1
|
Voznesenskaya A, Berggren PO, Ilegems E. Sustained heterologous gene expression in pancreatic islet organoids using adeno-associated virus serotype 8. Front Bioeng Biotechnol 2023; 11:1147244. [PMID: 37545890 PMCID: PMC10400289 DOI: 10.3389/fbioe.2023.1147244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 07/06/2023] [Indexed: 08/08/2023] Open
Abstract
Genetic modification of pancreatic islet organoids, assembled in vitro prior to transplantation is an emerging alternative to direct in vivo genetic manipulations for a number of clinical and research applications. We have previously shown that dispersion of islet cells followed by re-aggregation into islet organoids, or pseudoislets, allows for efficient transduction with viral vectors, while maintaining physiological functions of native islets. Among viruses currently used for genetic manipulations, adeno-associated viruses (AAVs) have the most attractive safety profile making them suitable for gene therapy applications. Studies reporting on pseudoislet transduction with AAVs are, however, lacking. Here, we have characterized in detail the performance of AAV serotype 8 in transduction of islet cells during pseudoislet formation in comparison with human adenovirus type 5 (AdV5). We have assessed such parameters as transduction efficiency, expression kinetics, and endocrine cell tropism of AAV8 alone or in combination with AdV5. Data provided within our study may serve as a reference point for future functional studies using AAVs for gene transfer to islet cell organoids and will facilitate further development of engineered pseudoislets of superior quality suitable for clinical transplantation.
Collapse
|
2
|
Qi H, Chen G, Huang Y, Si Z, Li J. Foxp3-modified bone marrow mesenchymal stem cells promotes liver allograft tolerance through the generation of regulatory T cells in rats. J Transl Med 2015; 13:274. [PMID: 26293578 PMCID: PMC4545923 DOI: 10.1186/s12967-015-0638-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 08/13/2015] [Indexed: 01/14/2023] Open
Abstract
Background The transcription factor forkhead box P3 (Foxp3) is a master regulatory gene necessary for the development and function of CD4+CD25+ regulatory T cells (Tregs). Mesenchymal stem cells (MSC) have recently emerged as promising candidates for cell-based immunosuppression/tolerance induction protocols. Thus, we hypothesized that MSC-based Foxp3 gene therapy would improve immunosuppressive capacity of MSC and induce donor-specific allograft tolerance in rat’s liver allograft model. Methods The present study utilized a lentivirus vector to overexpress the therapeutic gene Foxp3 on MSC. In vivo, Injections of 2 × 106 MSC, FUGW-MSC or Foxp3-MSC into the portal vein were carried out immediately after liver transplantation. Results Successful gene transfer of Foxp3 in MSC was achieved by lentivirus carrying Foxp3 and Foxp3-MSC engraftment in liver allograft was confirmed by fluorescence microscopy. Foxp3-MSC treatment significantly inhibited the proliferation of allogeneic ACI CD4+ T cells to splenocytes (SC) from the same donor strain or third-party BN rat compared with MSC. Foxp3-MSC suppressive effect on the proliferation of CD4+ T cells is contact dependent and associated with Programmed death ligand 1(PD-L1) upregulation in MSC. Co-culture of CD4+ T cells with Foxp3-MSC results in a shift towards a Tregs phenotype. More importantly, Foxp3-MSC monotherapy achieved donor-specific liver allograft tolerance and generated a state of CD4+CD25+Foxp3+ Tregs-dependent tolerance. Conclusion Foxp3-engineered MSC therapy seems to be a promising and attractive cell therapy approach for inducing immunosuppression or transplant tolerance.
Collapse
Affiliation(s)
- Haizhi Qi
- Department of Organ Transplantation, Second Xiangya Hospital, Central South University, 139 RenMin Road, Changsha, Hunan, China.
| | - Guangshun Chen
- Department of Organ Transplantation, Second Xiangya Hospital, Central South University, 139 RenMin Road, Changsha, Hunan, China.
| | - Yaxun Huang
- Department of Organ Transplantation, Second Xiangya Hospital, Central South University, 139 RenMin Road, Changsha, Hunan, China.
| | - Zhongzhou Si
- Department of Organ Transplantation, Second Xiangya Hospital, Central South University, 139 RenMin Road, Changsha, Hunan, China.
| | - Jiequn Li
- Department of Organ Transplantation, Second Xiangya Hospital, Central South University, 139 RenMin Road, Changsha, Hunan, China.
