1
|
Cuevas-Ocaña S, Yang JY, Aushev M, Schlossmacher G, Bear CE, Hannan NRF, Perkins ND, Rossant J, Wong AP, Gray MA. A Cell-Based Optimised Approach for Rapid and Efficient Gene Editing of Human Pluripotent Stem Cells. Int J Mol Sci 2023; 24:10266. [PMID: 37373413 PMCID: PMC10299534 DOI: 10.3390/ijms241210266] [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: 05/16/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Introducing or correcting disease-causing mutations through genome editing in human pluripotent stem cells (hPSCs) followed by tissue-specific differentiation provide sustainable models of multiorgan diseases, such as cystic fibrosis (CF). However, low editing efficiency resulting in extended cell culture periods and the use of specialised equipment for fluorescence activated cell sorting (FACS) make hPSC genome editing still challenging. We aimed to investigate whether a combination of cell cycle synchronisation, single-stranded oligodeoxyribonucleotides, transient selection, manual clonal isolation, and rapid screening can improve the generation of correctly modified hPSCs. Here, we introduced the most common CF mutation, ΔF508, into the CFTR gene, using TALENs into hPSCs, and corrected the W1282X mutation using CRISPR-Cas9, in human-induced PSCs. This relatively simple method achieved up to 10% efficiency without the need for FACS, generating heterozygous and homozygous gene edited hPSCs within 3-6 weeks in order to understand genetic determinants of disease and precision medicine.
Collapse
Affiliation(s)
- Sara Cuevas-Ocaña
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (G.S.); (N.D.P.); (M.A.G.)
- Biodiscovery Institute, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Jin Ye Yang
- Programme in Developmental & Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (J.Y.Y.); (J.R.); (A.P.W.)
| | - Magomet Aushev
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Biomedicine West Wing, Centre for Life, Times Square, Newcastle upon Tyne NE1 3BZ, UK;
| | - George Schlossmacher
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (G.S.); (N.D.P.); (M.A.G.)
| | - Christine E. Bear
- Programme in Molecular Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada;
| | - Nicholas R. F. Hannan
- Biodiscovery Institute, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Neil D. Perkins
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (G.S.); (N.D.P.); (M.A.G.)
| | - Janet Rossant
- Programme in Developmental & Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (J.Y.Y.); (J.R.); (A.P.W.)
| | - Amy P. Wong
- Programme in Developmental & Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (J.Y.Y.); (J.R.); (A.P.W.)
| | - Michael A. Gray
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (G.S.); (N.D.P.); (M.A.G.)
| |
Collapse
|
2
|
Garcia-Cañadas M, Sanchez-Luque FJ, Sanchez L, Rojas J, Garcia Perez JL. LINE-1 Retrotransposition Assays in Embryonic Stem Cells. Methods Mol Biol 2023; 2607:257-309. [PMID: 36449167 DOI: 10.1007/978-1-0716-2883-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The ongoing mobilization of active non-long terminal repeat (LTR) retrotransposons continues to impact the genomes of most mammals, including humans and rodents. Non-LTR retrotransposons mobilize using an intermediary RNA and a copy-and-paste mechanism termed retrotransposition. Non-LTR retrotransposons are subdivided into long and short interspersed elements (LINEs and SINEs, respectively), depending on their size and autonomy; while active class 1 LINEs (LINE-1s or L1s) encode the enzymatic machinery required to mobilize in cis, active SINEs use the enzymatic machinery of active LINE-1s to mobilize in trans. The mobilization mechanism used by LINE-1s/SINEs was exploited to develop ingenious plasmid-based retrotransposition assays in cultured cells, which typically exploit a reporter gene that can only be activated after a round of retrotransposition. Retrotransposition assays, in cis or in trans, are instrumental tools to study the biology of mammalian LINE-1s and SINEs. In fact, these and other biochemical/genetic assays were used to uncover that endogenous mammalian LINE-1s/SINEs naturally retrotranspose during early embryonic development. However, embryonic stem cells (ESCs) are typically used as a cellular model in these and other studies interrogating LINE-1/SINE expression/regulation during early embryogenesis. Thus, human and mouse ESCs represent an excellent model to understand how active retrotransposons are regulated and how their activity impacts the germline. Here, we describe robust and quantitative protocols to study human/mouse LINE-1 (in cis) and SINE (in trans) retrotransposition using (human and mice) ESCs. These protocols are designed to study the mobilization of active non-LTR retrotransposons in a cellular physiologically relevant context.
Collapse
Affiliation(s)
- Marta Garcia-Cañadas
- Pfizer-University of Granada-Andalusian Government Centre for Genomics and Oncological Research (GENYO), PTS Granada, Granada, Spain.
| | - Francisco J Sanchez-Luque
- Institute of Parasitology and Biomedicine "Lopez-Neyra" (IPBLN), Spanish National Research Council (CSIC), PTS Granada, Granada, Spain
| | - Laura Sanchez
- Pfizer-University of Granada-Andalusian Government Centre for Genomics and Oncological Research (GENYO), PTS Granada, Granada, Spain
| | - Johana Rojas
- Pfizer-University of Granada-Andalusian Government Centre for Genomics and Oncological Research (GENYO), PTS Granada, Granada, Spain
| | - Jose L Garcia Perez
- Pfizer-University of Granada-Andalusian Government Centre for Genomics and Oncological Research (GENYO), PTS Granada, Granada, Spain.
