1
|
Chen H, Xie Y, Zhang M, Huang J, Jiang W, Zhang R, Li C, Du X, Chen H, Nie Q, Liang S, Tan Q, Yang J, Jin M, Huang S, Kuang L, Su N, Qi H, Luo X, Xu X, Deng C, Chen L, Luo F. An Hsp70 promoter-based mouse for heat shock-induced gene modulation. J Mol Med (Berl) 2024; 102:693-707. [PMID: 38492027 DOI: 10.1007/s00109-024-02433-9] [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: 01/04/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
Physical therapy is extensively employed in clinical settings. Nevertheless, the absence of suitable animal models has resulted in an incomplete understanding of the in vivo mechanisms and cellular distribution that respond to physical stimuli. The objective of this research was to create a mouse model capable of indicating the cells affected by physical stimuli. In this study, we successfully established a mouse line based on the heat shock protein 70 (Hsp70) promoter, wherein the expression of CreERT2 can be induced by physical stimuli. Following stimulation of the mouse tail, ear, or cultured calvarias with heat shock (generated by heating, ultrasound, or laser), a distinct Cre-mediated excision was observed in cells stimulated by these physical factors with minimal occurrence of leaky reporter expression. The application of heat shock to Hsp70-CreERT2; FGFR2-P253R double transgenic mice or Hsp70-CreERT2 mice infected with AAV-BMP4 at calvarias induced the activation of Cre-dependent mutant FGFR2-P253R or BMP4 respectively, thereby facilitating the premature closure of cranial sutures or the repair of calvarial defects. This novel mouse line holds significant potential for investigating the underlying mechanisms of physical therapy, tissue repair and regeneration, lineage tracing, and targeted modulation of gene expression of cells in local tissue stimulated by physical factor at the interested time points. KEY MESSAGES: In the study, an Hsp70-CreERT2 transgenic mouse was generated for heat shock-induced gene modulation. Heat shock, ultrasound, and laser stimulation effectively activated Cre expression in Hsp70-CreERT2; reporter mice, which leads to deletion of floxed DNA sequence in the tail, ear, and cultured calvaria tissues of mice. Local laser stimuli on cultured calvarias effectively induce Fgfr2-P253R expression in Hsp70-mTmG-Fgfr2-P253R mice and result in accelerated premature closure of cranial suture. Heat shock activated AAV9-FLEX-BMP4 expression and subsequently promoted the repair of calvarial defect of Hsp70-CreERT2; Rosa26-mTmG mice.
Collapse
Affiliation(s)
- Hangang Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yangli Xie
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Mei Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Junlan Huang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Wanling Jiang
- Department of Chinese Medicine Rehabilitation, Chongqing Emergency Medical Center, Chongqing University Central Hospital), Chongqing, 400042, China
| | - Ruobin Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Can Li
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiaolan Du
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Hua Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Qiang Nie
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Sen Liang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Qiaoyan Tan
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Jing Yang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Min Jin
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Shuo Huang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Liang Kuang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Nan Su
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Huabing Qi
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Xiaoqing Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiaoling Xu
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chuxia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lin Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China.
| | - Fengtao Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
2
|
Rodgers TM, Muzzio N, Valero A, Ahmad I, Lüdtke TU, Moya SE, Romero G. Poly (β-amino Ester) Nanoparticles Modified with a Rabies Virus-derived peptide for the Delivery of ASCL1 Across a 3D In Vitro Model of the Blood Brain Barrier. ACS APPLIED NANO MATERIALS 2023; 6:6299-6311. [PMID: 37274933 PMCID: PMC10234607 DOI: 10.1021/acsanm.3c00651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Gene editing has emerged as a therapeutic approach to manipulate the genome for killing cancer cells, protecting healthy tissues, and improving immune response to a tumor. The gene editing tool achaete-scute family bHLH transcription factor 1 CRISPR guide RNA (ASCL1-gRNA) is known to restore neuronal lineage potential, promote terminal differentiation, and attenuate tumorigenicity in glioblastoma tumors. Here, we fabricated a polymeric nonviral carrier to encapsulate ASCL1-gRNA by electrostatic interactions and deliver it into glioblastoma cells across a 3D in vitro model of the blood-brain barrier (BBB). To mimic rabies virus (RV) neurotropism, gene-loaded poly (β-amino ester) nanoparticles are surface functionalized with a peptide derivative of rabies virus glycoprotein (RVG29). The capability of the obtained NPs, hereinafter referred to as RV-like NPs, to travel across the BBB, internalize into glioblastoma cells and deliver ASCL1-gRNA are investigated in a 3D BBB in vitro model through flow cytometry and CLSM microscopy. The formation of nicotinic acetylcholine receptors in the 3D BBB in vitro model is confirmed by immunochemistry. These receptors are known to bind to RVG29. Unlike Lipofectamine that primarily internalizes and transfects endothelial cells, RV-like NPs are capable to travel across the BBB, preferentially internalize glioblastoma cells and deliver ASCL1-gRNA at an efficiency of 10 % causing non-cytotoxic effects.
