1
|
Christoffers S, Seiler L, Wiebe E, Blume C. Possibilities and efficiency of MSC co-transfection for gene therapy. Stem Cell Res Ther 2024; 15:150. [PMID: 38783353 PMCID: PMC11119386 DOI: 10.1186/s13287-024-03757-6] [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: 02/12/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are not only capable of self-renewal, trans-differentiation, homing to damaged tissue sites and immunomodulation by secretion of trophic factors but are also easy to isolate and expand. Because of these characteristics, they are used in numerous clinical trials for cell therapy including immune and neurological disorders, diabetes, bone and cartilage diseases and myocardial infarction. However, not all trials have successful outcomes, due to unfavourable microenvironmental factors and the heterogenous nature of MSCs. Therefore, genetic manipulation of MSCs can increase their prospect. Currently, most studies focus on single transfection with one gene. Even though the introduction of more than one gene increases the complexity, it also increases the effectivity as different mechanism are triggered, leading to a synergistic effect. In this review we focus on the methodology and efficiency of co-transfection, as well as the opportunities and pitfalls of these genetically engineered cells for therapy.
Collapse
Affiliation(s)
- Sina Christoffers
- Institute for Technical Chemistry, Leibniz University Hannover, Callinstr. 3-5, 30167, Hannover, Germany.
- Cluster of Excellence Hearing4all, Hannover, Germany.
| | - Lisa Seiler
- Institute for Technical Chemistry, Leibniz University Hannover, Callinstr. 3-5, 30167, Hannover, Germany
| | - Elena Wiebe
- Institute for Technical Chemistry, Leibniz University Hannover, Callinstr. 3-5, 30167, Hannover, Germany
- Cluster of Excellence Hearing4all, Hannover, Germany
| | - Cornelia Blume
- Institute for Technical Chemistry, Leibniz University Hannover, Callinstr. 3-5, 30167, Hannover, Germany
- Cluster of Excellence Hearing4all, Hannover, Germany
| |
Collapse
|
2
|
Shams F, Pourjabbar B, Hashemi N, Farahmandian N, Golchin A, Nuoroozi G, Rahimpour A. Current progress in engineered and nano-engineered mesenchymal stem cells for cancer: From mechanisms to therapy. Biomed Pharmacother 2023; 167:115505. [PMID: 37716113 DOI: 10.1016/j.biopha.2023.115505] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023] Open
Abstract
Mesenchymal stem cells (MSCs), as self-renewing multipotent stromal cells, have been considered promising agents for cancer treatment. A large number of studies have demonstrated the valuable properties of MSC-based treatment, such as low immunogenicity and intrinsic tumor-trophic migratory properties. To enhance the potency of MSCs for therapeutic purposes, equipping MSCs with targeted delivery functions using genetic engineering is highly beneficial. Genetically engineered MSCs can express tumor suppressor agents such as pro-apoptotic, anti-proliferative, anti-angiogenic factors and act as ideal delivery vehicles. MSCs can also be loaded with nanoparticle drugs for increased efficacy and externally moderated targeting. Moreover, exosomes secreted by MSCs have important physiological properties, so they can contribute to intercellular communication and transfer cargo into targeted tumor cells. The precise role of genetically modified MSCs in tumor environments is still up for debate, but the beginning of clinical trials has been confirmed by promising results from preclinical investigations of MSC-based gene therapy for a wide range of malignancies. This review highlights the advanced techniques of engineering/nano-engineering and MSC-derived exosomes in tumor-targeted therapy.
Collapse
Affiliation(s)
- Forough Shams
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, 1968917313 Tehran, Iran
| | - Bahareh Pourjabbar
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nader Hashemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, 1968917313 Tehran, Iran
| | - Navid Farahmandian
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Golchin
- Cellular & Molecular Research Center, Cellular & Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia 57157993313, Iran; Department of Clinical Biochemistry & Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia 57157993313, Islamic Republic of Iran
| | - Ghader Nuoroozi
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Rahimpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Zhu X, Ma D, Yang B, An Q, Zhao J, Gao X, Zhang L. Research progress of engineered mesenchymal stem cells and their derived exosomes and their application in autoimmune/inflammatory diseases. Stem Cell Res Ther 2023; 14:71. [PMID: 37038221 PMCID: PMC10088151 DOI: 10.1186/s13287-023-03295-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 03/22/2023] [Indexed: 04/12/2023] Open
Abstract
Autoimmune/inflammatory diseases affect many people and are an important cause of global incidence and mortality. Mesenchymal stem cells (MSCs) have low immunogenicity, immune regulation, multidifferentiation and other biological characteristics, play an important role in tissue repair and immune regulation and are widely used in the research and treatment of autoimmune/inflammatory diseases. In addition, MSCs can secrete extracellular vesicles with lipid bilayer structures under resting or activated conditions, including exosomes, microparticles and apoptotic bodies. Among them, exosomes, as the most important component of extracellular vesicles, can function as parent MSCs. Although MSCs and their exosomes have the characteristics of immune regulation and homing, engineering these cells or vesicles through various technical means, such as genetic engineering, surface modification and tissue engineering, can further improve their homing and other congenital characteristics, make them specifically target specific tissues or organs, and improve their therapeutic effect. This article reviews the advanced technology of engineering MSCs or MSC-derived exosomes and its application in some autoimmune/inflammatory diseases by searching the literature published in recent years at home and abroad.
Collapse
Affiliation(s)
- Xueqing Zhu
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Baoqi Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Qi An
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Jingwen Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xinnan Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
4
|
Choi Y, Lee HK, Choi KC. Engineered adult stem cells: a promising tool for anti-cancer therapy. BMB Rep 2023; 56:71-77. [PMID: 36330711 PMCID: PMC9978368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Indexed: 02/24/2023] Open
Abstract
Cancers are one of the most dreaded diseases in human history and have been targeted by numerous trials including surgery, chemotherapy, radiation therapy, and anti-cancer drugs. Adult stem cells (ASCs), which can regenerate tissues and repair damage, have emerged as leading therapeutic candidates due to their homing ability toward tumor foci. Stem cells can precisely target malicious tumors, thereby minimizing the toxicity of normal cells and unfavorable side effects. ASCs, such as mesenchymal stem cells (MSCs), neural stem cells (NSCs), and hematopoietic stem cells (HSCs), are powerful tools for delivering therapeutic agents to various primary and metastatic cancers. Engineered ASCs act as a bridge between the tumor sites and tumoricidal reagents, producing therapeutic substances such as exosomes, viruses, and anti-cancer proteins encoded by several suicide genes. This review focuses on various anti-cancer therapies implemented via ASCs and summarizes the recent treatment progress and shortcomings. [BMB Reports 2023; 56(2): 71-77].
Collapse
Affiliation(s)
- Youngdong Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Hong Kyu Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea,Corresponding author. Tel: +82-43-261-3664; Fax: +82-43-267-3150; E-mail:
| |
Collapse
|
5
|
Choi Y, Lee HK, Choi KC. Engineered adult stem cells: a promising tool for anti-cancer therapy. BMB Rep 2023; 56:71-77. [PMID: 36330711 PMCID: PMC9978368 DOI: 10.5483/bmbrep.2022-0091] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/28/2022] [Accepted: 11/04/2022] [Indexed: 08/03/2023] Open
Abstract
Cancers are one of the most dreaded diseases in human history and have been targeted by numerous trials including surgery, chemotherapy, radiation therapy, and anti-cancer drugs. Adult stem cells (ASCs), which can regenerate tissues and repair damage, have emerged as leading therapeutic candidates due to their homing ability toward tumor foci. Stem cells can precisely target malicious tumors, thereby minimizing the toxicity of normal cells and unfavorable side effects. ASCs, such as mesenchymal stem cells (MSCs), neural stem cells (NSCs), and hematopoietic stem cells (HSCs), are powerful tools for delivering therapeutic agents to various primary and metastatic cancers. Engineered ASCs act as a bridge between the tumor sites and tumoricidal reagents, producing therapeutic substances such as exosomes, viruses, and anti-cancer proteins encoded by several suicide genes. This review focuses on various anti-cancer therapies implemented via ASCs and summarizes the recent treatment progress and shortcomings. [BMB Reports 2023; 56(2): 71-77].
Collapse
Affiliation(s)
- Youngdong Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Hong Kyu Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
6
|
Pishavar E, Oroojalian F, Salmasi Z, Hashemi E, Hashemi M. Recent advances of dendrimer in targeted delivery of drugs and genes to stem cells as cellular vehicles. Biotechnol Prog 2021; 37:e3174. [PMID: 33987965 DOI: 10.1002/btpr.3174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 05/02/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023]
Abstract
Stem cells can be used to repair dysfunctional and injured (or cancerous) tissues by delivering therapeutics. However, in comparison with other cells, it is harder to transfect drugs or genes into stem cells. Dendrimers have been considered as efficient vectors to deliver both genes and drugs to stem cells due to their unique properties including adjustable molecular weight and size, low toxicity, high loading capacity, and having multiple peripheral chemical agents which can be functionalized to improve deliverance efficiency. In this review, we discuss dendrimer-mediated drug and gene delivery to stem cells as cellular vehicles and the role of this strategy in treating a variety of disorders via regenerative medicine approaches.