- MRC Human Genetics Unit, Institute of Genetics and Cancer (IGC)/University of Edinburgh, Western General Hospital Campus, Edinburgh, UK.
| |
Collapse
|
3
|
Wongkhieo S, Numdee K, Lam EWF, Choowongkomon K, Kongsema M, Khongkow M. Liposomal Thiostrepton Formulation and Its Effect on Breast Cancer Growth Inhibition. J Pharm Sci 2021; 110:2508-2516. [PMID: 33515584 DOI: 10.1016/j.xphs.2021.01.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/30/2022]
Abstract
Forkhead box M1 (FOXM1) is known to play a role in breast cancer progression. FOXM1 inhibition becomes one of the strategies in developing the novel cancer therapy. Recently, thiostrepton has been recognized as a potent FOXM1 inhibitor. To improve its potential, we aimed to develop a nanodelivery system for thiostrepton. Here, liposome-encapsulated thiostrepton (TSLP) was developed. Physiochemical properties were characterized by TEM and dynamic light scattering technique. The biological activities were also evaluated, by cellular internalization, MTT assay, spheroid formation assay and RT-PCR. The result showed that the range sizes of TSLP were 152 ± 2 nm, polydispersity index (PdI) of 0.23 ± 0.02 and zeta potential of -20.2 ± 0.1 mV. As expected, TSLP showed a higher potential in reducing FOXM1 levels in MCF-7 cells than free thiostrepton. Additionally, TSLP significantly improved the efficiently and specificity of thiostrepton in reducing cell viability of MCF-7, but not of the fibroblast (HDFn) cells. Interestingly, TSLP had an ability to induce MCF-7 cell death in both 2D monolayer and 3D spheroid culture. In conclusions, TSLP could possibly be one of the potential developments using nano-delivery system to improve abilities and specificity of thiostrepton in breast cancer cell inhibition and death inducing, with decreasing non-specific toxicity.
Collapse
Affiliation(s)
- Sudtirak Wongkhieo
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand; National Nanotechnology Centre (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Katawut Numdee
- National Nanotechnology Centre (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Eric W F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Mesayamas Kongsema
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand.
| | - Mattaka Khongkow
- National Nanotechnology Centre (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand.
| |
Collapse
|
4
|
Structure-property relationships of d-mannitol-based cationic poly(amide triazoles) and their self-assembling complexes with DNA. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2019.109458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
5
|
Kongsema M, Wongkhieo S, Khongkow M, Lam EWF, Boonnoy P, Vongsangnak W, Wong-Ekkabut J. Molecular mechanism of Forkhead box M1 inhibition by thiostrepton in breast cancer cells. Oncol Rep 2019; 42:953-962. [PMID: 31322278 PMCID: PMC6667886 DOI: 10.3892/or.2019.7225] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 07/03/2019] [Indexed: 12/16/2022] Open
Abstract
Breast cancer is the most common type of malignancies in women worldwide, and genotoxic chemotherapeutic drugs are effective by causing DNA damage in cancer cells. However, >90% of patients with metastatic cancer are resistant to chemotherapy. The Forkhead box M1 (FOXM1) transcription factor plays a pivotal role in the resistance of breast cancer cells to chemotherapy by promoting DNA damage repair following genotoxic drug treatment. The aim of the present study was to investigate the inhibition of the FOXM1 protein by thiostrepton, a natural antibiotic produced by the Streptomyces species. Experimental studies were designed to examine the effectiveness of thiostrepton in downregulating FOXM1 mRNA expression and activity, leading to senescence and apoptosis of breast cancer cells. The cytotoxicity of thiostrepton in breast cancer was determined using cell viability assay. Additionally, thiostrepton treatment decreased the mRNA expression of cyclin B1 (CCNB1), a downstream target of FOXM1. The present results indicated that thiostrepton inhibited FOXM1 mRNA expression and its effect on CCNB1. Molecular dynamic simulations were performed to study the interactions between FOXM1-DNA and thiostrepton after molecular docking. The results revealed that the possible mechanism underlying the inhibitory effect of thiostrepton on FOXM1 function was by forming a tight complex with the DNA and FOXM1 via its binding domain. Collectively, these results indicated that thiostrepton is a specific and direct inhibitor of the FOXM1 protein in breast cancer. The findings of the present study may lead to the development of novel therapeutic strategies for breast cancer and help overcome resistance to conventional chemotherapeutic drugs.
