201
|
Spiegel A, Bachmann M, Jurado Jiménez G, Sarov M. CRISPR/Cas9-based knockout pipeline for reverse genetics in mammalian cell culture. Methods 2019; 164-165:49-58. [PMID: 31051255 DOI: 10.1016/j.ymeth.2019.04.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/04/2019] [Accepted: 04/24/2019] [Indexed: 12/14/2022] Open
Abstract
We present a straightforward protocol for reverse genetics in cultured mammalian cells, using CRISPR/Cas9-mediated homology-dependent repair (HDR) based insertion of a protein trap cassette, resulting in a termination of the endogenous gene expression. Complete loss of function can be achieved with monoallelic trap cassette insertion, as the second allele is frequently disrupted by an error-prone non-homologous end joining (NHEJ) mechanism. The method should be applicable to any expressed gene in most cell lines, including those with low HDR efficiency, as the knockout alleles can be directly selected for.
Collapse
Affiliation(s)
- Aleksandra Spiegel
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Mandy Bachmann
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Gabriel Jurado Jiménez
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Mihail Sarov
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany.
| |
Collapse
|
202
|
Ahmad N, Rahman M, Mukhtar Z, Zafar Y, Zhang B. A critical look on CRISPR‐based genome editing in plants. J Cell Physiol 2019; 235:666-682. [DOI: 10.1002/jcp.29052] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 02/12/2019] [Accepted: 06/06/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Niaz Ahmad
- Agricultural Biotechnology Division National Institute for Biotechnology & Genetic Engineering (NIBGE) Faisalabad Pakistan
| | - Mehboob‐ur Rahman
- Agricultural Biotechnology Division National Institute for Biotechnology & Genetic Engineering (NIBGE) Faisalabad Pakistan
| | - Zahid Mukhtar
- Agricultural Biotechnology Division National Institute for Biotechnology & Genetic Engineering (NIBGE) Faisalabad Pakistan
| | - Yusuf Zafar
- Pakistan Agriculture Research Council Islamabad Pakistan
| | - Baohong Zhang
- Department of Biology East Carolina University Greenville North Caroline
| |
Collapse
|
203
|
Xu X, Wan T, Xin H, Li D, Pan H, Wu J, Ping Y. Delivery of CRISPR/Cas9 for therapeutic genome editing. J Gene Med 2019; 21:e3107. [PMID: 31237055 DOI: 10.1002/jgm.3107] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/25/2019] [Accepted: 06/11/2019] [Indexed: 12/15/2022] Open
Abstract
The clustered, regularly-interspaced, short palindromic repeat (CRISPR)-associated nuclease 9 (CRISPR/Cas9) is emerging as a promising genome-editing tool for treating diseases in a precise way, and has been applied to a wide range of research in the areas of biology, genetics, and medicine. Delivery of therapeutic genome-editing agents provides a promising platform for the treatment of genetic disorders. Although viral vectors are widely used to deliver CRISPR/Cas9 elements with high efficiency, they suffer from several drawbacks, such as mutagenesis, immunogenicity, and off-target effects. Recently, non-viral vectors have emerged as another class of delivery carriers in terms of their safety, simplicity, and flexibility. In this review, we discuss the modes of CRISPR/Cas9 delivery, the barriers to the delivery process and the application of CRISPR/Cas9 system for the treatment of genetic disorders. We also highlight several representative types of non-viral vectors, including polymers, liposomes, cell-penetrating peptides, and other synthetic vectors, for the therapeutic delivery of CRISPR/Cas9 system. The applications of CRISPR/Cas9 in treating genetic disorders mediated by the non-viral vectors are also discussed.
Collapse
Affiliation(s)
- Xiaojie Xu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Tao Wan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Huhu Xin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Da Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Jun Wu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| |
Collapse
|
204
|
Zhao W, Hou X, Vick OG, Dong Y. RNA delivery biomaterials for the treatment of genetic and rare diseases. Biomaterials 2019; 217:119291. [PMID: 31255978 DOI: 10.1016/j.biomaterials.2019.119291] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/14/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022]
Abstract
Genetic and rare diseases (GARDs) affect more than 350 million patients worldwide and remain a significant challenge in the clinic. Hence, continuous efforts have been made to bridge the significant gap between the supply and demand of effective treatments for GARDs. Recent decades have witnessed the impressive progress in the fight against GARDs, with an improved understanding of the genetic origins of rare diseases and the rapid development in gene therapy providing a new avenue for GARD therapy. RNA-based therapeutics, such as RNA interference (RNAi), messenger RNA (mRNA) and RNA-involved genome editing technologies, demonstrate great potential as a therapy tool for treating genetic associated rare diseases. In the meantime, a variety of RNA delivery vehicles were established for boosting the widespread applications of RNA therapeutics. Among all the RNA delivery platforms which enable the systemic applications of RNAs, non-viral RNA delivery biomaterials display superior properties and a few biomaterials have been successfully exploited for achieving the RNA-based gene therapies on GARDs. In this review article, we focus on recent advances in the development of novel biomaterials for delivery of RNA-based therapeutics and highlight their applications to treat GARDs.
Collapse
Affiliation(s)
- Weiyu Zhao
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, United States
| | - Xucheng Hou
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, United States
| | - Olivia G Vick
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, United States; Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, United States; The Center for Clinical and Translational Science, The Ohio State University, Columbus, OH, 43210, United States; The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, United States; Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, 43210, United States; Department of Radiation Oncology, The Ohio State University, Columbus, OH, 43210, United States.
| |
Collapse
|
205
|
Sanzari I, Leone A, Ambrosone A. Nanotechnology in Plant Science: To Make a Long Story Short. Front Bioeng Biotechnol 2019; 7:120. [PMID: 31192203 PMCID: PMC6550098 DOI: 10.3389/fbioe.2019.00120] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/07/2019] [Indexed: 11/28/2022] Open
Abstract
This mini-review aims at gaining knowledge on basic aspects of plant nanotechnology. While in recent years the enormous progress of nanotechnology in biomedical sciences has revolutionized therapeutic and diagnostic approaches, the comprehension of nanoparticle-plant interactions, including uptake, mobilization and accumulation, is still in its infancy. Deeper studies are needed to establish the impact of nanomaterials (NMs) on plant growth and agro-ecosystems and to develop smart nanotechnology applications in crop improvement. Herein we provide a short overview of NMs employed in plant science and concisely describe key NM-plant interactions in terms of uptake, mobilization mechanisms, and biological effects. The major current applications in plants are reviewed also discussing the potential use of polymeric soft NMs which may open new and safer opportunities for smart delivery of biomolecules and for new strategies in plant genetic engineering, with the final aim to enhance plant defense and/or stimulate plant growth and development and, ultimately, crop production. Finally, we envisage that multidisciplinary collaborative approaches will be central to fill the knowledge gap in plant nanotechnology and push toward the use of NMs in agriculture and, more in general, in plant science research.