Collapse
Affiliation(s)
- Elham Pishavar
- Pharmacutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ezzat Hashemi
- Department of Neurology and Neurological Science, Stanford University, Stanford, California, USA
| | - Maryam Hashemi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Non-viral delivery systems of DNA into stem cells: Promising and multifarious actions for regenerative medicine. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101861] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
8
|
Damasceno PKF, de Santana TA, Santos GC, Orge ID, Silva DN, Albuquerque JF, Golinelli G, Grisendi G, Pinelli M, Ribeiro Dos Santos R, Dominici M, Soares MBP. Genetic Engineering as a Strategy to Improve the Therapeutic Efficacy of Mesenchymal Stem/Stromal Cells in Regenerative Medicine. Front Cell Dev Biol 2020; 8:737. [PMID: 32974331 PMCID: PMC7471932 DOI: 10.3389/fcell.2020.00737] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) have been widely studied in the field of regenerative medicine for applications in the treatment of several disease settings. The therapeutic potential of MSCs has been evaluated in studies in vitro and in vivo, especially based on their anti-inflammatory and pro-regenerative action, through the secretion of soluble mediators. In many cases, however, insufficient engraftment and limited beneficial effects of MSCs indicate the need of approaches to enhance their survival, migration and therapeutic potential. Genetic engineering emerges as a means to induce the expression of different proteins and soluble factors with a wide range of applications, such as growth factors, cytokines, chemokines, transcription factors, enzymes and microRNAs. Distinct strategies have been applied to induce genetic modifications with the goal to enhance the potential of MCSs. This review aims to contribute to the update of the different genetically engineered tools employed for MSCs modification, as well as the factors investigated in different fields in which genetically engineered MSCs have been tested.
Collapse
Affiliation(s)
- Patricia Kauanna Fonseca Damasceno
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.,Health Institute of Technology, SENAI CIMATEC, Salvador, Brazil
| | | | | | - Iasmim Diniz Orge
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.,Health Institute of Technology, SENAI CIMATEC, Salvador, Brazil
| | - Daniela Nascimento Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.,Health Institute of Technology, SENAI CIMATEC, Salvador, Brazil
| | | | - Giulia Golinelli
- Division of Oncology, Laboratory of Cellular Therapy, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Grisendi
- Division of Oncology, Laboratory of Cellular Therapy, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Pinelli
- Division of Plastic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Ricardo Ribeiro Dos Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.,Health Institute of Technology, SENAI CIMATEC, Salvador, Brazil.,National Institute of Science and Technology for Regenerative Medicine (INCT-REGENERA), Rio de Janeiro, Brazil
| | - Massimo Dominici
- Division of Oncology, Laboratory of Cellular Therapy, University of Modena and Reggio Emilia, Modena, Italy
| | - Milena Botelho Pereira Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.,Health Institute of Technology, SENAI CIMATEC, Salvador, Brazil.,National Institute of Science and Technology for Regenerative Medicine (INCT-REGENERA), Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Lee SJ, Kim JJ, Kang KY, Paik MJ, Lee G, Yee ST. Enhanced anti-tumor immunotherapy by silica-coated magnetic nanoparticles conjugated with ovalbumin. Int J Nanomedicine 2019; 14:8235-8249. [PMID: 31802864 PMCID: PMC6802490 DOI: 10.2147/ijn.s194352] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 07/27/2019] [Indexed: 12/15/2022] Open
Abstract
Background The effective induction of an antigen-specific T cell immune response through dendritic cell activation is one of the key goals of tumor immunotherapy. Methods In this study, efficient antigen-delivery carriers using silica-coated magnetic nanoparticles were designed and, their antigen-specific T cell immune response through dendritic cell activation investigated. Results The results showed that the silica-coated magnetic nanoparticles with conjugated ovalbumin enhanced the production of cytokines and antigen uptake in bone marrow-derived dendritic cells. Also, this induced an antigen-specific cytotoxic T lymphocyte (CTL) immune response and activated antigen-specific Th1 cell responses, including IL-2 and IFN-γ production and proliferation. We proved that the immune-stimulatory effects of silica-coated magnetic nanoparticles with conjugated ovalbumin were efficient in inhibiting of tumor growth in EG7-OVA (mouse lymphoma-expressing ovalbumin tumor-bearing mice model). Conclusion Therefore, the silica-coated magnetic nanoparticles with conjugated ovalbumin are expected to be useful as efficient anti-cancer immunotherapy agents.
Collapse
Affiliation(s)
- Sung-Ju Lee
- Department of Pharmacy, Sunchon National University, Suncheon 540-742, Republic of Korea
| | - Jong-Jin Kim
- Center for Self-assembly and Complexity, Institute for Basic Science (IBS), Pohang 37673, Republic of Korea
| | - Kyung-Yun Kang
- Suncheon Research Center for Natural Medicines, Suncheon, Republic of Korea
| | - Man-Jeong Paik
- Department of Pharmacy, Sunchon National University, Suncheon 540-742, Republic of Korea
| | - Gwang Lee
- Department of Physiology and Department of Biomedical Science, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sung-Tae Yee
- Department of Pharmacy, Sunchon National University, Suncheon 540-742, Republic of Korea
| |
Collapse
|
10
|
Gizaw M, Faglie A, Pieper M, Poudel S, Chou SF. The Role of Electrospun Fiber Scaffolds in Stem Cell Therapy for Skin Tissue Regeneration. MED ONE 2019; 4:e190002. [PMID: 30972372 PMCID: PMC6453140 DOI: 10.20900/mo.20190002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stem cell therapy has emerged as one of the topics in tissue engineering where undifferentiated and multipotent cells are strategically placed/ injected in tissue structure for cell regeneration. Over the years, stem cells have shown promising results in skin repairs for non-healing and/or chronic wounds. The addition of the stem cells around the wound site promotes signaling pathways for growth factors that regulate tissue reconstruction. However, injecting stem cells around the wound site has its drawbacks, including cell death due to lack of microenvironment cues. This particular issue is resolved when biomaterial scaffolds are involved in the cultivation and mechanical support of the stem cells. In this review, we describe the current models of stem cell therapy by injections and those that are done through cell cultures using electrospun fiber scaffolds. Electrospun fibers are considered as an ideal candidate for cell cultures due to their surface properties. Through the control of fiber morphology and fiber structure, cells are able to proliferate and differentiate into keratinocytes for skin tissue regeneration. Furthermore, we provide another perspective of using electrospun fibers and stem cells in a layer-by-layer structure for skin substitutes (dressing). Finally, electrospun fibers have the potential to incorporate bioactive agents to achieve controlled release properties, which is beneficial to the survival of the delivered stem cells or the recruitment of the cells. Overall, our work illustrates that electrospun fibers are ideal for stem cell cultures while serving as cell carriers for wound dressing materials.
Collapse
Affiliation(s)
- Mulugeta Gizaw
- Department of Mechanical Engineering, College of Engineering, The University of Texas at Tyler, Tyler, TX 75799, USA
| | - Addison Faglie
- Department of Mechanical Engineering, College of Engineering, The University of Texas at Tyler, Tyler, TX 75799, USA
| | - Martha Pieper
- Department of Mechanical Engineering, College of Engineering, The University of Texas at Tyler, Tyler, TX 75799, USA
| | - Sarju Poudel
- Department of Mechanical Engineering, College of Engineering, The University of Texas at Tyler, Tyler, TX 75799, USA
| | - Shih-Feng Chou
- Department of Mechanical Engineering, College of Engineering, The University of Texas at Tyler, Tyler, TX 75799, USA
| |
Collapse
|
11
|
Controlled Non-Viral Gene Delivery in Cartilage and Bone Repair: Current Strategies and Future Directions. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
12
|
Bansal R, Seth B, Tiwari S, Jahan S, Kumari M, Pant AB, Chaturvedi RK, Kumar P, Gupta KC. Hexadecylated linear PEI self-assembled nanostructures as efficient vectors for neuronal gene delivery. Drug Deliv Transl Res 2018; 8:1436-1449. [DOI: 10.1007/s13346-018-0517-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
13
|
Wu P, Chen H, Jin R, Weng T, Ho JK, You C, Zhang L, Wang X, Han C. Non-viral gene delivery systems for tissue repair and regeneration. J Transl Med 2018; 16:29. [PMID: 29448962 PMCID: PMC5815227 DOI: 10.1186/s12967-018-1402-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/07/2018] [Indexed: 12/11/2022] Open
Abstract
Critical tissue defects frequently result from trauma, burns, chronic wounds and/or surgery. The ideal treatment for such tissue loss is autografting, but donor sites are often limited. Tissue engineering (TE) is an inspiring alternative for tissue repair and regeneration (TRR). One of the current state-of-the-art methods for TRR is gene therapy. Non-viral gene delivery systems (nVGDS) have great potential for TE and have several advantages over viral delivery including lower immunogenicity and toxicity, better cell specificity, better modifiability, and higher productivity. However, there is no ideal nVGDS for TRR, hence, there is widespread research to improve their properties. This review introduces the basic principles and key aspects of commonly-used nVGDSs. We focus on recent advances in their applications, current challenges, and future directions.