Collapse
Affiliation(s)
- Mesayamas Kongsema
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Sudtirak Wongkhieo
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Mattaka Khongkow
- National Nanotechnology Centre (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Phansiri Boonnoy
- Computational Biomodelling Laboratory for Agricultural Science and Technology (CBLAST), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Wanwipa Vongsangnak
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Jirasak Wong-Ekkabut
- Computational Biomodelling Laboratory for Agricultural Science and Technology (CBLAST), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| |
Collapse
|
6
|
Wang J, Ren KF, Gao YF, Zhang H, Huang WP, Qian HL, Xu ZK, Ji J. Photothermal Spongy Film for Enhanced Surface-Mediated Transfection to Primary Cells. ACS APPLIED BIO MATERIALS 2019; 2:2676-2684. [DOI: 10.1021/acsabm.9b00358] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jing Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Ke-Feng Ren
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Yi-Fan Gao
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - He Zhang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Wei-Pin Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Hong-Lin Qian
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Zhi-Kang Xu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| |
Collapse
|
7
|
Kim CY, Hwang IK, Kang C, Chung EB, Jung CR, Oh H, Jeong YH, Moon SH, Kim JS, Hong KS, Park JH, Chung HM. Improved Transfection Efficiency and Metabolic Activity in Human Embryonic Stem Cell Using Non-Enzymatic Method. Int J Stem Cells 2018; 11:149-156. [PMID: 30173502 PMCID: PMC6285293 DOI: 10.15283/ijsc18037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/12/2018] [Accepted: 07/12/2018] [Indexed: 01/13/2023] Open
Abstract
Human embryonic stem cells (hESCs) are pluripotent cells widely used in conventional and regenerative medicine due to their ability to self-renew, proliferate and differentiate. Recently, genetic modification of stem cells using genome editing is the most advanced technique for treating hereditary diseases. Nevertheless, the low transfection efficiency of hESCs using enzymatic methods is still limited in in vitro preclinical research. To overcome these limitations, we have developed transfection methods using non-enzymatic treatments on hESCs. In this study, hESCs were transfected following enzymatic (TrypLE and trypsin) and non-enzymatic treatment ethylenediaminetetraacetic acid (EDTA) to increase transfection efficiency. Flow cytometric analysis using an enhanced green fluorescent protein vector showed a significantly increased transfection efficiency of EDTA method compared to standard enzyme method. In addition, the EDTA approach maintained stable cell viability and recovery rate of hESCs after transfection. Also, metabolic activity by using Extracellular Flux Analyzer revealed that EDTA method maintained as similar levels of cell functionality as normal group comparing with enzymatic groups. These results suggest that transfection using EDTA is a more efficient and safe substitute for transfection than the use of standard enzymatic methods.
Collapse
Affiliation(s)
- C-Yoon Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea.,Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - In-Kyu Hwang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Changhee Kang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Eun-Bin Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Cho-Rok Jung
- Gene Therapy Research Unit, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Hanseul Oh
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Young-Hoon Jeong
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Sung-Hwan Moon
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Jong Soo Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Ki-Sung Hong
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Jae-Hak Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Hyung-Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
8
|
Shi B, Xue M, Wang Y, Wang Y, Li D, Zhao X, Li X. An improved method for increasing the efficiency of gene transfection and transduction. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2018; 10:95-104. [PMID: 29755642 PMCID: PMC5943608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 04/11/2018] [Indexed: 06/08/2023]
Abstract
Transfection and transduction using lentivirus has gained attention in biomedical research. To date, how to reach the maximum transfection and viral transduction efficiency is still challenging. Here we compared the transfection and viral transduction efficiency using commercially available transfection reagents including FuGENE 6, Lipofectamine 2000 and Lipofectamine 3000 in different cell lines and primary cultured cells. Enhanced green fluorescent protein (EGFP) was clearly seen in Eppendorf tubes from harvested cells using Lipofectamine 3000 without using a microscope and UV activation. Strong expression of EGFP was observed in HEK293 cells, mouse primary cortical neurons and human umbilical vein endothelial cells (HUVECs) using confocal microscopy. Western blot showed the strongest EGFP expression using cell lysates from Lipofectamine 3000 transfected HEK293 cells and transduced HUVECs compared with Lipofectamine 2000 or FuGENE 6 reagents. Using Cx43 shRNA lentivirus combined with Lipofectamine 3000 transfection reagent, we can achieve about 90% Cx43 knockdown efficacy in HUVECs. Therefore, our results suggest that a much higher transfection and viral transduction efficiency can be attained by using Lipofectamine 3000 transfection reagent.
Collapse
Affiliation(s)
- Baomin Shi
- Department of General Surgery, Tongji Hospital, Tongji UniversityShanghai, China
| | - Mengzhou Xue
- Department of Neurorehabilitation, The Second Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Yi Wang
- Department of Ophthalmology, Taishan Medical UniversityTaian, China
| | - Yufeng Wang
- Department of General Surgery, Tongji Hospital, Tongji UniversityShanghai, China
| | - Davey Li
- University of WaterlooWaterloo, Canada
| | - Xiaomin Zhao
- Institute of Pharmacology, Taishan Medical UniversityTaian, China
| | - Xinbo Li
- Casey Eye Institute, Oregon Health & Science UniversityPortland, Oregon, USA
| |
Collapse
|
9
|
Heffernan C, Maurel P. Lentiviral Transduction of Rat Schwann Cells and Dorsal Root Ganglia Neurons for In Vitro Myelination Studies. Methods Mol Biol 2018; 1739:177-193. [PMID: 29546708 DOI: 10.1007/978-1-4939-7649-2_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Lentiviral transduction is a gene delivery method that provides numerous advantages over direct transfection and traditional retroviral or adenoviral delivery methods. It facilitates for the transduction of primary cells inherently difficult to transfect, delivers constructs of interest to nondividing as well as dividing cells, and permits the long-term expression of sizable DNA inserts (e.g., <7 kb). The study of peripheral nerve myelination at the molecular level has long benefited from the Schwann cells/dorsal root ganglia (DRG) neurons myelinating co-culture system. As this culture system takes about a month to develop and perform experiments with, lentiviral-delivered constructs can be used to manipulate gene expression in Schwann cells and DRG neurons, primary cells that are otherwise resilient to direct transfection. Here we present our protocol for lentiviral production and purification and subsequent infection of large numbers of Schwann cells and/or DRG neurons for the molecular study of peripheral nerve myelination in vitro.
