Politecnico di Milano
- I-20133 Milano
- Italy
| | - A. Lodesani
- Department of Physics
- Politecnico di Milano
- I-20133 Milano
- Italy
| | - A. Brambilla
- Department of Physics
- Politecnico di Milano
- I-20133 Milano
- Italy
| | - M. Finazzi
- Department of Physics
- Politecnico di Milano
- I-20133 Milano
- Italy
| | - L. Duò
- Department of Physics
- Politecnico di Milano
- I-20133 Milano
- Italy
| | - F. Ciccacci
- Department of Physics
- Politecnico di Milano
- I-20133 Milano
- Italy
| | - A. Picone
- Department of Physics
- Politecnico di Milano
- I-20133 Milano
- Italy
| |
Collapse
|
28
|
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: 2.0] [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
|