Collapse
Affiliation(s)
- Ilaria Sanzari
- Faculty of Engineering and the Environment, University of Southampton, Southampton, United Kingdom
| | | | | |
Collapse
|
206
|
Abstract
The CRISPR-Cas9 system has developed into a powerful platform for genome editing in various types of cells and tissues with single-nucleotide precision, but limited delivery options hamper its application in real-world settings. A new study by Shen et al. describes the use of an amphipathic peptide to deliver Cas9/sgRNA ribonucleoprotein complexes, leading to the disruption of GFP genes in cells and mice. Disruption of the Nrip1 gene in isolated pre-adipocytes led to a "browning" phenotype, pointing to new options in the fight against diabetes and obesity.
Collapse
Affiliation(s)
- Xingang Guan
- From the Department of Bioengineering, Center for Minimally Invasive Therapeutics at California Nanosystems Institute, UCLA, Los Angeles, California 90095.,the College of Medical Technology, Beihua University, Jilin city, Jilin 132013, China, and
| | - Zhimin Luo
- From the Department of Bioengineering, Center for Minimally Invasive Therapeutics at California Nanosystems Institute, UCLA, Los Angeles, California 90095.,the School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shanxi 710061, China
| | - Wujin Sun
- From the Department of Bioengineering, Center for Minimally Invasive Therapeutics at California Nanosystems Institute, UCLA, Los Angeles, California 90095,
| |
Collapse
|
207
|
Li Y, Chakraborty A, Chen J, Xu Q. Combinatorial Library of Light-Cleavable Lipidoid Nanoparticles for Intracellular Drug Delivery. ACS Biomater Sci Eng 2019; 5:2391-2398. [DOI: 10.1021/acsbiomaterials.9b00445] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Anirban Chakraborty
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Jinjin Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
208
|
Kowalski PS, Rudra A, Miao L, Anderson DG. Delivering the Messenger: Advances in Technologies for Therapeutic mRNA Delivery. Mol Ther 2019; 27:710-728. [PMID: 30846391 PMCID: PMC6453548 DOI: 10.1016/j.ymthe.2019.02.012] [Citation(s) in RCA: 712] [Impact Index Per Article: 118.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 12/18/2022] Open
Abstract
mRNA has broad potential as a therapeutic. Current clinical efforts are focused on vaccination, protein replacement therapies, and treatment of genetic diseases. The clinical translation of mRNA therapeutics has been made possible through advances in the design of mRNA manufacturing and intracellular delivery methods. However, broad application of mRNA is still limited by the need for improved delivery systems. In this review, we discuss the challenges for clinical translation of mRNA-based therapeutics, with an emphasis on recent advances in biomaterials and delivery strategies, and we present an overview of the applications of mRNA-based delivery for protein therapy, gene editing, and vaccination.
Collapse
Affiliation(s)
- Piotr S Kowalski
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Arnab Rudra
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Lei Miao
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
209
|
Koujah L, Shukla D, Naqvi AR. CRISPR-Cas based targeting of host and viral genes as an antiviral strategy. Semin Cell Dev Biol 2019; 96:53-64. [PMID: 30953741 DOI: 10.1016/j.semcdb.2019.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 12/23/2022]
Abstract
Viral infections in human are leading cause of mortality and morbidity across the globe. Several viruses (including HIV and Herpesvirus), have evolved ingenious strategies to evade host-immune system and persist life-long. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR-associated (Cas) is an ancient antiviral system recently discovered in bacteria that has shown tremendous potential as a precise, invariant genome editing tool. Using CRISPR-Cas based system to activate host defenses or genetic modification of viral genome can provide novel, exciting and successful antiviral mechanisms and treatment modalities. In this review, we will provide progress on the CRISPR-Cas based antiviral approaches that facilitate clearance of virus-infected cells and/or prohibit virus infection or replication. We will discuss on the possibilities of CRIPSR-Cas as prophylaxis and therapy in viral infections and review the challenges of this potent gene editing technology.
Collapse
Affiliation(s)
- Lulia Koujah
- Department of Microbiology and Immunology, University of Illinois at Chicago, IL, 60612, USA; Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL, 60612, USA
| | - Deepak Shukla
- Department of Microbiology and Immunology, University of Illinois at Chicago, IL, 60612, USA; Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL, 60612, USA.
| | - Afsar R Naqvi
- Mucosal Immunology Lab, College of Dentistry, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
210
|
Wu J, Zheng Y, Jiang S, Qu Y, Wei T, Zhan W, Wang L, Yu Q, Chen H. Two-in-One Platform for High-Efficiency Intracellular Delivery and Cell Harvest: When a Photothermal Agent Meets a Thermoresponsive Polymer. ACS APPLIED MATERIALS & INTERFACES 2019; 11:12357-12366. [PMID: 30859807 DOI: 10.1021/acsami.9b01586] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Efficient intracellular delivery of exogenous macromolecules is a key operation in biological research and for clinical applications. Moreover, under particular in vitro or ex vivo conditions, harvesting the engineered cells that maintain good viability is also important. However, none of the methods currently available is truly satisfactory in all respects. Herein, a "two-in-one" platform based on a polydopamine/poly( N-isopropylacrylamide) (PDA/PNIPAAm) hybrid film is developed, showing high efficiency in both cargo delivery and cell harvest without compromising cell viability. Due to the strong photothermal effect of PDA in response to near-infrared irradiation, this film can deliver diverse molecules to a number of cell types (including three hard-to-transfect cells) with an efficiency of ∼99% via membrane-disruption mechanism. Moreover, due to the thermoresponsive properties of PNIPAAm, the cells are harvested from the film without compromising viability by simply decreasing the temperature. A proof-of-concept experiment demonstrates that, using this platform, "recalcitrant" endothelial cells can be transfected by the functional ZNF580 gene and the harvested transfected cells can be recultured with high retention of viability and improved migration. In general, this "two-in-one" platform provides a reliable, universally applicable approach for both intracellular delivery and cell harvest in a highly efficient and nondestructive way, with great potential for use in a wide range of biomedical applications.
Collapse
Affiliation(s)
- Jingxian Wu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , 199 Ren'ai Road , Suzhou 215123 , P. R. China
| | - Yanjun Zheng
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , 199 Ren'ai Road , Suzhou 215123 , P. R. China
| | - Shuaibing Jiang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , 199 Ren'ai Road , Suzhou 215123 , P. R. China
| | - Yangcui Qu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , 199 Ren'ai Road , Suzhou 215123 , P. R. China
| | - Ting Wei
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , 199 Ren'ai Road , Suzhou 215123 , P. R. China
| | - Wenjun Zhan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , 199 Ren'ai Road , Suzhou 215123 , P. R. China
| | - Lei Wang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , 199 Ren'ai Road , Suzhou 215123 , P. R. China
| | - Qian Yu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , 199 Ren'ai Road , Suzhou 215123 , P. R. China
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , 199 Ren'ai Road , Suzhou 215123 , P. R. China
| |
Collapse
|
211
|
Improved Delivery of CRISPR/Cas9 System Using Magnetic Nanoparticles into Porcine Fibroblast. Mol Biotechnol 2019; 61:173-180. [PMID: 30560399 DOI: 10.1007/s12033-018-0145-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Genetically modified pigs play an important role in agriculture and biomedical research; hence, new efficient methods are needed to obtain genetically engineered cells and animals. The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas (CRISPR-associated) system represents an effective genome editing tool. It consists of two key molecules: single guide RNA (sgRNA) and the Cas9 endonuclease that can be introduced into the cells as one plasmid. Typical delivery methods for CRISPR/Cas9 components are limited by low transfection efficiency or toxic effects on cells. Here, we describe the use of magnetic nanoparticles and gradient magnetic field to improve delivery of CRISPR/Cas9 constructs into porcine fetal fibroblasts. Polyethylenimine-coated nanoparticles with magnetic iron oxide core were used to form magnetic plasmid DNA lipoplexes. CRISPR/Cas9 construct was prepared to induce site-specific cutting at the porcine H11 locus. Quantitative assessment of genomic cleavage by sequence trace decomposition demonstrated that the magnetofection efficiency was more than 3.5 times higher compared to the classic lipofection method. The Tracking of Indels by Decomposition web tool precisely determined the spectrum of indels that occurred. Simultaneously, no additional cytotoxicity associated with the utilization of magnetic nanoparticles was observed. Our results indicate that magnetofection enables effective delivery of the CRISPR/Cas9 construct into porcine fetal fibroblasts with low cell toxicity.