Collapse
Affiliation(s)
- Tina M Rodgers
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Nicolas Muzzio
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Andrea Valero
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Ikram Ahmad
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Tanja Ursula Lüdtke
- Soft Matter Nanotechnology, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramon 182, Donostia/San Sebastian, Gipuzkoa, 20014 Spain
| | - Sergio E Moya
- Soft Matter Nanotechnology, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramon 182, Donostia/San Sebastian, Gipuzkoa, 20014 Spain
| | - Gabriela Romero
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| |
Collapse
|
3
|
Chen WCW, Gaidukov L, Lai Y, Wu MR, Cao J, Gutbrod MJ, Choi GCG, Utomo RP, Chen YC, Wroblewska L, Kellis M, Zhang L, Weiss R, Lu TK. A synthetic transcription platform for programmable gene expression in mammalian cells. Nat Commun 2022; 13:6167. [PMID: 36257931 PMCID: PMC9579178 DOI: 10.1038/s41467-022-33287-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/13/2022] [Indexed: 12/24/2022] Open
Abstract
Precise, scalable, and sustainable control of genetic and cellular activities in mammalian cells is key to developing precision therapeutics and smart biomanufacturing. Here we create a highly tunable, modular, versatile CRISPR-based synthetic transcription system for the programmable control of gene expression and cellular phenotypes in mammalian cells. Genetic circuits consisting of well-characterized libraries of guide RNAs, binding motifs of synthetic operators, transcriptional activators, and additional genetic regulatory elements express mammalian genes in a highly predictable and tunable manner. We demonstrate the programmable control of reporter genes episomally and chromosomally, with up to 25-fold more activity than seen with the EF1α promoter, in multiple cell types. We use these circuits to program the secretion of human monoclonal antibodies and to control T-cell effector function marked by interferon-γ production. Antibody titers and interferon-γ concentrations significantly correlate with synthetic promoter strengths, providing a platform for programming gene expression and cellular function in diverse applications.
Collapse
Affiliation(s)
- William C W Chen
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA.
| | - Leonid Gaidukov
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Yong Lai
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ming-Ru Wu
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
| | - Jicong Cao
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael J Gutbrod
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA
| | - Gigi C G Choi
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Laboratory of Combinatorial Genetics and Synthetic Biology, School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Rachel P Utomo
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biochemistry, Wellesley College, Wellesley, MA, 02481, USA
| | - Ying-Chou Chen
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | | | - Manolis Kellis
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA
| | - Lin Zhang
- Pfizer Inc., Andover, MA, 01810, USA
| | - Ron Weiss
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Timothy K Lu
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
4
|
Redirection of the Transcription Factor SP1 to AT Rich Binding Sites by a Synthetic Adaptor Molecule. Helv Chim Acta 2021. [DOI: 10.1002/hlca.202100095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
5
|
Park G, Kang B, Park SV, Lee D, Oh SS. A unified computational view of DNA duplex, triplex, quadruplex and their donor-acceptor interactions. Nucleic Acids Res 2021; 49:4919-4933. [PMID: 33893806 PMCID: PMC8136788 DOI: 10.1093/nar/gkab285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 04/07/2021] [Accepted: 04/14/2021] [Indexed: 01/09/2023] Open
Abstract
DNA can assume various structures as a result of interactions at atomic and molecular levels (e.g., hydrogen bonds, π–π stacking interactions, and electrostatic potentials), so understanding of the consequences of these interactions could guide development of ways to produce elaborate programmable DNA for applications in bio- and nanotechnology. We conducted advanced ab initio calculations to investigate nucleobase model structures by componentizing their donor-acceptor interactions. By unifying computational conditions, we compared the independent interactions of DNA duplexes, triplexes, and quadruplexes, which led us to evaluate a stability trend among Watson–Crick and Hoogsteen base pairing, stacking, and even ion binding. For a realistic solution-like environment, the influence of water molecules was carefully considered, and the potassium-ion preference of G-quadruplex was first analyzed at an ab initio level by considering both base-base and ion-water interactions. We devised new structure factors including hydrogen bond length, glycosidic vector angle, and twist angle, which were highly effective for comparison between computationally-predicted and experimentally-determined structures; we clarified the function of phosphate backbone during nucleobase ordering. The simulated tendency of net interaction energies agreed well with that of real world, and this agreement validates the potential of ab initio study to guide programming of complicated DNA constructs.
