Journal of Stem Cells Research Development & Therapy Category: Medical Type: Short Review

Genetic Engineering of Mesenchymal Stem Cells to Improve Therapeutic Effects

Young Woo Eom#1, Yongdae Yoon#2, Joon Hyung Sohn3 and Soon Koo Baik4*
1 Cell therapy and tissue engineering center, Yonsei University Wonju College of Medicine, Wonju, Republic of korea
2 Regeneration medicine research center, Yonsei University Wonju College of Medicine, Wonju, Republic of korea
3 Central research laboratory, Yonsei University Wonju College of Medicine, Wonju, Republic of korea
4 Department of internal medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, 26426, Republic of korea

*Corresponding Author(s):
Soon Koo Baik
Department Of Internal Medicine, Yonsei University Wonju College Of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, 26426, Republic Of Korea
Tel:+82 337411223,
Fax:+82 337456782
Email:baiksk@yonsei.ac.kr
#Equal Contribution

Received Date: Sep 07, 2020
Accepted Date: Sep 10, 2020
Published Date: Sep 17, 2020

Abstract

Based on the therapeutic potential of MSCs, including homing to damaged sites, trans-differentiation, secretion of trophic factors, and immunomodulation, approximately 1,100 clinical studies have been registered in the ClinicalTrials.Gov, and several drugs using MSCs have been approved worldwide. However, despite their therapeutic potential, MSCs have not yet shown sufficient therapeutic effects in humans. Therefore, in order to increase the therapeutic potential of MSCs, methods such as MSC priming, genetic modification, Three-Dimensional (3-D) culture, and MSC-derived exosomes are being studied intensively. Among them, genetic modification increases the expression of therapeutic genes, leading to increased homing of MSCs to the damaged sites, increased engraftment rates, and increased survival durations of transplanted MSCs. It has been reported that genetically engineered MSCs can greatly increase their therapeutic effects. This review aims to provide an overview of the method of target gene delivery to MSCs and discuss the advantages and disadvantages of each method.

Keywords

Genetic modification; Mesenchymal stem cells; Non-viral vector; Viral vector

INTRODUCTION

Mesenchymal Stem Cells (MSCs) are multipotent stromal cells that can be isolated from various tissues, including bone marrow, adipose tissue, dental pulp, amniotic fluid, and umbilical cord [1-5]. According to the International Society for Cellular Therapy (ISCT), MSCs are defined as cells that can adhere to plastic, express CD73, CD90, and CD105 as cell surface antigens (≥ 95% positive), and differentiate into adipocytes, chondroblasts, and osteoblasts under in vitro differentiation conditions [6]. Potential therapeutic mechanisms of MSCs for the regenerative treatment of incurable diseases have been reported. First, MSCs possess a homing property, allowing them to adhere to damaged and tumor sites [7,8]. The homing effect of MSCs theoretically implies that, in clinical applications, MSCs can be delivered to the damaged area for injury repair using only intravascular transplantation of MSCs and not surgery. Second, although it has been reported that the ratio of differentiated cells to transplanted cells is very low [9], MSCs can directly differentiate into damaged cells, facilitating repair [10]. Third, MSCs have the ability to regulate immune responses [11-13] and can promote the regeneration of damaged tissues by regulating the activity of immune cells [14-16]. Fourth, it has been reported that MSCs can express various trophic factors, which can inhibit the activity of immune cells, inhibitor delay cell death in damaged sites, and promote progenitor/stem cell proliferation and differentiation into target cells [11,12,14,16-18]. Finally, MSCs are known to be hypoimmunogenic or immune-privileged, which allows allogeneic MSC transplantation across major histocompatibility barriers and the creation of off-the-shelf therapies consisting of MSCs grown in culture [19].

