1
|
Kalter N, Gulati S, Rosenberg M, Ayaz Q, Nguyen J, Wang S, Schroeder B, Li CY, Hendel A. Precise measurement of CRISPR genome editing outcomes through single-cell DNA sequencing. Mol Ther Methods Clin Dev 2025; 33:101449. [PMID: 40225018 PMCID: PMC11987616 DOI: 10.1016/j.omtm.2025.101449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 03/11/2025] [Indexed: 04/15/2025]
Abstract
Gene therapy for clinical applications necessitates a comprehensive, accurate, and precise measurement of gene-edited drug products. State-of-the-art pipelines for evaluating editing outcomes rely primarily on bulk sequencing approaches, which are limited to population-level assessment. Here, we leveraged Tapestri, a single-cell sequencing technology for an in-depth analysis of editing outcomes. Using this platform, we characterized the genotype of triple-edited cells simultaneously at more than 100 loci, including editing zygosity, structural variations, and cell clonality. Our findings revealed a unique editing pattern in nearly every edited cell, highlighting the importance of single-cell resolution measurement to ensure the highest safety standards.
Collapse
Affiliation(s)
- Nechama Kalter
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 529002, Israel
| | - Saurabh Gulati
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Michael Rosenberg
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 529002, Israel
| | - Qawer Ayaz
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Joanne Nguyen
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Shu Wang
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Benjamin Schroeder
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Chieh-Yuan Li
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Ayal Hendel
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 529002, Israel
| |
Collapse
|
2
|
Nam H, Han J, Yu J, Cho C, Kim D, Kim Y, Kim M, Kim J, Jo D, Bae S. Autophagy induction enhances homologous recombination-associated CRISPR-Cas9 gene editing. Nucleic Acids Res 2025; 53:gkaf258. [PMID: 40239991 PMCID: PMC11997770 DOI: 10.1093/nar/gkaf258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 02/24/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 (CRISPR-associated protein 9)-based gene editing via homologous recombination (HR) enables precise gene correction and insertion. However, its low efficiency poses a challenge due to the predominance of nonhomologous end-joining during DNA repair processes. Although numerous efforts have been made to boost HR efficiency, there remains a critical need to devise a novel method that can be universally applied across cell types and in vivo animals, which could ultimately facilitate therapeutic treatments. This study demonstrated that autophagy induction using different protocols, including nutrient deprivation or chemical treatment, significantly improved HR-associated gene editing at diverse genomic loci in mammalian cells. Notably, interacting cofactor proteins that bind to Cas9 under the autophagic condition have been identified, and autophagy induction could also enhance in vivo HR-associated gene editing in mice. These findings pave the way for effective gene correction or insertion for in vivo therapeutic treatments.
Collapse
Affiliation(s)
- Hye Jin Nam
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jun Hee Han
- Department of Chemistry, Hanyang University, Seoul 04673, Republic of Korea
| | - Jihyeon Yu
- Medical Research Center of Genomic Medicine Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Chang Sik Cho
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Dongha Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Young Eun Kim
- Center for Bioanalysis, Division of Chemical and Medical Metrology, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - Min Ji Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University, Seoul 03080, Republic of Korea
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Dong Hyun Jo
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sangsu Bae
- Medical Research Center of Genomic Medicine Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
3
|
Jia J, Hao Y, Zhang L, Cao X, An L, Wang H, Ma Q, Jin X, Ma X. Development and validation of optimized lentivirus-like particles for gene editing tool delivery with Gag-Only strategy. Eur J Med Res 2025; 30:242. [PMID: 40186294 PMCID: PMC11969815 DOI: 10.1186/s40001-025-02499-2] [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: 02/17/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND The development of gene editing tools such as CRISPR-Cas9 and base editors (BE) is critical for genetic diseases and cancer. Lentivirus-like particles (LVLPs) grows into an auspicious platform for delivering mRNA or ribonucleic proteins (RNPs) due to it integrates the advantage of viral and non-viral vectors. Current LVLP systems predominantly utilize HIV-Gag and Pol proteins. However, the reverse transcriptase and integrase of Pol, pose risks of genomic integration and potential tumorigenesis. Enhancing the safety of VLP system is essential. This study focuses on improving the LVLP to minimize these risks. METHODS We implemented a Gag-Only strategy, constructing LVLPs with HIV-Gag protein, thereby eliminating the integration risks linked to Pol. By leveraging the interactions between MS2-MCP (MS2 coat protein), PP7 and PP7 BP (PP7 binding protein), and the psi (HIV packaging signal) with HIV-Gag, we encapsulated PAMless andesine base editor (CE-8e-SpRY) mRNA and sgRNA targeting the PD1 start codon (ATG) into the LVLP. Using recombinant lentiviral vector technology, we developed a stable PD1-expressing 293T cell line (PD1-293T) to assess the editing efficiency of LVLP. RESULTS The psi-LVLP demonstrated effective packaging capabilities, achieving 15% base editing efficiency in 293T cells. By optimizing plasmid ratios, we observed increased CE-8e-SpRY mRNA copy numbers, with 30% base editing efficiency. Additionally, the integration of HDVrz (hepatitis delta virus ribozyme) and psi into sgRNA (HDVrz-psi-LVLP) substantially enhanced sgRNA copy numbers, resulting in approximately 50% base editing efficiency in 293T cells and 20% base editing efficiency in Jurkat cells. Mendelian randomization analyses revealed significant genetic correlations between PD1, B2M, CIITA, and TIGIT genes with various cancer risks. Furthermore, HDVrz-psi-LVLPs targeting the start codons of B2M, CIITA, and TIGIT exhibited high base editing activity in both Jurkat and 293T cells. CONCLUSION In conclusion, this optimized platform effectively encapsulates CE-8e-SpRY mRNA and sgRNA, achieving high editing efficiencies across multiple genes and cell types. We introduce a safer and more efficient gene editing tool delivery system by constructing LVLPs based on the Gag-Only strategy. Our study presents a promising implication for cancer immunotherapy.
Collapse
Affiliation(s)
- Jinlin Jia
- National Research Institute for Family Planning, Beijing, 100081, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- National Human Genetic Resources Center, Beijing, 100052, China
| | - Yanzhe Hao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 100052, China.
| | - Lu Zhang
- National Research Institute for Family Planning, Beijing, 100081, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- National Human Genetic Resources Center, Beijing, 100052, China
| | - Xiaofang Cao
- National Research Institute for Family Planning, Beijing, 100081, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- National Human Genetic Resources Center, Beijing, 100052, China
| | - Lisha An
- National Research Institute for Family Planning, Beijing, 100081, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- National Human Genetic Resources Center, Beijing, 100052, China
| | - Hu Wang
- National Research Institute for Family Planning, Beijing, 100081, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- National Human Genetic Resources Center, Beijing, 100052, China
| | - Qi Ma
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 100052, China
| | - Xiaohua Jin
- National Research Institute for Family Planning, Beijing, 100081, China.
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China.
- National Human Genetic Resources Center, Beijing, 100052, China.
| | - Xu Ma
- National Research Institute for Family Planning, Beijing, 100081, China.
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China.
- National Human Genetic Resources Center, Beijing, 100052, China.
| |
Collapse
|
4
|
Zhang Y, Zou W, Zhou Y, Chen J, Hu Y, Wu F. Pamoic acid and carbenoxolone specifically inhibit CRISPR/Cas9 in bacteria, mammalian cells, and mice in a DNA topology-specific manner. Genome Biol 2025; 26:75. [PMID: 40156040 PMCID: PMC11951523 DOI: 10.1186/s13059-025-03521-w] [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: 08/10/2024] [Accepted: 02/28/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Regulation of the target DNA cleavage activity of CRISPR/Cas has naturally evolved in a few bacteria or bacteriophages but is lacking in higher species. Thus, identification of bioactive agents and mechanisms that can suppress the activity of Cas9 is urgently needed to rebalance this new genetic pressure. RESULTS Here, we identify four specific inhibitors of Cas9 by screening 4607 compounds that could inhibit the endonuclease activity of Cas9 via three distinct mechanisms: substrate-competitive and protospacer adjacent motif (PAM)-binding site-occupation; substrate-targeting; and sgRNA-targeting mechanisms. These inhibitors inhibit, in a dose-dependent manner, the activity of Streptococcus pyogenes Cas9 (SpyCas9), Staphylococcus aureus Cas9 (SauCas9), and SpyCas9 nickase-based BE4 base editors in in vitro purified enzyme assays, bacteria, mammalian cells, and mice. Importantly, pamoic acid and carbenoxolone show DNA-topology selectivity and preferentially inhibit the cleavage of linear DNA compared with a supercoiled plasmid. CONCLUSIONS These pharmacologically selective inhibitors and new mechanisms offer new tools for controlling the DNA-topology selective activity of Cas9.
Collapse
Affiliation(s)
- Yuxuan Zhang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wentao Zou
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yueyang Zhou
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jiaqi Chen
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Youtian Hu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Fang Wu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
5
|
Ganesh I, Karthiga I, Murugan M, Rangarajalu K, Ballambattu VB, Ravikumar S. CRISPR/Cas-Based Prenatal Screening for Aneuploidy: Challenges and Opportunities for Early Diagnosis. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:610. [PMID: 40282900 PMCID: PMC12028914 DOI: 10.3390/medicina61040610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025]
Abstract
Aneuploidy is increasingly recognized globally as a common cause of miscarriage among expectant mothers. The existing prenatal screening techniques for detecting aneuploidy have several limitations. The ability to diagnose aneuploidy early in a non-invasive manner is not feasible with the current screening methods, as they may produce false positive or false negative results. Recently, the widely used gene editing tool CRISPR/Cas has shown great promise in diagnostics. This review summarizes the prenatal screening tests used in the first trimester to assess aneuploidy conditions. Additionally, we discuss the advantages and disadvantages of molecular diagnostic tests, including the benefits and challenges of CRISPR/Cas-based trisomy detection. Thus, the proposed prenatal screening using CRISPR/Cas could provide significant benefits to expectant mothers by potentially enabling the early diagnosis of trisomy, helping to prevent miscarriage and birth defects. Furthermore, it opens new avenues for research, allowing clinicians and researchers to develop, optimize, and implement CRISPR/Cas-based prenatal screening assays in the future.
Collapse
Affiliation(s)
- Irisappan Ganesh
- Department of Medical Biotechnology, Aarupadai Veedu Medical College & Hospital, Vinayaka Mission’s Research Foundation (Deemed to be University), Puducherry 607402, India; (I.G.); (M.M.)
| | - Ilangovan Karthiga
- Department of Biochemistry, Aarupadai Veedu Medical College & Hospital, Vinayaka Mission’s Research Foundation (Deemed to be University), Puducherry 607402, India; (I.K.); (K.R.)
| | - Manoranjani Murugan
- Department of Medical Biotechnology, Aarupadai Veedu Medical College & Hospital, Vinayaka Mission’s Research Foundation (Deemed to be University), Puducherry 607402, India; (I.G.); (M.M.)
| | - Kumar Rangarajalu
- Department of Biochemistry, Aarupadai Veedu Medical College & Hospital, Vinayaka Mission’s Research Foundation (Deemed to be University), Puducherry 607402, India; (I.K.); (K.R.)
| | - Vishnu Bhat Ballambattu
- Advisor—Medical Research & Publications, Aarupadai Veedu Medical College & Hospital, Vinayaka Mission’s Research Foundation (Deemed to be University), Puducherry 607402, India;
| | - Sambandam Ravikumar
- Department of Medical Biotechnology, Aarupadai Veedu Medical College & Hospital, Vinayaka Mission’s Research Foundation (Deemed to be University), Puducherry 607402, India; (I.G.); (M.M.)
| |
Collapse
|
6
|
Liu D, Cao D, Han R. Recent advances in therapeutic gene-editing technologies. Mol Ther 2025:S1525-0016(25)00200-X. [PMID: 40119516 DOI: 10.1016/j.ymthe.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/26/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
The advent of gene-editing technologies, particularly CRISPR-based systems, has revolutionized the landscape of biomedical research and gene therapy. Ongoing research in gene editing has led to the rapid iteration of CRISPR technologies, such as base and prime editors, enabling precise nucleotide changes without the need for generating harmful double-strand breaks (DSBs). Furthermore, innovations such as CRISPR fusion systems with DNA recombinases, DNA polymerases, and DNA ligases have expanded the size limitations for edited sequences, opening new avenues for therapeutic development. Beyond the CRISPR system, mobile genetic elements (MGEs) and epigenetic editors are emerging as efficient alternatives for precise large insertions or stable gene manipulation in mammalian cells. These advances collectively set the stage for next-generation gene therapy development. This review highlights recent developments of genetic and epigenetic editing tools and explores preclinical innovations poised to advance the field.
Collapse
Affiliation(s)
- Dongqi Liu
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Di Cao
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Renzhi Han
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
7
|
Padmaswari MH, Agrawal S, Nelson CE. Preclinical development of genome editing to treat Duchenne muscular dystrophy by exon skipping. J Neuromuscul Dis 2025:22143602251326993. [PMID: 40105473 DOI: 10.1177/22143602251326993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Duchenne muscular dystrophy (DMD) is caused by loss-of-function mutations to the gene encoding dystrophin. Restoring the reading frame of dystrophin by removing internal out-of-frame exons may address symptoms of DMD. Therefore, the principle of exon skipping has been at the center stage in drug development for Duchenne muscular dystrophy (DMD) over the past two decades. Antisense oligonucleotides (AONs) have proven effective in modulating splicing sites for exon skipping, resulting in the FDA approval of several drugs using this technique in recent years. However, due to the temporary nature of AON, researchers are actively exploring genome editing as a potential long-term, single-administration treatment. The advancements in genome-editing technology over the last decade have boosted this transition. While no clinical trials for exon skipping in DMD via genome editing have been conducted as of this writing, preclinical studies show encouraging results. This review describes the preclinical landscape of genome editing for exon skipping in DMD treatment. Along with highlighting the adaptability of genome editing in exon skipping, this review also describes delivery challenges and outlines future research directions that could set a new stage for enhanced therapeutic development in DMD.