| |
Collapse
|
3
|
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
|
4
|
Lee BW, Kim MH, Chae HY, Hwang HJ, Kang D, Ihm SH. Enhanced gene transfer to pancreatic islets using glucagon-like peptide-1. Transplant Proc 2013; 45:591-6. [PMID: 23498795 DOI: 10.1016/j.transproceed.2012.10.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 09/21/2012] [Accepted: 10/30/2012] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The efficient transfer of genes into intact islets is difficult since islets exist as clusters of differentiated cells with little replication potential. Cell proliferation in response to growth factors is known to be accompanied by loosening of cell-to-cell contacts and increasing paracellular permeability. In this study, we investigated whether gene delivery into intact islet cells was facilitated by modulating β-cell proliferation. METHODS Isolated rat islets were pretreated with glucagon-like peptide (GLP)-1 or human growth hormone for 24 hours, or with 300 mg/dL of glucose for 48 hours before transduction with a suboptimal dose of recombinant adenoviral vector expressing green fluorescent protein (GFP) and β-galactosidase (multiplicity of infection of 25). Transduction efficiency was assessed by measuring β-galactosidase activity and GFP expression using enzyme-linked immunosorbent assay, flow cytometry, and fluorescence microscopy. The numbers of 7-aminoactinomycin D-positive dead cells and 5-ethynyl-2-deoxyuridine (EdU)-positive proliferating cells were also monitored using flow cytometry and fluorescence microscopy. RESULTS The transduction efficiency of rat islet cells by a suboptimal dose of viral vector was significantly improved by GLP-1 pretreatment, accompanied by enhanced cell viability and cell proliferation. An increased GFP expression in islet cells after GLP-1 pretreatment was observed among the increased numbers of EdU-positive proliferating cells. CONCLUSION Pretreatment of rat islets with GLP-1 enhanced the transduction efficiency of an adenoviral vector, reducing viral dose burden while improving islet cell viability. From a therapeutic standpoint, genetic modification of pancreatic islets combined with GLP-1 pretreatment may be a promising option for ex vivo gene therapy prior to islet transplantation.
Collapse
Affiliation(s)
- B W Lee
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | | | | | | | | | | |
Collapse
|
5
|
Song L, Liu P, Han C, Liu Y, Zou W, Piao H, Wang Y, Liu J. A novel approach to facilitate dopaminergic neuron generation from stem-cells: The combination of genetic modification and signaling factors within a three-dimensional perfusion microbioreactor. Med Hypotheses 2013; 80:407-10. [DOI: 10.1016/j.mehy.2012.12.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 12/04/2012] [Accepted: 12/29/2012] [Indexed: 12/12/2022]
|
6
|
Gabriel E, Schievenbusch S, Kolossov E, Hengstler JG, Rotshteyn T, Bohlen H, Nierhoff D, Hescheler J, Drobinskaya I. Differentiation and selection of hepatocyte precursors in suspension spheroid culture of transgenic murine embryonic stem cells. PLoS One 2012; 7:e44912. [PMID: 23028675 PMCID: PMC3454367 DOI: 10.1371/journal.pone.0044912] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 08/09/2012] [Indexed: 01/08/2023] Open
Abstract
Embryonic stem cell-derived hepatocyte precursor cells represent a promising model for clinical transplantations to diseased livers, as well as for establishment of in vitro systems for drug metabolism and toxicology investigations. This study aimed to establish an in vitro culture system for scalable generation of hepatic progenitor cells. We used stable transgenic clones of murine embryonic stem cells possessing a reporter/selection vector, in which the enhanced green fluorescent protein- and puromycin N-acetyltransferase-coding genes are driven by a common alpha-fetoprotein gene promoter. This allowed for "live" monitoring and puromycin selection of the desired differentiating cell type possessing the activated alpha-fetoprotein gene. A rotary culture system was established, sequentially yielding initially partially selected hepatocyte lineage-committed cells, and finally, a highly purified cell population maintained as a dynamic suspension spheroid culture, which progressively developed the hepatic gene expression phenotype. The latter was confirmed by quantitative RT-PCR analysis, which showed a progressive up-regulation of hepatic genes during spheroid culture, indicating development of a mixed hepatocyte precursor-/fetal hepatocyte-like cell population. Adherent spheroids gave rise to advanced differentiated hepatocyte-like cells expressing hepatic proteins such as albumin, alpha-1-antitrypsin, cytokeratin 18, E-cadherin, and liver-specific organic anion transporter 1, as demonstrated by fluorescent immunostaining. A fraction of adherent cells was capable of glycogen storage and of reversible up-take of indocyanine green, demonstrating their hepatocyte-like functionality. Moreover, after transplantation of spheroids into the mouse liver, the spheroid-derived cells integrated into recipient. These results demonstrate that large-scale hepatocyte precursor-/hepatocyte-like cultures can be established for use in clinical trials, as well as in in vitro screening assays.