Collapse
Affiliation(s)
- Pan Wu
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Haojiao Chen
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Ronghua Jin
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Tingting Weng
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Jon Kee Ho
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Chuangang You
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Liping Zhang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China
| | - Xingang Wang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China.
| | - Chunmao Han
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
14
|
Long W, Yi Y, Chen S, Cao Q, Zhao W, Liu Q. Potential New Therapies for Pediatric Diffuse Intrinsic Pontine Glioma. Front Pharmacol 2017; 8:495. [PMID: 28790919 PMCID: PMC5525007 DOI: 10.3389/fphar.2017.00495] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/11/2017] [Indexed: 12/20/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is an extensively invasive malignancy with infiltration into other regions of the brainstem. Although large numbers of specific targeted therapies have been tested, no significant progress has been made in treating these high-grade gliomas. Therefore, the identification of new therapeutic approaches is of great importance for the development of more effective treatments. This article reviews the conventional therapies and new potential therapeutic approaches for DIPG, including epigenetic therapy, immunotherapy, and the combination of stem cells with nanoparticle delivery systems.
Collapse
Affiliation(s)
- Wenyong Long
- Department of Neurosurgery, Xiangya Hospital, Central South UniversityChangsha, China
| | - Yang Yi
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Shen Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Qi Cao
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, HoustonTX, United States
| | - Wei Zhao
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South UniversityChangsha, China
| |
Collapse
|
15
|
Raisin S, Belamie E, Morille M. Non-viral gene activated matrices for mesenchymal stem cells based tissue engineering of bone and cartilage. Biomaterials 2016; 104:223-37. [DOI: 10.1016/j.biomaterials.2016.07.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/14/2016] [Accepted: 07/16/2016] [Indexed: 12/22/2022]
|
16
|
Das J, Choi YJ, Yasuda H, Han JW, Park C, Song H, Bae H, Kim JH. Efficient delivery of C/EBP beta gene into human mesenchymal stem cells via polyethylenimine-coated gold nanoparticles enhances adipogenic differentiation. Sci Rep 2016; 6:33784. [PMID: 27677463 PMCID: PMC5039411 DOI: 10.1038/srep33784] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 09/02/2016] [Indexed: 01/04/2023] Open
Abstract
The controlled differentiation of stem cells via the delivery of specific genes encoding appropriate differentiation factors may provide useful models for regenerative medicine and aid in developing therapies for human patients. However, the majority of non-viral vectors are not efficient enough to manipulate difficult-to-transfect adult human stem cells in vitro. Herein, we report the first use of 25 kDa branched polyethylenimine-entrapped gold nanoparticles (AuPEINPs) and covalently bound polyethylenimine-gold nanoparticles (AuMUAPEINPs) as carriers for efficient gene delivery into human mesenchymal stem cells (hMSCs). We determined a functional application of these nanoparticles by transfecting hMSCs with the C/EBP beta gene, fused to EGFP, to induce adipogenic differentiation. Transfection efficacy with AuPEINPs and AuMUAPEINPs was 52.3% and 40.7%, respectively, which was 2.48 and 1.93 times higher than that by using Lipofectamine 2000. Luciferase assay results also demonstrated improved gene transfection efficiency of AuPEINPs/AuMUAPEINPs over Lipofectamine 2000 and polyethylenimine. Overexpression of exogenous C/EBP beta significantly enhanced adipogenesis in hMSCs as indicated by both of Oil Red O staining and mRNA expression analyses. Nanoparticle/DNA complexes exhibited favorable cytocompatibility in hMSCs. Taken together, AuPEINPs and AuMUAPEINPs potentially represent safe and highly efficient vehicles for gene delivery to control hMSC differentiation and for therapeutic gene delivery applications.
Collapse
Affiliation(s)
- Joydeep Das
- Dept. of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Yun-Jung Choi
- Dept. of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Hideyo Yasuda
- Dept. of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Jae Woong Han
- Dept. of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Chankyu Park
- Dept. of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Hyuk Song
- Dept. of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Hojae Bae
- Dept. of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul 143-701, South Korea
| | - Jin-Hoi Kim
- Dept. of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| |
Collapse
|
17
|
Sessions JW, Skousen CS, Price KD, Hanks BW, Hope S, Alder JK, Jensen BD. CRISPR-Cas9 directed knock-out of a constitutively expressed gene using lance array nanoinjection. SPRINGERPLUS 2016; 5:1521. [PMID: 27652094 PMCID: PMC5017990 DOI: 10.1186/s40064-016-3037-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 08/10/2016] [Indexed: 11/23/2022]
Abstract
Background CRISPR-Cas9 genome editing and labeling has emerged as an important tool in biologic research, particularly in regards to potential transgenic and gene therapy applications. Delivery of CRISPR-Cas9 plasmids to target cells is typically done by non-viral methods (chemical, physical, and/or electrical), which are limited by low transfection efficiencies or with viral vectors, which are limited by safety and restricted volume size. In this work, a non-viral transfection technology, named lance array nanoinjection (LAN), utilizes a microfabricated silicon chip to physically and electrically deliver genetic material to large numbers of target cells. To demonstrate its utility, we used the CRISPR-Cas9 system to edit the genome of isogenic cells. Two variables related to the LAN process were tested which include the magnitude of current used during plasmid attraction to the silicon lance array (1.5, 4.5, or 6.0 mA) and the number of times cells were injected (one or three times). Results Results indicate that most successful genome editing occurred after injecting three times at a current control setting of 4.5 mA, reaching a median level of 93.77 % modification. Furthermore, we found that genome editing using LAN follows a non-linear injection-dose response, meaning samples injected three times had modification rates as high as nearly 12 times analogously treated single injected samples. Conclusions These findings demonstrate the LAN’s ability to deliver genetic material to cells and indicate that successful alteration of the genome is influenced by a serial injection method as well as the electrical current settings.
Collapse
Affiliation(s)
- John W Sessions
- Department of Mechanical Engineering, Brigham Young University, Provo, UT USA
| | - Craig S Skousen
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT USA
| | - Kevin D Price
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT USA
| | - Brad W Hanks
- Department of Mechanical Engineering, Brigham Young University, Provo, UT USA
| | - Sandra Hope
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT USA
| | - Jonathan K Alder
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT USA
| | - Brian D Jensen
- Department of Mechanical Engineering, Brigham Young University, Provo, UT USA
| |
Collapse
|
18
|
Synthesis and characterization of a PAMAM-OH derivative containing an acid-labile β-thiopropionate bond for gene delivery. Int J Pharm 2016; 509:314-327. [PMID: 27260132 DOI: 10.1016/j.ijpharm.2016.05.060] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/24/2016] [Accepted: 05/27/2016] [Indexed: 12/23/2022]
Abstract
The present report describes the synthesis of a hydroxyl terminal PAMAM dendrimer (PAMAM-OH) derivative (PAMSPF). The hydroxyls of PAMAM-OH were attached to S-Methyl-l-cysteine (SMLC) via an acid-labile ester bond, named as β-thiopropionate bond, followed by modification with folic acid (FA) through a polyethylene glycol (PEG) linker. The degrees of attachment of SMLC and FA to the PAMAM-OH backbone were 83.9% and 12.8%, respectively. PAMSPF could condense DNA to form spherical nanoparticles with particle sizes of ∼200nm and remain stable in the presence of heparin and nuclease. The β-thiopropionate bond in PAMSPF was hydrolyzed completely and the DNA release rate was 95.8±3.3% after incubation under mildly acidic conditions at 37°C for 3h. PAMSPF/DNA was less cytotoxic to KB and HepG2 cells and exhibited a higher gene transfection efficiency than native PAMAM/DNA. The uptake assays showed that PAMSPF/DNA entered KB cells within 0.5h through folate receptor-mediated endocytosis and escaped from endosomes within 2h. In addition, PAMSPF/DNA displayed long circulation time along with excellent targeting of tumor sites in vivo. These findings demonstrate that PAMSPF is an excellent carrier for safe and effective gene delivery.