Collapse
|
212
|
Yin J, Hou S, Wang Q, Bao L, Liu D, Yue Y, Yao W, Gao X. Microenvironment-Responsive Delivery of the Cas9 RNA-Guided Endonuclease for Efficient Genome Editing. Bioconjug Chem 2019; 30:898-906. [PMID: 30802405 DOI: 10.1021/acs.bioconjchem.9b00022] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Successful and efficient delivery of Cas9 protein and gRNA into cells is critical for genome editing and its therapeutic application. In this study, we developed an improved supercharged polypeptide (SCP) mediated delivery system based on dithiocyclopeptide linker to realize the effective genome editing in tumor cells. The fusion protein Cas9-linker-SCP (Cas9-LS) forms positively charged complexes with gRNA in vitro to provide possibilities for gRNA delivery into cells. Under the microenvironment of tumor cells, the dithiocyclopeptide linker, containing matrix metalloproteinase 2 (MMP-2) sensitive sequence and an intramolecular disulfide bond, can be completely disconnected to promote the release of Cas9 protein with the nuclear localization sequence (NLS) in the cytoplasm and transfer to the cell nucleus for highly efficient genome editing, resulting in an obvious increase of indel efficiency in comparison to fusion protein without dithiocyclopeptide linker (Cas9-SCP). Furthermore, Cas9-LS shows no significant cytotoxicity and minimal hemolytic activity. We envision that the microenvironment-responsive Cas9 protein delivery system can facilitate more efficient genome editing in tumor cells.
Collapse
Affiliation(s)
- Jun Yin
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing 210009 , China
| | - Shan Hou
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing 210009 , China
| | - Qun Wang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing 210009 , China
| | - Lichen Bao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing 210009 , China
| | - Dingkang Liu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing 210009 , China
| | - Yali Yue
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing 210009 , China
| | - Wenbing Yao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing 210009 , China
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing 210009 , China
| |
Collapse
|
213
|
Chang J, Chen X, Glass Z, Gao F, Mao L, Wang M, Xu Q. Integrating Combinatorial Lipid Nanoparticle and Chemically Modified Protein for Intracellular Delivery and Genome Editing. Acc Chem Res 2019; 52:665-675. [PMID: 30586281 DOI: 10.1021/acs.accounts.8b00493] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The use of protein to precisely manipulate cell signaling is an effective approach for controlling cell fate and developing precision medicine. More recently, programmable nucleases, such as CRISPR/Cas9, have shown extremely high potency for editing genetic flow of mammalian cells, and for treating genetic disorders. The therapeutic potential of proteins with an intracellular target, however, is mostly challenged by their low cell impermeability. Therefore, a developing delivery system to transport protein to the site of action in a spatiotemporal controlled manner is of great importance to expand the therapeutic index of the protein. In this Account, we first summarize our most recent advances in designing combinatorial lipid nanoparticles with diverse chemical structures for intracellular protein delivery. By designing parallel Michael addition or ring-opening reaction of aliphatic amines, we have generated a combinatorial library of cationic lipids, and identified several leading nanoparticle formulations for intracellular protein delivery both in vitro and in vivo. Moreover, we optimized the chemical structure of lipids to control lipid degradation and protein release inside cells for CRISPR/Cas9 genome-editing protein delivery. In the second part of this Account, we survey our recent endeavor in developing a chemical approach to modify protein, in particular, coupled with the nanoparticle delivery platform, to improve protein delivery for targeted diseases treatment and genome editing. Chemical modification of protein is a useful tool to modulate protein function and to improve the therapeutic index of protein drugs. Herein, we mostly summarize our recent advances on designing chemical approaches to modify protein with following unique findings: (1) chemically modified protein shows selective turn-on activity based on the specific intracellular microenvironment, with which we were able to protein-based targeted cancer therapy; (2) the conjugation of hyaluronic acid (HA) to protein allows cancer cell surface receptor-targeted delivery of protein; (3) the introduction of nonpeptidic boronic acid into protein enabled cell nucleus targeted delivery; this is the first report that a nonpeptidic signal can direct protein to subcellular compartment; and (4) the fusion of protein with negatively supercharged green fluorescent protein (GFP) facilitates the self-assembly of protein with lipid nanoparticle for genome-editing protein delivery. At the end of the Account, we give a perspective of expanding the chemistry that could be integrated to design biocompatible lipid nanocarriers for protein delivery and genome editing in vitro and in vivo, as well as the chemical approaches that we can harness to modulate protein activity in live cells for targeted diseases treatment.
Collapse
Affiliation(s)
- Jin Chang
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecule Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
| | - Xianghan Chen
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecule Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
- College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241000, China
| | - Zachary Glass
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Feng Gao
- College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241000, China
| | - Lanqun Mao
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecule Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ming Wang
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecule Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
214
|
Tao Y, Hou X, Zuo F, Li X, Pang Y, Jiang G. Application of nanoparticle-based siRNA and CRISPR/Cas9 delivery systems in gene-targeted therapy. Nanomedicine (Lond) 2019; 14:511-514. [PMID: 30806159 DOI: 10.2217/nnm-2018-0522] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Yingkai Tao
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Xiaoyang Hou
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Fengmei Zuo
- College of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Xinxin Li
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Yanyu Pang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Guan Jiang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| |
Collapse
|
215
|
Li Y, Li AC, Xu Q. Intracellular Delivery of His-Tagged Genome-Editing Proteins Enabled by Nitrilotriacetic Acid-Containing Lipidoid Nanoparticles. Adv Healthc Mater 2019; 8:e1800996. [PMID: 30565897 PMCID: PMC6474682 DOI: 10.1002/adhm.201800996] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/26/2018] [Indexed: 12/26/2022]
Abstract
Protein- and peptide-based therapeutics with high tolerance and specificity along with low off-target effects and genetic risks have attracted tremendous attention over the last three decades. Herein, a new type of noncationic lipidoid nanoparticle (LNP) is reported for His-tagged protein delivery. Active lipidoids are synthesized by conjugating a nitrilotriacetic acid group with hydrophobic tails (EC16, O16B, and O17O) and nanoparticles are formulated in the presence of nickel ions and helper lipids (cholesterol, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine, and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000]). It is demonstrated that the newly developed LNPs are capable of delivering various His-tagged proteins including green fluorescent protein (GFP), (-30)GFP-Cre recombinase, and CRISPR/Cas9 ribonucleoprotein into mammalian cells.