Collapse
Affiliation(s)
- Elke Gabriel
- Institute of Neurophysiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| | | | | | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Dortmund, Germany
| | - Tamara Rotshteyn
- Institute of Neurophysiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| | | | - Dirk Nierhoff
- Gastroenterology and Hepatology Clinic, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| | - Irina Drobinskaya
- Institute of Neurophysiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| |
Collapse
|
7
|
The restoration of full-thickness cartilage defects with BMSCs and TGF-beta 1 loaded PLGA/fibrin gel constructs. Biomaterials 2010; 31:8964-73. [PMID: 20822812 DOI: 10.1016/j.biomaterials.2010.08.018] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Accepted: 08/07/2010] [Indexed: 01/18/2023]
Abstract
Poly(lactide-co-glycolide) (PLGA) sponge was filled with fibrin gel, bone marrow mesenchymal stem cells (BMSCs) and transforming growth factor-β1 (TGF-β1) to obtain a construct for cartilage restoration in vivo. The PLGA sponge lost its weight steadily in vitro, but degraded much faster in the construct of PLGA/fibrin gel/BMSCs implanted in the full-thickness cartilage defects. The in vivo degradation of the fibrin gel inside the construct was prolonged to 12 wk too. The CM-DiI labeled allogenic BMSCs were detectable after transplantation (implantation) into the defects for 12 wk by small animal in vivo fluorescence imaging and confocal laser scanning microscopy. In vivo repair experiments were firstly performed by implantation of the PLGA/fibrin gel/BMSCs and PLGA/BMSCs constructs into full-thickness cartilage defects (3 mm in diameter and 4 mm in depth) of New Zealand white rabbits for 12 wk. The defects implanted with the PLGA/fibrin gel/BMSCs constructs were filled with cartilage-like tissue containing collagen type II and glycosaminoglycans (GAGs), while those by the PLGA/BMSCs constructs were filled with fibrous-like tissues. To repair the defects of larger size (4 mm in diameter), addition of growth factors was mandatory as exemplified here by further loading of TGF-β1. Implantation of the PLGA/fibrin gel/BMSCs/TGF-β1 constructs into the full-thickness cartilage defects for 12 wk resulted in full restoration of the osteochondral tissue. The neo-cartilage integrated well with its surrounding cartilage and subchondral bone. Immunohistochemical and GAGs staining confirmed the similar distribution of collagen type II and GAGs in the regenerated cartilage as that of hyaline cartilage. The quantitative reverse transcription-polymerase chain reaction (qRT-PCR) revealed that the cartilage special genes were significantly up-regulated compared with those of the TGF-β1 absent constructs.
Collapse
|
8
|
Oishi K, Noguchi H, Yukawa H, Inoue M, Miyamoto Y, Iwata H, Hasegawa M, Hayashi S. Efficient transfection of sendai virus vector to mouse pancreatic stem cells in the floating state. Cell Transplant 2010; 19:893-900. [PMID: 20587148 DOI: 10.3727/096368910x509022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Sendai virus (SeV) vectors can efficiently introduce foreign genes without toxicity into various organs and are expected to be clinically applicable. We previously compared the transfectional efficiency of SeV and adenovirus (AdV) vectors by assessing the transfer of the green fluorescent protein (GFP) gene to pancreatic stem cells. Although the gene transfer efficiency was similar between these vectors, SeV vector had a lower toxicity in comparison to the AdV vector. In this study, we assessed the gene transfer efficiency of SeV vector in the floating state to pancreatic stem cells. The efficiency of gene transfer was much higher at all time points and at all concentrations in the floating state versus in the adhesion state. In addition, the pancreatic stem cells transfected with SeV in the floating state maintained their differentiation ability. These data suggest that SeV transfection to pancreatic stem cells in the floating state may be useful in gene transfer technology.