Collapse
|
19
|
Xu J, Li J, Lin S, Wu T, Huang H, Zhang K, Sun Y, Yeung KWK, Li G, Bian L. Nanocarrier‐Mediated Codelivery of Small Molecular Drugs and siRNA to Enhance Chondrogenic Differentiation and Suppress Hypertrophy of Human Mesenchymal Stem Cells. ADVANCED FUNCTIONAL MATERIALS 2016; 26:2463-2472. [DOI: 10.1002/adfm.201504070] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Cartilage loss is a leading cause of disability among adults, and effective therapy remains elusive. Human mesenchymal stem cells (hMSCs), which have demonstrated self‐renewal and multipotential differentiation, are a promising cell source for cartilage repair. However, the hypertrophic differentiation of the chondrogenically induced MSCs and resulting tissue calcification hinders the clinical translation of MSCs for cartilage repair. Here, a multifunctional nanocarrier based on quantum dots (QDs) is developed to enhance chondrogenic differentiation and suppress hypertrophy of hMSCs simultaneously. Briefly, the QDs are modified with β‐cyclodextrin (β‐CD) and RGD peptide. The resulting nanocarrier is capable of carrying hydrophobic small molecules such as kartogenin in the hydrophobic pockets of conjugated β‐CD to induce chondrogenic differentiation of hMSCs. Meanwhile, via electrostatic interaction the conjugated RGD peptides bind the cargo siRNA targeting Runx2, which is a key regulator of hMSC hypertrophy. Furthermore, due to the excellent photostability of QDs, hMSCs labeled with the nanocarrier can be tracked for up to 14 d after implantation in nude mice. Overall, this work demonstrates the potential of our nanocarrier for inducing and maintaining the chondrogenic phenotype and tracking hMSCs in vivo.
Collapse
Affiliation(s)
- Jianbin Xu
- Division of Biomedical Engineering Department of Mechanical and Automation Engineering The Chinese University of Hong Kong Shatin, New Territories 999077 Hong Kong P. R. China
| | - Jinming Li
- Division of Biomedical Engineering Department of Mechanical and Automation Engineering The Chinese University of Hong Kong Shatin, New Territories 999077 Hong Kong P. R. China
| | - Sien Lin
- Department of Orthopaedics and Traumatology Faculty of Medicine The Chinese University of Hong Kong Shatin, New Territories 999077 Hong Kong P. R. China
| | - Tianyi Wu
- Department of Orthopaedics and Traumatology Faculty of Medicine The Chinese University of Hong Kong Shatin, New Territories 999077 Hong Kong P. R. China
| | - Heqin Huang
- Division of Biomedical Engineering Department of Mechanical and Automation Engineering The Chinese University of Hong Kong Shatin, New Territories 999077 Hong Kong P. R. China
| | - Kunyu Zhang
- Division of Biomedical Engineering Department of Mechanical and Automation Engineering The Chinese University of Hong Kong Shatin, New Territories 999077 Hong Kong P. R. China
| | - Yuxin Sun
- Department of Orthopaedics and Traumatology Faculty of Medicine The Chinese University of Hong Kong Shatin, New Territories 999077 Hong Kong P. R. China
| | - Kelvin W. K. Yeung
- Department of Orthopaedics and Traumatology The University of Hong Kong Hong Kong P.R. China 999077
| | - Gang Li
- Department of Orthopaedics and Traumatology Faculty of Medicine The Chinese University of Hong Kong Shatin, New Territories 999077 Hong Kong P. R. China
| | - Liming Bian
- Division of Biomedical Engineering Department of Mechanical and Automation Engineering The Chinese University of Hong Kong Shatin, New Territories 999077 Hong Kong P. R. China
- Shun Hing Institute of Advanced Engineering The Chinese University of Hong Kong Hong Kong P.R.China 999077
- Shenzhen Research Institute The Chinese University of Hong Kong Hong Kong P.R.China 999077
- China Orthopedic Regenerative Medicine Group (CORMed) Hong Kong P.R.China 999077
| |
Collapse
|
20
|
Glucocorticoid Cell Priming Enhances Transfection Outcomes in Adult Human Mesenchymal Stem Cells. Mol Ther 2015; 24:331-341. [PMID: 26478250 DOI: 10.1038/mt.2015.195] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/12/2015] [Indexed: 12/11/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are one of the most widely researched stem cell types with broad applications from basic research to therapeutics, the majority of which require introduction of exogenous DNA. However, safety and scalability issues hinder viral delivery, while poor efficiency hinders nonviral gene delivery, particularly to hMSCs. Here, we present the use of a pharmacologic agent (glucocorticoid) to overcome barriers to hMSC DNA transfer to enhance transfection using three common nonviral vectors. Glucocorticoid priming significantly enhances transfection in hMSCs, demonstrated by a 3-fold increase in efficiency, 4-15-fold increase in transgene expression, and prolonged transgene expression when compared to transfection without glucocorticoids. These effects are dependent on glucocorticoid receptor binding and caused in part by maintenance of normal metabolic function and increased cellular (5-fold) and nuclear (6-10-fold) DNA uptake over hMSCs transfected without glucocorticoids. Results were consistent across five human donors and in cells up to passage five. Glucocorticoid cell priming is a simple and effective technique to significantly enhance nonviral transfection of hMSCs that should enhance their clinical use, accelerate new research, and decrease reliance on early passage cells.
Collapse
|
21
|
Park JS, Suryaprakash S, Lao YH, Leong KW. Engineering mesenchymal stem cells for regenerative medicine and drug delivery. Methods 2015; 84:3-16. [PMID: 25770356 PMCID: PMC4526354 DOI: 10.1016/j.ymeth.2015.03.002] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/19/2015] [Accepted: 03/02/2015] [Indexed: 12/14/2022] Open
Abstract
Researchers have applied mesenchymal stem cells (MSC) to a variety of therapeutic scenarios by harnessing their multipotent, regenerative, and immunosuppressive properties with tropisms toward inflamed, hypoxic, and cancerous sites. Although MSC-based therapies have been shown to be safe and effective to a certain degree, the efficacy remains low in most cases when MSC are applied alone. To enhance their therapeutic efficacy, researchers have equipped MSC with targeted delivery functions using genetic engineering, therapeutic agent incorporation, and cell surface modification. MSC can be genetically modified virally or non-virally to overexpress therapeutic proteins that complement their innate properties. MSC can also be primed with non-peptidic drugs or magnetic nanoparticles for enhanced efficacy and externally regulated targeting, respectively. Furthermore, MSC can be functionalized with targeting moieties to augment their homing toward therapeutic sites using enzymatic modification, chemical conjugation, or non-covalent interactions. These engineering techniques are still works in progress, requiring optimization to improve the therapeutic efficacy and targeting effectiveness while minimizing any loss of MSC function. In this review, we will highlight the advanced techniques of engineering MSC, describe their promise and the challenges of translation into clinical settings, and suggest future perspectives on realizing their full potential for MSC-based therapy.
Collapse
Affiliation(s)
- Ji Sun Park
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States
| | - Smruthi Suryaprakash
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States
| | - Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States.
| |
Collapse
|
22
|
Pang C, Brunelli A, Zhu C, Hristozov D, Liu Y, Semenzin E, Wang W, Tao W, Liang J, Marcomini A, Chen C, Zhao B. Demonstrating approaches to chemically modify the surface of Ag nanoparticles in order to influence their cytotoxicity and biodistribution after single dose acute intravenous administration. Nanotoxicology 2015; 10:129-39. [DOI: 10.3109/17435390.2015.1024295] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
23
|
Li KC, Hu YC. Cartilage tissue engineering: recent advances and perspectives from gene regulation/therapy. Adv Healthc Mater 2015; 4:948-68. [PMID: 25656682 DOI: 10.1002/adhm.201400773] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/10/2015] [Indexed: 12/16/2022]
Abstract
Diseases in articular cartilages affect millions of people. Despite the relatively simple biochemical and cellular composition of articular cartilages, the self-repair ability of cartilage is limited. Successful cartilage tissue engineering requires intricately coordinated interactions between matrerials, cells, biological factors, and phycial/mechanical factors, and still faces a multitude of challenges. This article presents an overview of the cartilage biology, current treatments, recent advances in the materials, biological factors, and cells used in cartilage tissue engineering/regeneration, with strong emphasis on the perspectives of gene regulation (e.g., microRNA) and gene therapy.
Collapse
Affiliation(s)
- Kuei-Chang Li
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| | - Yu-Chen Hu
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| |
Collapse
|
24
|
Muroski ME, Carnevale KJF, Riskowski RA, Strouse GF. Plasmid transfection in mammalian cells spatiotemporally tracked by a gold nanoparticle. ACS NANO 2015; 9:124-33. [PMID: 25494916 DOI: 10.1021/nn5060305] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Recent advances in cell transfection have suggested that delivery of a gene on a gold nanoparticle (AuNP) can enhance transfection efficiency. The mechanism of transfection is poorly understood, particularly when the gene is appended to a AuNP, as expression of the desired exogenous protein is dependent not only on the efficiency of the gene being taken into the cell but also on efficient endosomal escape and cellular processing of the nucleic acid. Design of a multicolor surface energy transfer (McSET) molecular beacon by independently dye labeling a linearized plasmid and short duplex DNA (sdDNA) appended to a AuNP allows spatiotemporal profiling of the transfection events, providing insight into package uptake, disassembly, and final plasmid expression. Delivery of the AuNP construct encapsulated in Lipofectamine2000 is monitored in Chinese hamster ovary cells using live-cell confocal microscopy. The McSET beacon signals the location and timing of the AuNP release and endosomal escape events for the plasmid and the sdDNA discretely, which are correlated with plasmid transcription by fluorescent protein expression within the cell. It is observed that delivery of the construct leads to endosomal release of the plasmid and sdDNA from the AuNP surface at different rates, prior to endosomal escape. Slow cytosolic diffusion of the nucleic acids is believed to be the limiting step for transfection, impacting the time-dependent expression of protein. The overall protein expression yield is enhanced when delivered on a AuNP, possibly due to better endosomal escape or lower degradation prior to endosomal escape.