Collapse
Affiliation(s)
- Gyuri Park
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Byunghwa Kang
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Soyeon V Park
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Donghwa Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea.,Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea.,Institute for Convergence Research and Education in Advanced Technology (I-CREATE), Yonsei University, Incheon 21983, South Korea
| | - Seung Soo Oh
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea.,Institute for Convergence Research and Education in Advanced Technology (I-CREATE), Yonsei University, Incheon 21983, South Korea.,School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| |
Collapse
|
6
|
Effect of Inducible BMP-7 Expression on the Osteogenic Differentiation of Human Dental Pulp Stem Cells. Int J Mol Sci 2021; 22:ijms22126182. [PMID: 34201124 PMCID: PMC8229115 DOI: 10.3390/ijms22126182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/31/2021] [Accepted: 06/04/2021] [Indexed: 12/19/2022] Open
Abstract
BMP-7 has shown inductive potential for in vitro osteogenic differentiation of mesenchymal stem cells, which are an ideal resource for regenerative medicine. Externally applied, recombinant BMP-7 was able to induce the osteogenic differentiation of DPSCs but based on our previous results with BMP-2, we aimed to study the effect of the tetracyclin-inducible BMP-7 expression on these cells. DPSC, mock, and DPSC-BMP-7 cell lines were cultured in the presence or absence of doxycycline, then alkaline phosphatase (ALP) activity, mineralization, and mRNA levels of different osteogenic marker genes were measured. In the DPSC-BMP-7 cell line, the level of BMP-7 mRNA significantly increased in the media supplemented with doxycycline, however, the expression of Runx2 and noggin genes was upregulated only after 21 days of incubation in the osteogenic medium with doxycycline. Moreover, while the examination of ALP activity showed reduced activity in the control medium containing doxycycline, the accumulation of minerals remained unchanged in the cultures. We have found that the induced BMP-7 expression failed to induce osteogenic differentiation of DPSCs. We propose three different mechanisms that may worth investigating for the engineering of expression systems that can be used for the induction of differentiation of mesenchymal stem cells.
Collapse
|
7
|
Gamboa L, Zamat AH, Kwong GA. Synthetic immunity by remote control. Theranostics 2020; 10:3652-3667. [PMID: 32206114 PMCID: PMC7069089 DOI: 10.7150/thno.41305] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 02/03/2020] [Indexed: 12/11/2022] Open
Abstract
Cell-based immunotherapies, such as T cells engineered with chimeric antigen receptors (CARs), have the potential to cure patients of disease otherwise refractory to conventional treatments. Early-on-treatment and long-term durability of patient responses depend critically on the ability to control the potency of adoptively transferred T cells, as overactivation can lead to complications like cytokine release syndrome, and immunosuppression can result in ineffective responses to therapy. Drugs or biologics (e.g., cytokines) that modulate immune activity are limited by mass transport barriers that reduce the local effective drug concentration, and lack site or target cell specificity that results in toxicity. Emerging technologies that enable site-targeted, remote control of key T cell functions - including proliferation, antigen-sensing, and target-cell killing - have the potential to increase treatment precision and safety profile. These technologies are broadly applicable to other immune cells to expand immune cell therapies across many cancers and diseases. In this review, we highlight the opportunities, challenges and the current state-of-the-art for remote control of synthetic immunity.