Collapse
Affiliation(s)
- Made Harumi Padmaswari
- Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
- Cell and Molecular Biology, University of Arkansas, Fayetteville, AR, USA
| | - Shilpi Agrawal
- Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Christopher E Nelson
- Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
- Cell and Molecular Biology, University of Arkansas, Fayetteville, AR, USA
| |
Collapse
|
8
|
Cao ML, Han RY, Chen SD, Zhao DY, Shi MY, Zou JH, Li L, Jiang HK. Gene Editing: An Effective Tool for the Future Treatment of Kidney Disease. J Inflamm Res 2025; 18:4001-4018. [PMID: 40125088 PMCID: PMC11927957 DOI: 10.2147/jir.s506760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Gene editing technology involves modifying target genes to alter genetic traits and generate new phenotypes. Beginning with zinc-finger nucleases (ZFN) and transcription activator-like effector nucleases (TALEN), the field has evolved through the advent of clustered regularly interspaced short palindromic repeats and CRISPR-associated protein (CRISPR-Cas) systems, and more recently to base editors (BE) and prime editors (PE). These innovations have provided deep insights into the molecular mechanisms of complex biological processes and have paved the way for novel therapeutic strategies for a range of diseases. Gene editing is now being applied in the treatment of both genetic and acquired kidney diseases, as well as in kidney transplantation and the correction of genetic mutations. This review explores the current applications of mainstream gene editing technologies in biology, with a particular emphasis on their roles in kidney disease research and treatment of. It also addresses the limitations and challenges associated with these technologies, while offering perspectives on their future potential in this field.
Collapse
Affiliation(s)
- Mei-Ling Cao
- Department of Neonatology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| | - Rui-Yi Han
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| | - Si-Da Chen
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People’s Republic of China
| | - Dan-Yang Zhao
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| | - Ming-Yue Shi
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| | - Jia-Hui Zou
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| | - Lei Li
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People’s Republic of China
| | - Hong-Kun Jiang
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| |
Collapse
|
9
|
Hunt JM, du Rand A, Verdon D, Clemance L, Loef E, Malhi C, Buttle B, Knapp DJ, Michaels YS, Garlick J, Dunbar PR, Purvis D, Feisst V, Sheppard H. Enhanced HDR-mediated correction of heterozygous COL7A1 mutations for recessive dystrophic epidermolysis bullosa. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102472. [PMID: 40027886 PMCID: PMC11872078 DOI: 10.1016/j.omtn.2025.102472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
Gene editing facilitated by homology-directed repair (HDR) holds great potential for treating monogenetic disorders such as recessive dystrophic epidermolysis bullosa (RDEB). However, low efficiency and variability between loci must be overcome for its widespread adoption into personalized therapies. To address these challenges, we developed a highly efficient and versatile gene editing strategy for RDEB that incorporates the small molecule inhibitor M3814 to enhance HDR. We focused on three RDEB causative COL7A1 mutations not previously targeted by existing gene therapies. Editing was achieved using Cas9-nuclease ribonucleoproteins with short single-stranded DNA donor templates, and outcomes were assessed with an Oxford Nanopore Technology sequencing analysis pipeline. We demonstrate precise genomic HDR rates of up to 75% of alleles in primary RDEB keratinocytes and 32% in fibroblasts. This approach restored collagen VII expression in up to 80% of keratinocytes within a bulk-edited population and resulted in correct collagen VII deposition in a 3D skin model. Additionally, at one locus we show that a dual Cas9-nickase strategy is less effective than Cas9-nuclease and prone to large on-target deletions. Our results demonstrate a significant advancement in the efficiency and consistency of HDR editing, potentially paving the way for more effective personalized gene therapies.
Collapse
Affiliation(s)
- John M.T. Hunt
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
- Maurice Wilkins Center, New Zealand
| | - Alex du Rand
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Daniel Verdon
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Leah Clemance
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Evert Loef
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Chloe Malhi
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Ben Buttle
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - David J.H.F. Knapp
- Institut de recherche en immunologie et en cancérologie (IRIC) and Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Yale S. Michaels
- Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB R2H 2A6, Canada
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Jonathan Garlick
- School of Dental Medicine, Tufts University, Boston, MA 02111, USA
| | - P. Rod Dunbar
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
- Maurice Wilkins Center, New Zealand
| | - Diana Purvis
- Te Whatu Ora Health New Zealand, Te Toka Tumai, Auckland 1023, New Zealand
| | - Vaughan Feisst
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Hilary Sheppard
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
- Maurice Wilkins Center, New Zealand
| |
Collapse
|
10
|
Chavhan RL, Jaybhaye SG, Hinge VR, Deshmukh AS, Shaikh US, Jadhav PK, Kadam US, Hong JC. Emerging applications of gene editing technologies for the development of climate-resilient crops. Front Genome Ed 2025; 7:1524767. [PMID: 40129518 PMCID: PMC11931038 DOI: 10.3389/fgeed.2025.1524767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/07/2025] [Indexed: 03/26/2025] Open
Abstract
Climate change threatens global crop yield and food security due to rising temperatures, erratic rainfall, and increased abiotic stresses like drought, heat, and salinity. Gene editing technologies, including CRISPR/Cas9, base editors, and prime editors, offer precise tools for enhancing crop resilience. This review explores the mechanisms of these technologies and their applications in developing climate-resilient crops to address future challenges. While CRISPR/enables targeted modifications of plant DNA, the base editors allow for direct base conversion without inducing double-stranded breaks, and the prime editors enable precise insertions, deletions, and substitutions. By understanding and manipulating key regulator genes involved in stress responses, such as DREB, HSP, SOS, ERECTA, HsfA1, and NHX; crop tolerance can be enhanced against drought, heat, and salt stress. Gene editing can improve traits related to root development, water use efficiency, stress response pathways, heat shock response, photosynthesis, membrane stability, ion homeostasis, osmotic adjustment, and oxidative stress response. Advancements in gene editing technologies, integration with genomics, phenomics, artificial intelligence (AI)/machine learning (ML) hold great promise. However, challenges such as off-target effects, delivery methods, and regulatory barriers must be addressed. This review highlights the potential of gene editing to develop climate-resilient crops, contributing to food security and sustainable agriculture.
Collapse
Affiliation(s)
- R. L. Chavhan
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - S. G. Jaybhaye
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - V. R. Hinge
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - A. S. Deshmukh
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - U. S. Shaikh
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - P. K. Jadhav
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - U. S. Kadam
- Division of Applied Life Science (BK21 Four), Division of Life Science, Plant Molecular Biology and Biotechnology Research Centre (PMBBRC), Gyeongsang National University, Jinju, Republic of Korea
| | - J. C. Hong
- Division of Applied Life Science (BK21 Four), Division of Life Science, Plant Molecular Biology and Biotechnology Research Centre (PMBBRC), Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
11
|
Pattali RK, Ornelas IJ, Nguyen CD, Xu D, Divekar NS, Nuñez NK. CRISPRoff epigenome editing for programmable gene silencing in human cell lines and primary T cells. Methods Enzymol 2025; 712:517-551. [PMID: 40121086 PMCID: PMC12052198 DOI: 10.1016/bs.mie.2025.01.010] [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] [Indexed: 03/25/2025]
Abstract
The advent of CRISPR-based technologies has enabled the rapid advancement of programmable gene manipulation in cells, tissues, and whole organisms. An emerging platform for targeted gene perturbation is epigenetic editing, the direct editing of chemical modifications on DNA and histones that ultimately results in repression or activation of the targeted gene. In contrast to CRISPR nucleases, epigenetic editors modulate gene expression without inducing DNA breaks or altering the genomic sequence of host cells. Recently, we developed the CRISPRoff epigenetic editing technology that simultaneously establishes DNA methylation and repressive histone modifications at targeted gene promoters. Transient expression of CRISPRoff and the accompanying single guide RNAs in mammalian cells results in transcriptional repression of targeted genes that is memorized heritably by cells through cell division and differentiation. Here, we describe our protocol for the delivery of CRISPRoff through plasmid DNA transfection, as well as the delivery of CRISPRoff mRNA, into transformed human cell lines and primary immune cells. We also provide guidance on evaluating target gene silencing and highlight key considerations when utilizing CRISPRoff for gene perturbations. Our protocols are broadly applicable to other CRISPR-based epigenetic editing technologies, as programmable genome manipulation tools continue to evolve rapidly.
Collapse
Affiliation(s)
- Rithu K Pattali
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Izaiah J Ornelas
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Carolyn D Nguyen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Da Xu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Nikita S Divekar
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - NunezJames K Nuñez
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States; Chan Zuckerberg Biohub San Francisco, San Francisco, CA, United States.
| |
Collapse
|
12
|
Barber HM, Pater AA, Gagnon KT, Damha MJ, O'Reilly D. Chemical engineering of CRISPR-Cas systems for therapeutic application. Nat Rev Drug Discov 2025; 24:209-230. [PMID: 39690326 DOI: 10.1038/s41573-024-01086-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 12/19/2024]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) technology has transformed molecular biology and the future of gene-targeted therapeutics. CRISPR systems comprise a CRISPR-associated (Cas) endonuclease and a guide RNA (gRNA) that can be programmed to guide sequence-specific binding, cleavage, or modification of complementary DNA or RNA. However, the application of CRISPR-based therapeutics is challenged by factors such as molecular size, prokaryotic or phage origins, and an essential gRNA cofactor requirement, which impact efficacy, delivery and safety. This Review focuses on chemical modification and engineering approaches for gRNAs to enhance or enable CRISPR-based therapeutics, emphasizing Cas9 and Cas12a as therapeutic paradigms. Issues that chemically modified gRNAs seek to address, including drug delivery, physiological stability, editing efficiency and off-target effects, as well as challenges that remain, are discussed.
Collapse
Affiliation(s)
- Halle M Barber
- Department of Chemistry, McGill University, Montreal, Quebec, Canada
| | - Adrian A Pater
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Keith T Gagnon
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | - Masad J Damha
- Department of Chemistry, McGill University, Montreal, Quebec, Canada.
| | - Daniel O'Reilly
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
13
|
de Franceschi L, Locatelli F, Rees D, Chabannon C, Dalle J, Rivella S, Iolascon A, Lobitz S, Abboud MR, de la Fuente J, Flevari P, Angelucci E, de Montalembert M. Selecting patients with sickle cell disease for gene addition or gene editing-based therapeutic approaches: Report on behalf of a joint EHA Specialized Working Group and EBMT Hemoglobinopathies Working Party consensus conference. Hemasphere 2025; 9:e70089. [PMID: 40084235 PMCID: PMC11904809 DOI: 10.1002/hem3.70089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/05/2024] [Accepted: 12/09/2024] [Indexed: 03/16/2025] Open
Abstract
Sickle cell disease (SCD) remains associated with reduced life expectancy and poor quality of life despite improvements observed in the last decades mostly related to comprehensive care, use of hydroxycarbamide, screening to identify patients at risk of strokes, and implementation of safe transfusion protocols. The course of the disease is highly variable, making it difficult to predict severity and response to therapy. Allogeneic hematopoietic stem cell transplantation potentially provides a cure with a relatively low rate of complications, but few patients have an HLA-identical sibling. The hopes of patients and healthcare providers have been raised after the initial excellent results of gene therapy studies. However, there is a strong contrast between the high expectations of families and patients and the limited availability of the product, which is technically complex and very expensive. In light of this consideration and of the limited data available on the long-term efficacy and toxicity of different gene therapy approaches, the European Hematology Association Red Cell & Iron Specialized Working Group (EHA SWG) and the hemoglobinopathy working part of the European Blood & Marrow Transplant (EBMT) Group have prioritized the development of recommendations for selection of patients with SCD who are good candidates for gene therapy. The decision-making algorithm was developed by a panel of experts in hemoglobinopathies and/or transplantation chosen by EHA SWG and EBMT, to discuss the selection of SCD patients for gene therapy and draw notes on the related clinical problems.