Collapse
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, US,
| | - Alice Chukun Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, US,
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, US,
| |
Collapse
|
216
|
Goeckel ME, Basgall EM, Lewis IC, Goetting SC, Yan Y, Halloran M, Finnigan GC. Modulating CRISPR gene drive activity through nucleocytoplasmic localization of Cas9 in S. cerevisiae. Fungal Biol Biotechnol 2019; 6:2. [PMID: 30766726 PMCID: PMC6360766 DOI: 10.1186/s40694-019-0065-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/10/2019] [Indexed: 01/28/2023] Open
Abstract
Background The bacterial CRISPR/Cas genome editing system has provided a major breakthrough in molecular biology. One use of this technology is within a nuclease-based gene drive. This type of system can install a genetic element within a population at unnatural rates. Combatting of vector-borne diseases carried by metazoans could benefit from a delivery system that bypasses traditional Mendelian laws of segregation. Recently, laboratory studies in fungi, insects, and even mice, have demonstrated successful propagation of CRISPR gene drives and the potential utility of this type of mechanism. However, current gene drives still face challenges including evolved resistance, containment, and the consequences of application in wild populations. Additional research into molecular mechanisms that would allow for control, titration, and inhibition of drive systems is needed. Results In this study, we use artificial gene drives in budding yeast to explore mechanisms to modulate nuclease activity of Cas9 through its nucleocytoplasmic localization. We examine non-native nuclear localization sequences (both NLS and NES) on Cas9 fusion proteins in vivo through fluorescence microscopy and genomic editing. Our results demonstrate that mutational substitutions to nuclear signals and combinatorial fusions can both modulate the level of gene drive activity within a population of cells. Conclusions These findings have implications for control of traditional nuclease-dependent editing and use of gene drive systems within other organisms. For instance, initiation of a nuclear export mechanism to Cas9 could serve as a molecular safeguard within an active gene drive to reduce or eliminate editing.
Collapse
Affiliation(s)
- Megan E Goeckel
- 1Department of Biochemistry and Molecular Biophysics, 141 Chalmers Hall, Kansas State University, Manhattan, KS 66506 USA
| | - Erianna M Basgall
- 1Department of Biochemistry and Molecular Biophysics, 141 Chalmers Hall, Kansas State University, Manhattan, KS 66506 USA
| | - Isabel C Lewis
- 1Department of Biochemistry and Molecular Biophysics, 141 Chalmers Hall, Kansas State University, Manhattan, KS 66506 USA
| | - Samantha C Goetting
- 1Department of Biochemistry and Molecular Biophysics, 141 Chalmers Hall, Kansas State University, Manhattan, KS 66506 USA
| | - Yao Yan
- 1Department of Biochemistry and Molecular Biophysics, 141 Chalmers Hall, Kansas State University, Manhattan, KS 66506 USA
| | - Megan Halloran
- 1Department of Biochemistry and Molecular Biophysics, 141 Chalmers Hall, Kansas State University, Manhattan, KS 66506 USA.,2Present Address: Department of Psychology, 106-B Kastle Hall, University of Kentucky, Lexington, KY 40506 USA
| | - Gregory C Finnigan
- 1Department of Biochemistry and Molecular Biophysics, 141 Chalmers Hall, Kansas State University, Manhattan, KS 66506 USA
| |
Collapse
|
217
|
Carboni V, Maaliki C, Alyami M, Alsaiari S, Khashab N. Synthetic Vehicles for Encapsulation and Delivery of CRISPR/Cas9 Gene Editing Machinery. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201800085] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Valentina Carboni
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials CenterKing Abdullah University of Science and Technology (KAUST) Thuwal 23955‐6900 Saudi Arabia
| | - Carine Maaliki
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials CenterKing Abdullah University of Science and Technology (KAUST) Thuwal 23955‐6900 Saudi Arabia
| | - Mram Alyami
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials CenterKing Abdullah University of Science and Technology (KAUST) Thuwal 23955‐6900 Saudi Arabia
| | - Shahad Alsaiari
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials CenterKing Abdullah University of Science and Technology (KAUST) Thuwal 23955‐6900 Saudi Arabia
| | - Niveen Khashab
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials CenterKing Abdullah University of Science and Technology (KAUST) Thuwal 23955‐6900 Saudi Arabia
| |
Collapse
|
218
|
Kim J, Koo BK, Yoon KJ. Modeling Host-Virus Interactions in Viral Infectious Diseases Using Stem-Cell-Derived Systems and CRISPR/Cas9 Technology. Viruses 2019; 11:v11020124. [PMID: 30704043 PMCID: PMC6409779 DOI: 10.3390/v11020124] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/14/2019] [Accepted: 01/23/2019] [Indexed: 02/06/2023] Open
Abstract
Pathologies induced by viral infections have undergone extensive study, with traditional model systems such as two-dimensional (2D) cell cultures and in vivo mouse models contributing greatly to our understanding of host-virus interactions. However, the technical limitations inherent in these systems have constrained efforts to more fully understand such interactions, leading to a search for alternative in vitro systems that accurately recreate in vivo physiology in order to advance the study of viral pathogenesis. Over the last decade, there have been significant technological advances that have allowed researchers to more accurately model the host environment when modeling viral pathogenesis in vitro, including induced pluripotent stem cells (iPSCs), adult stem-cell-derived organoid culture systems and CRISPR/Cas9-mediated genome editing. Such technological breakthroughs have ushered in a new era in the field of viral pathogenesis, where previously challenging questions have begun to be tackled. These include genome-wide analysis of host-virus crosstalk, identification of host factors critical for viral pathogenesis, and the study of viral pathogens that previously lacked a suitable platform, e.g., noroviruses, rotaviruses, enteroviruses, adenoviruses, and Zika virus. In this review, we will discuss recent advances in the study of viral pathogenesis and host-virus crosstalk arising from the use of iPSC, organoid, and CRISPR/Cas9 technologies.
Collapse
Affiliation(s)
- Jihoon Kim
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria.
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria.
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea.
| |
Collapse
|
219
|
Krasheninina OA, Apartsin EK, Fuentes E, Szulc A, Ionov M, Venyaminova AG, Shcharbin D, de la Mata FJ, Bryszewska M, Gόmez R. Complexes of Pro-Apoptotic siRNAs and Carbosilane Dendrimers: Formation and Effect on Cancer Cells. Pharmaceutics 2019; 11:pharmaceutics11010025. [PMID: 30634643 PMCID: PMC6359069 DOI: 10.3390/pharmaceutics11010025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 12/30/2018] [Accepted: 01/05/2019] [Indexed: 12/17/2022] Open
Abstract
This paper examines the complexation of anti-cancer small interfering RNAs (siRNAs) by cationic carbosilane dendrimers, and the interaction of the formed complexes with HeLa and HL-60 cancer cells. Stepwise formation of the complexes accompanied by the evolution of their properties has been observed through the increase of the charge ratio (dendrimer/siRNA). The complexes decrease the viability of both “easy-to-transfect” cells (HeLa) and “hard-to transfect” ones (HL-60), indicating a high potential of the cationic carbosilane dendrimers for siRNA delivery into tumor cells.