Collapse
Affiliation(s)
- Corey Heffernan
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Patrice Maurel
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA.
| |
Collapse
|
10
|
Fitzgerald M, Gibbs C, Shimpi AA, Deans TL. Adoption of the Q Transcriptional System for Regulating Gene Expression in Stem Cells. ACS Synth Biol 2017; 6:2014-2020. [PMID: 28776984 DOI: 10.1021/acssynbio.7b00149] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The field of mammalian synthetic biology seeks to engineer enabling technologies to create novel approaches for programming cells to probe, perturb, and regulate gene expression with unprecedented precision. To accomplish this, new genetic parts continue to be identified that can be used to build novel genetic circuits to re-engineer cells to perform specific functions. Here, we establish a new transcription-based genetic circuit that combines genes from the quinic acid sensing metabolism of Neorospora crassa and the bacterial Lac repressor system to create a new orthogonal genetic tool to be used in mammalian cells. This work establishes a novel genetic tool, called LacQ, that functions to regulate gene expression in Chinese hamster ovarian (CHO) cells, human embryonic kidney 293 (HEK293) cells, and in mouse embryonic stem (ES) cells.
Collapse
Affiliation(s)
- Michael Fitzgerald
- Department of Bioengineering, University of Utah , Salt Lake City, Utah 84112, United States
| | - Chelsea Gibbs
- Department of Bioengineering, University of Utah , Salt Lake City, Utah 84112, United States
| | - Adrian A Shimpi
- Department of Bioengineering, University of Utah , Salt Lake City, Utah 84112, United States
| | - Tara L Deans
- Department of Bioengineering, University of Utah , Salt Lake City, Utah 84112, United States
| |
Collapse
|
11
|
Khalil AS, Yu X, Xie AW, Fontana G, Umhoefer JM, Johnson HJ, Hookway TA, McDevitt TC, Murphy WL. Functionalization of microparticles with mineral coatings enhances non-viral transfection of primary human cells. Sci Rep 2017; 7:14211. [PMID: 29079806 PMCID: PMC5660152 DOI: 10.1038/s41598-017-14153-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/02/2017] [Indexed: 12/28/2022] Open
Abstract
Gene delivery to primary human cells is a technology of critical interest to both life science research and therapeutic applications. However, poor efficiencies in gene transfer and undesirable safety profiles remain key limitations in advancing this technology. Here, we describe a materials-based approach whereby application of a bioresorbable mineral coating improves microparticle-based transfection of plasmid DNA lipoplexes in several primary human cell types. In the presence of these mineral-coated microparticles (MCMs), we observed up to 4-fold increases in transfection efficiency with simultaneous reductions in cytotoxicity. We identified mechanisms by which MCMs improve transfection, as well as coating compositions that improve transfection in three-dimensional cell constructs. The approach afforded efficient transfection in primary human fibroblasts as well as mesenchymal and embryonic stem cells for both two- and three-dimensional transfection strategies. This MCM-based transfection is an advancement in gene delivery technology, as it represents a non-viral approach that enables highly efficient, localized transfection and allows for transfection of three-dimensional cell constructs.
Collapse
Affiliation(s)
- Andrew S Khalil
- Department of Biomedical Engineering-University of Wisconsin-Madison, Madison, WI, USA
| | - Xiaohua Yu
- Department of Orthopedics and Rehabilitation-University of Wisconsin-Madison, Madison, WI, USA
| | - Angela W Xie
- Department of Biomedical Engineering-University of Wisconsin-Madison, Madison, WI, USA
| | - Gianluca Fontana
- Department of Biomedical Engineering-University of Wisconsin-Madison, Madison, WI, USA
| | - Jennifer M Umhoefer
- Department of Biomedical Engineering-University of Wisconsin-Madison, Madison, WI, USA
| | - Hunter J Johnson
- Department of Biomedical Engineering-University of Wisconsin-Madison, Madison, WI, USA
| | - Tracy A Hookway
- Department of Bioengineering & Therapeutic Sciences-University of California, San Francisco, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology & Medicine-Gladstone Institutes, San Francisco, CA, USA
| | - Todd C McDevitt
- Department of Bioengineering & Therapeutic Sciences-University of California, San Francisco, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology & Medicine-Gladstone Institutes, San Francisco, CA, USA
| | - William L Murphy
- Department of Biomedical Engineering-University of Wisconsin-Madison, Madison, WI, USA.
- Department of Orthopedics and Rehabilitation-University of Wisconsin-Madison, Madison, WI, USA.
- The Materials Science Program-University of Wisconsin-Madison, Madison, WI, USA.