Based on the therapeutic potential of MSCs, more than 1,100 clinical trials using MSCs have been registered for various diseases (https://clinicaltrials.gov). However, despite the progress in basic and clinical studies using MSCs, MSC treatment has not yet shown sufficient therapeutic effects in humans. Therefore, to improve the therapeutic potential of MSCs, MSC priming [20-22], genetic modification [23-27], Three-Dimensional (3-D) culture [28-30], and MSC-derived exosomes [31-34] have been studied. Once delivered to the damaged site, MSCs release various factors that regulate the activity of inflammatory cells after exposure to inflammatory cytokines; this is followed by treatment of the damaged area. Therefore, the therapeutic effect of MSCs can be improved by pre-exposing them to inflammatory cytokines, such as IFN-g, TNF-a and IL-1b [22]. Three-dimensional culture of stem cells using various scaffolds has been reported to increase the proliferation and differentiation efficiencies of stem cells [28,30] and enhance their therapeutic effects in liver disease, peritonitis, kidney injury, and myocardial infarction [35-37]. Since various trophic factors secreted by MSCs exhibit therapeutic effects, their regenerative therapeutic effects can be increased by utilizing the MSC-derived secretome or exosomes. Since exosomes can be stored, controlled qualitatively, and administered repeatedly, they are an optimal factor that can be used for the treatment of acute diseases. One way to reliably improve the therapeutic effect of MSCs is to increase the expression of the target gene, which plays an important role in tissue regeneration. Therefore, in this review, we will discuss gene delivery methods into MSCs, which are known to have low transformation efficiencies, and discuss the production of functionally enhanced MSCs and their therapeutic efficacy.

GENE DELIVERY INTO MSCS

The ability of MSCs to home to the damaged and/or tumor site makes it possible to use MSCs as a vehicle for various therapeutic agents, including genes. The method of gene delivery to MSCs depends on whether viral or non-viral vector systems are used [38]. Retrovirus, lentivirus, adenovirus, and adeno-associated virus have been extensively used as viral vectors for gene delivery into MSCs [38-40]. In non-viral vector systems, single or combinations of cationic lipids, surfactants, peptides, polysaccharides, metals (gold, magnetic iron), and synthetic polymers have been used for genetic manipulation [41-43].

Adenoviral vectors 

Adenoviruses are non-enveloped viruses with icosahedral nucleocapsids, containing a double-stranded DNA genome. Adenoviruses are the most commonly used gene delivery vector because they have a wide host range and can infect both dividing and non-dividing cells [44,45]. The efficiency of gene delivery by adenoviruses is closely associated with the expression of Coxsackievirus and Adenovirus Receptors (CARs) on target cells [46]. Since MSCs express a very low level of CARs [47], gene delivery efficiency using adenovirus vectors is very low. To improve the efficiency of gene delivery by adenoviruses into MSCs, a capsid- and a fiber-modified adenovirus has been developed [46,48,49]. In addition, the initial robust expression of the newly introduced gene gradually declines in bone marrow-derived MSCs after 21 days; thus, this strategy could only be applied for the transient expression of target genes [50]. However, adenoviruses have high immunogenicity, which limits their use in gene therapy.

Lentiviral vectors 

Lentiviruses are a genus of retroviruses that contain a single-stranded RNA genome. After entering cells, lentiviral RNA is reverse-transcribed into double-stranded DNA, which can be integrated into the host genome, leading to insertional mutagenesis. Recently, non-integrating lentivirus vectors have been generated through alterations in the viral integrase or long terminal repeats and have been used for stable and safe gene delivery, resulting in long-term expression of the transgene [51]. Lentiviruses are one of the most widely used vectors in MSC-based gene therapy and have benefits such as a large genome size, high infection efficiency, and stable gene transfer [39,49]. In addition, lentiviruses can be transduced into non-dividing cells and persist for several generations. MSCs engineered with HSP70 by a lentiviral vector improved the survival and resistance to apoptosis in hypoxic and ischemic conditions without affecting the morphology, viability, or differentiation abilities of MSCs [52]. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-overexpressing MSCs induced apoptosis in cancer cell lines, including lung, colon, pleural mesothelioma and oral squamous cancer [53].

Retroviral vectors 

Retroviruses have a lipid envelope and a double-stranded RNA genome, which is reverse-transcribed to DNA that integrates into the host genome, resulting in insertional mutations. Despite the high tropism of retroviruses to host cells, there are many difficulties in using them for gene therapy, such as the absence of long-term transgene expression, ineffective transduction of MSCs, induction of insertional mutagenesis, and the requirement for administering high loads of vectors in several rounds to transduce host cells [39,46 49].

Adeno-associated virus-based vectors

Adeno-Associated Viruses (AAV) are considered an attractive gene therapy vector for the following reasons: despite their ubiquity in the human population, they have no association with any disease; most human tissues can be infected with AAV; AAV vectors are not capable of replication without a helper adenovirus; AAVs exist in an episomal form for long-term transgene expression; and AAV vectors have been shown to be nontoxic in clinical trials in humans [54-56]. However, the clinical applicability of AAV vectors has been limited due to their low transduction efficiency in MSCs [46,49].