Collapse
Affiliation(s)
- Olga A Krasheninina
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Evgeny K Apartsin
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Elena Fuentes
- Departamento de Química Orgánica y Química Inorgánica, UAH-IQAR, Universidad de Alcalá, 28805 Alcalá de Henares, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
| | - Aleksandra Szulc
- Department of General Biophysics, University of Lodz, 90-236 Lodz, Poland.
| | - Maksim Ionov
- Department of General Biophysics, University of Lodz, 90-236 Lodz, Poland.
| | - Alya G Venyaminova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of NASB, 220072 Minsk, Belarus.
| | - F Javier de la Mata
- Departamento de Química Orgánica y Química Inorgánica, UAH-IQAR, Universidad de Alcalá, 28805 Alcalá de Henares, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
- Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Madrid, Spain.
| | - Maria Bryszewska
- Department of General Biophysics, University of Lodz, 90-236 Lodz, Poland.
| | - Rafael Gόmez
- Departamento de Química Orgánica y Química Inorgánica, UAH-IQAR, Universidad de Alcalá, 28805 Alcalá de Henares, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
- Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Madrid, Spain.
| |
Collapse
|
220
|
CRISPR/Cas9 for Sickle Cell Disease: Applications, Future Possibilities, and Challenges. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1144:37-52. [PMID: 30715679 DOI: 10.1007/5584_2018_331] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sickle cell disease (SCD) is an inherited monogenic disorder resulting in serious mortality and morbidity worldwide. Although the disease was characterized more than a century ago, there are only two FDA approved medications to lessen disease severity, and a definitive cure available to all patients with SCD is lacking. Rapid and substantial progress in genome editing approaches have proven valuable as a curative option given plausibility to either correct the underlying mutation in patient-derived hematopoietic stem/progenitor cells (HSPCs), induce fetal hemoglobin expression to circumvent sickling of red blood cells (RBCs), or create corrected induced pluripotent stem cells (iPSCs) among other approaches. Recent discovery of CRISPR/Cas9 has not only revolutionized genome engineering but has also brought the possibility of translating these concepts into a clinically meaningful reality. Here we summarize genome engineering applications using CRISPR/Cas9, addressing challenges and future perspectives of CRISPR/Cas9 as a curative option for SCD.
Collapse
|
221
|
Sandoval IM, Collier TJ, Manfredsson FP. Design and Assembly of CRISPR/Cas9 Lentiviral and rAAV Vectors for Targeted Genome Editing. Methods Mol Biol 2019; 1937:29-45. [PMID: 30706388 DOI: 10.1007/978-1-4939-9065-8_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Clustered regularly interspaced short palindromic repeat (CRISPR/Cas) system has emerged as an extremely useful tool for biological research and as a potential technology for gene therapy approaches. CRISPR/Cas mediated genome editing can be used to easily and efficiently modify endogenous genes in a large variety of cells and organisms. Furthermore, a modified version of the Cas9 nuclease has been developed that can be used for regulation of endogenous gene expression and labeling of genomic loci, among other applications. This chapter provides an introduction to the basis of the technology and a detail protocol for the most classic application: gene inactivation by CRISPR/Cas9 nuclease system from Streptococcus pyogenes. This workflow can be easily adapted for other CRISPR systems and applications.
Collapse
Affiliation(s)
- Ivette M Sandoval
- Department of Translational Science & Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.
- Mercy Health Saint Mary's, Grand Rapids, MI, USA.
| | - Timothy J Collier
- Department of Translational Science & Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
- Mercy Health Saint Mary's, Grand Rapids, MI, USA
| | - Fredric P Manfredsson
- Department of Translational Science & Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
- Mercy Health Saint Mary's, Grand Rapids, MI, USA
| |
Collapse
|
222
|
Li Y, Bolinger J, Yu Y, Glass Z, Shi N, Yang L, Wang M, Xu Q. Intracellular delivery and biodistribution study of CRISPR/Cas9 ribonucleoprotein loaded bioreducible lipidoid nanoparticles. Biomater Sci 2019; 7:596-606. [DOI: 10.1039/c8bm00637g] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A combinatorial library of cationic lipidoids were used as nanocarriers for intracellular delivery of the CRISPR/Cas9 ribonucleoprotein complex.
Collapse
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Justin Bolinger
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Yingjie Yu
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Zachary Glass
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Nicola Shi
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Liu Yang
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Ming Wang
- Beijing National Laboratory for Molecular Sciences
- Key Laboratory of Analytical Chemistry for Living Biosystems
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing
| | - Qiaobing Xu
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| |
Collapse
|
223
|
Ewart DT, Peterson EJ, Steer CJ. Gene editing for inflammatory disorders. Ann Rheum Dis 2019; 78:6-15. [PMID: 30077989 DOI: 10.1136/annrheumdis-2018-213454] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/02/2018] [Accepted: 07/03/2018] [Indexed: 12/24/2022]
Abstract
Technology for precise and efficient genetic editing is constantly evolving and is now capable of human clinical applications. Autoimmune and inflammatory diseases are chronic, disabling, sometimes life-threatening, conditions that feature heritable components. Both primary genetic lesions and the inflammatory pathobiology underlying these diseases represent fertile soil for new therapies based on the capabilities of gene editing. The ability to orchestrate precise targeted modifications to the genome will likely enable cell-based therapies for inflammatory diseases such as monogenic autoinflammatory disease, acquired autoimmune disease and for regenerative medicine in the setting of an inflammatory environment. Here, we discuss recent advances in genome editing and their evolving applications in immunoinflammatory diseases. Strengths and limitations of older genetic modification tools are compared with CRISPR/Cas9, base editing, RNA editing, targeted activators and repressors of transcription and targeted epigenetic modifiers. Commonly employed delivery vehicles to target cells or tissues of interest with genetic modification machinery, including viral, non-viral and cellular vectors, are described. Finally, applications in animal and human models of inflammatory diseases are discussed. Use of chimeric autoantigen receptor T cells, correction of monogenic diseases with genetically edited haematopoietic stem and progenitor cells, engineering of induced pluripotent stem cells and ex vivo expansion and modification of regulatory T cells for a range of chronic inflammatory diseases are reviewed.
Collapse
Affiliation(s)
- David T Ewart
- Division of Rheumatic and Autoimmune Diseases, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Erik J Peterson
- Division of Rheumatic and Autoimmune Diseases, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Clifford J Steer
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
224
|
Givens BE, Naguib YW, Geary SM, Devor EJ, Salem AK. Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing Therapeutics. AAPS J 2018; 20:108. [PMID: 30306365 PMCID: PMC6398936 DOI: 10.1208/s12248-018-0267-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/18/2018] [Indexed: 12/17/2022] Open
Abstract
The recent progress in harnessing the efficient and precise method of DNA editing provided by CRISPR/Cas9 is one of the most promising major advances in the field of gene therapy. However, the development of safe and optimally efficient delivery systems for CRISPR/Cas9 elements capable of achieving specific targeting of gene therapy to the location of interest without off-target effects is a primary challenge for clinical therapeutics. Nanoparticles (NPs) provide a promising means to meet such challenges. In this review, we present the most recent advances in developing innovative NP-based delivery systems that efficiently deliver CRISPR/Cas9 constructs and maximize their effectiveness.