- The Stem Cell and Regenerative Medicine Center-University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
12
|
Macia A, Widmann TJ, Heras SR, Ayllon V, Sanchez L, Benkaddour-Boumzaouad M, Muñoz-Lopez M, Rubio A, Amador-Cubero S, Blanco-Jimenez E, Garcia-Castro J, Menendez P, Ng P, Muotri AR, Goodier JL, Garcia-Perez JL. Engineered LINE-1 retrotransposition in nondividing human neurons. Genome Res 2016; 27:335-348. [PMID: 27965292 PMCID: PMC5340962 DOI: 10.1101/gr.206805.116] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 12/01/2016] [Indexed: 12/21/2022]
Abstract
Half the human genome is made of transposable elements (TEs), whose ongoing activity continues to impact our genome. LINE-1 (or L1) is an autonomous non-LTR retrotransposon in the human genome, comprising 17% of its genomic mass and containing an average of 80-100 active L1s per average genome that provide a source of inter-individual variation. New LINE-1 insertions are thought to accumulate mostly during human embryogenesis. Surprisingly, the activity of L1s can further impact the somatic human brain genome. However, it is currently unknown whether L1 can retrotranspose in other somatic healthy tissues or if L1 mobilization is restricted to neuronal precursor cells (NPCs) in the human brain. Here, we took advantage of an engineered L1 retrotransposition assay to analyze L1 mobilization rates in human mesenchymal (MSCs) and hematopoietic (HSCs) somatic stem cells. Notably, we have observed that L1 expression and engineered retrotransposition is much lower in both MSCs and HSCs when compared to NPCs. Remarkably, we have further demonstrated for the first time that engineered L1s can retrotranspose efficiently in mature nondividing neuronal cells. Thus, these findings suggest that the degree of somatic mosaicism and the impact of L1 retrotransposition in the human brain is likely much higher than previously thought.
Collapse
Affiliation(s)
- Angela Macia
- Department of Genomic Medicine and Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain
| | - Thomas J Widmann
- Department of Genomic Medicine and Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain
| | - Sara R Heras
- Department of Genomic Medicine and Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain
| | - Veronica Ayllon
- Department of Oncology, GENYO, Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain
| | - Laura Sanchez
- Department of Genomic Medicine and Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain
| | - Meriem Benkaddour-Boumzaouad
- Department of Genomic Medicine and Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain
| | - Martin Muñoz-Lopez
- Department of Genomic Medicine and Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain
| | - Alejandro Rubio
- Department of Genomic Medicine and Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain
| | - Suyapa Amador-Cubero
- Department of Genomic Medicine and Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain
| | - Eva Blanco-Jimenez
- Department of Genomic Medicine and Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain
| | | | - Pablo Menendez
- Department of Oncology, GENYO, Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain.,Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Instituciò Catalana Recerca Estudis Avançats (ICREA), 08036 Barcelona, Spain
| | - Philip Ng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Alysson R Muotri
- Department of Pediatrics/Rady Children's Hospital San Diego, University of California San Diego, La Jolla, California 92093, USA
| | - John L Goodier
- McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Jose L Garcia-Perez
- Department of Genomic Medicine and Centre for Genomics and Oncology (Pfizer-University of Granada and Andalusian Regional Government), PTS Granada, 18016 Granada, Spain.,Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| |
Collapse
|
13
|
Niakan S, Heidari B, Akbari G, Nikousefat Z. Comparison of Different Electroporation Parameters on Transfection Efficiency of Sheep Testicular Cells. CELL JOURNAL 2016; 18:425-37. [PMID: 27602325 PMCID: PMC5011331 DOI: 10.22074/cellj.2016.4571] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 11/28/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Electroporation can be a highly efficient method for introducing the foreign genetic materials into the targeted cells for transient and/or permanent genetic modification. Considering the application of this technique as a very efficient method for drug, oligonucleotide, antibody and plasmid delivery for clinical applications and production of transgenic animals, the present study aimed to optimize the transfection efficiency of sheep testicular cells including spermatogonial stem cells (SSCs) via electroporation. MATERIALS AND METHODS This study is an experimental research conducted in Biotechnology Research Center (Avicenna Research Institute, Tehran, Iran) from September 2013 to March 2014. Following isolation and propagation of one-month lamb testicular cells (SSCs and somatic testicular cells including; Sertoli, Leydig, and myoid cells), the effect of different electroporation parameters including total voltages (280, 320, and 350 V), burst durations (10, 8, and 5 milliseconds), burst modes (single or double) and addition of dimethyl sulfoxide (DMSO) were evaluated on transfection efficiency, viability rate and mean fluorescent intensity (MFI) of sheep testicular cells. RESULTS The most transfection efficiency was obtained in 320 V/8 milliseconds/single burst group in transduction medium with and without DMSO. There was a significantly inverse correlation between transfection efficiency with application of both following parameters: addition of DMSO and double burst. After transfection, the highest and lowest viability rates of testicular cells were demonstrated in 320 V/8 milliseconds with transduction medium without DMSO and 350 V/5 milliseconds in medium containing DMSO. Ad- dition of DMSO to transduction medium in all groups significantly decreased the viability rate. The comparison of gene expression indicated that Sertoli and SSCs had the most fluorescence intensity in 320 V/double burst/DMSO positive. However, myoid and Leydig cells showed the maximum expression in 320 V/single burst and/or 350 V/double burst/ DMSO positive. CONCLUSION We optimized the electroporation method for transfection of sheep testicular cells and recommended the application of 320 V/8 milliseconds/single pulse/DMSO negative for transduction of plasmid vector into these cells. Among testicular cells, the most external gene expression was demonstrated in SSC population.
