1
|
Kim S, Lee EW, Oh DB, Seo J. BAP1 controls mesenchymal stem cell migration by inhibiting the ERK signaling pathway. BMB Rep 2024; 57:250-255. [PMID: 37964637 PMCID: PMC11139679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/19/2023] [Accepted: 11/09/2023] [Indexed: 11/16/2023] Open
Abstract
Due to their stem-like characteristics and immunosuppressive properties, Mesenchymal stem cells (MSCs) offer remarkable potential in regenerative medicine. Much effort has been devoted to enhancing the efficacy of MSC therapy by enhancing MSC migration. In this study, we identified deubiquitinase BRCA1- associated protein 1 (BAP1) as an inhibitor of MSC migration. Using deubiquitinase siRNA library screening based on an in vitro wound healing assay, we found that silencing BAP1 significantly augmented MSC migration. Conversely, BAP1 overexpression reduced the migration and invasion capabilities of MSCs. BAP1 depletion in MSCs upregulates ERK phosphorylation, thereby increasing the expression of the migration factor, osteopontin. Further examination revealed that BAP1 interacts with phosphorylated ERK1/2, deubiquitinating their ubiquitins, and thus attenuating the ERK signaling pathway. Overall, our study highlights the critical role of BAP1 in regulating MSC migration through its deubiquitinase activity, and suggests a novel approach to improve the therapeutic potential of MSCs in regenerative medicine. [BMB Reports 2024; 57(5): 250-255].
Collapse
Affiliation(s)
- Seobin Kim
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Biosystems and Bioengineering, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Eun-Woo Lee
- Metabolic Disease Research Center, KRIBB, Daejeon 34141, Korea
- Department of Functional Genomics, UST, Daejeon 34113, Korea
| | - Doo-Byoung Oh
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Biosystems and Bioengineering, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Jinho Seo
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Biosystems and Bioengineering, University of Science and Technology (UST), Daejeon 34113, Korea
| |
Collapse
|
2
|
Ji W, Wang W, Li P, Liu Y, Zhang B, Qi F. sFgl2 gene-modified MSCs regulate the differentiation of CD4 + T cells in the treatment of autoimmune hepatitis. Stem Cell Res Ther 2023; 14:316. [PMID: 37924141 PMCID: PMC10625288 DOI: 10.1186/s13287-023-03550-x] [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: 10/18/2022] [Accepted: 10/27/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Autoimmune hepatitis (AIH) is a T-cell-mediated autoimmune liver disease that can lead to liver injury and has a poor long-term prognosis. Mesenchymal stromal cells (MSCs) have immunosuppressive effects and can treat AIH. CD4+ T cells express the unique inhibitory Fcγ receptor (FcγRIIB), which is the only receptor for the immunosuppressive factor soluble fibrinogen-like protein 2 (sFgl2). This study aimed to examine the therapeutic effect of sFgl2 gene-modified MSCs (sFgl2-MSCs) on AIH. METHODS MSCs were obtained from the inguinal fat of mice and cocultured with CD4+ T cells sorted from mouse spleens. FcγRIIB expression on CD4+ T cells was determined by flow cytometry. sFgl2 expression in MSCs transfected with lentiviral vectors carrying the Fgl2 gene and a green fluorescent protein-encoding sequence was determined by enzyme-linked immunosorbent assay. The percentages of Th1 cells Th17 cells and regulatory T cells (Tregs) were determined by flow cytometry And the levels of p-SHP2 and p-SMAD2/3 were detected by Western blotting after the cells were cocultured with MSCs for 72 h. After locating MSCs by in vivo imaging Con A-induced experimental AIH mice were randomly divided into 4 groups and administered different treatments. After 24 h histopathological scores liver function and cytokine levels were examined and the proportions of CD4+ T cells CD8+ T cells Tregs Th17 cells and Th1 cells in the spleen and liver were determined by flow cytometry. In addition immunohistochemical staining was used to detect the liver infiltration of T-bet-, Foxp3- and RORγ-positive cells. RESULTS FcγRIIB expression on CD4+ T cells was upregulated after coculture with MSCs. After coculture with sFgl2-MSCs, the proportion of Tregs among CD4+ T cells increased, the proportion of Th17 and Th1 cells decreased, and the levels of p-SHP2 and p-SMAD2/3 increased. In vivo, sFgl2-MSCs significantly improved liver function, decreased liver necrosis area, decreased tumor necrosis factor-α, interleukin (IL)-1β and IL-6 expression, increased IL-10 expression, reduced liver infiltration of CD4+ T and CD8+ T cells, increased the proportion of Tregs and reduced the proportions of Th17 and Th1 cells in mice. CONCLUSION By promoting Tregs differentiation and inhibiting Th17 and Th1 cell differentiation, sFgl2 gene-modified MSCs have a more powerful therapeutic effect on Con A-induced experimental AIH and may represent a strategy for the clinical treatment of AIH.
Collapse
Affiliation(s)
- Wenbin Ji
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Weiwei Wang
- Department of General Surgery, Tianjin Medical University Baodi Clinical College, Guangchuan Road, Baodi, Tianjin, 301800, China
| | - Peiyuan Li
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Yanhong Liu
- Department of General Surgery, Tianjin Union Medical Center, Tianjin, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Baotong Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Feng Qi
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
3
|
Kováč J, Priščáková P, Gbelcová H, Heydari A, Žiaran S. Bioadhesive and Injectable Hydrogels and Their Correlation with Mesenchymal Stem Cells Differentiation for Cartilage Repair: A Mini-Review. Polymers (Basel) 2023; 15:4228. [PMID: 37959908 PMCID: PMC10648146 DOI: 10.3390/polym15214228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/06/2023] [Accepted: 10/17/2023] [Indexed: 11/15/2023] Open
Abstract
Injectable bioadhesive hydrogels, known for their capacity to carry substances and adaptability in processing, offer great potential across various biomedical applications. They are especially promising in minimally invasive stem cell-based therapies for treating cartilage damage. This approach harnesses readily available mesenchymal stem cells (MSCs) to differentiate into chondrocytes for cartilage regeneration. In this review, we investigate the relationship between bioadhesion and MSC differentiation. We summarize the fundamental principles of bioadhesion and discuss recent trends in bioadhesive hydrogels. Furthermore, we highlight their specific applications in conjunction with stem cells, particularly in the context of cartilage repair. The review also encompasses a discussion on testing methods for bioadhesive hydrogels and direct techniques for differentiating MSCs into hyaline cartilage chondrocytes. These approaches are explored within both clinical and laboratory settings, including the use of genetic tools. While this review offers valuable insights into the interconnected aspects of these topics, it underscores the need for further research to fully grasp the complexities of their relationship.
Collapse
Affiliation(s)
- Ján Kováč
- Medical Vision, Záhradnícka 55, 821 08 Bratislava, Slovakia; (J.K.); (P.P.); (H.G.); (A.H.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Petra Priščáková
- Medical Vision, Záhradnícka 55, 821 08 Bratislava, Slovakia; (J.K.); (P.P.); (H.G.); (A.H.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Helena Gbelcová
- Medical Vision, Záhradnícka 55, 821 08 Bratislava, Slovakia; (J.K.); (P.P.); (H.G.); (A.H.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Abolfazl Heydari
- Medical Vision, Záhradnícka 55, 821 08 Bratislava, Slovakia; (J.K.); (P.P.); (H.G.); (A.H.)