Collapse
Affiliation(s)
- Koichi Oishi
- Department of Advanced Medicine in Biotechnology and Robotics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Oishi K, Noguchi H, Saito H, Yukawa H, Miyamoto Y, Murase K, Hayashi S. Cell labeling with a novel contrast agent of magnetic resonance imaging. Cell Transplant 2010; 19:887-92. [PMID: 20587147 DOI: 10.3727/096368910x509013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell therapy is a proven and efficient method for treating multiple diseases. For both basic research and clinical practice, the development of noninvasive in vivo imaging methods is essential for monitoring the trafficking or homing of transplanted cells. One attractive approach for the effective imaging of transplanted cells is the efficient labeling of cells with a contrast agent. In this study, we developed a novel contrast agent of magnetic resonance imaging (MRI), TMADM-02. TMADM-02 was efficiently transduced into cells without toxicity. However, the aggregation of TMADM-02 was observed because of its low stability in culture medium. Therefore, TMADM-02 may have led to a false-positive test result. In future studies, we should verify not only the efficiency of labeling cells but also the stability of the contrast agent of MRI for clinical applications.
Collapse
Affiliation(s)
- Koichi Oishi
- Department of Advanced Medicine in Biotechnology and Robotics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|
10
|
Nonviral gene delivery to mesenchymal stem cells using cationic liposomes for gene and cell therapy. J Biomed Biotechnol 2010; 2010:735349. [PMID: 20625411 PMCID: PMC2896879 DOI: 10.1155/2010/735349] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Accepted: 04/06/2010] [Indexed: 11/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) hold a great promise for application in several therapies due to their unique biological characteristics. In order to harness their full potential in cell-or gene-based therapies it might be advantageous to enhance some of their features through gene delivery strategies. Accordingly, we are interested in developing an efficient and safe methodology to genetically engineer human bone marrow MSC (BM MSC), enhancing their therapeutic efficacy in Regenerative Medicine. The plasmid DNA delivery was optimized using a cationic liposome-based reagent. Transfection efficiencies ranged from ~2% to ~35%, resulting from using a Lipid/DNA ratio of 1.25 with a transgene expression of 7 days. Importantly, the number of plasmid copies in different cell passages was quantified for the first time and ~20,000 plasmid copies/cell were obtained independently of cell passage. As transfected MSC have shown high viabilities (>90%) and recoveries (>52%) while maintaining their multipotency, this might be an advantageous transfection strategy when the goal is to express a therapeutic gene in a safe and transient way.
Collapse
|
11
|
Wang W, Li B, Li Y, Jiang Y, Ouyang H, Gao C. In vivo restoration of full-thickness cartilage defects by poly(lactide-co-glycolide) sponges filled with fibrin gel, bone marrow mesenchymal stem cells and DNA complexes. Biomaterials 2010; 31:5953-65. [PMID: 20488531 DOI: 10.1016/j.biomaterials.2010.04.029] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 04/14/2010] [Indexed: 10/19/2022]
Abstract
A composite construct comprising of bone marrow mesenchymal stem cells (BMSCs), plasmid DNA encoding transforming growth factor-beta1 (pDNA-TGF-beta1), fibrin gel and poly (lactide-co-glycolide) (PLGA) sponge was designed and employed to repair articular cartilage defects. To improve the gene transfection efficiency, a cationized chitosan derivative N,N,N-trimethyl chitosan chloride (TMC) was employed as the vector. The TMC/DNA complexes had a transfection efficiency of 9% to BMSCs and showed heterogeneous TGF-beta1 expression in a 10-day culture period in vitro. In vivo culture of the composite constructs was performed by implantation into full-thickness cartilage defects of New Zealand white rabbit joints, using the constructs absence of pDNA-TGF-beta1 or BMSCs as controls. Heterogeneous expression of TGF-beta1 in vivo was detected at 4 weeks, but its level was decreased in comparison with that of 2 weeks. After implantation for 12 weeks, the cartilage defects were successfully repaired by the composite constructs of the experimental group, and the neo-cartilage integrated well with its surrounding tissue and subchondral bone. Immunohistochemical and glycosaminoglycans (GAGs) staining confirmed the similar amount and distribution of collagen type II and GAGs in the regenerated cartilage as that of hyaline cartilage. The cartilage special genes expressed in the neo-tissue were closer to those of the normal cartilage. An overall score of 2.83 was obtained according to Wakitani's standard. By contrast, only part of the defects was repaired by the pDNA-TGF-beta1 absence constructs, and no cartilage repair but fibrous tissue was found for the BMSCs absence constructs. Therefore, combination of the PLGA sponge/fibrin gel scaffold with BMSCs and gene therapy is an effective method to restore cartilage defects and may have a great potential for practical applications in the near future.