Collapse
Affiliation(s)
- Sarah Niakan
- Department of Clinical Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Banafsheh Heidari
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ghasem Akbari
- Department of Clinical Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Nikousefat
- Department of Clinical Science, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| |
Collapse
|
14
|
Reibetanz U, Hübner D, Jung M, Liebert UG, Claus C. Influence of Growth Characteristics of Induced Pluripotent Stem Cells on Their Uptake Efficiency for Layer-by-Layer Microcarriers. ACS NANO 2016; 10:6563-6573. [PMID: 27362252 DOI: 10.1021/acsnano.6b00999] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Induced pluripotent stem cells (iPSCs) have the ability to differentiate into any specialized somatic cell type, which makes them an attractive tool for a wide variety of scientific approaches, including regenerative medicine. However, their pluripotent state and their growth in compact colonies render them difficult to access and, therefore, restrict delivery of specific agents for cell manipulation. Thus, our investigation focus was set on the evaluation of the capability of layer-by-layer (LbL) designed microcarriers to serve as a potential drug delivery system to iPSCs, as they offer several appealing advantages. Most notably, these carriers allow for the transport of active agents in a protected environment and for a rather specific delivery through surface modifications. As we could show, charge and mode of LbL carrier application as well as the size of the iPSC colonies determine the interaction with and the uptake rate by iPSCs. None of the examined conditions had an influence on iPSC colony properties such as colony morphology and size or maintenance of pluripotent properties. An overall interaction rate of LbL carriers with iPSCs of up to 20% was achieved. Those data emphasize the applicability of LbL carriers for stem cell research. Additionally, the potential use of LbL carriers as a promising delivery tool for iPSCs was contrasted to viral particles and liposomes. The identified differences among those delivery tools have substantiated our major conclusion that LbL carrier uptake rate is influenced by characteristic features of the iPSC colonies (most notably colony size) in addition to their surface charges.
Collapse
Affiliation(s)
- Uta Reibetanz
- Institute for Medical Physics and Biophysics, Faculty of Medicine, University of Leipzig , 04107 Leipzig, Germany
| | - Denise Hübner
- Institute of Virology, University of Leipzig , 04103 Leipzig, Germany
| | - Matthias Jung
- Department of Psychiatry, University of Halle-Wittenberg , Halle, Germany
| | - Uwe Gerd Liebert
- Institute of Virology, University of Leipzig , 04103 Leipzig, Germany
| | - Claudia Claus
- Institute of Virology, University of Leipzig , 04103 Leipzig, Germany
| |
Collapse
|
15
|
Nii T, Kohara H, Marumoto T, Sakuma T, Yamamoto T, Tani K. Single-Cell-State Culture of Human Pluripotent Stem Cells Increases Transfection Efficiency. Biores Open Access 2016; 5:127-36. [PMID: 27257519 PMCID: PMC4876534 DOI: 10.1089/biores.2016.0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Efficient gene transfer into human pluripotent stem cells (hPSCs) holds great promise for regenerative medicine and pharmaceutical development. In the past decade, various methods were developed for gene transfer into hPSCs; however, hPSCs form tightly packed colonies, making gene transfer difficult. In this study, we established a stable culture method of hPSCs at a single-cell state to reduce cell density and investigated gene transfection efficiency followed by gene editing efficiency. hPSCs cultured in a single-cell state were transfected using nonliposomal transfection reagents with plasmid DNA or mRNA encoding enhanced green fluorescent protein. We found that most cells (DNA > 90%; mRNA > 99%) were transfected without the loss of undifferentiated PSC marker expression or pluripotency. Moreover, we demonstrated an efficient gene editing method using transcription activator-like effector nucleases (TALENs) targeting the adenomatous polyposis coli (APC) gene. Our new method may improve hPSC gene transfer techniques, thus facilitating their use for human regenerative medicine.