- Polymer Institute of the Slovak Academy of Sciences, Dúbravská Cesta 9, 845 41 Bratislava, Slovakia
| | - Stanislav Žiaran
- Medical Vision, Záhradnícka 55, 821 08 Bratislava, Slovakia; (J.K.); (P.P.); (H.G.); (A.H.)
- Department of Urology, Faculty of Medicine, Comenius University, Limbová 5, 833 05 Bratislava, Slovakia
| |
Collapse
|
4
|
Eom Y, Eom SY, Lee J, Hwang S, Won J, Kim H, Chung S, Kim HJ, Lee MY. Therapeutic Effects and Underlying Mechanism of SOCS-com Gene-Transfected ADMSCs in Pressure Ulcer Mouse Models. Cells 2023; 12:1840. [PMID: 37508509 PMCID: PMC10378383 DOI: 10.3390/cells12141840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Although the proportion of ulcer patients with medical problems among the elderly has increased with the extension of human life expectancy, treatment efficiency is drastically low, incurring substantial social costs. MSCs have independent regeneration potential, making them useful in clinical trials of difficult-to-treat diseases. In particular, ADMSCs are promising in the stem cell therapy industry as they can be obtained in vast amounts using non-invasive methods. Furthermore, studies are underway to enhance the regeneration potential of ADMSCs using cytokines, growth factors, and gene delivery to generate highly functional ADMSCs. In this study, key regulators of wound healing, SOCS-1, -3, and -5, were combined to maximize the regenerative potential of ADMSCs in pressure ulcer treatments. After transfecting SOCS-1, -3, -5, and SOCS-com into ADMSCs using a non-viral method, the expression of the inflammatory factors TNF-alpha, INF-gamma, and IL-10 was confirmed. ADMSCs transfected with SOCS-com showed decreased overall expression of inflammatory factors and increased expression of anti-inflammatory factors. Based on these results, we implanted ADMSCs transfected with SOCS-com into a pressure ulcer mouse model to observe their subsequent wound-healing effects. Notably, SOCS-com improved wound closure in ulcers, and reconstruction of the epidermis and dermis was observed. The healing mechanism of ADMSCs transfected with SOCS-com was examined by RNA sequencing. Gene analysis results confirmed that expression changes occurred in genes of key regulators of wound healing, such as chemokines, MMP-1, 9, CSF-2, and IL-33, and that such genetic changes enhanced wound healing in ulcers. Based on these results, we demonstrate the potential of ADMSCs transfected with SOCS-com as an ulcer treatment tool.
Collapse
Affiliation(s)
- Youngsic Eom
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Republic of Korea
| | - So Young Eom
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Jeonghwa Lee
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Saeyeon Hwang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 34943, Republic of Korea
| | - Jihee Won
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Hyunsoo Kim
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hye Joung Kim
- Institute of Chemical Engineering Convergence System, Korea University, Seoul 02841, Republic of Korea
| | - Mi-Young Lee
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Republic of Korea
| |
Collapse
|
5
|
Zhou H, He Y, Xiong W, Jing S, Duan X, Huang Z, Nahal GS, Peng Y, Li M, Zhu Y, Ye Q. MSC based gene delivery methods and strategies improve the therapeutic efficacy of neurological diseases. Bioact Mater 2023; 23:409-437. [PMCID: PMC9713256 DOI: 10.1016/j.bioactmat.2022.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/08/2022] [Accepted: 11/13/2022] [Indexed: 12/05/2022] Open
|
6
|
Potočnik T, Maček Lebar A, Kos Š, Reberšek M, Pirc E, Serša G, Miklavčič D. Effect of Experimental Electrical and Biological Parameters on Gene Transfer by Electroporation: A Systematic Review and Meta-Analysis. Pharmaceutics 2022; 14:pharmaceutics14122700. [PMID: 36559197 PMCID: PMC9786189 DOI: 10.3390/pharmaceutics14122700] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
The exact mechanisms of nucleic acid (NA) delivery with gene electrotransfer (GET) are still unknown, which represents a limitation for its broader use. Further, not knowing the effects that different experimental electrical and biological parameters have on GET additionally hinders GET optimization, resulting in the majority of research being performed using a trial-and-error approach. To explore the current state of knowledge, we conducted a systematic literature review of GET papers in in vitro conditions and performed meta-analyses of the reported GET efficiency. For now, there is no universal GET strategy that would be appropriate for all experimental aims. Apart from the availability of the required electroporation device and electrodes, the choice of an optimal GET approach depends on parameters such as the electroporation medium; type and origin of cells; and the size, concentration, promoter, and type of the NA to be transfected. Equally important are appropriate controls and the measurement or evaluation of the output pulses to allow a fair and unbiased evaluation of the experimental results. Since many experimental electrical and biological parameters can affect GET, it is important that all used parameters are adequately reported to enable the comparison of results, as well as potentially faster and more efficient experiment planning and optimization.
Collapse
Affiliation(s)
- Tjaša Potočnik
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, 1000 Ljubljana, Slovenia
| | - Alenka Maček Lebar
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, 1000 Ljubljana, Slovenia
| | - Špela Kos
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloška cesta 2, 1000 Ljubljana, Slovenia
| | - Matej Reberšek
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, 1000 Ljubljana, Slovenia
| | - Eva Pirc
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, 1000 Ljubljana, Slovenia
| | - Gregor Serša
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloška cesta 2, 1000 Ljubljana, Slovenia
| | - Damijan Miklavčič
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
7
|
Lin Y, Zhou HC, Chen N, Ren Y, Gao R, Li Q, Deng Y, Han X, Zhang X, Xiang AP, Guo B, Liu C, Ren J. Unveiling the improved targeting migration of mesenchymal stem cells with CXC chemokine receptor 3-modification using intravital NIR-II photoacoustic imaging. J Nanobiotechnology 2022; 20:307. [PMID: 35764961 PMCID: PMC9238014 DOI: 10.1186/s12951-022-01513-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/14/2022] [Indexed: 12/13/2022] Open
Abstract
Background Therapy with genetically modified mesenchymal stem cells (MSCs) has clinical translation promise. Optimizing the targeting migratory ability of MSCs relies on accurate imaging of the distribution and extravasation kinetics of MSCs, and the corresponding imaging results could be used to predict therapeutic outcomes and guide the optimization of the treatment program. Among the different imaging modalities, second near-infrared (NIR-II) optical-resolution photoacoustic microscopy (OR-PAM) has merits, including a fine resolution, a deep penetration, a high sensitivity, and a large signal-to-background ratio. It would be an ideal candidate for precise monitoring of MSCs, although it has not been tested for this purpose so far. Results Penetrating peptide-decorated conjugated polymer nanoparticles (TAT-CPNPs) with strong NIR-II absorbance were used to label chemokine-receptor genetically modified MSCs, which were subsequently evaluated under intravital NIR-II OR-PAM regarding their targeting migratory ability. Based on the upregulation of chemokine (C-X-C motif) ligand 10 in the inflamed ears of contact hypersensitivity mice, MSCs with overexpression of corresponding receptor, chemokine (C-X-C motif) receptor 3 (Cxcr3) were successfully generated (MSCCxcr3). TAT-CPNPs labeling enabled NIR-II photoacoustic imaging to discern MSCCxcr3 covered by 1.2 cm of chicken breast tissue. Longitudinal OR-PAM imaging revealed enhanced inflammation-targeting migration of MSCCxcr3 over time attributed to Cxcr3 gene modification, which was further validated by histological analysis. Conclusions TAT-CPNPs-assisted NIR-II PA imaging is promising for monitoring distribution and extravasation kinetics of MSCs, which would greatly facilitate optimizing MSC-based therapy. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01513-7.