Collapse
Affiliation(s)
- Megan E Muroski
- Department of Chemistry and Biochemistry, Florida State University , Tallahassee, Florida 32306-4390, United States
| | | | | | | |
Collapse
|
25
|
Muroski ME, Morgan TJ, Levenson CW, Strouse GF. A gold nanoparticle pentapeptide: gene fusion to induce therapeutic gene expression in mesenchymal stem cells. J Am Chem Soc 2014; 136:14763-71. [PMID: 25198921 DOI: 10.1021/ja505190q] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSC) have been identified as having great potential as autologous cell therapeutics to treat traumatic brain injury and spinal injury as well as neuronal and cardiac ischemic events. All future clinical applications of MSC cell therapies must allow the MSC to be harvested, transfected, and induced to express a desired protein or selection of proteins to have medical benefit. For the full potential of MSC cell therapy to be realized, it is desirable to systematically alter the protein expression of therapeutically beneficial biomolecules in harvested MSC cells with high fidelity in a single transfection event. We have developed a delivery platform on the basis of the use of a solid gold nanoparticle that has been surface modified to produce a fusion containing a zwitterionic, pentapeptide designed from Bax inhibiting peptide (Ku70) to enhance cellular uptake and a linearized expression vector to induce enhanced expression of brain-derived neurotrophic factor (BDNF) in rat-derived MSCs. Ku70 is observed to effect >80% transfection following a single treatment of femur bone marrow isolated rat MSCs with efficiencies for the delivery of a 6.6 kbp gene on either a Au nanoparticle (NP) or CdSe/ZnS quantum dot (QD). Gene expression is observed within 4 d by optical measurements, and secretion is observed within 10 d by Western Blot analysis. The combination of being able to selectively engineer the NP, to colocalize biological agents, and to enhance the stability of those agents has provided the strong impetus to utilize this novel class of materials to engineer primary MSCs.
Collapse
Affiliation(s)
- Megan E Muroski
- Department of Chemistry and Biochemistry, 95 Chieftan Way, Florida State University , Tallahassee, Florida 32306-4390, United States
| | | | | | | |
Collapse
|
26
|
Bartman CM, Egelston J, Ren X, Das R, Phiel CJ. A simple and efficient method for transfecting mouse embryonic stem cells using polyethylenimine. Exp Cell Res 2014; 330:178-85. [PMID: 25102378 DOI: 10.1016/j.yexcr.2014.07.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/12/2014] [Accepted: 07/15/2014] [Indexed: 12/24/2022]
Abstract
Mouse embryonic stem cells (ESCs) can be transfected by electroporation, liposomal reagents, and viral transduction methods. The cationic polymer polyethylenimine (PEI) has been shown to transfect a variety of differentiated mammalian cell types, including mouse ESCs, but existing methods require the use of additional equipment that is not readily accessible to most labs. Here we describe conditions that permit for the efficient transfection of mouse ESCs with low cytotoxicity and without the need for specialized equipment. Our goal was to devise a protocol for the PEI-mediated transfection of mouse ESCs that was comparable in ease to commercial transfection reagents. For these studies, we compared PEI transfection efficiency and cytotoxicity to a well-known liposomal transfection reagent, Lipofectamine2000(™) (LF2K), using fluorescence microscopy, flow cytometry, cell viability assays, and Western blotting. We provide evidence that PEI transfection of mouse ESCs compares favorably to LF2K. Our optimized protocol for efficient transfection of mouse ESCs with PEI is detailed in this report.
Collapse
Affiliation(s)
- Colleen M Bartman
- Department of Integrative Biology, University of Colorado Denver, S4111, 1201 Fifth Street, Denver, CO 80204, United States
| | - Jennifer Egelston
- Department of Integrative Biology, University of Colorado Denver, S4111, 1201 Fifth Street, Denver, CO 80204, United States
| | - Xiaojun Ren
- Department of Chemistry, University of Colorado Denver, 1201 Fifth Street, Denver, CO 80204, United States
| | - Raibatak Das
- Department of Integrative Biology, University of Colorado Denver, S4111, 1201 Fifth Street, Denver, CO 80204, United States
| | - Christopher J Phiel
- Department of Integrative Biology, University of Colorado Denver, S4111, 1201 Fifth Street, Denver, CO 80204, United States.
| |
Collapse
|
27
|
Multi-modal transfection agent based on monodisperse magnetic nanoparticles for stem cell gene delivery and tracking. Biomaterials 2014; 35:7239-47. [DOI: 10.1016/j.biomaterials.2014.05.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 05/04/2014] [Indexed: 01/30/2023]
|
28
|
Wang Y, Cui H, Li K, Sun C, Du W, Cui J, Zhao X, Chen W. A magnetic nanoparticle-based multiple-gene delivery system for transfection of porcine kidney cells. PLoS One 2014; 9:e102886. [PMID: 25048709 PMCID: PMC4105564 DOI: 10.1371/journal.pone.0102886] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 06/24/2014] [Indexed: 01/13/2023] Open
Abstract
Superparamagnetic nanoparticles are promising candidates for gene delivery into mammalian somatic cells and may be useful for reproductive cloning using the somatic cell nuclear transfer technique. However, limited investigations of their potential applications in animal genetics and breeding, particularly multiple-gene delivery by magnetofection, have been performed. Here, we developed a stable, targetable and convenient system for delivering multiple genes into the nuclei of porcine somatic cells using magnetic Fe3O4 nanoparticles as gene carriers. After surface modification by polyethylenimine, the spherical magnetic Fe3O4 nanoparticles showed strong binding affinity for DNA plasmids expressing the genes encoding a green (DNAGFP) or red (DNADsRed) fluorescent protein. At weight ratios of DNAGFP or DNADsRed to magnetic nanoparticles lower than or equal to 10∶1 or 5∶1, respectively, the DNA molecules were completely bound by the magnetic nanoparticles. Atomic force microscopy analyses confirmed binding of the spherical magnetic nanoparticles to stretched DNA strands up to several hundred nanometers in length. As a result, stable and efficient co-expression of GFP and DsRed in porcine kidney PK-15 cells was achieved by magnetofection. The results presented here demonstrate the potential application of magnetic nanoparticles as an attractive delivery system for animal genetics and breeding studies.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Zhongguancun Haidian District, Beijing, China
| | - Haixin Cui
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Zhongguancun Haidian District, Beijing, China
- * E-mail:
| | - Kui Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Haidian District, Beijing, China
| | - Changjiao Sun
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Zhongguancun Haidian District, Beijing, China
| | - Wei Du
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Zhongguancun Haidian District, Beijing, China
| | - Jinhui Cui
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Zhongguancun Haidian District, Beijing, China
| | - Xiang Zhao
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Zhongguancun Haidian District, Beijing, China
| | - Wenjie Chen
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Zhongguancun Haidian District, Beijing, China
| |
Collapse
|
29
|
Yang HN, Park JS, Jeon SY, Park W, Na K, Park KH. The effect of quantum dot size and poly(ethylenimine) coating on the efficiency of gene delivery into human mesenchymal stem cells. Biomaterials 2014; 35:8439-49. [PMID: 24985737 DOI: 10.1016/j.biomaterials.2014.06.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 06/10/2014] [Indexed: 01/13/2023]
Abstract
Quantum dot (QDs) have been employed as bioimaging agents and delivery vehicles for gene therapeutics in several types of cells. In this study, we fabricated multiple QD bundled nanoparticles (NPs) to investigate the effect of QD size and poly(ethylenimine) (PEI) coating on the efficiency of gene delivery into human mesenchymal stem cells (hMSCs). Several types of QDs, which exhibit different ranges of particle size and fluorescence when employed, were coated with PEI to alter their negative charges and to enable them to be bundled into larger particles. Using specific wavelengths of QDs for bioimaging, gene-complexed QD bundled NPs were easily detected in the hMSCs using several different methods such as fluorescence-activated cell sorter, confocal laser scanning microscopy, and in vivo optical imaging. These PEI-coated, bundled QD NPs exhibited significantly higher gene transfection efficacy than single-type QDs. Particularly, the largest QD bundled NPs examined, QD655, had a much higher uptake capability and greater gene expression ability than the other QD NPs (QD525, QD565, and QD605). We believe that our findings help to enrich knowledge of design considerations that will aid in the engineering of QD NPs for stem cell application in the future.