Non-viral vectors

Plasmids, which are non-viral vectors, have been considered as another suitable candidate for gene delivery into MSCs because they can be easily produced and have low immunogenicity [46,57,58]. Unlike viral vectors, conventional transfection methods such as lipofection, magnetofection, and electroporation are combined to deliver non-viral vectors into host cells. However, the efficiency of gene delivery into MSCs is very low compared to that of viral vectors. Moreover, transfection reagents and/or procedures can increase the cytotoxicity of MSCs, leading to cell death or senescence [58-62]. Recently, Helledie et al., reported that electroporation is a superior gene delivery method for lipofection in MSCs without causing loss of proliferation and differentiation potentials, while lipofection with Lipofectamine2000 decreased proliferation rate and increased cell death in MSCs [61]. They described a simple and reliable electroporation protocol that resulted in a transfection efficiency of up to 90% compared to most viral methods, but the absolute transfection efficiency was approximately 35%. Recently, a novel method for efficient gene delivery into MSCs has been developed based on Therapeutic Ultrasound (TUS) [63]. MSCs were transfected with plasmids encoding hemopexin-like domain fragment (PEX), an inhibitor of angiogenesis, using low intensity and moderate frequency TUS. MSCs transfected with TUS-mediated PEX expressed biologically active PEX without loss of stemness and homing capabilities and subsequently inhibited 70% of prostate tumor growth in a mouse model [63].

CONCLUSION

The homing effect of MSCs to the damaged and/or tumor site makes it possible to use MSCs as a transport vesicle for various therapeutic agents, including genes. MSCs equipped with these therapeutic agents not only have important therapeutic effects, but also act predominantly in only the damaged site, reducing the expected frequency of side effects resulting from the nonselective action of the drug. For gene delivery into MSCs, viral and non-viral vectors have been studied, and genetically engineered MSCs have been reported to significantly improve their therapeutic effects in regenerative medicine and cancer treatment. Viral vectors have disadvantages such as high immunogenicity and insertional mutagenesis, but have the advantages of high transfection efficiency and long-term gene expression. Conversely, gene delivery with non-viral vectors has a low transfection efficiency and transient expression of target genes. Therefore, based on the disease being treated, different types of vectors must be used to suit the therapeutic purpose. Further, new methods must be developed to make use of the advantages of each vector and to compensate for the disadvantages. If such research is conducted in the future, it is expected that not only will the therapeutic effect of MSCs be enhanced, but the application of MSCs to various diseases can greatly improve the quality of life of patients.

FUNDING

This work was supported by a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (HI17C1365), and the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Korean government, the Ministry of Education (NRF-2017R1D1A1A02019212).

COMPETING INTEREST

The authors declare that they have no competing interests.