Collapse
Affiliation(s)
- Yuejun Lin
- Department of Ultrasound, Laboratory of Novel Optoacoustic/Ultrasonic Imaging, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Hui-Chao Zhou
- Department of Ultrasound, Laboratory of Novel Optoacoustic/Ultrasonic Imaging, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Ningbo Chen
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yaguang Ren
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Rongkang Gao
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qiaojia Li
- Department of Ultrasound, Laboratory of Novel Optoacoustic/Ultrasonic Imaging, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yiwen Deng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Xuejiao Han
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Bing Guo
- School of Science and Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen, 518055, China.
| | - Chengbo Liu
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Jie Ren
- Department of Ultrasound, Laboratory of Novel Optoacoustic/Ultrasonic Imaging, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
8
|
Zhao R, Jin X, Li A, Xu B, Shen Y, Wang W, Huang J, Zhang Y, Li X. Precise Diabetic Wound Therapy: PLS Nanospheres Eliminate Senescent Cells via DPP4 Targeting and PARP1 Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104128. [PMID: 34738744 PMCID: PMC8728814 DOI: 10.1002/advs.202104128] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Indexed: 05/14/2023]
Abstract
Diabetic ulcers, a difficult problem faced by clinicians, are strongly associated with an increase in cellular senescence. Few empirical studies have focused on exploring a targeted strategy to cure diabetic wounds by eliminating senescent fibroblasts (SFs) and reducing side effects. In this study, poly-l-lysine/sodium alginate (PLS) is modified with talabostat (PT100) and encapsulates a PARP1 plasmid (PARP1@PLS-PT100) for delivery to target the dipeptidyl peptidase 4 (DPP4) receptor and eliminate SFs. PARP1@PLS-PT100 releases encapsulated plasmids, displaying high selectivity for SFs over normal fibroblasts by targeting the DPP4 receptor, decreasing senescence-associated secretory phenotypes (SASPs), and stimulating the secretion of anti-inflammatory factors. Furthermore, the increased apoptosis of SFs and the disappearance of cellular senescence alleviates SASPs, accelerates re-epithelialization and collagen deposition, and significantly induces macrophage M2 polarization, which mediates tissue repair and the inflammatory response. This innovative strategy has revealed the previously undefined role of PARP1@PLS-PT100 in promoting diabetic wound healing, suggesting its therapeutic potential in refractory wound repair.
Collapse
Affiliation(s)
- Renliang Zhao
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Xiangyun Jin
- Department of Orthopedic TraumaRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127P. R. China
| | - Ang Li
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Bitong Xu
- Department of SpineCenter for Orthopaedic SurgeryThe Third Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Yifan Shen
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Wei Wang
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Jinghuan Huang
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yadong Zhang
- Department of SpineCenter for Orthopaedic SurgeryThe Third Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Xiaolin Li
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| |
Collapse
|
9
|
Kozisek T, Hamann A, Samuelson L, Fudolig M, Pannier AK. Comparison of promoter, DNA vector, and cationic carrier for efficient transfection of hMSCs from multiple donors and tissue sources. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:81-93. [PMID: 34513295 PMCID: PMC8413668 DOI: 10.1016/j.omtn.2021.06.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/25/2021] [Indexed: 01/22/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are primary cells with high clinical relevance that could be enhanced through genetic modification. However, gene delivery, particularly through nonviral routes, is inefficient. To address the shortcomings of nonviral gene delivery to hMSCs, our lab has previously demonstrated that pharmacological "priming" of hMSCs with clinically approved drugs can increase transfection in hMSCs by modulating transfection-induced cytotoxicity. However, even with priming, hMSC transfection remains inefficient for clinical applications. This work takes a complementary approach to addressing the challenges of transfecting hMSCs by systematically investigating key transfection parameters for their effect on transgene expression. Specifically, we investigated two promoters (cytomegalovirus [CMV] and elongation factor 1 alpha), four DNA vectors (plasmid, plasmid with no F1 origin, minicircle, and mini-intronic plasmid), two cationic carriers (Lipofectamine 3000 and Turbofect), and four donors of hMSCs from two tissues (adipose and bone marrow) for efficient hMSC transfection. Following systematic comparison of each variable, we identified adipose-derived hMSCs transfected with mini-intronic plasmids containing the CMV promoter delivered using Lipofectamine 3000 as the parameters that produced the highest transfection levels. The data presented in this work can guide the development of other hMSC transfection systems with the goal of producing clinically relevant, genetically modified hMSCs.
Collapse
Affiliation(s)
- Tyler Kozisek
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Andrew Hamann
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Luke Samuelson
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Miguel Fudolig
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Angela K. Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
10
|
How Far Are Non-Viral Vectors to Come of Age and Reach Clinical Translation in Gene Therapy? Int J Mol Sci 2021; 22:ijms22147545. [PMID: 34299164 PMCID: PMC8304344 DOI: 10.3390/ijms22147545] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/10/2021] [Indexed: 01/14/2023] Open
Abstract
Efficient delivery of genetic material into cells is a critical process to translate gene therapy into clinical practice. In this sense, the increased knowledge acquired during past years in the molecular biology and nanotechnology fields has contributed to the development of different kinds of non-viral vector systems as a promising alternative to virus-based gene delivery counterparts. Consequently, the development of non-viral vectors has gained attention, and nowadays, gene delivery mediated by these systems is considered as the cornerstone of modern gene therapy due to relevant advantages such as low toxicity, poor immunogenicity and high packing capacity. However, despite these relevant advantages, non-viral vectors have been poorly translated into clinical success. This review addresses some critical issues that need to be considered for clinical practice application of non-viral vectors in mainstream medicine, such as efficiency, biocompatibility, long-lasting effect, route of administration, design of experimental condition or commercialization process. In addition, potential strategies for overcoming main hurdles are also addressed. Overall, this review aims to raise awareness among the scientific community and help researchers gain knowledge in the design of safe and efficient non-viral gene delivery systems for clinical applications to progress in the gene therapy field.