Collapse
Affiliation(s)
- Takenobu Nii
- Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University , Fukuoka, Japan
| | - Hiroshi Kohara
- Project Division of ALA Advanced Medical Research, The Institute of Medical Science, The University of Tokyo , Tokyo, Japan
| | - Tomotoshi Marumoto
- Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.; Department of Advanced Molecular and Cell Therapy, Kyushu University Hospital, Fukuoka, Japan
| | - Tetsushi Sakuma
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University , Hiroshima, Japan
| | - Takashi Yamamoto
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University , Hiroshima, Japan
| | - Kenzaburo Tani
- Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.; Project Division of ALA Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.; Department of Advanced Molecular and Cell Therapy, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
16
|
Liu H, Ren C, Zhu B, Wang L, Liu W, Shi J, Lin J, Xia X, Zeng F, Chen J, Jiang X. High-Efficient Transfection of Human Embryonic Stem Cells by Single-Cell Plating and Starvation. Stem Cells Dev 2016; 25:477-91. [PMID: 26772602 DOI: 10.1089/scd.2015.0301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nowadays, the low efficiency of small interfering RNA (siRNA) or plasmid DNA (pDNA) transfection is a critical issue in genetic manipulation of human embryonic stem (hES) cells. Development of an efficient transfection method for delivery of siRNAs and plasmids into hES cells becomes more and more imperative. In this study, we tried to modify the traditional transfection protocol by introducing two crucial processes, single-cell plating and starvation, to increase the transfection efficiency in hES cells. Furthermore, we comparatively examined the transfection efficiency of some commercially available siRNA or pDNA transfection reagents in hES cells. Our results showed that the new developed method markedly enhanced the transfection efficiency without influencing the proliferation and pluripotency of hES cells. Lipofectamine RNAiMAX exhibited much higher siRNA transfection efficiency than the other reagents, and FuGENE HD was identified as the best suitable reagent for efficient pDNA transfection of hES cells among the tested reagents.
Collapse
Affiliation(s)
- Hui Liu
- 1 Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University , Changsha, People's Republic of China
| | - Caiping Ren
- 1 Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University , Changsha, People's Republic of China
| | - Bin Zhu
- 1 Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University , Changsha, People's Republic of China
| | - Lei Wang
- 1 Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University , Changsha, People's Republic of China
| | - Weidong Liu
- 1 Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University , Changsha, People's Republic of China
| | - Jia Shi
- 1 Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University , Changsha, People's Republic of China
| | - Jianxing Lin
- 1 Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University , Changsha, People's Republic of China
| | - Xiaomeng Xia
- 2 Department of Gynecology and Obstetrics, the Second Xiangya Hospital, Central South University , Changsha, People's Republic of China
| | - Fei Zeng
- 3 Department of Gynecology and Obstetrics, the Third Xiangya Hospital, Central South University , Changsha, People's Republic of China
| | - Jiawen Chen
- 1 Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University , Changsha, People's Republic of China
| | - Xingjun Jiang
- 4 Department of Neurosurgery, Xiangya Hospital, Central South University , Changsha, People's Republic of China
| |
Collapse
|
17
|
Qian X, Kim JK, Tong W, Villa-Diaz LG, Krebsbach PH. DPPA5 Supports Pluripotency and Reprogramming by Regulating NANOG Turnover. Stem Cells 2015; 34:588-600. [PMID: 26661329 DOI: 10.1002/stem.2252] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 09/19/2015] [Accepted: 10/18/2015] [Indexed: 01/16/2023]
Abstract
Although a specific group of transcription factors such as OCT4, SOX2, and NANOG are known to play essential roles in pluripotent stem cell (PSC) self-renewal, pluripotency, and reprogramming, other factors and the key signaling pathways regulating these important properties are not completely understood. Here, we demonstrate that the PSC marker Developmental Pluripotency Associated 5 (DPPA5) plays an important role in human PSC (hPSC) self-renewal and cell reprogramming in feeder-free conditions. Compared to hPSCs grown on mouse embryonic fibroblasts, cells cultured on feeder-free substrates, such as Matrigel, Laminin-511, Vitronectin, or the synthetic polymer poly[2-(methacryloyloxy) ethyl dimethyl-(3-sulfopropyl) ammonium hydroxide], had significantly higher DPPA5 gene expression and protein levels. Overexpression of DPPA5 in hPSCs increased NANOG protein levels via a post-transcriptional mechanism. Coimmunoprecipitation, protein stability assays, and quantitative RT-PCR, demonstrated that DPPA5 directly interacted, stabilized, and enhanced the function of NANOG in hPSCs. Additionally, DPPA5 increased the reprogramming efficiency of human somatic cells to induced pluripotent stem cells (hiPSCs). Our study provides new insight into the function of DPPA5 and NANOG regulation in hPSCs.
Collapse
Affiliation(s)
- Xu Qian
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Jin Koo Kim
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Wilbur Tong
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Luis G Villa-Diaz
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Paul H Krebsbach
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
18
|
Loh XJ, Wu YL. Cationic star copolymers based on β-cyclodextrins for efficient gene delivery to mouse embryonic stem cell colonies. Chem Commun (Camb) 2015; 51:10815-8. [DOI: 10.1039/c5cc03686k] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A cationic star copolymer with a β-cyclodextrin core was developed for nonviral gene transfer to mouse embryonic stem cells (mESCs).