Collapse
Affiliation(s)
- Wei Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | | | | | | | | | | |
Collapse
|
12
|
Zhi ZL, Liu B, Jones PM, Pickup JC. Polysaccharide Multilayer Nanoencapsulation of Insulin-Producing β-Cells Grown as Pseudoislets for Potential Cellular Delivery of Insulin. Biomacromolecules 2010; 11:610-6. [DOI: 10.1021/bm901152k] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Zheng-liang Zhi
- Metabolic Unit, King’s College London School of Medicine, Guy’s Hospital, London, United Kingdom, and Diabetes Research Group, School of Biomedical and Health Sciences, King’s College London, Guy’s Campus, London, United Kingdom
| | - Bo Liu
- Metabolic Unit, King’s College London School of Medicine, Guy’s Hospital, London, United Kingdom, and Diabetes Research Group, School of Biomedical and Health Sciences, King’s College London, Guy’s Campus, London, United Kingdom
| | - Peter M Jones
- Metabolic Unit, King’s College London School of Medicine, Guy’s Hospital, London, United Kingdom, and Diabetes Research Group, School of Biomedical and Health Sciences, King’s College London, Guy’s Campus, London, United Kingdom
| | - John C Pickup
- Metabolic Unit, King’s College London School of Medicine, Guy’s Hospital, London, United Kingdom, and Diabetes Research Group, School of Biomedical and Health Sciences, King’s College London, Guy’s Campus, London, United Kingdom
| |
Collapse
|
13
|
Park JS, Na K, Woo DG, Yang HN, Kim JM, Kim JH, Chung HM, Park KH. Non-viral gene delivery of DNA polyplexed with nanoparticles transfected into human mesenchymal stem cells. Biomaterials 2009; 31:124-32. [PMID: 19818490 DOI: 10.1016/j.biomaterials.2009.09.023] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Accepted: 09/07/2009] [Indexed: 10/20/2022]
Abstract
Human mesenchymal stem cells (hMSCs) represent a potent target for gene delivery for both stem cell differentiation applications and clinical therapies. However, it has, thus far, proven difficult to develop delivery vehicles that increase the efficiency of gene delivery to hMSCs, due to several problematic issues. We have evaluated different vehicles with regard to the efficiency with which they deliver hMSCs and enhance the ability to deliver a reporter gene. In this study, a non-viral gene delivery system using nanoparticles was designed, with emphasis placed on the ability of the system to mediate high levels of gene expression into stem cells. Via polyplexing with polyethylenimine (PEI), the cell-uptake ability of the nanoparticles was enhanced for both in vitro and in vivo culture systems. In experiments with PEI/pNDA polyplexed with nanoparticles, the expression of green fluorescent protein (GFP) with this vehicle was noted in up to 75% of hMSCs 2 days after transfection, and GFP gene expression was detected via Western blotting, flow cytometric analysis, and immunofluorescence using a confocal laser microscope after transfection.
Collapse
Affiliation(s)
- Ji S Park
- Department of Biomedical Science, College of Life Science, CHA University 606-16, Yeoksam 1-dong, Kangnam-gu, Seoul 135-081, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Tissue engineering is a newly emerging biomedical technology and methodology to assist and accelerate the regeneration and repairing of defective and damaged tissues based on the natural healing potentials of patients themselves. For the new therapeutic strategy, it is indispensable to provide cells with a local environment that enhances and regulates their proliferation and differentiation for cell-based tissue regeneration. Biomaterial technology plays an important role in the creation of this cell environment. For example, the biomaterial scaffolds and the drug delivery system (DDS) of biosignalling molecules have been investigated to enhance the proliferation and differentiation of cell potential for tissue regeneration. In addition, the scaffold and DDS technologies contribute to develop the basic research of stem cell biology and medicine as well as obtain a large number of cells with a high quality for cell transplantation therapy. A technology to genetically engineer cells for their functional manipulation is also useful for cell research and therapy. Several examples of tissue engineering applications with the cell scaffold and DDS of growth factors and genes are introduced to emphasize the significance of biomaterial technology in new therapeutic and research fields.
Collapse
Affiliation(s)
- Yasuhiko Tabata
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|