REFERENCES

  1. Bunnell BA, Flaat M, Gagliardi C, Patel B, Ripoll C (2008) Adipose-derived stem cells: Isolation, expansion and differentiation. Methods 45: 115-120.
  2. Gregory CA, Prockop DJ, Spees JL (2005) Non-hematopoietic bone marrow stem cells: molecular control of expansion and differentiation. Exp Cell Res 306: 330-335.
  3. Liu J, Yu F, Sun Y, Jiang B, Zhang W, et al. (2015) Concise reviews: Characteristics and potential applications of human dental tissue-derived mesenchymal stem cells. Stem Cells 33: 627-638.
  4. Nagamura-Inoue T, He H (2014) Umbilical cord-derived mesenchymal stem cells: Their advantages and potential clinical utility. World J Stem Cells 6: 195-202.
  5. Spitzhorn LS, Rahman MS, Schwindt L, Ho HT, Wruck W, et al. (2017) Isolation and Molecular Characterization of Amniotic Fluid-Derived Mesenchymal Stem Cells Obtained from Caesarean Sections. Stem Cells Int 2017: 5932706.
  6. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, et al. (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8: 315-317.
  7. Chapel A, Bertho JM, Bensidhoum M, Fouillard L, Young RG, et al. (2003) Mesenchymal stem cells home to injured tissues when co-infused with hematopoietic cells to treat a radiation-induced multi-organ failure syndrome. J Gene Med 5: 1028-1038.
  8. Sordi V, Malosio ML, Marchesi F, Mercalli A, Melzi R, et al. (2005) Bone marrow mesenchymal stem cells express a restricted set of functionally active chemokine receptors capable of promoting migration to pancreatic islets. Blood 106: 419-427.
  9. Xagorari A, Siotou E, Yiangou M, Tsolaki E, Bougiouklis D, et al. (2013) Protective effect of mesenchymal stem cell-conditioned medium on hepatic cell apoptosis after acute liver injury. Int J Clin Exp Pathol 6: 831-840.
  10. Sato Y, Araki H, Kato J, Nakamura K, Kawano Y, et al. (2005) Human mesenchymal stem cells xenografted directly to rat liver are differentiated into human hepatocytes without fusion. Blood 106: 756-763.
  11. Ghannam S, Bouffi C, Djouad F, Jorgensen C, Noel D (2010) Immunosuppression by mesenchymal stem cells: mechanisms and clinical applications. Stem Cell Res Ther 1: 2.
  12. Sato K, Ozaki K, Oh I, Meguro A, Hatanaka K, et al. (2007) Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood 109: 228-234.
  13. Spaggiari GM, Abdelrazik H, Becchetti F, Moretta L (2009) MSCs inhibit monocyte-derived DC maturation and function by selectively interfering with the generation of immature DCs: central role of MSC-derived prostaglandin E2. Blood 113 :6576-6583.
  14. Jo H, Eom YW, Kim HS, Park HJ, Kim HM, et al. (2018) Regulatory Dendritic Cells Induced by Mesenchymal Stem Cells Ameliorate Dextran Sodium Sulfate-Induced Chronic Colitis in Mice. Gut Liver 12: 664-673.
  15. Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, et al. (2009) Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med 15: 42-49.
  16. Park HJ, Kim J, Saima FT, Rhee KJ, Hwang S, et al. (2018) Adipose-derived stem cells ameliorate colitis by suppression of inflammasome formation and regulation of M1-macrophage population through prostaglandin E2. Biochem Biophys Res Commun 498: 988-995.
  17. Caplan AI, Dennis JE (2006) Mesenchymal stem cells as trophic mediators. J Cell Biochem 98:1076-1084.
  18. Kupcova Skalnikova H (2013) Proteomic techniques for characterisation of mesenchymal stem cell secretome. Biochimie 95: 2196-2211.
  19. Wang D, Zhang H, Liang J, Li X, Feng X, et al. (2013) Allogeneic mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus: 4 years of experience. Cell Transplant 22: 2267-2277.
  20. Giuliani M, Bennaceur-Griscelli A, Nanbakhsh A, Oudrhiri N, Chouaib S, et al. (2014) TLR ligands stimulation protects MSC from NK killing. Stem Cells 32: 290-300.
  21. Krampera M, Cosmi L, Angeli R, Pasini A, Liotta F, et al. (2006) Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells 24: 386-398.
  22. Sharma RR, Pollock K, Hubel A, McKenna D (2014) Mesenchymal stem or stromal cells: a review of clinical applications and manufacturing practices. Transfusion 54: 1418-1437.