Collapse
Affiliation(s)
- Han Na Yang
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Acecore, 502 Yatap-dong Bundang-gu, Seongam-si, Gyeonggi-do, Republic of Korea
| | - Ji Sun Park
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Acecore, 502 Yatap-dong Bundang-gu, Seongam-si, Gyeonggi-do, Republic of Korea
| | - Su Yeon Jeon
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Acecore, 502 Yatap-dong Bundang-gu, Seongam-si, Gyeonggi-do, Republic of Korea
| | - Wooram Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Kun Na
- Department of Biotechnology, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Keun-Hong Park
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Acecore, 502 Yatap-dong Bundang-gu, Seongam-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
30
|
Fouriki A, Dobson J. Oscillating magnet array-based nanomagnetic gene transfection of human mesenchymal stem cells. Nanomedicine (Lond) 2014; 9:989-97. [DOI: 10.2217/nnm.13.74] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aim: In this work, the potential of nanomagnetic transfection of primary human mesenchymal stem cells (hMSCs) and the effects of a novel nonviral oscillating magnet array system in enhancing transfection efficiency were investigated. Materials & methods: Green fluorescent protein plasmids coupled to magnetic nanoparticles (MNPs) were introduced onto hMSCs in culture. Magnetic fields generated by arrays of neodymium iron boron magnets positioned below the culture plates direct the MNP/DNA complexes into contact with the cells. The magnet arrays were oscillated, promoting more efficient endocytosis via mechanical stimulation. Green fluorescent protein expression, cell viability and stem cell surface markers were assayed. Results: MNP/DNA complexes were delivered into hMSCs, and the oscillating magnet array system appears to improve transfection efficiency as well as cell viability. The expression of hMSC-specific cell surface markers was unaffected. Conclusion: Nonviral transfection using MNPs and oscillating magnet arrays offers a more efficient and ‘cell-friendly’ method of transfecting hMSCs than other nonviral techniques, while preserving their stem cell characteristics. Original submitted 8 March 2012; Revised submitted 12 February 2013
Collapse
Affiliation(s)
- Angeliki Fouriki
- Institute for Science & Technology in Medicine, Keele University, Thornburrow Drive, Hartshill, Stoke-on-Trent, ST4 7QB, UK
| | - Jon Dobson
- Institute for Science & Technology in Medicine, Keele University, Thornburrow Drive, Hartshill, Stoke-on-Trent, ST4 7QB, UK
- J Crayton Pruitt Family Department of Biomedical Engineering, Department of Materials Science & Engineering, & the Institute for Cell Engineering & Regenerative Medicine University of Florida, PO Box 116131, Gainesville, FL 32611, USA
| |
Collapse
|
31
|
Recent progresses in gene delivery-based bone tissue engineering. Biotechnol Adv 2013; 31:1695-706. [DOI: 10.1016/j.biotechadv.2013.08.015] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 07/24/2013] [Accepted: 08/19/2013] [Indexed: 12/18/2022]
|
32
|
Seo SJ, Kim TH, Choi SJ, Park JH, Wall IB, Kim HW. Gene delivery techniques for adult stem cell-based regenerative therapy. Nanomedicine (Lond) 2013; 8:1875-91. [DOI: 10.2217/nnm.13.165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Over the past decade, stem cells have been considered to be a promising resource to cure and regenerate damaged or diseased tissues with research extending from basic studies to clinical application. Furthermore, genetically modified stem cells have the potential to reduce tumorigenic risks and achieve safe tissue formation. Recent advances in genetic modification of stem cells have rendered these cells more accessible and stable. The successful genetic modification of stem cells relies heavily on designing vector systems, either viral or nonviral vectors, which can efficiently deliver therapeutic genes to the cells with minimum toxicity. Currently, viral vectors showing high transfection efficiencies still raise safety issues, whereas safer nonviral vectors exhibit extremely poor transfection in stem cells. Here, we attempt to review and discuss the main factors raising concern in previous reports, and devise strategies to solve the issues in gene delivery systems for successful stem cell-targeting regenerative therapy.
Collapse
Affiliation(s)
- Seog-Jin Seo
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan 330–714, South Korea
| | - Tae-Hyun Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan 330–714, South Korea
- Department of Nanobiomedical Science & BK21 plus NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 330–714, South Korea
| | - Seong-Jun Choi
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan 330–714, South Korea
| | - Jeong-Hui Park
- Department of Nanobiomedical Science & BK21 plus NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 330–714, South Korea
- Department of Biochemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| | - Ivan B Wall
- Department of Nanobiomedical Science & BK21 plus NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 330–714, South Korea
- Department of Biochemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| | - Hae-Won Kim
- Department of Biomaterials Science, College of Dentistry, Dankook University Cheonan 330–714, South Korea
| |
Collapse
|
33
|
Dai T, Yang E, Sun Y, Zhang L, Zhang L, Shen N, Li S, Liu L, Xie Y, Wu S, Gao Z. Preparation and drug release mechanism of CTS-TAX-NP-MSCs drug delivery system. Int J Pharm 2013; 456:186-94. [DOI: 10.1016/j.ijpharm.2013.07.070] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 07/02/2013] [Accepted: 07/19/2013] [Indexed: 12/17/2022]
|
34
|
Auffinger B, Morshed R, Tobias A, Cheng Y, Ahmed AU, Lesniak MS. Drug-loaded nanoparticle systems and adult stem cells: a potential marriage for the treatment of malignant glioma? Oncotarget 2013; 4:378-96. [PMID: 23594406 PMCID: PMC3717302 DOI: 10.18632/oncotarget.937] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Despite all recent advances in malignant glioma research, only modest progress has been achieved in improving patient prognosis and quality of life. Such a clinical scenario underscores the importance of investing in new therapeutic approaches that, when combined with conventional therapies, are able to effectively eradicate glioma infiltration and target distant tumor foci. Nanoparticle-loaded delivery systems have recently arisen as an exciting alternative to improve targeted anti-glioma drug delivery. As drug carriers, they are able to efficiently protect the therapeutic agent and allow for sustained drug release. In addition, their surface can be easily manipulated with the addition of special ligands, which are responsible for enhancing tumor-specific nanoparticle permeability. However, their inefficient intratumoral distribution and failure to target disseminated tumor burden still pose a big challenge for their implementation as a therapeutic option in the clinical setting. Stem cell-based delivery of drug-loaded nanoparticles offers an interesting option to overcome such issues. Their ability to incorporate nanoparticles and migrate throughout interstitial barriers, together with their inherent tumor-tropic properties and synergistic anti-tumor effects make these stem cell carriers a good fit for such combined therapy. In this review, we will describe the main nanoparticle delivery systems that are presently available in preclinical and clinical studies. We will discuss their mechanisms of targeting, current delivery methods, attractive features and pitfalls. We will also debate the potential applications of stem cell carriers loaded with therapeutic nanoparticles in anticancer therapy and why such an attractive combined approach has not yet reached clinical trials.
Collapse
Affiliation(s)
- Brenda Auffinger
- Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
35
|
Wang C, Ning L, Wang H, Lu Z, Li X, Fan X, Wang X, Liu Y. A peptide-mediated targeting gene delivery system for malignant glioma cells. Int J Nanomedicine 2013; 8:3631-40. [PMID: 24101872 PMCID: PMC3790891 DOI: 10.2147/ijn.s44990] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant glioma. Although there has been considerable progress in treatment strategies, the prognosis of many patients with GBM remains poor. In this work, polyethylenimine (PEI) and the VTWTPQAWFQWV (VTW) peptide were modified and synthesized into GBM-targeting nanoparticles. The transfection efficiency of U-87 (human glioblastoma) cells was evaluated using fluorescence microscopy and flow cytometry. Cell internalization was investigated to verify the nanoparticle delivery into the cytoplasm. Results showed that the methods of polymer conjugation and the amount of VTW peptide were important factors to polymer synthesis and transfection. The PEI-VTW20 nanoparticles increased the transfection efficiency significantly. This report describes the use of VTW peptide-based PEI nanoparticles for intracellular gene delivery in a GBM cell-specific manner.
Collapse
Affiliation(s)
- Chuanwei Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China ; Brain Science Research Institute of Shandong University, Jinan, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Malakooty Poor E, Baghaban Eslaminejad M, Gheibi N, Bagheri F, Atyabi F. Chitosan–pDNA nanoparticle characteristics determine the transfection efficacy of gene delivery to human mesenchymal stem cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2013; 42:376-84. [DOI: 10.3109/21691401.2013.832685] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
37
|
|
38
|
Wegman F, Oner FC, Dhert WJA, Alblas J. Non-viral gene therapy for bone tissue engineering. Biotechnol Genet Eng Rev 2013; 29:206-20. [PMID: 24568281 DOI: 10.1080/02648725.2013.801227] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The possibilities of using gene therapy for bone regeneration have been extensively investigated. Improvements in the design of new transfection agents, combining vectors and delivery/release systems to diminish cytotoxicity and increase transfection efficiencies have led to several successful in vitro, ex vivo and in vivo strategies. These include growth factor or short interfering ribonucleic acid (siRNA) delivery, or even enzyme replacement therapies, and have led to increased osteogenic differentiation and bone formation in vivo. These results provide optimism to consider use in humans with some of these gene-delivery strategies in the near future.