Collapse
Affiliation(s)
- Lucia de Franceschi
- Department of Engineering for Innovative MedicineUniversity of VeronaVeronaItaly
- Azienda Ospedaliera Universitaria integrata di VeronaVeronaItaly
| | - Franco Locatelli
- IRCCS Bambino Gesù Children's HospitalCatholic University of the Sacred HeartRomeItaly
| | - David Rees
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences Medicine, King's College London, and Department of Haematological MedicineKing's College HospitalLondonUK
| | - Christian Chabannon
- Institut Paoli‐Calmettes Comprehensive Cancer Center and Module Biotherapies du Centre d'Investigations Cliniques de Marseille, INSERM‐Aix‐Marseille Université AP‐HM‐IPCCBT‐1409MarseilleFrance
| | - Jean‐Hugues Dalle
- Pediatric Hematology and Immunoloy Department, Robert‐Debré Academic HospitalGHU AP‐HP Nord Université Paris CitéParisFrance
| | - Stefano Rivella
- Department of PediatricsHematology, The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Penn Institute for RNA InnovationUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Institute for Regenerative Medicine (IRM)University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli Federico IINaplesItaly
- CEINGE Biotecnologie AvanzateNaplesItaly
| | - Stephan Lobitz
- Pediatric Hematology & Oncology, Gemeinschaftsklinikum MittelrheinKoblenzGermany
| | - Miguel R. Abboud
- Department of Pediatrics and Adolescent MedicineAmerican University of BeirutBeirutLebanon
| | - Josu de la Fuente
- Department of Immunology and InflammationCentre for Haematology, Imperial College LondonLondonUK
- Department of PaediatricsImperial College Healthcare NHS TrustLondonUK
| | - Pagona Flevari
- Thalassemia Unit—Center of Expertise in Haemoglobinopathies, Laiko General HospitalAthensGreece
| | - Emanuele Angelucci
- UO Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Mariane de Montalembert
- Department of General Pediatrics and Pediatric Infectious Diseases, Sickle Cell Center, Necker‐Enfants Malades Hospital, Assistance Publique—Hôpitaux de Paris (AP‐HP)Université Paris CitéParisFrance
- Laboratory of Excellence GR‐ExParisFrance
| |
Collapse
|
14
|
Dahl-Jessen M, Terkelsen T, Bak RO, Jensen UB. Characterization of the role of spatial proximity of DNA double-strand breaks in the formation of CRISPR-Cas9-induced large structural variations. Genome Res 2025; 35:231-241. [PMID: 39805705 PMCID: PMC11874742 DOI: 10.1101/gr.278575.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Structural variations (SVs) play important roles in genetic diversity, evolution, and carcinogenesis and are, as such, important for human health. However, it remains unclear how spatial proximity of double-strand breaks (DSBs) affects the formation of SVs. To investigate if spatial proximity between two DSBs affects DNA repair, we used data from 3C experiments (Hi-C, ChIA-PET, and ChIP-seq) to identify highly interacting loci on six different chromosomes. The target regions correlate with the borders of megabase-sized topologically associated domains (TADs), and we used CRISPR-Cas9 nuclease and pairs of single guide RNAs (sgRNAs) against these targets to generate DSBs in both K562 cells and H9 human embryonic stem cells (hESCs). Droplet digital PCR (ddPCR) was used to quantify the resulting recombination events, and high-throughput sequencing was used to analyze the chimeric junctions created between the two DSBs. We observe a significantly higher formation frequency of deletions and inversions with DSBs in proximity compared with deletions and inversions with DSBs not in proximity in K562 cells. Additionally, our results suggest that DSB proximity may affect the ligation of chimeric deletion junctions. Taken together, spatial proximity between DSBs is a significant predictor of large-scale deletion and inversion frequency induced by CRISPR-Cas9 in K562 cells. This finding has implications for understanding SVs in the human genome and for the future application of CRISPR-Cas9 in gene editing and the modeling of rare SVs.
Collapse
Affiliation(s)
- Mikkel Dahl-Jessen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Thorkild Terkelsen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark;
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
- Department for Clinical Genetics, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Rasmus O Bak
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
- Aarhus Institute of Advanced Studies (AIAS), Aarhus University, 8000 Aarhus, Denmark
| | - Uffe Birk Jensen
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
- Department for Clinical Genetics, Aarhus University Hospital, 8200 Aarhus, Denmark
| |
Collapse
|
15
|
Su KC, Radul E, Maier NK, Tsang MJ, Goul C, Moodie B, Marescal O, Keys HR, Cheeseman IM. Functional genetics reveals modulators of antimicrotubule drug sensitivity. J Cell Biol 2025; 224:e202403065. [PMID: 39570287 PMCID: PMC11590752 DOI: 10.1083/jcb.202403065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 09/04/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
Microtubules play essential roles in diverse cellular processes and are important pharmacological targets for treating human disease. Here, we sought to identify cellular factors that modulate the sensitivity of cells to antimicrotubule drugs. We conducted a genome-wide CRISPR/Cas9-based functional genetics screen in human cells treated with the microtubule-destabilizing drug nocodazole or the microtubule-stabilizing drug paclitaxel. We further conducted a focused secondary screen to test drug sensitivity for ∼1,400 gene targets across two distinct human cell lines and to additionally test sensitivity to the KIF11 inhibitor, STLC. These screens defined gene targets whose loss enhances or suppresses sensitivity to antimicrotubule drugs. In addition to gene targets whose loss sensitized cells to multiple compounds, we observed cases of differential sensitivity to specific compounds and differing requirements between cell lines. Our downstream molecular analysis further revealed additional roles for established microtubule-associated proteins and identified new players in microtubule function.
Collapse
Affiliation(s)
- Kuan-Chung Su
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Elena Radul
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Nolan K. Maier
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mary-Jane Tsang
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Claire Goul
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Océane Marescal
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Heather R. Keys
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Iain M. Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
16
|
Wang Z, Qi X, Ren X, Lin Y, Zeng F, Wang Q. Synthetic evolution of Saccharomyces cerevisiae for biomanufacturing: Approaches and applications. MLIFE 2025; 4:1-16. [PMID: 40026576 PMCID: PMC11868838 DOI: 10.1002/mlf2.12167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/19/2024] [Accepted: 12/19/2024] [Indexed: 03/05/2025]
Abstract
The yeast Saccharomyces cerevisiae is a well-studied unicellular eukaryote with a significant role in the biomanufacturing of natural products, biofuels, and bulk and value-added chemicals, as well as the principal model eukaryotic organism utilized for fundamental research. Robust tools for building and optimizing yeast chassis cells were made possible by the quick development of synthetic biology, especially in engineering evolution. In this review, we focused on methods and tools from synthetic biology that are used to design and engineer S. cerevisiae's evolution. A detailed discussion was held regarding transcriptional regulation, template-dependent and template-free approaches. Furthermore, the applications of evolved S. cerevisiae were comprehensively summarized. These included improving environmental stress tolerance and raising cell metabolic performance in the production of biofuels and bulk and value-added chemicals. Finally, the future considerations were briefly discussed.
Collapse
Affiliation(s)
- Zhen Wang
- College of Science & TechnologyHebei Agricultural UniversityCangzhouChina
| | - Xianni Qi
- Key Laboratory of Engineering Biology for Low‐carbon Manufacturing, Tianjin Institute of Industrial BiotechnologyChinese Academy of SciencesTianjinChina
- National Center of Technology Innovation for Synthetic BiologyTianjinChina
| | - Xinru Ren
- College of Science & TechnologyHebei Agricultural UniversityCangzhouChina
| | - Yuping Lin
- Key Laboratory of Engineering Biology for Low‐carbon Manufacturing, Tianjin Institute of Industrial BiotechnologyChinese Academy of SciencesTianjinChina
| | - Fanli Zeng
- College of Life SciencesHebei Agricultural UniversityBaodingChina
| | - Qinhong Wang
- Key Laboratory of Engineering Biology for Low‐carbon Manufacturing, Tianjin Institute of Industrial BiotechnologyChinese Academy of SciencesTianjinChina
- National Center of Technology Innovation for Synthetic BiologyTianjinChina
| |
Collapse
|
17
|
Doctor Y, Sanghvi M, Mali P. A Manual for Genome and Transcriptome Engineering. IEEE Rev Biomed Eng 2025; 18:250-267. [PMID: 39514364 PMCID: PMC11875898 DOI: 10.1109/rbme.2024.3494715] [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] [Indexed: 11/16/2024]
Abstract
Genome and transcriptome engineering have emerged as powerful tools in modern biotechnology, driving advancements in precision medicine and novel therapeutics. In this review, we provide a comprehensive overview of the current methodologies, applications, and future directions in genome and transcriptome engineering. Through this, we aim to provide a guide for tool selection, critically analyzing the strengths, weaknesses, and best use cases of these tools to provide context on their suitability for various applications. We explore standard and recent developments in genome engineering, such as base editors and prime editing, and provide insight into tool selection for change of function (knockout, deletion, insertion, substitution) and change of expression (repression, activation) contexts. Advancements in transcriptome engineering are also explored, focusing on established technologies like antisense oligonucleotides (ASOs) and RNA interference (RNAi), as well as recent developments such as CRISPR-Cas13 and adenosine deaminases acting on RNA (ADAR). This review offers a comparison of different approaches to achieve similar biological goals, and consideration of high-throughput applications that enable the probing of a variety of targets. This review elucidates the transformative impact of genome and transcriptome engineering on biological research and clinical applications that will pave the way for future innovations in the field.
Collapse
Affiliation(s)
| | | | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, CA 92039, USA
| |
Collapse
|
18
|
Bonowicz K, Jerka D, Piekarska K, Olagbaju J, Stapleton L, Shobowale M, Bartosiński A, Łapot M, Bai Y, Gagat M. CRISPR-Cas9 in Cardiovascular Medicine: Unlocking New Potential for Treatment. Cells 2025; 14:131. [PMID: 39851560 PMCID: PMC11763404 DOI: 10.3390/cells14020131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain a significant global health challenge, with many current treatments addressing symptoms rather than the genetic roots of these conditions. The advent of CRISPR-Cas9 technology has revolutionized genome editing, offering a transformative approach to targeting disease-causing mutations directly. This article examines the potential of CRISPR-Cas9 in the treatment of various CVDs, including atherosclerosis, arrhythmias, cardiomyopathies, hypertension, and Duchenne muscular dystrophy (DMD). The technology's ability to correct single-gene mutations with high precision and efficiency positions it as a groundbreaking tool in cardiovascular therapy. Recent developments have extended the capabilities of CRISPR-Cas9 to include mitochondrial genome editing, a critical advancement for addressing mitochondrial dysfunctions often linked to cardiovascular disorders. Despite its promise, significant challenges remain, including off-target effects, ethical concerns, and limitations in delivery methods, which hinder its translation into clinical practice. This article also explores the ethical and regulatory considerations surrounding gene editing technologies, emphasizing the implications of somatic versus germline modifications. Future research efforts should aim to enhance the accuracy of CRISPR-Cas9, improve delivery systems for targeted tissues, and ensure the safety and efficacy of treatments in the long term. Overcoming these obstacles could enable CRISPR-Cas9 to not only treat but also potentially cure genetically driven cardiovascular diseases, heralding a new era in precision medicine for cardiovascular health.
Collapse
Affiliation(s)
- Klaudia Bonowicz
- Department of Histology and Embryology and Vascular Biology Student Research Club, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.B.); (D.J.); (K.P.); (J.O.); (L.S.); (M.S.)
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Płock, 09-402 Płock, Poland; (A.B.); (M.Ł.)
| | - Dominika Jerka
- Department of Histology and Embryology and Vascular Biology Student Research Club, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.B.); (D.J.); (K.P.); (J.O.); (L.S.); (M.S.)
| | - Klaudia Piekarska
- Department of Histology and Embryology and Vascular Biology Student Research Club, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.B.); (D.J.); (K.P.); (J.O.); (L.S.); (M.S.)
| | - Janet Olagbaju
- Department of Histology and Embryology and Vascular Biology Student Research Club, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.B.); (D.J.); (K.P.); (J.O.); (L.S.); (M.S.)
| | - Laura Stapleton
- Department of Histology and Embryology and Vascular Biology Student Research Club, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.B.); (D.J.); (K.P.); (J.O.); (L.S.); (M.S.)
| | - Munirat Shobowale
- Department of Histology and Embryology and Vascular Biology Student Research Club, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.B.); (D.J.); (K.P.); (J.O.); (L.S.); (M.S.)
| | - Andrzej Bartosiński
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Płock, 09-402 Płock, Poland; (A.B.); (M.Ł.)
| | - Magdalena Łapot
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Płock, 09-402 Płock, Poland; (A.B.); (M.Ł.)
| | - Yidong Bai
- Department of Cell Systems and Anatomy, UT Health, Long School of Medicine, San Antonio, TX 78229, USA;
| | - Maciej Gagat
- Department of Histology and Embryology and Vascular Biology Student Research Club, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland; (K.B.); (D.J.); (K.P.); (J.O.); (L.S.); (M.S.)
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Płock, 09-402 Płock, Poland; (A.B.); (M.Ł.)
| |
Collapse
|
19
|
Egawa M, Uno N, Komazaki R, Ohkame Y, Yamazaki K, Yoshimatsu C, Ishizu Y, Okano Y, Miyamoto H, Osaki M, Suzuki T, Hosomichi K, Aizawa Y, Kazuki Y, Tomizuka K. Generation of Monosomy 21q Human iPS Cells by CRISPR/Cas9-Mediated Interstitial Megabase Deletion. Genes Cells 2025; 30:e13184. [PMID: 39581190 DOI: 10.1111/gtc.13184] [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: 09/16/2024] [Revised: 11/05/2024] [Accepted: 11/09/2024] [Indexed: 11/26/2024]
Abstract
Missing an entire chromosome or chromosome arm in normal diploid cells has a deleterious impact on cell viability, which may contribute to the development of specific birth defects. Nevertheless, the effects of chromosome loss in human cells have remained unexplored due to the lack of suitable model systems. Here, we developed an efficient, selection-free approach to generate partial monosomy in human induced pluripotent stem cells (iPSCs). The introduction of Cas9 proteins and a pair of gRNAs induces over megabase-sized interstitial chromosomal deletions. Using human chromosome 21 (HSA21) as a model, partial monosomy 21q (PM21q) iPSC lines with deletions ranging from 4.5 to 27.9 Mb were isolated. A 33.6 Mb deletion, encompassing all protein-coding genes on 21q, was also achieved, establishing the first 21q monosomy human iPSC line. Transcriptome and proteome analyses revealed that the abundances of mRNA and protein encoded by the majority of genes in the monosomic regions are half of the diploid expression level, indicating an absence of dosage compensation. The ability to generate customized partial monosomy cell lines on an isogenic, karyotypically normal background should facilitate the gain of novel insights into the impact of chromosome loss on cellular fitness.