Collapse
|
11
|
Attia N, Mashal M, Puras G, Pedraz JL. Mesenchymal Stem Cells as a Gene Delivery Tool: Promise, Problems, and Prospects. Pharmaceutics 2021; 13:843. [PMID: 34200425 PMCID: PMC8229096 DOI: 10.3390/pharmaceutics13060843] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/24/2021] [Accepted: 05/31/2021] [Indexed: 12/13/2022] Open
Abstract
The cell-based approach in gene therapy arises as a promising strategy to provide safe, targeted, and efficient gene delivery. Owing to their unique features, as homing and tumor-tropism, mesenchymal stem cells (MSCs) have recently been introduced as an encouraging vehicle in gene therapy. Nevertheless, non-viral transfer of nucleic acids into MSCs remains limited due to various factors related to the main stakeholders of the process (e.g., nucleic acids, carriers, or cells). In this review, we have summarized the main types of nucleic acids used to transfect MSCs, the pros and cons, and applications of each. Then, we have emphasized on the most efficient lipid-based carriers for nucleic acids to MSCs, their main features, and some of their applications. While a myriad of studies have demonstrated the therapeutic potential for engineered MSCs therapy in various illnesses, optimization for clinical use is an ongoing challenge. On the way of improvement, genetically modified MSCs have been combined with various novel techniques and tools (e.g., exosomes, spheroids, 3D-Bioprinting, etc.,) aiming for more efficient and safe applications in biomedicine.
Collapse
Affiliation(s)
- Noha Attia
- Laboratory of Pharmaceutics, NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (N.A.); (M.M.)
- Department of Basic Sciences, The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda
- The Center of Research and Evaluation, The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda
- Histology and Cell Biology Department, Faculty of Medicine, University of Alexandria, Alexandria 21561, Egypt
| | - Mohamed Mashal
- Laboratory of Pharmaceutics, NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (N.A.); (M.M.)
- The Center of Research and Evaluation, The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda
| | - Gustavo Puras
- Laboratory of Pharmaceutics, NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (N.A.); (M.M.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
- Laboratory of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
| | - Jose Luis Pedraz
- Laboratory of Pharmaceutics, NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (N.A.); (M.M.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
- Laboratory of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
| |
Collapse
|
12
|
Minicircles for Investigating and Treating Arthritic Diseases. Pharmaceutics 2021; 13:pharmaceutics13050736. [PMID: 34067675 PMCID: PMC8156692 DOI: 10.3390/pharmaceutics13050736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/15/2021] [Indexed: 01/22/2023] Open
Abstract
Gene delivery systems have become an essential component of research and the development of therapeutics for various diseases. Minicircles are non-viral vectors with promising characteristics for application in a variety of fields. With their minimal size, minicircles exhibit relatively high safety and efficient delivery of genes of interest into cells. Cartilage tissue lacks the natural ability to heal, making it difficult to treat osteoarthritis (OA) and rheumatoid arthritis (RA), which are the two main types of joint-related disease. Although both OA and RA affect the joint, RA is an autoimmune disease, while OA is a degenerative joint condition. Gene transfer using minicircles has also been used in many studies regarding cartilage and its diseased conditions. In this review, we summarize the cartilage-, OA-, and RA-based studies that have used minicircles as the gene delivery system.
Collapse
|
13
|
Harman RM, Marx C, Van de Walle GR. Translational Animal Models Provide Insight Into Mesenchymal Stromal Cell (MSC) Secretome Therapy. Front Cell Dev Biol 2021; 9:654885. [PMID: 33869217 PMCID: PMC8044970 DOI: 10.3389/fcell.2021.654885] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
The therapeutic potential of the mesenchymal stromal cell (MSC) secretome, consisting of all molecules secreted by MSCs, is intensively studied. MSCs can be readily isolated, expanded, and manipulated in culture, and few people argue with the ethics of their collection. Despite promising pre-clinical studies, most MSC secretome-based therapies have not been implemented in human medicine, in part because the complexity of bioactive factors secreted by MSCs is not completely understood. In addition, the MSC secretome is variable, influenced by individual donor, tissue source of origin, culture conditions, and passage. An increased understanding of the factors that make up the secretome and the ability to manipulate MSCs to consistently secrete factors of biologic importance will improve MSC therapy. To aid in this goal, we can draw from the wealth of information available on secreted factors from MSC isolated from veterinary species. These translational animal models will inspire efforts to move human MSC secretome therapy from bench to bedside.
Collapse
Affiliation(s)
| | | | - Gerlinde R. Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
14
|
Florian M, Wang JP, Deng Y, Souza-Moreira L, Stewart DJ, Mei SHJ. Gene engineered mesenchymal stem cells: greater transgene expression and efficacy with minicircle vs. plasmid DNA vectors in a mouse model of acute lung injury. Stem Cell Res Ther 2021; 12:184. [PMID: 33726829 PMCID: PMC7962085 DOI: 10.1186/s13287-021-02245-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/25/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) and in its severe form, acute respiratory distress syndrome (ARDS), results in increased pulmonary vascular inflammation and permeability and is a major cause of mortality in many critically ill patients. Although cell-based therapies have shown promise in experimental ALI, strategies are needed to enhance the potency of mesenchymal stem cells (MSCs) to develop more effective treatments. Genetic modification of MSCs has been demonstrated to significantly improve the therapeutic benefits of these cells; however, the optimal vector for gene transfer is not clear. Given the acute nature of ARDS, transient transfection is desirable to avoid off-target effects of long-term transgene expression, as well as the potential adverse consequences of genomic integration. METHODS Here, we explored whether a minicircle DNA (MC) vector containing human angiopoietin 1 (MC-ANGPT1) can provide a more effective platform for gene-enhanced MSC therapy of ALI/ARDS. RESULTS At 24 h after transfection, nuclear-targeted electroporation using an MC-ANGPT1 vector resulted in a 3.7-fold greater increase in human ANGPT1 protein in MSC conditioned media compared to the use of a plasmid ANGPT1 (pANGPT1) vector (2048 ± 567 pg/mL vs. 552.1 ± 33.5 pg/mL). In the lipopolysaccharide (LPS)-induced ALI model, administration of pANGPT1 transfected MSCs significantly reduced bronchoalveolar lavage (BAL) neutrophil counts by 57%, while MC-ANGPT1 transfected MSCs reduced it by 71% (p < 0.001) by Holm-Sidak's multiple comparison test. Moreover, compared to pANGPT1, the MC-ANGPT1 transfected MSCs significantly reduced pulmonary inflammation, as observed in decreased levels of proinflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), and macrophage inflammatory protein-2 (MIP-2). pANGPT1-transfected MSCs significantly reduced BAL albumin levels by 71%, while MC-ANGPT1-transfected MSCs reduced it by 85%. CONCLUSIONS Overall, using a minicircle vector, we demonstrated an efficient and sustained expression of the ANGPT1 transgene in MSCs and enhanced the therapeutic effect on the ALI model compared to plasmid. These results support the potential benefits of MC-ANGPT1 gene enhancement of MSC therapy to treat ARDS.