Collapse
Affiliation(s)
- Lena Gamboa
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Ali H. Zamat
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Gabriel A. Kwong
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Georgia Immunoengineering Consortium, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
8
|
Choi J, Bae T, Byambasuren N, Park SH, Jo CH, Kim D, Hur JK, Hwang NS. CRISPR-Cpf1 Activation of Endogenous BMP4 Gene for Osteogenic Differentiation of Umbilical-Cord-Derived Mesenchymal Stem Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:309-316. [PMID: 32021879 PMCID: PMC6994413 DOI: 10.1016/j.omtm.2019.12.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022]
Abstract
The CRISPR systems provide powerful genome-editing tools for wide applications in biological and medical research fields. However, the safety issue due to off-target effects of CRISPR has been one of the major hindrances of its application to regenerative medicine. The conventional CRISPR system has the intrinsic danger of inducing unpredictable mutations at non-targeted genomic loci via erroneous double-strand DNA breaks (DSBs). In this study, we demonstrate a safety-enhanced application of a recently discovered CRISPR-Cpf1 for targeted gene activation, without DNA double-strand break, to facilitate osteogenic differentiation of human umbilical-cord-derived mesenchymal stem cells (UC-MSCs). To this end, we developed a catalytically inactive AsCpf1 fused to tripartite transcription activator domain (dAsCpf1-VPR) that can induce upregulation of targeted gene expression in mammalian cells. We observed that the CRISPR-dAsCpf1-VPR activator can be applied to enhance the osteogenic differentiation of human UC-MSCs, via increasing the expression level of endogenous BMP4 gene. The results suggested that the CRISPR-Cpf1 activator provides versatile methods applicable for bone regeneration and regenerative medicine.
Collapse
Affiliation(s)
- Jaehoon Choi
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, Korea
| | - Taegeun Bae
- Department of Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Ninj Byambasuren
- Interdisciplinary Program in Bioengineering, Institute of Bio Engineering, Seoul National University, Seoul, Korea
| | - Seong-Ho Park
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Chris H Jo
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Dokyoung Kim
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Korea.,Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea.,Center for Converging Humanities, Kyung Hee University, Seoul, Korea.,Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Kyung Hee University, Seoul, Korea
| | - Junho K Hur
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea.,Department of Pathology, College of Medicine, Kyung Hee Unversity, Seoul, Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, Korea.,Interdisciplinary Program in Bioengineering, Institute of Bio Engineering, Seoul National University, Seoul, Korea
| |
Collapse
|
9
|
Lee JH, Park SJ, Choi JW. Electrical Property of Graphene and Its Application to Electrochemical Biosensing. NANOMATERIALS 2019; 9:nano9020297. [PMID: 30791566 PMCID: PMC6409852 DOI: 10.3390/nano9020297] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/13/2019] [Accepted: 02/18/2019] [Indexed: 12/27/2022]
Abstract
Graphene, a single atom thick layer of two-dimensional closely packed honeycomb carbon lattice, and its derivatives have attracted much attention in the field of biomedical, due to its unique physicochemical properties. The valuable physicochemical properties, such as high surface area, excellent electrical conductivity, remarkable biocompatibility and ease of surface functionalization have shown great potentials in the applications of graphene-based bioelectronics devices, including electrochemical biosensors for biomarker analysis. In this review, we will provide a selective overview of recent advances on synthesis methods of graphene and its derivatives, as well as its application to electrochemical biosensor development. We believe the topics discussed here are useful, and able to provide a guideline in the development of novel graphene and on graphene-like 2-dimensional (2D) materials based biosensors in the future.
Collapse
Affiliation(s)
- Jin-Ho Lee
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Korea.
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| | - Soo-Jeong Park
- Research Center for Disease Biophysics of Sogang-Harvard, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Korea.
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Korea.
- Research Center for Disease Biophysics of Sogang-Harvard, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Korea.
| |
Collapse
|
10
|
Opportunities in biotechnology. J Biotechnol 2018; 282:38-45. [DOI: 10.1016/j.jbiotec.2018.06.303] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/28/2018] [Accepted: 06/05/2018] [Indexed: 12/21/2022]
|
11
|
|