Collapse
Affiliation(s)
- Masaya Egawa
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Narumi Uno
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Rina Komazaki
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yusuke Ohkame
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
| | - Kyotaro Yamazaki
- Chromosome Engineering Research Group, the Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
- Department of Homeostatic Regulation, National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki, Japan
| | - Chihiro Yoshimatsu
- Department of Chromosome Biomedical Engineering, Integrated Medical Sciences, Graduate School of Medical Sciences, Tottori University, Tottori, Japan
| | - Yuki Ishizu
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yusaku Okano
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Hitomaru Miyamoto
- Department of Chromosome Biomedical Engineering, Integrated Medical Sciences, Graduate School of Medical Sciences, Tottori University, Tottori, Japan
| | - Mitsuhiko Osaki
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Teruhiko Suzuki
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kazuyoshi Hosomichi
- Laboratory of Computational Genomics, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yasunori Aizawa
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
- Kanagawa Institute of Industrial Science and Technology, Ebina, Japan
| | - Yasuhiro Kazuki
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
- Chromosome Engineering Research Group, the Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
- Department of Chromosome Biomedical Engineering, Integrated Medical Sciences, Graduate School of Medical Sciences, Tottori University, Tottori, Japan
| | - Kazuma Tomizuka
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
20
|
Sousa AA, Hemez C, Lei L, Traore S, Kulhankova K, Newby GA, Doman JL, Oye K, Pandey S, Karp PH, McCray PB, Liu DR. Systematic optimization of prime editing for the efficient functional correction of CFTR F508del in human airway epithelial cells. Nat Biomed Eng 2025; 9:7-21. [PMID: 38987629 PMCID: PMC11754097 DOI: 10.1038/s41551-024-01233-3] [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: 12/28/2023] [Accepted: 06/12/2024] [Indexed: 07/12/2024]
Abstract
Prime editing (PE) enables precise and versatile genome editing without requiring double-stranded DNA breaks. Here we describe the systematic optimization of PE systems to efficiently correct human cystic fibrosis (CF) transmembrane conductance regulator (CFTR) F508del, a three-nucleotide deletion that is the predominant cause of CF. By combining six efficiency optimizations for PE-engineered PE guide RNAs, the PEmax architecture, the transient expression of a dominant-negative mismatch repair protein, strategic silent edits, PE6 variants and proximal 'dead' single-guide RNAs-we increased correction efficiencies for CFTR F508del from less than 0.5% in HEK293T cells to 58% in immortalized bronchial epithelial cells (a 140-fold improvement) and to 25% in patient-derived airway epithelial cells. The optimizations also resulted in minimal off-target editing, in edit-to-indel ratios 3.5-fold greater than those achieved by nuclease-mediated homology-directed repair, and in the functional restoration of CFTR ion channels to over 50% of wild-type levels (similar to those achieved via combination treatment with elexacaftor, tezacaftor and ivacaftor) in primary airway cells. Our findings support the feasibility of a durable one-time treatment for CF.
Collapse
Affiliation(s)
- Alexander A Sousa
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Colin Hemez
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Lei Lei
- Stead Family Department of Pediatrics and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Soumba Traore
- Stead Family Department of Pediatrics and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Katarina Kulhankova
- Stead Family Department of Pediatrics and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jordan L Doman
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Keyede Oye
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Smriti Pandey
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Philip H Karp
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, IA, USA
| | - Paul B McCray
- Stead Family Department of Pediatrics and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
21
|
Xu W, Zhang S, Qin H, Yao K. From bench to bedside: cutting-edge applications of base editing and prime editing in precision medicine. J Transl Med 2024; 22:1133. [PMID: 39707395 DOI: 10.1186/s12967-024-05957-3] [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: 09/25/2024] [Accepted: 12/08/2024] [Indexed: 12/23/2024] Open
Abstract
CRISPR-based gene editing technology theoretically allows for precise manipulation of any genetic target within living cells, achieving the desired sequence modifications. This revolutionary advancement has fundamentally transformed the field of biomedicine, offering immense clinical potential for treating and correcting genetic disorders. In the treatment of most genetic diseases, precise genome editing that avoids the generation of mixed editing byproducts is considered the ideal approach. This article reviews the current progress of base editors and prime editors, elaborating on specific examples of their applications in the therapeutic field, and highlights opportunities for improvement. Furthermore, we discuss the specific performance of these technologies in terms of safety and efficacy in clinical applications, and analyze the latest advancements and potential directions that could influence the future development of genome editing technologies. Our goal is to outline the clinical relevance of this rapidly evolving scientific field and preview a roadmap for successful DNA base editing therapies for the treatment of hereditary or idiopathic diseases.
Collapse
Affiliation(s)
- Weihui Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Shiyao Zhang
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China.
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China.
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China.
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China.
| |
Collapse
|
22
|
Shembrey C, Yang R, Casan J, Hu W, Chen H, Singh GJ, Sadras T, Prasad K, Shortt J, Johnstone RW, Trapani JA, Ekert PG, Fareh M. Principles of CRISPR-Cas13 mismatch intolerance enable selective silencing of point-mutated oncogenic RNA with single-base precision. SCIENCE ADVANCES 2024; 10:eadl0731. [PMID: 39693429 DOI: 10.1126/sciadv.adl0731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/14/2024] [Indexed: 12/20/2024]
Abstract
Single-nucleotide variants (SNVs) are extremely prevalent in human cancers, although most of these remain clinically unactionable. The programmable RNA nuclease CRISPR-Cas13 has been deployed to specifically target oncogenic RNAs. However, silencing oncogenic SNVs with single-base precision remains extremely challenging due to the intrinsic mismatch tolerance of Cas13. Here, we show that introducing synthetic mismatches at precise positions of the spacer sequence enables de novo design of guide RNAs [CRISPR RNAs (crRNAs)] with strong preferential silencing of point-mutated transcripts. We applied these design principles to effectively silence the oncogenic KRAS G12 hotspot, NRAS G12D and BRAF V600E transcripts with minimal off-target silencing of the wild-type transcripts, underscoring the adaptability of this platform to silence various SNVs. Unexpectedly, the SNV-selective crRNAs harboring mismatched nucleotides reduce the promiscuous collateral activity of the RfxCas13d ortholog. These findings demonstrate that the CRISPR-Cas13 system can be reprogrammed to target mutant transcripts with single-base precision, showcasing the tremendous potential of this tool in personalized transcriptome editing.
Collapse
Affiliation(s)
- Carolyn Shembrey
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
| | - Ray Yang
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
| | - Joshua Casan
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
| | - Wenxin Hu
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
| | - Honglin Chen
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
| | - Gurjeet J Singh
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
| | - Teresa Sadras
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
| | - Krishneel Prasad
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
| | - Jake Shortt
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
- School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia
| | - Ricky W Johnstone
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
| | - Joseph A Trapani
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
| | - Paul G Ekert
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales, Sydney, Kensington, NSW, Australia
- UNSW Centre for Childhood Cancer Research, University of New South Wales, Sydney, Kensington, NSW, Australia
| | - Mohamed Fareh
- Rosie Lew Program in Immunotherapy and Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne 3052 Australia
| |
Collapse
|
23
|
Ursch LT, Müschen JS, Ritter J, Klermund J, Bernard BE, Kolb S, Warmuth L, Andrieux G, Miller G, Jiménez-Muñoz M, Theis FJ, Boerries M, Busch DH, Cathomen T, Schumann K. Modulation of TCR stimulation and pifithrin-α improve the genomic safety profile of CRISPR-engineered human T cells. Cell Rep Med 2024; 5:101846. [PMID: 39637860 PMCID: PMC11722128 DOI: 10.1016/j.xcrm.2024.101846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/24/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
CRISPR-engineered chimeric antigen receptor (CAR) T cells are at the forefront of novel cancer treatments. However, several reports describe the occurrence of CRISPR-induced chromosomal aberrations. So far, measures to increase the genomic safety of T cell products focused mainly on the components of the CRISPR-Cas9 system and less on T cell-intrinsic features, such as their massive expansion after T cell receptor (TCR) stimulation. Here, we describe driving forces of indel formation in primary human T cells. Increased T cell activation and proliferation speed correlate with larger deletions. Editing of non-activated T cells reduces the risk of large deletions with the downside of reduced knockout efficiencies. Alternatively, the addition of the small-molecule pifithrin-α limits large deletions, chromosomal translocations, and aneuploidy in a p53-independent manner while maintaining the functionality of CRISPR-engineered T cells, including CAR T cells. Controlling T cell activation and pifithrin-α treatment are easily implementable strategies to improve the genomic integrity of CRISPR-engineered T cells.
Collapse
Affiliation(s)
- Laurenz T Ursch
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Jule S Müschen
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Julia Ritter
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Julia Klermund
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106 Freiburg, Germany; Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, 79106 Freiburg, Germany
| | - Bettina E Bernard
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Saskia Kolb
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Linda Warmuth
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Gregor Miller
- Core Facility Statistical Consulting, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Marina Jiménez-Muñoz
- Core Facility Statistical Consulting, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; School of Computing, Information and Technology, Technical University of Munich, 85748 Garching, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg, 79106 Freiburg, Germany
| | - Dirk H Busch
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany; German Center for Infection Research, Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Munich, 81675 Munich, Germany
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106 Freiburg, Germany; Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Kathrin Schumann
- Technical University of Munich (TUM), School of Medicine and Health, Department of Preclinical Medicine, Institute for Medical Microbiology, Immunology and Hygiene, 81675 Munich, Germany; TUM, Institute for Advanced Study, 85748 Garching, Germany.
| |
Collapse
|
24
|
Georgakopoulou A, Li C, Kiem HP, Lieber A. In vitro and in vivo expansion of CD33/HBG promoter-edited HSPCs with Mylotarg. Mol Ther Methods Clin Dev 2024; 32:101343. [PMID: 39429723 PMCID: PMC11490927 DOI: 10.1016/j.omtm.2024.101343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024]
Abstract
We developed an in vivo HSC gene therapy approach that consists of HSC mobilization and intravenous injection of HSC-tropic HDAd vectors. To achieve therapeutically relevant numbers of corrected cells, we incorporated in vivo expansion of transduced cells. We used an HDAd vector for a multiplex adenine base editing approach to (1) remove the region within CD33 that is recognized by gemtuzumab ozogamicin (GO) (Mylotarg), and (2) create therapeutic edits within the HBG1/2 promoters to reactivate γ-globin/HbF. In vitro studies with HDAd-transduced human CD34+ cells showed editing of both targeted sites and a 2- to 3-fold GO-mediated expansion of edited erythroid/myeloid progenitors. After erythroid in vitro differentiation, up to 40% of erythrocytes were HbF positive. For in vivo studies, mice were transplanted with human CD34+ cells. After engraftment, HSCs were mobilized with G-CSF/AMD3100 followed by an intravenous HDAd injection and GO-mediated in vivo selection. Two months later, editing in human cells within the bone marrow was significantly higher in GO-treated mice. The percentage of HbF+ human erythroid cells was 2.5-fold greater compared with untreated mice. These data indicate that in vivo GO selection can increase edited erythroid cells.
Collapse
Affiliation(s)
- Aphrodite Georgakopoulou
- University of Washington, Department of Medicine, Division of Medical Genetics, Seattle, WA 98195, USA
| | - Chang Li
- University of Washington, Department of Medicine, Division of Medical Genetics, Seattle, WA 98195, USA
| | | | - André Lieber
- University of Washington, Department of Medicine, Division of Medical Genetics, Seattle, WA 98195, USA
| |
Collapse
|
25
|
Masi S, Dalpiaz H, Borghi C. Gene editing of angiotensin for blood pressure management. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2024; 23:200323. [PMID: 39258007 PMCID: PMC11382036 DOI: 10.1016/j.ijcrp.2024.200323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 09/12/2024]
Abstract
Arterial hypertension has remained the world's leading cause of morbidity and mortality for more than 20 years. While early Genome-Wide Association Studies raised the hypothesis that a precision medicine approach could be implemented in the treatment of hypertension, the large number of single nucleotide polymorphisms that were found to be associated with blood pressure and their limited impact on the blood pressure values have initially hampered these expectations. With the development and refinement of gene-editing and RNA-based approaches allowing selective and organ-specific modulation of critical systems involved in blood pressure regulation, a renewed interest in genetic treatments for hypertension has emerged. The CRISPR-Cas9 system, antisense oligonucleotides (ASO) and small interfering RNA (siRNA) have been used to specifically target the hepatic angiotensinogen (AGT) production, with the scope of safely but effectively reducing the activation of the renin-angiotensin system, ultimately leading to an effective reduction of the blood pressure with extremely simplified treatment regimens that involve weekly, monthly or even once-in-life injection of the drugs. Among the various approaches, siRNA and ASO that reduce hepatic AGT production are in advanced development, with phase I and II clinical trials showing their safety and effectiveness. In the current manuscript, we review the mode of action of these new approaches to hypertension treatment, discussing the results of the clinical trials and their potential to revolutionize the management of hypertension.