Collapse
Affiliation(s)
- Maria Florian
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Jia-Pey Wang
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Yupu Deng
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | - Duncan J Stewart
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
- The Ottawa Hospital, Ottawa, ON, Canada
- University of Ottawa, Ottawa, ON, Canada
| | | |
Collapse
|
15
|
Cha EB, Shin KK, Seo J, Oh DB. Antibody-secreting macrophages generated using CpG-free plasmid eliminate tumor cells through antibody-dependent cellular phagocytosis. BMB Rep 2020. [PMID: 32438971 PMCID: PMC7473480 DOI: 10.5483/bmbrep.2020.53.8.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The non-viral delivery of genes into macrophages, known as hard-to-transfect cells, is a challenge. In this study, the microporation of a CpG-free and small plasmid (pCGfd-GFP) showed high transfection efficiency, sustainable transgene expression, and good cell viability in the transfections of Raw 264.7 and primary bone marrow-derived macrophages. The non-viral method using the pCGfd vector encoding anti-EGFR single-chain Fv fused with Fc (scFv-Fc) generated the macrophages secreting anti-EGFR scFv-Fc. These macrophages effectively phagocytized tumor cells expressing EGFR through the antibody-dependent mechanism, as was proved by experiments using EGFR-knockout tumor cells. Finally, peri-tumoral injections of anti-EGFR scFv-Fc-secreting macrophages were shown to inhibit tumor growth in the xeno-graft mouse model.
Collapse
Affiliation(s)
- Eun Bi Cha
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Keun Koo Shin
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Jinho Seo
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Doo-Byoung Oh
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Korea
| |
Collapse
|
16
|
Han JH, Han S, Jeong IS, Cheon SH, Kim SW. Minicircle-based GCP-2 ex vivo gene therapy enhanced the reepithelialization and angiogenic capacity. J Tissue Eng Regen Med 2020; 14:829-839. [PMID: 32336047 DOI: 10.1002/term.3049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 04/06/2020] [Accepted: 04/15/2020] [Indexed: 12/18/2022]
Abstract
Recently, minicircle (MC)-based cell therapy has been emerging as a novel technology for nonviral genetic modification. In this study, we investigated the characteristics of granulocyte chemotactic protein-2 (GCP-2)-overexpressing fibroblasts (GCP-2/MC) using MC microporation technology, as well as its therapeutic mechanism in wound healing. GCP-2 parent plasmid and MC containing GCP-2 were generated. Human dermal fibroblasts (HDF) were transfected with MC containing GCP-2. Quantitative reverse transcription polymerase chain reaction (qRT-PCR), scratch wound assay, and in vivo wound healing assay were performed. Gene and protein expression analysis revealed that GCP-2/MC highly expressed epithelialization growth factor, epidermal growth factor (EGF), chemokines, GCP-2, interleukin (IL)-8, as well as wound healing-associated genes such as insulin growth factor (IGF)-1 and hepatocyte growth factor (HGF). An in vitro scratch wound closure and matrigel tube formation assays demonstrated that the culture medium derived from GCP-2/MC substantially accelerated the wound closure and matrigel network formation. Wounds in nude mice were created by skin excisions followed by injections of GCP-2/MC. Results showed high cell survival potential and that GCP-2/MC transplantation highly accelerated skin wound closure by increasing reepithelialization, capillary density, and enhancing angiogenic factors, suggesting direct benefits for cutaneous closure. Taken together, these data suggest that MC-based GCP-2 overexpression could be a promising alternative strategy for promoting wound healing.
Collapse
Affiliation(s)
- Ju Hye Han
- Institute for Bio-Medical Convergence, Department of Medicine, Catholic Kwandong University College of Medicine, Gangneung, Republic of Korea
| | - Seongho Han
- Department of Family Medicine, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - In Sil Jeong
- Institute for Bio-Medical Convergence, Department of Medicine, Catholic Kwandong University College of Medicine, Gangneung, Republic of Korea
| | - Sae Hee Cheon
- Department of Dental Hygiene, Masan University, Masan, South Korea
| | - Sung-Whan Kim
- Institute for Bio-Medical Convergence, Department of Medicine, Catholic Kwandong University College of Medicine, Gangneung, Republic of Korea
| |
Collapse
|
17
|
Nie WB, Zhang D, Wang LS. Growth Factor Gene-Modified Mesenchymal Stem Cells in Tissue Regeneration. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1241-1256. [PMID: 32273686 PMCID: PMC7105364 DOI: 10.2147/dddt.s243944] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/10/2020] [Indexed: 12/13/2022]
Abstract
There have been marked changes in the field of stem cell therapeutics in recent years, with many clinical trials having been conducted to date in an effort to treat myriad diseases. Mesenchymal stem cells (MSCs) are the cell type most frequently utilized in stem cell therapeutic and tissue regenerative strategies, and have been used with excellent safety to date. Unfortunately, these MSCs have limited ability to engraft and survive, reducing their clinical utility. MSCs are able to secrete growth factors that can support the regeneration of tissues, and engineering MSCs to express such growth factors can improve their survival, proliferation, differentiation, and tissue reconstructing abilities. As such, it is likely that such genetically modified MSCs may represent the next stage of regenerative therapy. Indeed, increasing volumes of preclinical research suggests that such modified MSCs expressing growth factors can effectively treat many forms of tissue damage. In the present review, we survey recent approaches to producing and utilizing growth factor gene-modified MSCs in the context of tissue repair and discuss its prospects for clinical application.
Collapse
Affiliation(s)
- Wen-Bo Nie
- Department of Rehabilitation Sciences, School of Nursing, Jilin University, Changchun, People's Republic of China
| | - Dan Zhang
- Department of Rehabilitation Sciences, School of Nursing, Jilin University, Changchun, People's Republic of China
| | - Li-Sheng Wang
- Department of Rehabilitation Sciences, School of Nursing, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
18
|
Huang D, Ren J, Li R, Guan C, Feng Z, Bao B, Wang W, Zhou C. Tooth Regeneration: Insights from Tooth Development and Spatial-Temporal Control of Bioactive Drug Release. Stem Cell Rev Rep 2020; 16:41-55. [PMID: 31834583 PMCID: PMC6987083 DOI: 10.1007/s12015-019-09940-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Tooth defect and tooth loss are common clinical diseases in stomatology. Compared with the traditional oral restoration treatment, tooth regeneration has unique advantages and is currently the focus of oral biomedical research. It is known that dozens of cytokines/growth factors and other bioactive factors are expressed in a spatial-temporal pattern during tooth development. On the other hand, the technology for spatial-temporal control of drug release has been intensively studied and well developed recently, making control release of these bioactive factors mimicking spatial-temporal pattern more feasible than ever for the purpose of tooth regeneration. This article reviews the research progress on the tooth development and discusses the future of tooth regeneration in the context of spatial-temporal release of developmental factors.