Collapse
Affiliation(s)
- Xian Jun Loh
- Institute of Materials Research and Engineering
- A*STAR (Agency for Science, Technology and Research)
- Singapore 117602
- Singapore
- Department of Materials Science and Engineering
| | - Yun-Long Wu
- School of Pharmaceutical Sciences
- Xiamen University
- Xiamen
- P. R. China
| |
Collapse
|
19
|
Ochoa GP, Sesma JZ, Díez MA, Díaz-Tahoces A, Avilés-Trigeros M, Grijalvo S, Eritja R, Fernández E, Pedraz JL. A novel formulation based on 2,3-di(tetradecyloxy)propan-1-amine cationic lipid combined with polysorbate 80 for efficient gene delivery to the retina. Pharm Res 2014; 31:1665-75. [PMID: 24449439 DOI: 10.1007/s11095-013-1271-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/19/2013] [Indexed: 11/28/2022]
Abstract
PURPOSE The aim of the present study was to evaluate the potential application of a novel formulation based on a synthesized cationic lipid 2,3-di(tetradecyloxy)propan-1-amine, combined with polysorbate 80 to deliver the pCMS-EGFP plasmid into the rat retina. METHODS We elaborated lipoplexes by mixing the formulation containing the cationic lipid and the polysorbate 80 with the plasmid at different cationic lipid/DNA ratios (w/w). Resulted lipoplexes were characterized in terms of size, charge, and capacity to condense, protect and release the DNA. In vitro transfection studies were performed in HEK-293 and ARPE-19 cells. Formulations were also tested in vivo by monitoring the expression of the EGFP after intravitreal and subretinal injections in rat eyes. RESULTS At 2/1 cationic lipid/DNA mass ratio, the resulted lipoplexes had 200 nm of hydrodynamic diameter; were positive charged, spherical, protected DNA against enzymatic digestion and transfected efficiently HEK-293 and ARPE-19 cultured cells exhibiting lower cytotoxicity than LipofectamineTM 2000. Subretinal administrations transfected mainly photoreceptors and retinal pigment epithelial cells; whereas intravitreal injections produced a more uniform distribution of transfection through the inner part of the retina. CONCLUSIONS These results hold great expectations for other gene delivery formulations based on this cationic lipid for retinal gene therapy purposes.
Collapse
|
20
|
Preparation of cell-culturing glass surfaces that release branched polyethyleneimine triggered by thiol-disulfide exchange. Colloids Surf B Biointerfaces 2013; 103:360-5. [PMID: 23261556 DOI: 10.1016/j.colsurfb.2012.10.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 10/18/2012] [Accepted: 10/18/2012] [Indexed: 11/21/2022]
Abstract
To develop a chemical stimulus-responsive substrate for culturing cells, polyethyleneimine (PEI) having a pyridyl disulfide moiety was attached via disulfide linkages to a glass coverslip modified with a silane coupling agent having a thiol group. The surface modification was confirmed by X-ray photoelectron spectroscopy and zeta potential analysis. The obtained surface exhibited sufficiently high cell adhesiveness. Zeta potential measurements revealed that the PEI derivatives were released from the surface through thiol-disulfide exchange when the modified glass coverslip was immersed in a neutral pH buffer containing cysteine. The cell viability assay demonstrated that this chemical stimulus was substantially nontoxic to 293T cells. Because PEI is a widely used transfection reagent, this functional glass coverslip would be potentially useful as an experimental platform for reverse transfection.
Collapse
|
21
|
Jin S, Yao H, Krisanarungson P, Haukas A, Ye K. Porous membrane substrates offer better niches to enhance the Wnt signaling and promote human embryonic stem cell growth and differentiation. Tissue Eng Part A 2012; 18:1419-30. [PMID: 22429220 DOI: 10.1089/ten.tea.2011.0474] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Human embryonic stem cells (hESCs) require specific niches for adhesion, expansion, and lineage-specific differentiation. In this study, we showed that a membrane substrate offers better tissue niches for hESC attachment, spreading, proliferation, and differentiation. The cell doubling time was shortened from 46.3±5.7 h for hESCs grown on solid substrates to 25.6±2.6 h for those on polyester (PE) membrane substrates with pore size of 0.4 μm. In addition, we observed an increase of approximately five- to ninefold of definitive endoderm marker gene expression in hESCs differentiated on PE or polyethylene terephthalate membrane substrates. Global gene expression analysis revealed upregulated expressions of a number of extracellular matrix and cell adhesion molecules in hESCs grown on membrane substrates. Further, an enhanced nuclear translocation of β-catenin was detected in these cells. These observations suggested the augmentation of Wnt signaling in hESCs grown on membrane substrates. These results also demonstrated that a membrane substrate can offer better physicochemical cues for enhancing in vitro hESC attachment, proliferation, and differentiation.
Collapse
Affiliation(s)
- Sha Jin
- Biomedical Engineering Program, College of Engineering, University of Arkansas, 700 Research Center Blvd., Fayetteville, AR 72701, USA.
| | | | | | | | | |
Collapse
|
22
|
Ji Q, Yamazaki T, Hanagata N, Lee MV, Hill JP, Ariga K. Silica-based gene reverse transfection: an upright nanosheet network for promoted DNA delivery to cells. Chem Commun (Camb) 2012; 48:8496-8. [DOI: 10.1039/c2cc34289h] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
23
|
Arnaoutova I, George J, Kleinman HK, Benton G. Basement Membrane Matrix (BME) has Multiple Uses with Stem Cells. Stem Cell Rev Rep 2011; 8:163-9. [DOI: 10.1007/s12015-011-9278-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
24
|
Tang L, Yang Y, Bai T, Liu W. Robust MeO2MA/vinyl-4,6-diamino-1,3,5-triazine copolymer hydrogels-mediated reverse gene transfection and thermo-induced cell detachment. Biomaterials 2011; 32:1943-9. [DOI: 10.1016/j.biomaterials.2010.11.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 11/02/2010] [Indexed: 01/17/2023]
|