Collapse
Affiliation(s)
- Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Hermann Dalpiaz
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Claudio Borghi
- Hypertension and Cardiovascular Disease Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40126, Bologna, Italy
- Cardiovascular Medicine Unit, Heart-Chest-Vascular Department, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40126, Bologna, Italy
| |
Collapse
|
26
|
Sapozhnikov DM, Szyf M. Genetic confounds of transgenerational epigenetic inheritance in mice. Epigenetics 2024; 19:2318519. [PMID: 38369744 PMCID: PMC10878023 DOI: 10.1080/15592294.2024.2318519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 02/07/2024] [Indexed: 02/20/2024] Open
Abstract
Transgenerational epigenetic inheritance in mammals remains a controversial phenomenon. A recent study by Takahashi et al. provides evidence for this mode of inheritance in mice by using a CRISPR/Cas9-based epigenetic editing technique to modify DNA methylation levels at specific promoters and then demonstrating the inheritance of the gain in methylation in offspring. In this technical commentary, we argue that the method used in the original study inherently amplifies the likelihood of genetic changes that thereafter lead to the heritability of epigenetic changes. We provide evidence that genetic changes from multiple sources do indeed occur in these experiments and explore several avenues by which these changes could be causal to the apparent inheritance of epigenetic changes. We conclude a genetic basis of inheritance cannot be ruled out and thus transgenerational epigenetic inheritance has not been adequately established by the original study.
Collapse
Affiliation(s)
- Daniel M. Sapozhnikov
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Moshe Szyf
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
27
|
Steinbeck BJ, Gao XD, McElroy AN, Pandey S, Doman JL, Riddle MJ, Xia L, Chen W, Eide CR, Lengert AH, Han SW, Blazar BR, Wandall HH, Dabelsteen S, Liu DR, Tolar J, Osborn MJ. Twin Prime Editing Mediated Exon Skipping/Reinsertion for Restored Collagen VII Expression in Recessive Dystrophic Epidermolysis Bullosa. J Invest Dermatol 2024; 144:2764-2777.e9. [PMID: 38763174 PMCID: PMC12050016 DOI: 10.1016/j.jid.2024.04.013] [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: 08/28/2023] [Revised: 04/12/2024] [Accepted: 04/26/2024] [Indexed: 05/21/2024]
Abstract
Gene editing nucleases, base editors, and prime editors are potential locus-specific genetic treatment strategies for recessive dystrophic epidermolysis bullosa; however, many recessive dystrophic epidermolysis bullosa COL7A1 pathogenic nucleotide variations (PNVs) are unique, making the development of personalized editing reagents challenging. A total of 270 of the ∼320 COL7A1 epidermolysis bullosa PNVs reside in exons that can be skipped, and antisense oligonucleotides and gene editing nucleases have been used to create in-frame deletions. Antisense oligonucleotides are transient, and nucleases generate deleterious double-stranded DNA breaks and uncontrolled mixtures of allele products. We developed a twin prime editing strategy using the PEmax and recently evolved PE6 prime editors and dual prime editing guide RNAs flanking COL7A1 exon 5. Prime editing-mediated deletion of exon 5 with a homozygous premature stop codon was achieved in recessive dystrophic epidermolysis bullosa fibroblasts, keratinocytes, and induced pluripotent stem cells with minimal double-stranded DNA breaks, and collagen type VII protein was restored. Twin prime editing can replace the target exon with recombinase attachment sequences, and we exploited this to reinsert a normal copy of exon 5 using the Bxb1 recombinase. These findings demonstrate that twin prime editing can facilitate locus-specific, predictable, in-frame deletions and sequence replacement with few double-stranded DNA breaks as a strategy that may enable a single therapeutic agent to treat multiple recessive dystrophic epidermolysis bullosa patient cohorts.
Collapse
Affiliation(s)
- Benjamin J Steinbeck
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xin D Gao
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Amber N McElroy
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Smriti Pandey
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Jordan L Doman
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Megan J Riddle
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lily Xia
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Weili Chen
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cindy R Eide
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andre H Lengert
- Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Sang Won Han
- Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Bruce R Blazar
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hans H Wandall
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Sally Dabelsteen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Jakub Tolar
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mark J Osborn
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
28
|
Xu D, Besselink S, Ramadoss GN, Dierks PH, Lubin JP, Pattali RK, Brim JI, Christenson AE, Colias PJ, Ornelas IJ, Nguyen CD, Chasins SE, Conklin BR, Nuñez JK. Programmable epigenome editing by transient delivery of CRISPR epigenome editor ribonucleoproteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625496. [PMID: 39651312 PMCID: PMC11623636 DOI: 10.1101/2024.11.26.625496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Programmable epigenome editors modify gene expression in mammalian cells by altering the local chromatin environment at target loci without inducing DNA breaks. However, the large size of CRISPR-based epigenome editors poses a challenge to their broad use in biomedical research and as future therapies. Here, we present Robust ENveloped Delivery of Epigenome-editor Ribonucleoproteins (RENDER) for transiently delivering programmable epigenetic repressors (CRISPRi, DNMT3A-3L-dCas9, CRISPRoff) and activator (TET1-dCas9) as ribonucleoprotein complexes into human cells to modulate gene expression. After rational engineering, we show that RENDER induces durable epigenetic silencing of endogenous genes across various human cell types, including primary T cells. Additionally, we apply RENDER to epigenetically repress endogenous genes in human stem cell-derived neurons, including the reduction of the neurodegenerative disease associated V337M-mutated Tau protein. Together, our RENDER platform advances the delivery of CRISPR-based epigenome editors into human cells, broadening the use of epigenome editing in fundamental research and therapeutic applications.
Collapse
|
29
|
Cullot G, Aird EJ, Schlapansky MF, Yeh CD, van de Venn L, Vykhlyantseva I, Kreutzer S, Mailänder D, Lewków B, Klermund J, Montellese C, Biserni M, Aeschimann F, Vonarburg C, Gehart H, Cathomen T, Corn JE. Genome editing with the HDR-enhancing DNA-PKcs inhibitor AZD7648 causes large-scale genomic alterations. Nat Biotechnol 2024:10.1038/s41587-024-02488-6. [PMID: 39604565 DOI: 10.1038/s41587-024-02488-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024]
Abstract
The DNA-PKcs inhibitor AZD7648 enhances CRISPR-Cas9-directed homology-directed repair efficiencies, with potential for clinical utility, but its possible on-target consequences are unknown. We found that genome editing with AZD7648 causes frequent kilobase-scale and megabase-scale deletions, chromosome arm loss and translocations. These large-scale chromosomal alterations evade detection through typical genome editing assays, prompting caution in deploying AZD7648 and reinforcing the need to investigate multiple types of potential editing outcomes.
Collapse
Affiliation(s)
- Grégoire Cullot
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland.
| | - Eric J Aird
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Moritz F Schlapansky
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Charles D Yeh
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Lilly van de Venn
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Iryna Vykhlyantseva
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Susanne Kreutzer
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Dominic Mailänder
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Bohdan Lewków
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Julia Klermund
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Montellese
- CSL Behring Research, Bern, Switzerland
- Swiss Institute for Translational Medicine sitem-insel, Bern, Switzerland
| | - Martina Biserni
- CSL Behring Research, Bern, Switzerland
- Swiss Institute for Translational Medicine sitem-insel, Bern, Switzerland
| | - Florian Aeschimann
- CSL Behring Research, Bern, Switzerland
- Swiss Institute for Translational Medicine sitem-insel, Bern, Switzerland
| | - Cédric Vonarburg
- CSL Behring Research, Bern, Switzerland
- Swiss Institute for Translational Medicine sitem-insel, Bern, Switzerland
| | - Helmuth Gehart
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jacob E Corn
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
30
|
Degtev D, Bravo J, Emmanouilidi A, Zdravković A, Choong OK, Liz Touza J, Selfjord N, Weisheit I, Francescatto M, Akcakaya P, Porritt M, Maresca M, Taylor D, Sienski G. Engineered PsCas9 enables therapeutic genome editing in mouse liver with lipid nanoparticles. Nat Commun 2024; 15:9173. [PMID: 39511150 PMCID: PMC11544209 DOI: 10.1038/s41467-024-53418-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
Clinical implementation of therapeutic genome editing relies on efficient in vivo delivery and the safety of CRISPR-Cas tools. Previously, we identified PsCas9 as a Type II-B family enzyme capable of editing mouse liver genome upon adenoviral delivery without detectable off-targets and reduced chromosomal translocations. Yet, its efficacy remains insufficient with non-viral delivery, a common challenge for many Cas9 orthologues. Here, we sought to redesign PsCas9 for in vivo editing using lipid nanoparticles. We solve the PsCas9 ribonucleoprotein structure with cryo-EM and characterize it biochemically, providing a basis for its rational engineering. Screening over numerous guide RNA and protein variants lead us to develop engineered PsCas9 (ePsCas9) with up to 20-fold increased activity across various targets and preserved safety advantages. We apply the same design principles to boost the activity of FnCas9, an enzyme phylogenetically relevant to PsCas9. Remarkably, a single administration of mRNA encoding ePsCas9 and its guide formulated with lipid nanoparticles results in high levels of editing in the Pcsk9 gene in mouse liver, a clinically relevant target for hypercholesterolemia treatment. Collectively, our findings introduce ePsCas9 as a highly efficient, and precise tool for therapeutic genome editing, in addition to the engineering strategy applicable to other Cas9 orthologues.
Collapse
Affiliation(s)
- Dmitrii Degtev
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden.
| | - Jack Bravo
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Aikaterini Emmanouilidi
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden
| | - Aleksandar Zdravković
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden
| | - Oi Kuan Choong
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden
| | - Julia Liz Touza
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden
| | - Niklas Selfjord
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden
| | - Isabel Weisheit
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden
| | - Margherita Francescatto
- Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden
| | - Pinar Akcakaya
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden
| | - Michelle Porritt
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden
| | - Marcello Maresca
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden.
| | - David Taylor
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA.
- Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX, 78712, USA.
- LIVESTRONG Cancer Institutes, Dell Medical School, Austin, TX, 78712, USA.
| | - Grzegorz Sienski
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
31
|
Hu W, Kumar A, Ahmed SF, Qi S, Ma DKG, Chen H, Singh GJ, Casan JML, Haber M, Voskoboinik I, McKay MR, Trapani JA, Ekert PG, Fareh M. Single-base tiled screen unveils design principles of PspCas13b for potent and off-target-free RNA silencing. Nat Struct Mol Biol 2024; 31:1702-1716. [PMID: 38951623 PMCID: PMC11564092 DOI: 10.1038/s41594-024-01336-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/15/2024] [Indexed: 07/03/2024]
Abstract
The development of precise RNA-editing tools is essential for the advancement of RNA therapeutics. CRISPR (clustered regularly interspaced short palindromic repeats) PspCas13b is a programmable RNA nuclease predicted to offer superior specificity because of its 30-nucleotide spacer sequence. However, its design principles and its on-target, off-target and collateral activities remain poorly characterized. Here, we present single-base tiled screening and computational analyses that identify key design principles for potent and highly selective RNA recognition and cleavage in human cells. We show that the de novo design of spacers containing guanosine bases at precise positions can greatly enhance the catalytic activity of inefficient CRISPR RNAs (crRNAs). These validated design principles (integrated into an online tool, https://cas13target.azurewebsites.net/ ) can predict highly effective crRNAs with ~90% accuracy. Furthermore, the comprehensive spacer-target mutagenesis revealed that PspCas13b can tolerate only up to four mismatches and requires ~26-nucleotide base pairing with the target to activate its nuclease domains, highlighting its superior specificity compared to other RNA or DNA interference tools. On the basis of this targeting resolution, we predict an extremely low probability of PspCas13b having off-target effects on other cellular transcripts. Proteomic analysis validated this prediction and showed that, unlike other Cas13 orthologs, PspCas13b exhibits potent on-target activity and lacks collateral effects.
Collapse
Affiliation(s)
- Wenxin Hu
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Amit Kumar
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Diagnostic Genomics, Monash Health Pathology, Monash Medical Centre, Clayton, Victoria, Australia
| | - Syed Faraz Ahmed
- Department of Electrical and Electronic Engineering, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Shijiao Qi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - David K G Ma
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Honglin Chen
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Gurjeet J Singh
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Joshua M L Casan
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Ilia Voskoboinik
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Matthew R McKay
- Department of Electrical and Electronic Engineering, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Joseph A Trapani
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul G Ekert
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Mohamed Fareh
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
32
|
Cai AJ, Gao K, Zhang F, Jiang YW. Recent advances and current status of gene therapy for epilepsy. World J Pediatr 2024; 20:1115-1137. [PMID: 39395088 DOI: 10.1007/s12519-024-00843-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/05/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Epilepsy is a common neurological disorder with complex pathogenic mechanisms, and refractory epilepsy often lacks effective treatments. Gene therapy is a promising therapeutic option, with various preclinical experiments achieving positive results, some of which have progressed to clinical studies. DATA SOURCES This narrative review was conducted by searching for papers published in PubMed/MEDLINE with the following single and/or combination keywords: epilepsy, children, neurodevelopmental disorders, genetics, gene therapy, vectors, transgenes, receptors, ion channels, micro RNAs (miRNAs), clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas)9 (CRISPR/Cas9), expression regulation, optogenetics, chemical genetics, mitochondrial epilepsy, challenges, ethics, and disease models. RESULTS Currently, gene therapy research in epilepsy primarily focuses on symptoms attenuation mediated by viral vectors such as adeno-associated virus and other types. Advances in gene therapy technologies, such as CRISPR/Cas9, have provided a new direction for epilepsy treatment. However, the clinical application still faces several challenges, including issues related to vectors, models, expression controllability, and ethical considerations. CONCLUSIONS Here, we summarize the relevant research and clinical advances in gene therapy for epilepsy and outline the challenges facing its clinical application. In addition to the shortcomings inherent in gene therapy components, the reconfiguration of excitatory and inhibitory properties in epilepsy treatment is a delicate process. On-demand, cell-autonomous treatments and multidisciplinary collaborations may be crucial in addressing these issues. Understanding gene therapy for epilepsy will help clinicians gain a clearer perception of the research progress and challenges, guiding the design of future clinical protocols and research decisions.