Collapse
Affiliation(s)
- Delan Huang
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jianhan Ren
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Runze Li
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chenyu Guan
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Zhicai Feng
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Baicheng Bao
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Weicai Wang
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chen Zhou
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
19
|
Liu GW, Johnson SL, Jain R, Peeler DJ, Shankland SJ, Pun SH. Optimized nonviral gene delivery for primary urinary renal progenitor cells to enhance cell migration. J Biomed Mater Res A 2019; 107:2718-2725. [PMID: 31404486 DOI: 10.1002/jbm.a.36775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 07/31/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023]
Abstract
Progressive loss of glomerular podocytes during kidney disease leads to irreversible kidney failure, and is exacerbated by the fact that podocytes are terminally differentiated epithelial cells and unable to proliferate. Regeneration of lost podocytes must therefore derive from nonpodocyte sources. Human urine-derived renal progenitor cells (uRPCs) are attractive podocyte progenitors for cell therapy applications due to their availability from patient urine and ability to migrate to injured glomeruli and differentiate into de novo podocytes after intravenous administration. Because gene delivery has emerged as an important strategy to augment the functionality and survival of cell therapies prior to injection, in this work we optimized nonviral gene delivery conditions (cell density, DNA dose, % FBS, and transfection material composition) to primary uRPCs. Using the cationic polymer-peptide conjugate VIPER for gene delivery and the Sleeping Beauty transposon/transposase constructs for gene integration, we optimized transfection parameters to achieve efficient transgene expression (up to 55% transfected cells) and stable transgene expression (>65% integration efficiency) lasting up to 10 days. With these methods, we transfected uRPCs to overexpress CXCR4, an important chemokine receptor that mediates uRPC migration to the kidneys after intravenous injection, and demonstrate that CXCR4-uRPCs exhibit enhanced migration compared to mock-transfected cells.
Collapse
Affiliation(s)
- Gary W Liu
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington
| | - Soren L Johnson
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington
| | - Ritika Jain
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington
| | - David J Peeler
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington
| | - Stuart J Shankland
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, Washington
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington
| |
Collapse
|
20
|
Hamann A, Nguyen A, Pannier AK. Nucleic acid delivery to mesenchymal stem cells: a review of nonviral methods and applications. J Biol Eng 2019; 13:7. [PMID: 30675180 PMCID: PMC6339289 DOI: 10.1186/s13036-019-0140-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/07/2019] [Indexed: 12/13/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) are multipotent stem cells that can be isolated and expanded from many tissues, and are being investigated for use in cell therapies. Though MSC therapies have demonstrated some success, none have been FDA approved for clinical use. MSCs lose stemness ex vivo, decreasing therapeutic potential, and face additional barriers in vivo, decreasing therapeutic efficacy. Culture optimization and genetic modification of MSCs can overcome these barriers. Viral transduction is efficient, but limited by safety concerns related to mutagenicity of integrating viral vectors and potential immunogenicity of viral antigens. Nonviral delivery methods are safer, though limited by inefficiency and toxicity, and are flexible and scalable, making them attractive for engineering MSC therapies. Main text Transfection method and nucleic acid determine efficiency and expression profile in transfection of MSCs. Transfection methods include microinjection, electroporation, and nanocarrier delivery. Microinjection and electroporation are efficient, but are limited by throughput and toxicity. In contrast, a variety of nanocarriers have been demonstrated to transfer nucleic acids into cells, however nanocarrier delivery to MSCs has traditionally been inefficient. To improve efficiency, plasmid sequences can be optimized by choice of promoter, inclusion of DNA targeting sequences, and removal of bacterial elements. Instead of DNA, RNA can be delivered for rapid protein expression or regulation of endogenous gene expression. Beyond choice of nanocarrier and nucleic acid, transfection can be optimized by priming cells with media additives and cell culture surface modifications to modulate barriers of transfection. Media additives known to enhance MSC transfection include glucocorticoids and histone deacetylase inhibitors. Culture surface properties known to modulate MSC transfection include substrate stiffness and specific protein coating. If nonviral gene delivery to MSCs can be sufficiently improved, MSC therapies could be enhanced by transfection for guided differentiation and reprogramming, transplantation survival and directed homing, and secretion of therapeutics. We discuss utilized delivery methods and nucleic acids, and resulting efficiency and outcomes, in transfection of MSCs reported for such applications. Conclusion Recent developments in transfection methods, including nanocarrier and nucleic acid technologies, combined with chemical and physical priming of MSCs, may sufficiently improve transfection efficiency, enabling scalable genetic engineering of MSCs, potentially bringing effective MSC therapies to patients.
Collapse
Affiliation(s)
- Andrew Hamann
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, 231 L.W. Chase Hall, Lincoln, NE 68583-0726 USA
| | - Albert Nguyen
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, 231 L.W. Chase Hall, Lincoln, NE 68583-0726 USA
| | - Angela K Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, 231 L.W. Chase Hall, Lincoln, NE 68583-0726 USA
| |
Collapse
|
21
|
Leishmania treatment and prevention: Natural and synthesized drugs. Eur J Med Chem 2018; 160:229-244. [DOI: 10.1016/j.ejmech.2018.10.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 12/31/2022]
|
22
|
Serra J, Alves CPA, Brito L, Monteiro GA, Cabral JMS, Prazeres DMF, da Silva CL. Engineering of Human Mesenchymal Stem/Stromal Cells with Vascular Endothelial Growth Factor-Encoding Minicircles for Angiogenic Ex Vivo Gene Therapy. Hum Gene Ther 2018; 30:316-329. [PMID: 30200778 DOI: 10.1089/hum.2018.154] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Peripheral artery disease (PAD) is a debilitating and prevalent condition characterized by blockage of the arteries, leading to limb amputation in more severe cases. Mesenchymal stem/stromal cells (MSC) are known to have intrinsic regenerative properties that can be potentiated by the introduction of pro-angiogenic genes such as the vascular endothelial growth factor (VEGF). Herein, the use of human bone marrow MSC transiently transfected with minicircles encoding for VEGF is proposed as an ex vivo gene therapy strategy to enhance angiogenesis in PAD patients. The VEGF gene was cloned in minicircle and conventional plasmid vectors and used to transfect bone marrow-derived MSC ex vivo. VEGF expression was evaluated both by quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. The number of VEGF transcripts following MSC transfection with minicircles increased 130-fold relative to the expression in non-transfected MSC, whereas for the plasmid (pVAX1)-based transfection, the increase was 50-fold. Compared to the VEGF basal levels secreted by MSC (11.1 ± 3.4 pg/1,000 cells/day), significantly higher values were detected by enzyme-linked immunosorbent assay after both minicircle and pVAX1 transfection (644.8 ± 82.5 and 508.3 ± 164.0 pg/1,000 cells/day, respectively). The VEGF overexpression improved the angiogenic potential of MSC in vitro, as confirmed by endothelial cell tube formation and cell migration assays, without affecting the expansion potential ex vivo, as well as multilineage differentiation capacity or immunophenotype of MSC. Although preclinical in vivo studies are required, these results suggest that minicircle-mediated VEGF gene delivery, combined with the unique properties of human MSC, could represent a promising ex vivo gene therapy approach for an improved angiogenesis in the context of PAD.