  23. Du Z, Wei C, Yan J, Han B, Zhang M, et al. (2013) Mesenchymal stem cells overexpressing C-X-C chemokine receptor type 4 improve early liver regeneration of small-for-size liver grafts. Liver Transpl 19: 215-225.
  24. Herberg S, Shi X, Johnson MH, Hamrick MW, Isales CM, et al. (2013) Stromal cell-derived factor-1beta mediates cell survival through enhancing autophagy in bone marrow-derived mesenchymal stem cells. PLoS One 8: e58207.
  25. Jang YO, Cho MY, Yun CO, Baik SK, Park KS, et al. (2016) Effect of Function-Enhanced Mesenchymal Stem Cells Infected With Decorin-Expressing Adenovirus on Hepatic Fibrosis. Stem Cells Transl Med 5: 1247-1256.
  26. Mirotsou M, Zhang Z, Deb A, Zhang L, Gnecchi M, et al. (2007) Secreted frizzled related protein 2 (Sfrp2) is the key Akt-mesenchymal stem cell-released paracrine factor mediating myocardial survival and repair. Proc Natl Acad Sci U S A 104: 1643-1648.
  27. Sun C, Li DG, Chen YW, Chen YW, Wang BC, et al. (2008) Transplantation of urokinase-type plasminogen activator gene-modified bone marrow-derived liver stem cells reduces liver fibrosis in rats. J Gene Med 10: 855-866.
  28. Arufe MC, De la Fuente A, Fuentes-Boquete I, De Toro FJ, Blanco FJ (2009) Differentiation of synovial CD-105(+) human mesenchymal stem cells into chondrocyte-like cells through spheroid formation. J Cell Biochem 108: 145-155.
  29. Ji R, Zhang N, You N, Li Q, Liu W, et al. (2012) The differentiation of MSCs into functional hepatocyte-like cells in a liver biomatrix scaffold and their transplantation into liver-fibrotic mice. Biomaterials 33: 8995-9008.
  30. Zhang X, Hu MG, Pan K, Li CH, Liu R (2016) 3D Spheroid Culture Enhances the Expression of Antifibrotic Factors in Human Adipose-Derived MSCs and Improves Their Therapeutic Effects on Hepatic Fibrosis. Stem Cells Int 2016: 4626073.
  31. Chen TS, Lai RC, Lee MM, Choo AB, Lee CN, et al. (2010) Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs. Nucleic Acids Res 38: 215-224.
  32. Gnecchi M, He H, Liang OD, Melo LG, Morello F, et al. (2005) Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med 11: 367-368.
  33. Gnecchi M, Danieli P, Malpasso G, Ciuffreda MC (2016) Paracrine Mechanisms of Mesenchymal Stem Cells in Tissue Repair. Methods Mol Biol 1416: 123-146.
  34. Lai RC, Tan SS, Teh BJ, Sze SK, Arslan F, et al. (2012) Proteolytic Potential of the MSC Exosome Proteome: Implications for an Exosome-Mediated Delivery of Therapeutic Proteasome. Int J Proteomics 2012: 971907.
  35. Bartosh TJ, Ylostalo JH, Mohammadipoor A, Bazhanov N, Coble K, et al. (2010) Aggregation of human mesenchymal stromal cells (MSCs) into 3D spheroids enhances their antiinflammatory properties. Proc Natl Acad Sci USA 107: 13724-13729.
  36. Wang CC, Chen CH, Hwang SM, Lin WW, Huang CH, et al. (2009) Spherically symmetric mesenchymal stromal cell bodies inherent with endogenous extracellular matrices for cellular cardiomyoplasty. Stem Cells 27: 724-732.
  37. Zhao X, Qiu X, Zhang Y, Zhang S, Gu X, Guo H (2016) Three-Dimensional Aggregates Enhance the Therapeutic Effects of Adipose Mesenchymal Stem Cells for Ischemia-Reperfusion Induced Kidney Injury in Rats. Stem Cells Int 2016: 9062638.
  38. Marofi F, Vahedi G, Biglari A, Esmaeilzadeh A, Athari SS (2017) Mesenchymal Stromal/Stem Cells: A New Era in the Cell-Based Targeted Gene Therapy of Cancer. Front Immunol 8: 1770.
  39. Nowakowski A, Andrzejewska A, Janowski M, Walczak P, Lukomska B (2013) Genetic engineering of stem cells for enhanced therapy. Acta Neurobiol Exp (Wars) 73: 1-18.
  40. Oggu GS, Sasikumar S, Reddy N, Ella KKR, Rao CM, et al. (2017) Gene Delivery Approaches for Mesenchymal Stem Cell Therapy: Strategies to Increase Efficiency and Specificity. Stem Cell Rev Rep 13: 725-740.
  41. Foldvari M, Chen DW, Nafissi N, Calderon D, Narsineni L, et al. (2016) Non-viral gene therapy: Gains and challenges of non-invasive administration methods. J Control Release 240: 165-190.
  42. Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, et al. (2007) Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol 2: 751-760.