Collapse
Affiliation(s)
- Ao-Jie Cai
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Henan Province, Zhengzhou, 450052, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| | - Fan Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| | - Yu-Wu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China.
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China.
- Children Epilepsy Center, Peking University First Hospital, Beijing, China.
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China.
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
33
|
Fang M, Zhang R, Wang C, Liu Z, Fei M, Tang B, Yang H, Sun D. Engineering probiotic Escherichia coli Nissle 1917 to block transfer of multiple antibiotic resistance genes by exploiting a type I CRISPR-Cas system. Appl Environ Microbiol 2024; 90:e0081124. [PMID: 39254327 PMCID: PMC11497782 DOI: 10.1128/aem.00811-24] [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: 04/27/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
Many multidrug-resistant (MDR) bacteria have evolved through the accumulation of antibiotic resistance genes (ARGs). Although the potential risk of probiotics as reservoirs of ARGs has been recognized, strategies for blocking the transfer of ARGs while using probiotics have rarely been explored. The probiotic Escherichia coli Nissle 1917 (EcN) has long been used for treating intestinal diseases. Here, we demonstrate frequent transfer of ARGs into EcN both in vitro and in vivo, raising concerns about its potential risk of accumulating antibiotic resistance. Given that no CRISPR-Cas system was found in natural EcN, we integrated the type I-E CRISPR-Cas3 system derived from E. coli BW25113 into EcN. The engineered EcN was able to efficiently cleave multiple ARGs [i.e., mcr-1, blaNDM-1, and tet(X)] encoding enzymes for degrading last-resort antibiotics. Through co-incubation of EcN expressing Cas3-Cascade and that expressing Cas9, we showed that the growth of the former strain outcompeted the latter strain, demonstrating a better clinical application prospect of EcN expressing the type I-E CRISPR-Cas3 system. In the intestine of a model animal (i.e., zebrafish), the engineered EcN exhibited immunity against the transfer of CRISPR-targeted ARGs. Our work equips EcN with immunity against the transfer of multiple ARGs by exploiting the exogenous type I-E CRISPR-Cas3 system, thereby reducing the risk of the spread of ARGs while using it as a probiotic chassis for generating living therapeutics. IMPORTANCE To reduce the development of antibiotic resistance, probiotics have been considered as a substitute for antibiotics. However, probiotics themselves are reservoirs of antibiotic resistance genes (ARGs). This study introduces a new strategy for limiting the spread of ARGs by engineering the typical probiotic strain Escherichia coli Nissle 1917 (EcN), which has been used for treating intestinal diseases and developed as living therapeutics. We also demonstrate that the type I CRISPR-Cas system imposes a lower growth burden than the type II CRISPR-Cas system, highlighting its promising clinical application potential. Our work not only provides a new strategy for restricting the transfer of ARGs while using probiotics but also enriches the genetic engineering toolbox of EcN, paving the way for the safe use and development of probiotics as living therapeutics.
Collapse
Affiliation(s)
- Mengdie Fang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ruiting Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Chenyu Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Zhizhi Liu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Mingyue Fei
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Biao Tang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Hua Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Dongchang Sun
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| |
Collapse
|
34
|
Falah G, Sharvit L, Atzmon G. CRISPR-Cas9 mediated d3GHR knockout in HEK293 cells: Revealing the longevity associated isoform stress resilience. Exp Gerontol 2024; 196:112586. [PMID: 39303817 DOI: 10.1016/j.exger.2024.112586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
The Growth Hormone Receptor (GHR) gene encodes a protein that is essential for mediating the biological effects of growth hormone (GH). A series of molecular events are set off when GH binds to its receptor, resulting in a variety of physiological reactions linked to development, growth, and metabolism. Recently a particular genetic variation, within the GHR gene that is labeled as the "d3GHR," which lacks exon 3 was associated with longevity. This specific deletion isoform was connected to changes in the structure of the GHR protein, which may have an impact on the GHR's function. To test in vitro the advantage of the d3 carrier that may link to longevity, we employed the CRISPR/Cas9 technique to produce two isoforms: the homozygotes isoform (d3/d3) and the heterozygotes isoform (d3/fl) using HEK293 cell line. The CRISPR editing effectiveness was >85 %, indicating that we had successfully built the Cas9-gRNA complex that is appropriate for the GHR gene. The viability of the resulted isoform cells was examined under three environmental stressors that mimic some aging processes. In addition, we examined the GHR signaling pathway by selecting potential downstream genes in the GHR signaling cascade. The results show that heterozygotes cells demonstrated higher survival rates under UV radiation compared with the WT cells (87 % compared with 67 % for the WT cells when exposed to 2 min of UV radiation), and in fasting conditions, the d3GHR cells showed a 15 % greater viability than the WT cells. Moreover, the baseline expression levels (without intervention) of the IGF1 and JAK/STAT genes signaling pathways significantly declined in the homozygotes cells compared with the WT (p < 0.05). This noteworthy finding might offer a practical approach to test illness prevention and give the scientific community critical new insights on mechanism associated with lifespan.
Collapse
Affiliation(s)
- Ghadeer Falah
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Lital Sharvit
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Gil Atzmon
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel; Departments of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
35
|
Raimondi F, Siow KM, Wrona D, Fuster-García C, Pastukhov O, Schmitz M, Bargsten K, Kissling L, Swarts DC, Andrieux G, Cathomen T, Modlich U, Jinek M, Siler U, Reichenbach J. Gene editing of NCF1 loci is associated with homologous recombination and chromosomal rearrangements. Commun Biol 2024; 7:1291. [PMID: 39384978 PMCID: PMC11464842 DOI: 10.1038/s42003-024-06959-z] [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: 09/30/2022] [Accepted: 09/24/2024] [Indexed: 10/11/2024] Open
Abstract
CRISPR-based genome editing of pseudogene-associated disorders, such as p47phox-deficient chronic granulomatous disease (p47 CGD), is challenged by chromosomal rearrangements due to presence of multiple targets. We report that interactions between highly homologous sequences that are localized on the same chromosome contribute substantially to post-editing chromosomal rearrangements. We successfully employed editing approaches at the NCF1 gene and its pseudogenes, NCF1B and NCF1C, in a human cell line model of p47 CGD and in patient-derived human hematopoietic stem and progenitor cells. Upon genetic engineering, a droplet digital PCR-based method identified cells with altered copy numbers, spanning megabases from the edited loci. We attributed the high aberration frequency to the interaction between repetitive sequences and their predisposition to recombination events. Our findings emphasize the need for careful evaluation of the target-specific genomic context, such as the presence of homologous regions, whose instability can constitute a risk factor for chromosomal rearrangements upon genome editing.
Collapse
Affiliation(s)
- Federica Raimondi
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich (Schlieren Campus), Schlieren, Switzerland
| | - Kah Mun Siow
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich (Schlieren Campus), Schlieren, Switzerland
| | - Dominik Wrona
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich (Schlieren Campus), Schlieren, Switzerland
| | - Carla Fuster-García
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Freiburg, Germany
| | - Oleksandr Pastukhov
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich (Schlieren Campus), Schlieren, Switzerland
| | - Michael Schmitz
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Katja Bargsten
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Lucas Kissling
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Daan C Swarts
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Geoffroy Andrieux
- Institute for Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ute Modlich
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich (Schlieren Campus), Schlieren, Switzerland
| | - Martin Jinek
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Ulrich Siler
- School of Life Sciences, Institute for Pharma Technology, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland
| | - Janine Reichenbach
- Division of Gene and Cell Therapy, Institute for Regenerative Medicine, University of Zurich (Schlieren Campus), Schlieren, Switzerland.
- Department of Somatic Gene Therapy, University Children's Hospital Zurich, Zurich, Switzerland.
- The Competence Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
36
|
Kumar A, Combe E, Mougené L, Zoulim F, Testoni B. Applications of CRISPR/Cas as a Toolbox for Hepatitis B Virus Detection and Therapeutics. Viruses 2024; 16:1565. [PMID: 39459899 PMCID: PMC11512240 DOI: 10.3390/v16101565] [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: 09/08/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Hepatitis B virus (HBV) infection remains a significant global health challenge, leading to chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma (HCC). Covalently closed circular DNA (cccDNA) and integrated HBV DNA are pivotal in maintaining viral persistence. Recent advances in CRISPR/Cas technology offer innovative strategies to inhibit HBV by directly targeting both cccDNA and integrated HBV DNA or indirectly by degrading HBV RNAs or targeting host proteins. This review provides a comprehensive overview of the latest advancements in using CRISPR/Cas to inhibit HBV, with a special highlight on newer non-double-strand (non-DSB) break approaches. Beyond the canonical use of CRISPR/Cas for target inhibition, we discuss additional applications, including HBV diagnosis and developing models to understand cccDNA biology, highlighting the diverse use of this technology in the HBV field.
Collapse
Affiliation(s)
- Anuj Kumar
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR 5286, 69008 Lyon, France; (A.K.); (E.C.); (L.M.); (F.Z.)
- The Lyon Hepatology Institute EVEREST, 69003 Lyon, France
| | - Emmanuel Combe
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR 5286, 69008 Lyon, France; (A.K.); (E.C.); (L.M.); (F.Z.)
- The Lyon Hepatology Institute EVEREST, 69003 Lyon, France
| | - Léa Mougené
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR 5286, 69008 Lyon, France; (A.K.); (E.C.); (L.M.); (F.Z.)
- The Lyon Hepatology Institute EVEREST, 69003 Lyon, France
| | - Fabien Zoulim
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR 5286, 69008 Lyon, France; (A.K.); (E.C.); (L.M.); (F.Z.)
- The Lyon Hepatology Institute EVEREST, 69003 Lyon, France
- Hepatology Department, Hospices Civils de Lyon (HCL), Croix-Rousse Hospital, 69004 Lyon, France
- University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France
| | - Barbara Testoni
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR 5286, 69008 Lyon, France; (A.K.); (E.C.); (L.M.); (F.Z.)
- The Lyon Hepatology Institute EVEREST, 69003 Lyon, France
| |
Collapse
|
37
|
Liu Z, Zhang W, Wang H, Shangguan P, Pan T, Yang Y, Zhang Y, Mao X, Liu Y, Zhang Q. Engineered CRISPR RNA improves the RNA cleavage efficiency of hfCas13X. FEBS Lett 2024; 598:2438-2449. [PMID: 39327223 DOI: 10.1002/1873-3468.15025] [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: 04/12/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024]
Abstract
As the most compact variant in the Cas13 family, CRISPR-Cas13X holds considerable promise for gene therapy applications. The development of high-fidelity Cas13X (hfCas13X) mutants has enhanced the safety profile for in vivo applications. However, a notable reduction in on-target cleavage efficiency accompanies the diminished collateral cleavage activity in hfCas13X. In this study, we obtained two engineered crRNA mutants that notably enhance the on-target cleavage efficiency of hfCas13X. Furthermore, we have identified a novel crRNA structure that consistently augments the on-target cleavage efficiency of hfCas13X across various cellular environments, without significant enhancement of its collateral activity. These findings collectively enrich the gene-editing toolkit, presenting a more effective hfCas13X system for future research and application.
Collapse
Affiliation(s)
- Zehui Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, China
| | - Wenxia Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, China
| | - Haili Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, China
| | - Pingping Shangguan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, China
| | - Tong Pan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, China
| | - Yimu Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, China
| | - Yi Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, China
| | - Xi Mao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, China
| | - Yingle Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, China
| | - Qi Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, China
| |
Collapse
|
38
|
Chen X, Moran Torres JP, Tedjai SVK, Lugones LG, Wösten HAB. Functional analysis of FlbA-regulated transcription factor genes in Aspergillus niger using a multiplexed CRISPRoff system. Int J Biol Macromol 2024; 277:134326. [PMID: 39089555 DOI: 10.1016/j.ijbiomac.2024.134326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/31/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
FlbA of Aspergillus niger (indirectly) regulates 36 transcription factor (TF) genes. As a result, it promotes sporulation and represses vegetative growth, protein secretion and lysis. In this study, the functions of part of the FlbA-regulated TF genes were studied by using CRISPRoff. This system was recently introduced as an epigenetic tool for modulating gene expression in A. niger. A plasmid encompassing an optimized CRISPRoff system as well as a library of sgRNA genes that target the promoters of the 36 FlbA-regulated TF genes was introduced in A. niger. Out of 24 transformants that exhibited a sporulation phenotype, 12 and 18 strains also showed a biomass and secretion phenotype, respectively. The transforming sgRNAs, and thus the genes responsible for the phenotypes, were identified from five of the transformants. The results show that the genes dofA, dofB, dofC, dofD, and socA are involved in sporulation and extracellular enzyme activity, while dofA and socA also play roles in biomass formation. Overall, this study shows that the multiplexed CRISPRoff system can be effectively used for functional analysis of genes in a fungus.