Collapse
Affiliation(s)
- Joana Serra
- 1 Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia P A Alves
- 1 Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Universidade de Lisboa, Lisboa, Portugal
| | - Liliana Brito
- 1 Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Universidade de Lisboa, Lisboa, Portugal
| | - Gabriel A Monteiro
- 1 Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Universidade de Lisboa, Lisboa, Portugal.,2 The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim M S Cabral
- 1 Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Universidade de Lisboa, Lisboa, Portugal.,2 The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Duarte Miguel F Prazeres
- 1 Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L da Silva
- 1 Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Universidade de Lisboa, Lisboa, Portugal.,2 The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
23
|
Hamilton AM, Foster PJ, Ronald JA. Evaluating Nonintegrating Lentiviruses as Safe Vectors for Noninvasive Reporter-Based Molecular Imaging of Multipotent Mesenchymal Stem Cells. Hum Gene Ther 2018; 29:1213-1225. [DOI: 10.1089/hum.2018.111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Amanda M. Hamilton
- Imaging Research Laboratories, Robarts Research Institute, London, Canada
| | - Paula J. Foster
- Imaging Research Laboratories, Robarts Research Institute, London, Canada
- Medical Biophysics, University of Western Ontario, London, Canada
| | - John A. Ronald
- Imaging Research Laboratories, Robarts Research Institute, London, Canada
- Medical Biophysics, University of Western Ontario, London, Canada
- Lawson Health Research Institute, London, Canada
| |
Collapse
|
24
|
He XT, Wang J, Li X, Yin Y, Sun HH, Chen FM. The Critical Role of Cell Homing in Cytotherapeutics and Regenerative Medicine. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800098] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Xiao-Tao He
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Jia Wang
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Xuan Li
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Hai-Hua Sun
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Fa-Ming Chen
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| |
Collapse
|
25
|
Mesenchymal Stem Cells in Primary Sjögren's Syndrome: Prospective and Challenges. Stem Cells Int 2018; 2018:4357865. [PMID: 30305818 PMCID: PMC6165618 DOI: 10.1155/2018/4357865] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/20/2018] [Accepted: 09/02/2018] [Indexed: 02/07/2023] Open
Abstract
Primary Sjögren's syndrome (pSS) is a chronic systemic inflammatory autoimmune disease characterized by lymphocytic infiltrates in exocrine glands. Current approaches do not control harmful autoimmune attacks or prevent irreversible damage and have considerable side effects. Mesenchymal stem cells (MSCs) have been effective in the treatment of several autoimmune diseases. The objective of this review is to illustrate the potential therapeutic role of MSCs in pSS. We summarize the recent advances in what is known about their immunomodulatory function and therapeutic applications in pSS. MSC transfusion can suppress autoimmunity and restore salivary gland secretory function in mouse models and patients with pSS by inducing regulatory T cells, suppressing Th1, Th17, and T follicular helper cell responses. In addition, MSCs can differentiate into salivary epithelial cells, presenting an option as a suitable alternative treatment. We also discuss current bioengineering methods which improve functions of MSCs for pSS. However, there remain many challenges to overcome before their wide clinical application.
Collapse
|
26
|
Tian Y, Zhou M, Shi H, Gao S, Xie G, Zhu M, Wu M, Chen J, Niu Z. Integration of Cell-Penetrating Peptides with Rod-like Bionanoparticles: Virus-Inspired Gene-Silencing Technology. NANO LETTERS 2018; 18:5453-5460. [PMID: 30091612 DOI: 10.1021/acs.nanolett.8b01805] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Inspired by the high gene transfer efficiency of viral vectors and to avoid side effects, we present here a 1D rod-like gene-silencing vector based on a plant virus. By decorating the transacting activator of transduction (TAT) peptide on the exterior surface, the TAT-modified tobacco mosaic virus (TMV) achieves a tunable isoelectric point (from ∼3.5 to ∼9.6) depending on the TAT dose. In addition to enhanced cell internalization, this plant virus-based vector (TMV-TAT) acquired endo/lysosomal escape capacity without inducing lysosomal damage, resulting in both high efficiency and low cytotoxicity. By loading silencer green fluorescent protein (GFP) siRNA onto the TMV-TAT vector (siRNA@TMV-TAT) and interfering with GFP-expressing mouse epidermal stem cells (ESCs/GFP) in vitro, the proportion of GFP-positive cells could be knocked down to levels even lower than 15% at a concentration of ∼100% cell viability. Moreover, by interfering with GFP-expressing highly metastatic hepatocellular carcinoma (MHCC97-H/GFP) tumors in vivo, treatment with siRNA@TMV-TAT complexes for 10 days achieved a GFP-negative rate as high as 80.8%. This work combines the high efficiency of viral vectors and the safety of nonviral vectors and may provide a promising strategy for gene-silencing technology.
Collapse
Affiliation(s)
- Ye Tian
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials , Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , 29 Zhongguancun East Road , Beijing 100190 , P. R. China
| | - Mengxue Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , No. 19(B) Yuquan Road , Beijing 100049 , P. R. China
| | - Haigang Shi
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials , Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , 29 Zhongguancun East Road , Beijing 100190 , P. R. China
| | - Sijia Gao
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials , Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , 29 Zhongguancun East Road , Beijing 100190 , P. R. China
| | - Guocheng Xie
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials , Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , 29 Zhongguancun East Road , Beijing 100190 , P. R. China
| | - Meng Zhu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials , Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , 29 Zhongguancun East Road , Beijing 100190 , P. R. China
| | - Man Wu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials , Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , 29 Zhongguancun East Road , Beijing 100190 , P. R. China
| | - Jun Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , No. 19(B) Yuquan Road , Beijing 100049 , P. R. China
| | - Zhongwei Niu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials , Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , 29 Zhongguancun East Road , Beijing 100190 , P. R. China
- School of Future Technology , University of Chinese Academy of Sciences , No.19(A) Yuquan Road , Beijing 100049 , P. R. China
| |
Collapse
|
27
|
Abstract
An ideal gene carrier requires an excellent gating system to efficiently load, protect, deliver, and release environmentally sensitive nucleic acids on demand. Presented in this communication is a polymersome with a "boarding gate" and a "debarkation gate" in the membrane to complete the above important missions. This dually gated polymersome is self-assembled from a block copolymer, poly(ethylene oxide)- block-poly[ N-isopropylacrylamide- stat-7-(2-methacryloyloxyethoxy)-4-methylcoumarin- stat-2-(diethylamino)ethyl methacrylate] [PEO- b-P(NIPAM- stat-CMA- stat-DEA)]. The hydrophilic PEO chains form the coronas of the polymersome, whereas the temperature and pH-sensitive P(NIPAM- stat-CMA- stat-DEA) block forms the dually gated heterogeneous membrane. The temperature-controlled "boarding gate" can be opened at room temperature for facile encapsulation of siRNA and plasmid DNA into polymersomes directly in aqueous solution. The "debarkation gate" can be triggered by proton sponge effect for intracellular release. Biological studies confirmed the successful encapsulation of siRNA and plasmid DNA, efficient in vitro and in vivo gene transfection, and the expression of green fluorescent protein (GFP) from GFP-encoding plasmid, suggesting that this kind of polymersome with a dual gating system can serve as an excellent biomacromolecular shuttle for gene delivery and other biological applications.