Collapse
Affiliation(s)
- Fiona Wegman
- a Department of Orthopaedics , UMC Utrecht , Utrecht , The Netherlands
| | | | | | | |
Collapse
|
39
|
Galactosylated chitosan oligosaccharide nanoparticles for hepatocellular carcinoma cell-targeted delivery of adenosine triphosphate. Int J Mol Sci 2013. [PMID: 23899789 DOI: 10.3390/ijm5140815755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Nanoparticles composed of galactosylated chitosan oligosaccharide (Gal-CSO) and adenosine triphosphate (ATP) were prepared for hepatocellular carcinoma cell-specific uptake, and the characteristics of Gal-CSO/ATP nanoparticles were evaluated. CSO/ATP nanoparticles were prepared as a control. The average diameter and zeta potential of Gal-CSO/ATP nanoparticles were 51.03 ± 3.26 nm and 30.50 ± 1.25 mV, respectively, suggesting suitable properties for a drug delivery system. Subsequently, the cytotoxicity of Gal-CSO/ATP nanoparticles were examined by the methyl tetrazolium (MTT) assay, and the half maximal inhibitory concentration (IC50) values were calculated with HepG2 (human hepatocellular carcinoma cell line) cells. The results showed that the cytotoxic effect of nanoparticles on HepG2 cells was low. In the meantime, it was also found that the Gal-CSO/ATP nanoparticles could be uptaken by HepG2 cells, due to expression of the asialoglycoprotein receptor (ASGP-R) on their surfaces. The presented results indicate that the Gal-CSO nanoparticles might be very attractive to be used as an intracellular drug delivery carrier for hepatocellular carcinoma cell targeting, thus warranting further in vivo or clinical investigations.
Collapse
|
40
|
Zhu XL, Du YZ, Yu RS, Liu P, Shi D, Chen Y, Wang Y, Huang FF. Galactosylated chitosan oligosaccharide nanoparticles for hepatocellular carcinoma cell-targeted delivery of adenosine triphosphate. Int J Mol Sci 2013; 14:15755-66. [PMID: 23899789 PMCID: PMC3759884 DOI: 10.3390/ijms140815755] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 07/07/2013] [Accepted: 07/23/2013] [Indexed: 01/14/2023] Open
Abstract
Nanoparticles composed of galactosylated chitosan oligosaccharide (Gal-CSO) and adenosine triphosphate (ATP) were prepared for hepatocellular carcinoma cell-specific uptake, and the characteristics of Gal-CSO/ATP nanoparticles were evaluated. CSO/ATP nanoparticles were prepared as a control. The average diameter and zeta potential of Gal-CSO/ATP nanoparticles were 51.03 ± 3.26 nm and 30.50 ± 1.25 mV, respectively, suggesting suitable properties for a drug delivery system. Subsequently, the cytotoxicity of Gal-CSO/ATP nanoparticles were examined by the methyl tetrazolium (MTT) assay, and the half maximal inhibitory concentration (IC50) values were calculated with HepG2 (human hepatocellular carcinoma cell line) cells. The results showed that the cytotoxic effect of nanoparticles on HepG2 cells was low. In the meantime, it was also found that the Gal-CSO/ATP nanoparticles could be uptaken by HepG2 cells, due to expression of the asialoglycoprotein receptor (ASGP-R) on their surfaces. The presented results indicate that the Gal-CSO nanoparticles might be very attractive to be used as an intracellular drug delivery carrier for hepatocellular carcinoma cell targeting, thus warranting further in vivo or clinical investigations.
Collapse
Affiliation(s)
- Xiu Liang Zhu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, China; E-Mails: (X.L.Z.); (D.S.); (Y.C.); (Y.W.); (F.F.H.)
| | - Yong Zhong Du
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; E-Mails: (Y.Z.D.); (P.L.)
| | - Ri Sheng Yu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, China; E-Mails: (X.L.Z.); (D.S.); (Y.C.); (Y.W.); (F.F.H.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel./Fax: +86-571-8820-8439
| | - Ping Liu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; E-Mails: (Y.Z.D.); (P.L.)
| | - Dan Shi
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, China; E-Mails: (X.L.Z.); (D.S.); (Y.C.); (Y.W.); (F.F.H.)
| | - Ying Chen
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, China; E-Mails: (X.L.Z.); (D.S.); (Y.C.); (Y.W.); (F.F.H.)
| | - Ying Wang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, China; E-Mails: (X.L.Z.); (D.S.); (Y.C.); (Y.W.); (F.F.H.)
| | - Fang Fang Huang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, China; E-Mails: (X.L.Z.); (D.S.); (Y.C.); (Y.W.); (F.F.H.)
| |
Collapse
|
41
|
Li X, Zhao Q, Qiu L. Smart ligand: aptamer-mediated targeted delivery of chemotherapeutic drugs and siRNA for cancer therapy. J Control Release 2013; 171:152-62. [PMID: 23777885 DOI: 10.1016/j.jconrel.2013.06.006] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 06/06/2013] [Accepted: 06/08/2013] [Indexed: 11/27/2022]
Abstract
Aptamers are a class of oligonucleotides that can specifically bind to different targets with high affinity. Since their discovery in 1980s, aptamers have attracted considerable interests in medical applications. So far, initial research using aptamers as delivery systems has produced exciting results. In this review, we summarize recent progress in aptamer-mediated chemotherapeutic drug and siRNA delivery systems in tumor treatment. With regard to chemotherapeutic drugs, the 2 main methods for targeted delivery using aptamers are as follows: aptamer-drug systems (in which aptamers directly deliver the drug both as a carrier and as a ligand) and aptamer-nanoparticles systems (in which nanoparticles function together with aptamers for targeted delivery of drugs). For delivery of siRNA, aptamers can be utilized by the following ways to facilitate targeting: (1) linked by a connector; (2) form a chimera; and (3) combined with nanoparticles. In co-delivery system, the advantages associated with the use of aptamers are beginning to become apparent also. Here, the challenges and new perspectives in the field of aptamer-mediated delivery have been discussed.
Collapse
Affiliation(s)
- Xin Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | | | | |
Collapse
|
42
|
Knipe JM, Peters JT, Peppas NA. Theranostic agents for intracellular gene delivery with spatiotemporal imaging. NANO TODAY 2013; 8:21-38. [PMID: 23606894 PMCID: PMC3627379 DOI: 10.1016/j.nantod.2012.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Gene therapy is the modification of gene expression to treat a disease. However, efficient intracellular delivery and monitoring of gene therapeutic agents is an ongoing challenge. Use of theranostic agents with suitable targeted, controlled delivery and imaging modalities has the potential to greatly advance gene therapy. Inorganic nanoparticles including magnetic nanoparticles, gold nanoparticles, and quantum dots have been shown to be effective theranostic agents for the delivery and spatiotemporal tracking of oligonucleotides in vitro and even a few cases in vivo. Major concerns remain to be addressed including cytotoxicity, particularly of quantum dots; effective dosage of nanoparticles for optimal theranostic effect; development of real-time in vivo imaging; and further improvement of gene therapy efficacy.
Collapse
Affiliation(s)
- Jennifer M. Knipe
- Department of Chemical Engineering, C0400, The University of Texas at Austin, Austin, TX 78712 (USA)
| | - Jonathan T. Peters
- Department of Chemical Engineering, C0400, The University of Texas at Austin, Austin, TX 78712 (USA)
| | - Nicholas A. Peppas
- Department of Chemical Engineering, C0400, The University of Texas at Austin, Austin, TX 78712 (USA)
- Department of Biomedical Engineering, C0800, The University of Texas at Austin, Austin, TX 78712 (USA)
- College of Pharmacy, C0400, The University of Texas at Austin, Austin, TX 78712 (USA)
| |
Collapse
|
43
|
Hou S, Ma H, Ji Y, Hou W, Jia N. A calcium phosphate nanoparticle-based biocarrier for efficient cellular delivery of antisense oligodeoxynucleotides. ACS APPLIED MATERIALS & INTERFACES 2013; 5:1131-1136. [PMID: 23323641 DOI: 10.1021/am3028926] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Antisense oligodeoxynucleotides (ASODNs) can bind to some specific RNA of survivin can prevent the mRNA translation at the genetic level, which will inhibit survivin expression and make the cancer cells apoptosis. However, the ASODNs-based therapies are hampered by their instability to cellular nuclease and their weak intracellular penetration. Here we reported a calcium phosphate (CP)-based carrier to achieve efficient delivery of ASODNs into cells. In this study, we used a facile microemulsion approach to prepare spherical and porous ASODNs-CP nanoparticles (ASODNS-CPNPs) with the size of 50-70 nm in diameter, and their structure, morphology and composition were characterized by TEM, XRD, FTIR, ICP and DLS, UV-Vis spectroscopy and agarose gel electrophoresis. The results indicated that the nanoparticles have a high ASODNs loading capacity. Furthermore, cellular uptake and delivery efficiency of the ASODNS-CPNPs, as well as cellular apoptosis induced by the ASODNs doping into the calcium phosphate nanoparticles, were investigated by confocal laser scanning microscopy, biological TEM, flow cytometry, and MTT assay. Efficient intracellular delivery of the nanoparticles was observed. All these results suggested that the prepared calcium phosphate nanoparticles could be used as a promising biocarrier for delivery of ASODNs.