Collapse
Affiliation(s)
- Xiaoyi Chen
- Microbiology, Department of Biology, Utrecht University, Padualaan 8, 3584, CH, Utrecht, the Netherlands.
| | - Juan P Moran Torres
- Microbiology, Department of Biology, Utrecht University, Padualaan 8, 3584, CH, Utrecht, the Netherlands.
| | - S Vyanjan K Tedjai
- Microbiology, Department of Biology, Utrecht University, Padualaan 8, 3584, CH, Utrecht, the Netherlands.
| | - Luis G Lugones
- Microbiology, Department of Biology, Utrecht University, Padualaan 8, 3584, CH, Utrecht, the Netherlands.
| | - Han A B Wösten
- Microbiology, Department of Biology, Utrecht University, Padualaan 8, 3584, CH, Utrecht, the Netherlands.
| |
Collapse
|
39
|
Regan SB, Medhi D, White TB, Jiang YZ, Jia S, Deng Q, Jasin M. Megabase-scale loss of heterozygosity provoked by CRISPR-Cas9 DNA double-strand breaks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615517. [PMID: 39386534 PMCID: PMC11463394 DOI: 10.1101/2024.09.27.615517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Harnessing DNA double-strand breaks (DSBs) is a powerful approach for gene editing, but it may provoke loss of heterozygosity (LOH), which predisposes to tumorigenesis. To interrogate this risk, we developed a two- color flow cytometry-based system (Flo-LOH), detecting LOH in ∼5% of cells following a DSB. After this initial increase, cells with LOH decrease due to a competitive disadvantage with parental cells, but if isolated, they stably propagate. Segmental loss from terminal deletions with de novo telomere addition and nonreciprocal translocations is observed as well as whole chromosome loss, especially following a centromeric DSB. LOH spans megabases distal from the DSB, but also frequently tens of megabases centromere-proximal. Inhibition of microhomology-mediated end joining massively increases LOH, which is synergistically increased with concomitant inhibition of canonical nonhomologous end joining. The capacity for large-scale LOH must therefore be considered when using DSB-based gene editing, especially in conjunction with end joining inhibition.
Collapse
|
40
|
Pattali RK, Ornelas IJ, Nguyen CD, Xu D, Divekar NS, Nuñez JK. CRISPRoff epigenetic editing for programmable gene silencing in human cells without DNA breaks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612111. [PMID: 39345634 PMCID: PMC11429707 DOI: 10.1101/2024.09.09.612111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The advent of CRISPR-based technologies has enabled the rapid advancement of programmable gene manipulation in cells, tissues, and whole organisms. An emerging platform for targeted gene perturbation is epigenetic editing, the direct editing of chemical modifications on DNA and histones that ultimately results in repression or activation of the targeted gene. In contrast to CRISPR nucleases, epigenetic editors modulate gene expression without inducing DNA breaks or altering the genomic sequence of host cells. Recently, we developed the CRISPRoff epigenetic editing technology that simultaneously establishes DNA methylation and repressive histone modifications at targeted gene promoters. Transient expression of CRISPRoff and the accompanying single guide RNAs in mammalian cells results in transcriptional repression of targeted genes that is memorized heritably by cells through cell division and differentiation. Here, we describe our protocol for the delivery of CRISPRoff through plasmid DNA transfection, as well as the delivery of CRISPRoff mRNA, into transformed human cell lines and primary immune cells. We also provide guidance on evaluating target gene silencing and highlight key considerations when utilizing CRISPRoff for gene perturbations. Our protocols are broadly applicable to other CRISPR-based epigenetic editing technologies, as programmable genome manipulation tools continue to evolve rapidly.
Collapse
Affiliation(s)
- Rithu K. Pattali
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Izaiah J. Ornelas
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Carolyn D. Nguyen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Da Xu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Nikita S. Divekar
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - James K. Nuñez
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, California, USA
| |
Collapse
|
41
|
Livraghi L, Hanly JJ, Evans E, Wright CJ, Loh LS, Mazo-Vargas A, Kamrava K, Carter A, van der Heijden ESM, Reed RD, Papa R, Jiggins CD, Martin A. A long noncoding RNA at the cortex locus controls adaptive coloration in butterflies. Proc Natl Acad Sci U S A 2024; 121:e2403326121. [PMID: 39213180 PMCID: PMC11388343 DOI: 10.1073/pnas.2403326121] [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: 02/16/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Evolutionary variation in the wing pigmentation of butterflies and moths offers striking examples of adaptation by crypsis and mimicry. The cortex locus has been independently mapped as the locus controlling color polymorphisms in 15 lepidopteran species, suggesting that it acts as a genomic hotspot for the diversification of wing patterns, but functional validation through protein-coding knockouts has proven difficult to obtain. Our study unveils the role of a long noncoding RNA (lncRNA) which we name ivory, transcribed from the cortex locus, in modulating color patterning in butterflies. Strikingly, ivory expression prefigures most melanic patterns during pupal development, suggesting an early developmental role in specifying scale identity. To test this, we generated CRISPR mosaic knock-outs in five nymphalid butterfly species and show that ivory mutagenesis yields transformations of dark pigmented scales into white or light-colored scales. Genotyping of Vanessa cardui germline mutants associates these phenotypes to small on-target deletions at the conserved first exon of ivory. In contrast, cortex germline mutant butterflies with confirmed null alleles lack any wing phenotype and exclude a color patterning role for this adjacent gene. Overall, these results show that a lncRNA gene acts as a master switch of color pattern specification and played key roles in the adaptive diversification of wing patterns in butterflies.
Collapse
Affiliation(s)
- Luca Livraghi
- Department of Biological Sciences, The George Washington University, Washington, DC20052
- Department of Zoology, University of Cambridge, CambridgeCB2 3EJ, United Kingdom
| | - Joseph J. Hanly
- Department of Biological Sciences, The George Washington University, Washington, DC20052
- Department of Biology, Duke University, Durham, NC27708
- Smithsonian Tropical Research Institute, Gamboa, Panama
| | - Elizabeth Evans
- Department of Biology, University of Puerto Rico at Río Piedras, San Juan00925, Puerto Rico
| | - Charlotte J. Wright
- Department of Zoology, University of Cambridge, CambridgeCB2 3EJ, United Kingdom
- Tree of Life, Wellcome Sanger Institute, CambridgeCB10 1RQ, United Kingdom
| | - Ling S. Loh
- Department of Biological Sciences, The George Washington University, Washington, DC20052
| | - Anyi Mazo-Vargas
- Department of Biological Sciences, The George Washington University, Washington, DC20052
- Department of Biology, Duke University, Durham, NC27708
| | - Kiana Kamrava
- Department of Biological Sciences, The George Washington University, Washington, DC20052
| | - Alexander Carter
- Department of Biological Sciences, The George Washington University, Washington, DC20052
| | - Eva S. M. van der Heijden
- Department of Zoology, University of Cambridge, CambridgeCB2 3EJ, United Kingdom
- Tree of Life, Wellcome Sanger Institute, CambridgeCB10 1RQ, United Kingdom
| | - Robert D. Reed
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY14853
| | - Riccardo Papa
- Department of Biology, University of Puerto Rico at Río Piedras, San Juan00925, Puerto Rico
- Comprehensive Cancer Center, University of Puerto Rico, San Juan00925, Puerto Rico
- Molecular Sciences and Research Center, University of Puerto Rico, San Juan00926, Puerto Rico
- Dipartimento di Scienze Chimiche della Vita e della Sostenibilità Ambientale, Università di Parma, Parma43124, Italy
| | - Chris D. Jiggins
- Department of Zoology, University of Cambridge, CambridgeCB2 3EJ, United Kingdom
| | - Arnaud Martin
- Department of Biological Sciences, The George Washington University, Washington, DC20052
| |
Collapse
|
42
|
Hosseini SY, Mallick R, Mäkinen P, Ylä-Herttuala S. Insights into Prime Editing Technology: A Deep Dive into Fundamentals, Potentials, and Challenges. Hum Gene Ther 2024; 35:649-668. [PMID: 38832869 DOI: 10.1089/hum.2024.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024] Open
Abstract
As the most versatile and precise gene editing technology, prime editing (PE) can establish a durable cure for most human genetic disorders. Several generations of PE have been developed based on an editor machine or prime editing guide RNA (pegRNA) to achieve any kind of genetic correction. However, due to the early stage of development, PE complex elements need to be optimized for more efficient editing. Smart optimization of editor proteins as well as pegRNA has been contemplated by many researchers, but the universal PE machine's current shortcomings remain to be solved. The modification of PE elements, fine-tuning of the host genes, manipulation of epigenetics, and blockage of immune responses could be used to reach more efficient PE. Moreover, the host factors involved in the PE process, such as repair and innate immune system genes, have not been determined, and PE cell context dependency is still poorly understood. Regarding the large size of the PE elements, delivery is a significant challenge and the development of a universal viral or nonviral platform is still far from complete. PE versions with shortened variants of reverse transcriptase are still too large to fit in common viral vectors. Overall, PE faces challenges in optimization for efficiency, high context dependency during the cell cycling, and delivery due to the large size of elements. In addition, immune responses, unpredictability of outcomes, and off-target effects further limit its application, making it essential to address these issues for broader use in nonpersonalized gene editing. Besides, due to the limited number of suitable animal models and computational modeling, the prediction of the PE process remains challenging. In this review, the fundamentals of PE, including generations, potential, optimization, delivery, in vivo barriers, and the future landscape of the technology are discussed.
Collapse
Affiliation(s)
- Seyed Younes Hosseini
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Bacteriology and Virology Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Petri Mäkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
43
|
Bailey SM, Kunkel SR, Bedford JS, Cornforth MN. The Central Role of Cytogenetics in Radiation Biology. Radiat Res 2024; 202:227-259. [PMID: 38981612 DOI: 10.1667/rade-24-00038.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/23/2024] [Indexed: 07/11/2024]
Abstract
Radiation cytogenetics has a rich history seldom appreciated by those outside the field. Early radiobiology was dominated by physics and biophysical concepts that borrowed heavily from the study of radiation-induced chromosome aberrations. From such studies, quantitative relationships between biological effect and changes in absorbed dose, dose rate and ionization density were codified into key concepts of radiobiological theory that have persisted for nearly a century. This review aims to provide a historical perspective of some of these concepts, including evidence supporting the contention that chromosome aberrations underlie development of many, if not most, of the biological effects of concern for humans exposed to ionizing radiations including cancer induction, on the one hand, and tumor eradication on the other. The significance of discoveries originating from these studies has widened and extended far beyond their original scope. Chromosome structural rearrangements viewed in mitotic cells were first attributed to the production of breaks by the radiations during interphase, followed by the rejoining or mis-rejoining among ends of other nearby breaks. These relatively modest beginnings eventually led to the discovery and characterization of DNA repair of double-strand breaks by non-homologous end joining, whose importance to various biological processes is now widely appreciated. Two examples, among many, are V(D)J recombination and speciation. Rapid technological advancements in cytogenetics, the burgeoning fields of molecular radiobiology and third-generation sequencing served as a point of confluence between the old and new. As a result, the emergent field of "cytogenomics" now becomes uniquely positioned for the purpose of more fully understanding mechanisms underlying the biological effects of ionizing radiation exposure.
Collapse
Affiliation(s)
- Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Stephen R Kunkel
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, Texas
| | - Joel S Bedford
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Michael N Cornforth
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
44
|
Zhang Y, Xia H, Peng W, Liu L, Liu L, Yang P. Application of Repetitive Sequences in Fish Cell Depletion as a Target for the CRISPR/Cas9 System. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:639-648. [PMID: 38833200 DOI: 10.1007/s10126-024-10328-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/27/2024] [Indexed: 06/06/2024]
Abstract
Specific cell depletion is a common means to study the physiological function of cell lineages and tissue regeneration. However, 100% depletion is difficult to achieve with existing cell depletion strategies. With the increasing maturity of CRISPR/Cas9 technology, it is increasingly used for the depletion of various cells. However, even with this technology, it is difficult to complete the depletion of specific gene knockout cells. For this reason, cell depletion with the use of repetitive sequences as the target of CRISPR/Cas9 was explored using zebrafish. All cells were used as the target cells for the first set of experiments. The results showed that injection of a mixture of DANA-gRNA and Cas9 mRNA into zygotes resulted in substantial cell apoptosis. Cells are almost invisible in the embryonic animal pole during the dome stage. The activities of the caspase-3 and caspase-9 proteins and the mRNA level of the P53 gene were significantly increased. Then, primordial germ cells (PGCs) in embryos were used as the target cells in subsequent experiments. To specifically knock out PGCs, we injected the mix of DANA-gRNA, pkop: Cas9 plasmid (the kop promotor allows Cas9 expression only in PGCs), and eGFP-nos3'UTR mRNA into zebrafish fertilized eggs. The results revealed that the activity of the caspase-3 protein was significantly increased, and the mRNA levels of P53, ku70, and ku80 were significantly upregulated, while the number of PGCs decreased gradually. Few PGCs labeled with GFP could be seen 20 h post-fertilization (hpf), and no PGCs could be seen at the germinal ridge 24 hpf. Therefore, the combination of CRISPR/Cas9 technology and repetitive sequences can achieve efficient cell depletion regardless of whether there is generalized expression or expression in specific cells. These results indicate that it is feasible to eliminate cells by using repeat sequences as CRISPR/Cas9 system target sites.