Collapse
Affiliation(s)
- Brittany E Givens
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
- Department of Chemical and Biochemical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Youssef W Naguib
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Sean M Geary
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Eric J Devor
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA.
- Department of Chemical and Biochemical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
225
|
Yamatsugu K, Kawashima SA, Kanai M. Leading approaches in synthetic epigenetics for novel therapeutic strategies. Curr Opin Chem Biol 2018; 46:10-17. [DOI: 10.1016/j.cbpa.2018.03.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/12/2018] [Accepted: 03/17/2018] [Indexed: 12/20/2022]
|
226
|
Brioschi M, Banfi C. The application of gene silencing in proteomics: from laboratory to clinic. Expert Rev Proteomics 2018; 15:717-732. [PMID: 30205712 DOI: 10.1080/14789450.2018.1521275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Since the completion of genome sequencing, gene silencing technologies have emerged as powerful tools to study gene functions in various biological processes, both in vivo and in vitro. Moreover, they have also been proposed as therapeutic agents to inhibit selected genes in a variety of pathological conditions, such as cancer, neurodegenerative, and cardiovascular diseases. Area covered: This review summarizes the mechanisms of action and applications of genome editing tools, from RNA interference to clustered regularly interspaced short palindromic repeats-based systems, in research and in clinics. We describe their essential role in high-throughput genetic screens and, in particular, in functional proteomics studies, to identify diagnostic markers and therapeutic targets. Indeed, gene silencing and proteomics have been extensively integrated to study global proteome changes, posttranslational modifications, and protein-protein interactions. Expert commentary: Functional proteomics approaches that leverage gene silencing tools have been successfully applied to examine the role of several genes in various contexts, leading to a deeper knowledge of biological pathways and disease mechanisms. Recent developments of gene silencing tools have improved their performance, also in terms of off-targets effects reduction, paving the way for a wider therapeutic application of these systems.
Collapse
Affiliation(s)
- Maura Brioschi
- a Unit of Proteomics , Centro Cardiologico Monzino IRCCS , Milano , Italy
| | - Cristina Banfi
- a Unit of Proteomics , Centro Cardiologico Monzino IRCCS , Milano , Italy
| |
Collapse
|
227
|
Shen Y, Cohen JL, Nicoloro SM, Kelly M, Yenilmez B, Henriques F, Tsagkaraki E, Edwards YJK, Hu X, Friedline RH, Kim JK, Czech MP. CRISPR-delivery particles targeting nuclear receptor-interacting protein 1 ( Nrip1) in adipose cells to enhance energy expenditure. J Biol Chem 2018; 293:17291-17305. [PMID: 30190322 DOI: 10.1074/jbc.ra118.004554] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/22/2018] [Indexed: 12/26/2022] Open
Abstract
RNA-guided, engineered nucleases derived from the prokaryotic adaptive immune system CRISPR-Cas represent a powerful platform for gene deletion and editing. When used as a therapeutic approach, direct delivery of Cas9 protein and single-guide RNA (sgRNA) could circumvent the safety issues associated with plasmid delivery and therefore represents an attractive tool for precision genome engineering. Gene deletion or editing in adipose tissue to enhance its energy expenditure, fatty acid oxidation, and secretion of bioactive factors through a "browning" process presents a potential therapeutic strategy to alleviate metabolic disease. Here, we developed "CRISPR-delivery particles," denoted CriPs, composed of nano-size complexes of Cas9 protein and sgRNA that are coated with an amphipathic peptide called Endo-Porter that mediates entry into cells. Efficient CRISPR-Cas9-mediated gene deletion of ectopically expressed GFP by CriPs was achieved in multiple cell types, including a macrophage cell line, primary macrophages, and primary pre-adipocytes. Significant GFP loss was also observed in peritoneal exudate cells with minimum systemic toxicity in GFP-expressing mice following intraperitoneal injection of CriPs containing Gfp-targeting sgRNA. Furthermore, disruption of a nuclear co-repressor of catabolism, the Nrip1 gene, in white adipocytes by CriPs enhanced adipocyte browning with a marked increase of uncoupling protein 1 (UCP1) expression. Of note, the CriP-mediated Nrip1 deletion did not produce detectable off-target effects. We conclude that CriPs offer an effective Cas9 and sgRNA delivery system for ablating targeted gene products in cultured cells and in vivo, providing a potential therapeutic strategy for metabolic disease.
Collapse
Affiliation(s)
| | | | | | - Mark Kelly
- From the Program in Molecular Medicine and
| | | | | | - Emmanouela Tsagkaraki
- From the Program in Molecular Medicine and.,the Molecular Basis of Human Disease Graduate Program, School of Sciences, Faculty of Medicine, University of Crete, P.O. Box 2208, Heraklion, Crete 71003, Greece
| | | | - Xiaodi Hu
- the Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Randall H Friedline
- the Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Jason K Kim
- From the Program in Molecular Medicine and.,the Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | | |
Collapse
|
228
|
Mueller K, Carlson-Stevermer J, Saha K. Increasing the precision of gene editing in vitro, ex vivo, and in vivo. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018; 7:83-90. [PMID: 31086832 PMCID: PMC6510258 DOI: 10.1016/j.cobme.2018.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
New gene editing tools like CRISPR-Cas9 enable precision genome engineering within cell lines, primary cells, and model organisms, with some formulations now entering the clinic. "Precision" applies to various aspects of gene editing, and can be tailored for each application. Here we review recent advances in four types of precision in gene editing: 1) increased DNA cutting precision (e.g., on-target:off-target nuclease specificity), 2) increased on-target knock-in of sequence variants and transgenes (e.g., increased homology-directed repair), 3) increased transcriptional control of edited genes, and 4) increased specificity in delivery to a specific cell or tissue. Design of next-generation gene and cell therapies will likely exploit a combination of these advances.
Collapse
Affiliation(s)
- Katherine Mueller
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Jared Carlson-Stevermer
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Krishanu Saha
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA
| |
Collapse
|
229
|
Li Y, Yang T, Yu Y, Shi N, Yang L, Glass Z, Bolinger J, Finkel IJ, Li W, Xu Q. Combinatorial library of chalcogen-containing lipidoids for intracellular delivery of genome-editing proteins. Biomaterials 2018; 178:652-662. [PMID: 29549971 DOI: 10.1016/j.biomaterials.2018.03.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/19/2022]
Abstract
Protein based therapeutics with high specificities and low off-target effects are used for transient and accurate manipulation of cell functions. However, developing safe and efficient carriers for intracellular delivery of active therapeutic proteins is a long-standing challenge. Here we report a combinatorial library of chalcogen (O, S, Se) containing lipidoid nanoparticles (LNPs) as efficient nanocarriers for intracellular delivery of negatively supercharged Cre recombinase ((-30)GFP-Cre) and anionic Cas9:single-guide RNA (Cas9:sgRNA) ribonucleoprotein (RNP) for genome editing. The structure-activity relationship between the lipidoids and intracellular protein delivery efficiencies was explored and it was demonstrated that the newly developed LNPs are effective for gene recombination in vivo.