  43. Tsutsui JM, Xie F, Porter RT (2004) The use of microbubbles to target drug delivery. Cardiovasc Ultrasound 2: 23.
  44. Graham FL, Prevec L (1991) Manipulation of adenovirus vectors. Methods Mol Biol 7: 109-128.
  45. Trapnell BC, Gorziglia M (1994) Gene therapy using adenoviral vectors. Curr Opin Biotechnol 5: 617-625.
  46. Chira S, Jackson CS, Oprea I, Ozturk F, Pepper MS, et al. (2015) Progresses towards safe and efficient gene therapy vectors. Oncotarget 6: 30675-30703.
  47. Suzuki T, Kawamura K, Li Q, Okamoto S, Tada Y, et al. (2014) Mesenchymal stem cells are efficiently transduced with adenoviruses bearing type 35-derived fibers and the transduced cells with the IL-28A gene produces cytotoxicity to lung carcinoma cells co-cultured. BMC Cancer 14: 713.
  48. Hammer K, Kazcorowski A, Liu L, Behr M, Schemmer P, et al. (2015) Engineered adenoviruses combine enhanced oncolysis with improved virus production by mesenchymal stromal carrier cells. Int J Cancer 137: 978-990.
  49. Nayerossadat N, Maedeh T, Ali PA (2012) Viral and nonviral delivery systems for gene delivery. Adv Biomed Res 1: 27.
  50. Knaan-Shanzer S, van de Watering MJ, van der Velde I, Goncalves MA, Valerio D, et al. (2005) Endowing human adenovirus serotype 5 vectors with fiber domains of species B greatly enhances gene transfer into human mesenchymal stem cells. Stem Cells 23: 1598-1607.
  51. Shaw A, Cornetta K (2014) Design and Potential of Non-Integrating Lentiviral Vectors. Biomedicines 2: 14-35.
  52. McGinley L, McMahon J, Strappe P, Barry F, Murphy M, et al. (2011) Lentiviral vector mediated modification of mesenchymal stem cells & enhanced survival in an in vitro model of ischaemia. Stem Cell Res Ther 2: 12.
  53. Yuan ZQ, Kolluri KK, Sage EK, Gowers KH, Janes SM (2015) Mesenchymal stromal cell delivery of full-length tumor necrosis factor-related apoptosis-inducing ligand is superior to soluble type for cancer therapy. Cytotherapy 17: 885-896.
  54. Brown N, Song L, Kollu NR, Hirsch ML (2017) Adeno-Associated Virus Vectors and Stem Cells: Friends or Foes? Hum Gene Ther 28: 450-463.
  55. Samulski RJ, Muzyczka N (2014) AAV-Mediated Gene Therapy for Research and Therapeutic Purposes. Annu Rev Virol 1: 427-451.
  56. Xiao X, Li J, Samulski RJ (1996) Efficient long-term gene transfer into muscle tissue of immunocompetent mice by adeno-associated virus vector. J Virol 70: 8098-8108.
  57. Feng B, Chen L (2009) Review of mesenchymal stem cells and tumors: executioner or coconspirator? Cancer Biother Radiopharm 24: 717-721.
  58. Hu YL, Fu YH, Tabata Y, Gao JQ (2010) Mesenchymal stem cells: A promising targeted-delivery vehicle in cancer gene therapy. J Control Release 147: 154-162.
  59. Aluigi M, Fogli M, Curti A, Isidori A, Gruppioni E, et al. (2006) Nucleofection is an efficient nonviral transfection technique for human bone marrow-derived mesenchymal stem cells. Stem Cells 24: 454-461.
  60. Haleem-Smith H, Derfoul A, Okafor C, Tuli R, Olsen D, et al. (2005) Optimization of high-efficiency transfection of adult human mesenchymal stem cells in vitro. Mol Biotechnol 30: 9-20.
  61. Helledie T, Nurcombe V, Cool SM (2008) A simple and reliable electroporation method for human bone marrow mesenchymal stem cells. Stem Cells Dev 17: 837-848.
  62. Uchida E, Mizuguchi H, Ishii-Watabe A, Hayakawa T (2002) Comparison of the efficiency and safety of non-viral vector-mediated gene transfer into a wide range of human cells. Biol Pharm Bull 25: 891-897.
  63. Haber T, Baruch L, Machluf M (2017) Ultrasound-Mediated Mesenchymal Stem Cells Transfection as a Targeted Cancer Therapy Platform. Sci Rep 7: 42046.

Citation: Eom YW, Yoon Y, Sohn JH, Baik SK (2020) Genetic Engineering of Mesenchymal Stem Cells to Improve Therapeutic Effects. J Stem Cell Res Dev Ther 6: 049.

Copyright: © 2020  Young Woo Eom#, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.


Herald Scholarly Open Access is a leading, internationally publishing house in the fields of Sciences. Our mission is to provide an access to knowledge globally.



© 2024, Copyrights Herald Scholarly Open Access. All Rights Reserved!