Collapse
Affiliation(s)
- Shenglei Hou
- The Education Ministry Key Laboratory of Resource Chemistry, Department of Chemistry, Life and Environmental Science College, Shanghai Normal University, Shanghai 200234, China
| | | | | | | | | |
Collapse
|
44
|
Roy SG, Acharya R, Chatterji U, De P. RAFT polymerization of methacrylates containing a tryptophan moiety: controlled synthesis of biocompatible fluorescent cationic chiral polymers with smart pH-responsiveness. Polym Chem 2013. [DOI: 10.1039/c2py20821k] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
45
|
Elsler S, Schetting S, Schmitt G, Kohn D, Madry H, Cucchiarini M. Effective, safe nonviral gene transfer to preserve the chondrogenic differentiation potential of human mesenchymal stem cells. J Gene Med 2012; 14:501-11. [PMID: 22711470 DOI: 10.1002/jgm.2644] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Genetic modification of mesenchymal stem cells (MSCs) comprises a promising tool to generate cell- and gene-based platforms for regenerative approaches of articular cartilage repair. In the present study, we systematically screened a panel of 15 nonviral compounds for their ability to promote safe, efficient and durable gene expression in human bone marrow-derived MSCs (hMSCS) without impeding their commitment towards chondrogenic differentiation. METHODS Primary hMSCs were transfected with plasmid vectors carrying sequences for the Photinus pyralis luciferase Escherichia coli β-galactosidase, or human insulin-like growth factor I via 15 nonviral formulations. Transgene expression and transfection efficiencies were monitored for each component in parallel with the effects on cell viability and cytotoxicity. Upon optimization, the most promising reagent was then evaluated for a possible influence on the chondrogenic potential of hMSCs. RESULTS Among all formulations tested, GeneJammer® gave the best results for transgene expression and transfection efficacy (25-14% from days 2-21 in monolayer cultures and 35% in 21-day aggregate cultures), allowing for high levels of viability (92-94%) and modest cytotoxicity (< 12%). Most notably, the application of this reagent did not affect the potential of the cells for chondrogenic differentiation when maintained in long-term (21 days) three-dimensional (aggregate) cultures. CONCLUSIONS The data indicate that safe, efficient transgene expression can be achieved in hMSCs over time using the nonviral GeneJammer® compound, showing promise for future therapeutic settings aiming to treat human articular cartilage disorders.
Collapse
Affiliation(s)
- Sebastian Elsler
- Center of Experimental Orthopaedics, Saarland University Medical Center, Saarland University, Homburg/Saar, Germany
| | | | | | | | | | | |
Collapse
|
46
|
Park JS, Yang HN, Woo DG, Jeon SY, Do HJ, Huh SH, Kim NH, Kim JH, Park KH. Exogenous Nurr1 gene expression in electrically-stimulated human MSCs and the induction of neurogenesis. Biomaterials 2012; 33:7300-8. [DOI: 10.1016/j.biomaterials.2012.06.069] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 06/25/2012] [Indexed: 02/06/2023]
|
47
|
Yang HN, Park JS, Woo DG, Jeon SY, Park KH. Transfection of VEGF(165) genes into endothelial progenitor cells and in vivo imaging using quantum dots in an ischemia hind limb model. Biomaterials 2012; 33:8670-84. [PMID: 22921925 DOI: 10.1016/j.biomaterials.2012.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 08/05/2012] [Indexed: 10/28/2022]
Abstract
Endothelial progenitor cells (EPCs) were transfected with fluorescently labeled quantum dot nanoparticles (QD NPs) with or without VEGF(165) plasmid DNA (pDNA) to probe the EPCs after in vivo transplantation and to test whether they presented as differentiated endothelial cells (ECs). Bare QD NPs and QD NPs coated with PEI or PEI + VEGF(165) genes were characterized by dynamic light scattering, scanning electron microscopy, and atomic force microscopy. Transfection of EPCs with VEGF(165) led to the expression of specific genes and proteins for mature ECs. A hind limb ischemia model was generated in nude mice, and VEGF(165) gene-transfected EPCs were transplanted intramuscularly into the ischemic limbs. At 28 days after transplantation, the VEGF(165) gene-transfected EPCs significantly increased the number of differentiated ECs compared with the injection of medium or bare EPCs without VEGF(165) genes. Laser Doppler imaging revealed that blood perfusion levels were increased significantly by VEGF(165) gene-transfected EPCs compared to EPCs without VEGF(165). Moreover, the transplantation of VEGF(165) gene-transfected EPCs increased the specific gene and protein expression levels of mature EC markers and angiogenic factors in the animal model.
Collapse
Affiliation(s)
- Han Na Yang
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Acecore, 502 Yatap-dong Bundang-gu, Seongnam-si, Republic of Korea
| | | | | | | | | |
Collapse
|
48
|
SOX9 gene plus heparinized TGF-β 3 coated dexamethasone loaded PLGA microspheres for inducement of chondrogenesis of hMSCs. Biomaterials 2012; 33:7151-63. [PMID: 22795539 DOI: 10.1016/j.biomaterials.2012.06.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 06/15/2012] [Indexed: 11/20/2022]
Abstract
Microparticulated types of scaffolds have been widely applied in stem cell therapy and the tissue engineering field for the regeneration of wound tissues. During application of simple genes or growth factors and cell delivery vehicles, we designed a method that employs dexamethsone loaded PLGA microspheres consisting of polyplexed SOX9 genes plus heparinized TGF-β 3 on the surface of polymeric microspheres prepared using a layer-by-layer (LbL) method. The fabrication of the polyplexed SOX9 genes plus heparinized TGF-β 3 and their subsequent coating onto dexamethsone loaded PLGA microspheres represents a method for functionalization of the polymeric matrix. The use of SOX9 gene plus heparinized TGF-β 3 coated dexamethsone loaded PLGA microspheres was evaluated to determine their potential as both gene carriers and cell delivery vehicle. By adhesion of hMSCs onto SOX9 gene plus heparinized TGF-β 3 coated dexamethsone loaded PLGA microspheres, the chondrogenesis-related specific genes of collagen type II were increased 30 times comparing to control. Also, the specific extracellular matrix of glycosaminoglycan (GAG) production of hMSCs adhered onto SOX9 gene plus heparinized TGF-β 3 coated dexamethasone loaded PLGA microspheres increased more 2.5 times than control group. Not only in vitro culture but in vivo results, the specific genes of COMP, aggrecan, collagen type II, and SOX9 showed much more gene expressions such as 20, 15, 10, 8 times.
Collapse
|
49
|
Mesenchymal stem cells: a potential targeted-delivery vehicle for anti-cancer drug, loaded nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2012; 9:174-84. [PMID: 22772046 DOI: 10.1016/j.nano.2012.06.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 06/04/2012] [Indexed: 12/22/2022]
Abstract
UNLABELLED The targeted delivery of anticancer agents is a promising field in anticancer therapy. Mesenchymal stem cells (MSCs) have inherent tumor-tropic and migratory properties, which allow them to serve as vehicles for targeted drug delivery systems for isolated tumors and metastatic diseases. MSCs have been successfully studied and discussed as a vehicle for cancer gene therapy. However, MSCs have not yet been discussed adequately as a potential vehicle for traditional anticancer drugs. In this review, we will examine the potential of MSCs as a targeted-delivery vehicle for anticancer drug-loaded nanoparticles (NPs), summarize various challenges, and discuss possible solutions for these challenges. FROM THE CLINICAL EDITOR In this review, the feasibility of mesenchymal stem cell-based targeted delivery of anticancer agents is discussed.
Collapse
|
50
|
Zhai W, He C, Wu L, Zhou Y, Chen H, Chang J, Zhang H. Degradation of hollow mesoporous silica nanoparticles in human umbilical vein endothelial cells. J Biomed Mater Res B Appl Biomater 2012; 100:1397-403. [DOI: 10.1002/jbm.b.32711] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 02/20/2012] [Accepted: 03/11/2012] [Indexed: 12/14/2022]
|