Collapse
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Tao Yang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, PR China
| | - Yingjie Yu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Nicola Shi
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Liu Yang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Zachary Glass
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Justin Bolinger
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Isaac James Finkel
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Wenhan Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
230
|
Damen M, Groenen AJJ, van Dongen SFM, Nolte RJM, Scholte BJ, Feiters MC. Transfection by cationic gemini lipids and surfactants. MEDCHEMCOMM 2018; 9:1404-1425. [PMID: 30288217 PMCID: PMC6148748 DOI: 10.1039/c8md00249e] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/11/2018] [Indexed: 12/13/2022]
Abstract
Diseases that are linked to defective genes or mutations can in principle be cured by gene therapy, in which damaged or absent genes are either repaired or replaced by new DNA in the nucleus of the cell. Related to this, disorders associated with elevated protein expression levels can be treated by RNA interference via the delivery of siRNA to the cytoplasm of cells. Polynucleotides can be brought into cells by viruses, but this is not without risk for the patient. Alternatively, DNA and RNA can be delivered by transfection, i.e. by non-viral vector systems such as cationic surfactants, which are also referred to as cationic lipids. In this review, recent progress on cationic lipids as transfection vectors will be discussed, with special emphasis on geminis, surfactants with 2 head groups and 2 tails connected by a spacer.
Collapse
Affiliation(s)
- M Damen
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - A J J Groenen
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - S F M van Dongen
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - R J M Nolte
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - B J Scholte
- Departments of Pediatric pulmonology and Cell Biology , Erasmus MC, P. O. Box 2040 , 3000 CA Rotterdam , The Netherlands
| | - M C Feiters
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| |
Collapse
|
231
|
Chen X, Gonçalves MAFV. DNA, RNA, and Protein Tools for Editing the Genetic Information in Human Cells. iScience 2018; 6:247-263. [PMID: 30240615 PMCID: PMC6137403 DOI: 10.1016/j.isci.2018.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/23/2018] [Accepted: 07/31/2018] [Indexed: 12/27/2022] Open
Abstract
Solving the structure of DNA in 1953 has unleashed a tour de force in molecular biology that has illuminated how the genetic information stored in DNA is copied and flows downstream into RNA and proteins. Currently, increasingly powerful technologies permit not only reading and writing DNA in vitro but also editing the genetic instructions in cells from virtually any organism. Editing specific genomic sequences in living cells has been particularly accelerated with the introduction of programmable RNA-guided nucleases (RGNs) based on prokaryotic CRISPR adaptive immune systems. The repair of chromosomal breaks made by RGNs with donor DNA patches results in targeted genome editing involving the introduction of specific genetic changes at predefined genomic positions. Hence, donor DNAs, guide RNAs, and nuclease proteins, each representing the molecular entities underlying the storage, transmission, and expression of genetic information, are, once delivered into cells, put to work as agents of change of that very same genetic text. Here, after providing an outline of the programmable nuclease-assisted genome editing field, we review the increasingly diverse range of DNA, RNA, and protein components (e.g., nucleases and "nickases") that, when brought together, underlie RGN-based genome editing in eukaryotic cells.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, Leiden 2333 ZC, the Netherlands
| | - Manuel A F V Gonçalves
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, Leiden 2333 ZC, the Netherlands.
| |
Collapse
|
232
|
Abstract
The diversity, modularity, and efficacy of CRISPR-Cas systems are driving a biotechnological revolution. RNA-guided Cas enzymes have been adopted as tools to manipulate the genomes of cultured cells, animals, and plants, accelerating the pace of fundamental research and enabling clinical and agricultural breakthroughs. We describe the basic mechanisms that set the CRISPR-Cas toolkit apart from other programmable gene-editing technologies, highlighting the diverse and naturally evolved systems now functionalized as biotechnologies. We discuss the rapidly evolving landscape of CRISPR-Cas applications, from gene editing to transcriptional regulation, imaging, and diagnostics. Continuing functional dissection and an expanding landscape of applications position CRISPR-Cas tools at the cutting edge of nucleic acid manipulation that is rewriting biology.
Collapse
Affiliation(s)
- Gavin J Knott
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Jennifer A Doudna
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
- Department of Chemistry, University of California, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
233
|
Stewart MP, Langer R, Jensen KF. Intracellular Delivery by Membrane Disruption: Mechanisms, Strategies, and Concepts. Chem Rev 2018; 118:7409-7531. [PMID: 30052023 PMCID: PMC6763210 DOI: 10.1021/acs.chemrev.7b00678] [Citation(s) in RCA: 456] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intracellular delivery is a key step in biological research and has enabled decades of biomedical discoveries. It is also becoming increasingly important in industrial and medical applications ranging from biomanufacture to cell-based therapies. Here, we review techniques for membrane disruption-based intracellular delivery from 1911 until the present. These methods achieve rapid, direct, and universal delivery of almost any cargo molecule or material that can be dispersed in solution. We start by covering the motivations for intracellular delivery and the challenges associated with the different cargo types-small molecules, proteins/peptides, nucleic acids, synthetic nanomaterials, and large cargo. The review then presents a broad comparison of delivery strategies followed by an analysis of membrane disruption mechanisms and the biology of the cell response. We cover mechanical, electrical, thermal, optical, and chemical strategies of membrane disruption with a particular emphasis on their applications and challenges to implementation. Throughout, we highlight specific mechanisms of membrane disruption and suggest areas in need of further experimentation. We hope the concepts discussed in our review inspire scientists and engineers with further ideas to improve intracellular delivery.
Collapse
Affiliation(s)
- Martin P. Stewart
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
- The Koch Institute for Integrative Cancer Research,
Massachusetts Institute of Technology, Cambridge, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
- The Koch Institute for Integrative Cancer Research,
Massachusetts Institute of Technology, Cambridge, USA
| | - Klavs F. Jensen
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
| |
Collapse
|
234
|
Blighe K, DeDionisio L, Christie KA, Chawes B, Shareef S, Kakouli-Duarte T, Chao-Shern C, Harding V, Kelly RS, Castellano L, Stebbing J, Lasky-Su JA, Nesbit MA, Moore CBT. Gene editing in the context of an increasingly complex genome. BMC Genomics 2018; 19:595. [PMID: 30086710 PMCID: PMC6081867 DOI: 10.1186/s12864-018-4963-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/26/2018] [Indexed: 12/15/2022] Open
Abstract
The reporting of the first draft of the human genome in 2000 brought with it much hope for the future in what was felt as a paradigm shift toward improved health outcomes. Indeed, we have now mapped the majority of variation across human populations with landmark projects such as 1000 Genomes; in cancer, we have catalogued mutations across the primary carcinomas; whilst, for other diseases, we have identified the genetic variants with strongest association. Despite this, we are still awaiting the genetic revolution in healthcare to materialise and translate itself into the health benefits for which we had hoped. A major problem we face relates to our underestimation of the complexity of the genome, and that of biological mechanisms, generally. Fixation on DNA sequence alone and a 'rigid' mode of thinking about the genome has meant that the folding and structure of the DNA molecule -and how these relate to regulation- have been underappreciated. Projects like ENCODE have additionally taught us that regulation at the level of RNA is just as important as that at the spatiotemporal level of chromatin.In this review, we chart the course of the major advances in the biomedical sciences in the era pre- and post the release of the first draft sequence of the human genome, taking a focus on technology and how its development has influenced these. We additionally focus on gene editing via CRISPR/Cas9 as a key technique, in particular its use in the context of complex biological mechanisms. Our aim is to shift the mode of thinking about the genome to that which encompasses a greater appreciation of the folding of the DNA molecule, DNA- RNA/protein interactions, and how these regulate expression and elaborate disease mechanisms.Through the composition of our work, we recognise that technological improvement is conducive to a greater understanding of biological processes and life within the cell. We believe we now have the technology at our disposal that permits a better understanding of disease mechanisms, achievable through integrative data analyses. Finally, only with greater understanding of disease mechanisms can techniques such as gene editing be faithfully conducted.