Collapse
Affiliation(s)
- Fangyingkai Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital , Tongji University School of Medicine , 301 Middle Yanchang Road , Shanghai 200072 , China
- Department of Polymeric Materials, School of Materials Science and Engineering , Tongji University , 4800 Caoan Road , Shanghai 201804 , China
| | - Jingyi Gao
- Department of Polymeric Materials, School of Materials Science and Engineering , Tongji University , 4800 Caoan Road , Shanghai 201804 , China
| | - Jiangang Xiao
- Department of Polymeric Materials, School of Materials Science and Engineering , Tongji University , 4800 Caoan Road , Shanghai 201804 , China
| | - Jianzhong Du
- Department of Orthopaedics, Shanghai Tenth People's Hospital , Tongji University School of Medicine , 301 Middle Yanchang Road , Shanghai 200072 , China
- Department of Polymeric Materials, School of Materials Science and Engineering , Tongji University , 4800 Caoan Road , Shanghai 201804 , China
| |
Collapse
|
28
|
Ranganath SH. Bioengineered cellular and cell membrane-derived vehicles for actively targeted drug delivery: So near and yet so far. Adv Drug Deliv Rev 2018; 132:57-80. [PMID: 29935987 DOI: 10.1016/j.addr.2018.06.012] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/31/2018] [Accepted: 06/18/2018] [Indexed: 12/16/2022]
Abstract
Cellular carriers for drug delivery are attractive alternatives to synthetic nanoparticles owing to their innate homing/targeting abilities. Here, we review molecular interactions involved in the homing of Mesenchymal stem cells (MSCs) and other cell types to understand the process of designing and engineering highly efficient, actively targeting cellular vehicles. In addition, we comprehensively discuss various genetic and non-genetic strategies and propose futuristic approaches of engineering MSC homing using micro/nanotechnology and high throughput small molecule screening. Most of the targeting abilities of a cell come from its plasma membrane, thus, efforts to harness cell membranes as drug delivery vehicles are gaining importance and are highlighted here. We also recognize and report the lack of detailed characterization of cell membranes in terms of safety, structural integrity, targeting functionality, and drug transport. Finally, we provide insights on future development of bioengineered cellular and cell membrane-derived vesicles for successful clinical translation.
Collapse
Affiliation(s)
- Sudhir H Ranganath
- Bio-INvENT Lab, Department of Chemical Engineering, Siddaganga Institute of Technology, B.H. Road, Tumakuru, 572103, Karnataka, India.
| |
Collapse
|
29
|
Li B, Guo W, Zhang F, Liu M, Wang S, Liu Z, Xiang S, Zeng Y. Synthesis and evaluation of L-arabinose-based cationic glycolipids as effective vectors for pDNA and siRNA in vitro. PLoS One 2017; 12:e0180276. [PMID: 28672000 PMCID: PMC5495346 DOI: 10.1371/journal.pone.0180276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/13/2017] [Indexed: 01/14/2023] Open
Abstract
Glycolipids might become a new type of promising non-viral gene delivery systems because of their low cytotoxicity, structural diversity, controllable aqua- and lipo-solubility, appropriate density and distribution of positive charges, high transfer efficiency and potential targeting function. In this study, four kinds of L-arabinose-based cationic glycolipids (Ara-DiC12MA, Ara-DiC14MA, Ara-DiC16MA and Ara-DiC18MA) containing quaternary ammonium as hydrophilic headgroup and two alkane chains as hydrophobic domain were synthesized and characterized. They were observed to have strong affinities for plasmid DNA (pDNA) and siRNA, the pDNA can be completely condensed at N/P ratio less than 2, and the siRNA can be completely retarded at N/P ratio less than 3. The dynamic light scattering (DLS) experiment and atomic force microscopy (AFM) experiment demonstrated that cationic lipids and their lipoplexes possessed suitable particle sizes with near-spherical shape and proper ζ-potentials for cell transfection. The Ara-DiC16MA liposome was found to have good transfection efficacy in HEK293, PC-3 and Mat cells compared with other three kinds of liposomes, and also maintain low cytotoxicity and better uptake capability in vitro. Furthermore, the gene silencing assay showed that Ara-DiC14MA and Ara-DiC16MA liposomes have demonstrated effective delivery and higher gene knockdown activity (>80%) in the above mentioned cells than Lipofectamine 2000. These results indicated Ara-DiC16MA can be developed for efficient and low toxic gene delivery.
Collapse
Affiliation(s)
- Bo Li
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, Hunan, P. R. China
| | - Wanrong Guo
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, Hunan, P. R. China
| | - Fan Zhang
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, Hunan, P. R. China
| | - Meiyan Liu
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, Hunan, P. R. China
| | - Shang Wang
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan, P. R. China
| | - Zhonghua Liu
- The National &Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, P.R. China
| | - Shuanglin Xiang
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan, P. R. China
| | - Youlin Zeng
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, Hunan, P. R. China
- * E-mail:
| |
Collapse
|
30
|
Park N, Rim YA, Jung H, Kim J, Yi H, Kim Y, Jang Y, Jung SM, Lee J, Kwok SK, Park SH, Ju JH. Etanercept-Synthesising Mesenchymal Stem Cells Efficiently Ameliorate Collagen-Induced Arthritis. Sci Rep 2017; 7:39593. [PMID: 28084468 PMCID: PMC5234034 DOI: 10.1038/srep39593] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 11/25/2016] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have multiple properties including anti-inflammatory and immunomodulatory effects in various disease models and clinical treatments. These beneficial effects, however, are sometimes inconsistent and unpredictable. For wider and proper application, scientists sought to improve MSC functions by engineering. We aimed to invent a novel method to produce synthetic biological drugs from engineered MSCs. We investigated the anti-arthritic effect of engineered MSCs in a collagen-induced arthritis (CIA) model. Biologics such as etanercept are the most successful drugs used in anti-cytokine therapy. Biologics are made of protein components, and thus can be theoretically produced from cells including MSCs. MSCs were transfected with recombinant minicircles encoding etanercept (trade name, Enbrel), which is a tumour necrosis factor α blocker currently used to treat rheumatoid arthritis. We confirmed minicircle expression in MSCs in vitro based on GFP. Etanercept production was verified from the conditioned media. We confirmed that self-reproduced etanercept was biologically active in vitro. Arthritis subsided more efficiently in CIA mice injected with mcTNFR2MSCs than in those injected with conventional MSCs or etanercept only. Although this novel strategy is in a very early conceptual stage, it seems to represent a potential alternative method for the delivery of biologics and engineering MSCs.
Collapse
Affiliation(s)
- Narae Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Yeri Alice Rim
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Hyerin Jung
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Juryun Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Hyoju Yi
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.,Department of Medicine, Institute for Stem Cell Biology and Regenerative Medicine, and Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Youngkyun Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.,Department of Medicine, Institute for Stem Cell Biology and Regenerative Medicine, and Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Yeonsue Jang
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Jennifer Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Ji Hyeon Ju
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| |
Collapse
|