Collapse
Affiliation(s)
- K Blighe
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, USA.
- Department of Cancer Studies and Molecular Medicine, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, LE2 7LX, UK.
- Bill Lyons Informatics Centre, UCL Cancer Institute, University College London, WC1E 6DD, London, UK.
| | - L DeDionisio
- Avellino Laboratories, Menlo Park, CA, 94025, USA
| | - K A Christie
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, BT52 1SA, UK
| | - B Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - S Shareef
- University of Raparin, Ranya, Kurdistan Region, Iraq
| | - T Kakouli-Duarte
- Institute of Technology Carlow, Department of Science and Health, Kilkenny Road, Carlow, Ireland
| | - C Chao-Shern
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, BT52 1SA, UK
- Avellino Laboratories, Menlo Park, CA, 94025, USA
| | - V Harding
- Imperial College London, Division of Cancer, Department of Surgery and Cancer, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - R S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, USA
| | - L Castellano
- Imperial College London, Division of Cancer, Department of Surgery and Cancer, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
- JMS Building, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| | - J Stebbing
- Imperial College London, Division of Cancer, Department of Surgery and Cancer, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - J A Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, USA
| | - M A Nesbit
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, BT52 1SA, UK
| | - C B T Moore
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, BT52 1SA, UK.
- Avellino Laboratories, Menlo Park, CA, 94025, USA.
| |
Collapse
|
235
|
Kumar TR. Fshb Knockout Mouse Model, Two Decades Later and Into the Future. Endocrinology 2018; 159:1941-1949. [PMID: 29579177 PMCID: PMC5888209 DOI: 10.1210/en.2018-00072] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/02/2018] [Indexed: 12/20/2022]
Abstract
In 1997, nearly 20 years ago, we reported the phenotypes of follicle-stimulating hormone (FSH) β (Fshb) null mice. Since then, these mice have been useful for various physiological and genetic studies in reproductive and skeletal biology. In a 2009 review titled "FSHβ Knockout Mouse Model: A Decade Ago and Into the Future," I summarized the need for and what led to the development of an FSH-deficient mouse model and its applications, including delineation of the emerging extragonadal roles of FSH in bone cells by using this genetic model. These studies opened up exciting avenues of research on osteoporosis and now extend into those on adiposity in postmenopausal women. Here, I summarize the progress made with this mouse model since 2009 with regard to FSH rerouting in vivo, deciphering the role of N-glycosylation on FSHβ, roles of FSH in somatic-germ cell interactions in gonads, and provide a road map that is anticipated to emerge in the near future. Undoubtedly, the next 10 years should be an even more exciting time to explore the fertile area of FSH biology and its implications for basic and clinical reproductive physiology research.
Collapse
Affiliation(s)
- T Rajendra Kumar
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado at Denver, Anschutz Medical Campus, Aurora, Colorado
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Colorado at Denver, Anschutz Medical Campus, Aurora, Colorado
- Correspondence: T. Rajendra Kumar, PhD, Edgar L. and Patricia M. Makowski Professor, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, 12700 East 19th Avenue, RC-2 Complex, 15-3000B, Aurora, Colorado 80045. E-mail:
| |
Collapse
|
236
|
Li L, Hu S, Chen X. Non-viral delivery systems for CRISPR/Cas9-based genome editing: Challenges and opportunities. Biomaterials 2018; 171:207-218. [PMID: 29704747 DOI: 10.1016/j.biomaterials.2018.04.031] [Citation(s) in RCA: 262] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/13/2018] [Accepted: 04/14/2018] [Indexed: 02/06/2023]
Abstract
In recent years, CRISPR (clustered regularly interspaced short palindromic repeat)/Cas (CRISPR-associated) genome editing systems have become one of the most robust platforms in basic biomedical research and therapeutic applications. To date, efficient in vivo delivery of the CRISPR/Cas9 system to the targeted cells remains a challenge. Although viral vectors have been widely used in the delivery of the CRISPR/Cas9 system in vitro and in vivo, their fundamental shortcomings, such as the risk of carcinogenesis, limited insertion size, immune responses and difficulty in large-scale production, severely limit their further applications. Alternative non-viral delivery systems for CRISPR/Cas9 are urgently needed. With the rapid development of non-viral vectors, lipid- or polymer-based nanocarriers have shown great potential for CRISPR/Cas9 delivery. In this review, we analyze the pros and cons of delivering CRISPR/Cas9 systems in the form of plasmid, mRNA, or protein and then discuss the limitations and challenges of CRISPR/Cas9-based genome editing. Furthermore, current non-viral vectors that have been applied for CRISPR/Cas9 delivery in vitro and in vivo are outlined in details. Finally, critical obstacles for non-viral delivery of CRISPR/Cas9 system are highlighted and promising strategies to overcome these barriers are proposed.
Collapse
Affiliation(s)
- Ling Li
- Department of PET Center, Xiangya Hospital, Central South University, Changsha, 410008, China; Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Shuo Hu
- Department of PET Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
237
|
Sato-Dahlman M, Wirth K, Yamamoto M. Role of Gene Therapy in Pancreatic Cancer-A Review. Cancers (Basel) 2018; 10:E103. [PMID: 29614005 PMCID: PMC5923358 DOI: 10.3390/cancers10040103] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 03/27/2018] [Accepted: 03/30/2018] [Indexed: 01/05/2023] Open
Abstract
Mortality from pancreatic ductal adenocarcinoma (PDAC) has remained essentially unchanged for decades and its relative contribution to overall cancer death is projected to only increase in the coming years. Current treatment for PDAC includes aggressive chemotherapy and surgical resection in a limited number of patients, with median survival of optimal treatment rather dismal. Recent advances in gene therapies offer novel opportunities for treatment, even in those with locally advanced disease. In this review, we summarize emerging techniques to the design and administration of virotherapy, synthetic vectors, and gene-editing technology. Despite these promising advances, shortcomings continue to exist and here will also be highlighted those approaches to overcoming obstacles in current laboratory and clinical research.
Collapse
Affiliation(s)
| | - Keith Wirth
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
- Surgery BTR, MMC 195, 8195F, 420 Delaware St SE, Minneapolis, MN 55455, USA.
| |
Collapse
|