Collapse
Affiliation(s)
- Yunsheng Zhang
- Hunan Provincial Key Laboratory for Health Aquaculture and Product Processing in Dongting Lake Area, Hunan Engineering Research Center of Aquatic Organism Resources and Environmental Ecology, Zoology Key Laboratory of Hunan Higher Education, Changde Research Center for Agricultural Biomacromolecule, Innovation Team of Microbial Technology, College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, Hunan, China.
| | - Hu Xia
- Hunan Provincial Key Laboratory for Health Aquaculture and Product Processing in Dongting Lake Area, Hunan Engineering Research Center of Aquatic Organism Resources and Environmental Ecology, Zoology Key Laboratory of Hunan Higher Education, Changde Research Center for Agricultural Biomacromolecule, Innovation Team of Microbial Technology, College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, Hunan, China
| | - Wei Peng
- Hunan Provincial Key Laboratory for Health Aquaculture and Product Processing in Dongting Lake Area, Hunan Engineering Research Center of Aquatic Organism Resources and Environmental Ecology, Zoology Key Laboratory of Hunan Higher Education, Changde Research Center for Agricultural Biomacromolecule, Innovation Team of Microbial Technology, College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, Hunan, China
| | - Lanhai Liu
- Changde Fishery Administration Station, Changde, China
| | - Liangguo Liu
- Hunan Provincial Key Laboratory for Health Aquaculture and Product Processing in Dongting Lake Area, Hunan Engineering Research Center of Aquatic Organism Resources and Environmental Ecology, Zoology Key Laboratory of Hunan Higher Education, Changde Research Center for Agricultural Biomacromolecule, Innovation Team of Microbial Technology, College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, Hunan, China
| | - Pinhong Yang
- Hunan Provincial Key Laboratory for Health Aquaculture and Product Processing in Dongting Lake Area, Hunan Engineering Research Center of Aquatic Organism Resources and Environmental Ecology, Zoology Key Laboratory of Hunan Higher Education, Changde Research Center for Agricultural Biomacromolecule, Innovation Team of Microbial Technology, College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, Hunan, China
| |
Collapse
|
45
|
Ferreira da Silva J, Tou CJ, King EM, Eller ML, Rufino-Ramos D, Ma L, Cromwell CR, Metovic J, Benning FMC, Chao LH, Eichler FS, Kleinstiver BP. Click editing enables programmable genome writing using DNA polymerases and HUH endonucleases. Nat Biotechnol 2024:10.1038/s41587-024-02324-x. [PMID: 39039307 PMCID: PMC11751136 DOI: 10.1038/s41587-024-02324-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 06/24/2024] [Indexed: 07/24/2024]
Abstract
Genome editing technologies based on DNA-dependent polymerases (DDPs) could offer several benefits compared with other types of editors to install diverse edits. Here, we develop click editing, a genome writing platform that couples the advantageous properties of DDPs with RNA-programmable nickases to permit the installation of a range of edits, including substitutions, insertions and deletions. Click editors (CEs) leverage the 'click'-like bioconjugation ability of HUH endonucleases with single-stranded DNA substrates to covalently tether 'click DNA' (clkDNA) templates encoding user-specifiable edits at targeted genomic loci. Through iterative optimization of the modular components of CEs and their clkDNAs, we demonstrate the ability to install precise genome edits with minimal indels in diverse immortalized human cell types and primary fibroblasts with precise editing efficiencies of up to ~30%. Editing efficiency can be improved by rapidly screening clkDNA oligonucleotides with various modifications, including repair-evading substitutions. Click editing is a precise and versatile genome editing approach for diverse biological applications.
Collapse
Affiliation(s)
- Joana Ferreira da Silva
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Connor J Tou
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Biological Engineering Program, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emily M King
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Biological and Biomedical Sciences Program, Harvard University, Boston, MA, USA
| | - Madeline L Eller
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - David Rufino-Ramos
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Linyuan Ma
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Christopher R Cromwell
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Jasna Metovic
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Friederike M C Benning
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Luke H Chao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Florian S Eichler
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Benjamin P Kleinstiver
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Pathology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Ge W, Gou S, Zhao X, Jin Q, Zhuang Z, Zhao Y, Liang Y, Ouyang Z, Liu X, Chen F, Shi H, Yan H, Wu H, Lai L, Wang K. In vivo evaluation of guide-free Cas9-induced safety risks in a pig model. Signal Transduct Target Ther 2024; 9:184. [PMID: 39025833 PMCID: PMC11258294 DOI: 10.1038/s41392-024-01905-1] [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: 09/14/2023] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/20/2024] Open
Abstract
The CRISPR/Cas9 system has shown great potential for treating human genetic diseases through gene therapy. However, there are concerns about the safety of this system, specifically related to the use of guide-free Cas9. Previous studies have shown that guide-free Cas9 can induce genomic instability in vitro. However, the in vivo safety risks associated with guide-free Cas9 have not been evaluated, which is necessary for the development of gene therapy in clinical settings. In this study, we used doxycycline-inducible Cas9-expressing pigs to evaluate the safety risks of guide-free Cas9 in vivo. Our findings demonstrated that expression of guide-free Cas9 could induce genomic damages and transcriptome changes in vivo. The severity of the genomic damages and transcriptome changes were correlate with the expression levels of Cas9 protein. Moreover, prolonged expression of Cas9 in pigs led to abnormal phenotypes, including a significant decrease in body weight, which may be attributable to genomic damage-induced nutritional absorption and metabolic dysfunction. Furthermore, we observed an increase in whole-genome and tumor driver gene mutations in pigs with long-term Cas9 expression, raising the risk of tumor occurrence. Our in vivo evaluation of guide-free Cas9 in pigs highlights the necessity of considering and monitoring the detrimental effects of Cas9 alone as genome editing via the CRISPR/Cas9 system is implemented in clinical gene therapy. This research emphasizes the importance of further study and implementation of safety measures to ensure the successful and safe application of the CRISPR/Cas9 system in clinical practice.
Collapse
Affiliation(s)
- Weikai Ge
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China
- Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen, 529020, China
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Shixue Gou
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Xiaozhu Zhao
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Qin Jin
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, 510530, China
| | - Zhenpeng Zhuang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China
| | - Yu Zhao
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China
- Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen, 529020, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, 510530, China
| | - Yanhui Liang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, 510530, China
| | - Zhen Ouyang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China
- Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen, 529020, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, 510530, China
| | - Xiaoyi Liu
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Fangbing Chen
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China
- Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen, 529020, China
| | - Hui Shi
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China
| | - Haizhao Yan
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China
- Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen, 529020, China
| | - Han Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China.
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, 510530, China.
| | - Liangxue Lai
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China.
- Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen, 529020, China.
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, 510530, China.
| | - Kepin Wang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
- Sanya institute of Swine resource, Hainan Provincial Research Center of Laboratory Animals, Sanya, 572000, China.
- Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen, 529020, China.
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, 510530, China.
| |
Collapse
|
47
|
Vinceti A, Iannuzzi RM, Boyle I, Trastulla L, Campbell CD, Vazquez F, Dempster JM, Iorio F. A benchmark of computational methods for correcting biases of established and unknown origin in CRISPR-Cas9 screening data. Genome Biol 2024; 25:192. [PMID: 39030569 PMCID: PMC11264729 DOI: 10.1186/s13059-024-03336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 07/10/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND CRISPR-Cas9 dropout screens are formidable tools for investigating biology with unprecedented precision and scale. However, biases in data lead to potential confounding effects on interpretation and compromise overall quality. The activity of Cas9 is influenced by structural features of the target site, including copy number amplifications (CN bias). More worryingly, proximal targeted loci tend to generate similar gene-independent responses to CRISPR-Cas9 targeting (proximity bias), possibly due to Cas9-induced whole chromosome-arm truncations or other genomic structural features and different chromatin accessibility levels. RESULTS We benchmarked eight computational methods, rigorously evaluating their ability to reduce both CN and proximity bias in the two largest publicly available cell-line-based CRISPR-Cas9 screens to date. We also evaluated the capability of each method to preserve data quality and heterogeneity by assessing the extent to which the processed data allows accurate detection of true positive essential genes, established oncogenetic addictions, and known/novel biomarkers of cancer dependency. Our analysis sheds light on the ability of each method to correct biases under different scenarios. AC-Chronos outperforms other methods in correcting both CN and proximity biases when jointly processing multiple screens of models with available CN information, whereas CRISPRcleanR is the top performing method for individual screens or when CN information is not available. In addition, Chronos and AC-Chronos yield a final dataset better able to recapitulate known sets of essential and non-essential genes. CONCLUSIONS Overall, our investigation provides guidance for the selection of the most appropriate bias-correction method, based on its strengths, weaknesses and experimental settings.
Collapse
Affiliation(s)
| | | | | | - Lucia Trastulla
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | | | | | | | - Francesco Iorio
- Computational Biology Research Centre, Human Technopole, Milan, Italy.
| |
Collapse
|
48
|
Loret C, Pauset A, Faye PA, Prouzet-Mauleon V, Pyromali I, Nizou A, Miressi F, Sturtz F, Favreau F, Turcq B, Lia AS. CRISPR Base Editing to Create Potential Charcot-Marie-Tooth Disease Models with High Editing Efficiency: Human Induced Pluripotent Stem Cell Harboring SH3TC2 Variants. Biomedicines 2024; 12:1550. [PMID: 39062123 PMCID: PMC11274897 DOI: 10.3390/biomedicines12071550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) represent a powerful tool to investigate neuropathological disorders in which the cells of interest are inaccessible, such as in the Charcot-Marie-Tooth disease (CMT), the most common inherited peripheral neuropathy. Developing appropriate cellular models becomes crucial in order to both study the disease's pathophysiology and test new therapeutic approaches. The generation of hiPS cellular models for disorders caused by a single nucleotide variation has been significantly improved following the development of CRISPR-based editing tools. In this study, we efficiently and quickly generated, by CRISPR editing, the two first hiPSCs cellular models carrying alterations involved in CMT4C, also called AR-CMTde-SH3TC2. This subtype of CMT is associated with alterations in the SH3TC2 gene and represents the most prevalent form of autosomal recessive demyelinating CMT. We aimed to develop models for two different SH3TC2 nonsense variants, c.211C>T, p.Gln71* and the most common AR-CMTde-SH3TC2 alteration, c.2860C>T, p.Arg954*. First, in order to determine the best CRISPR strategy to adopt on hiPSCs, we first tested a variety of sgRNAs combined with a selection of recent base editors using the conveniently cultivable and transfectable HEK-293T cell line. The chosen CRISPR base-editing strategy was then applied to hiPSCs derived from healthy individuals to generate isogenic CMT disease models with up to 93% editing efficiency. For point mutation generation, we first recommend to test your strategies on alternative cell line such as HEK-293T before hiPSCs to evaluate a variety of sgRNA-BE combinations, thus boosting the chance of achieving edited cellular clones with the hard-to-culture and to transfect hiPSCs.
Collapse
Affiliation(s)
- Camille Loret
- University of Limoges, NeurIT UR 20218, GEIST Institute, F-87000 Limoges, France; (P.-A.F.); (I.P.); (A.N.); (F.M.); (F.S.); (F.F.); (A.-S.L.)
| | - Amandine Pauset
- University of Bordeaux, CRISP'edit, TBMCore UAR CNRS 3427, US Inserm 005, F-33000 Bordeaux, France (V.P.-M.); (B.T.)
- University of Bordeaux, Modeling Transformation and Resistance in Leukemia, BRIC Inserm U1312, F-33000 Bordeaux, France
| | - Pierre-Antoine Faye
- University of Limoges, NeurIT UR 20218, GEIST Institute, F-87000 Limoges, France; (P.-A.F.); (I.P.); (A.N.); (F.M.); (F.S.); (F.F.); (A.-S.L.)
- CHU Limoges, Department of Biochemistry and Molecular Genetics, F-87000 Limoges, France
| | - Valérie Prouzet-Mauleon
- University of Bordeaux, CRISP'edit, TBMCore UAR CNRS 3427, US Inserm 005, F-33000 Bordeaux, France (V.P.-M.); (B.T.)
- University of Bordeaux, Modeling Transformation and Resistance in Leukemia, BRIC Inserm U1312, F-33000 Bordeaux, France
| | - Ioanna Pyromali
- University of Limoges, NeurIT UR 20218, GEIST Institute, F-87000 Limoges, France; (P.-A.F.); (I.P.); (A.N.); (F.M.); (F.S.); (F.F.); (A.-S.L.)
| | - Angélique Nizou
- University of Limoges, NeurIT UR 20218, GEIST Institute, F-87000 Limoges, France; (P.-A.F.); (I.P.); (A.N.); (F.M.); (F.S.); (F.F.); (A.-S.L.)
| | - Federica Miressi
- University of Limoges, NeurIT UR 20218, GEIST Institute, F-87000 Limoges, France; (P.-A.F.); (I.P.); (A.N.); (F.M.); (F.S.); (F.F.); (A.-S.L.)
| | - Franck Sturtz
- University of Limoges, NeurIT UR 20218, GEIST Institute, F-87000 Limoges, France; (P.-A.F.); (I.P.); (A.N.); (F.M.); (F.S.); (F.F.); (A.-S.L.)
- CHU Limoges, Department of Biochemistry and Molecular Genetics, F-87000 Limoges, France
| | - Frédéric Favreau
- University of Limoges, NeurIT UR 20218, GEIST Institute, F-87000 Limoges, France; (P.-A.F.); (I.P.); (A.N.); (F.M.); (F.S.); (F.F.); (A.-S.L.)
- CHU Limoges, Department of Biochemistry and Molecular Genetics, F-87000 Limoges, France
| | - Béatrice Turcq
- University of Bordeaux, CRISP'edit, TBMCore UAR CNRS 3427, US Inserm 005, F-33000 Bordeaux, France (V.P.-M.); (B.T.)
- University of Bordeaux, Modeling Transformation and Resistance in Leukemia, BRIC Inserm U1312, F-33000 Bordeaux, France
| | - Anne-Sophie Lia
- University of Limoges, NeurIT UR 20218, GEIST Institute, F-87000 Limoges, France; (P.-A.F.); (I.P.); (A.N.); (F.M.); (F.S.); (F.F.); (A.-S.L.)
- CHU Limoges, Department of Biochemistry and Molecular Genetics, F-87000 Limoges, France
- CHU Limoges, Department of Bioinformatics, F-87000 Limoges, France
| |
Collapse
|
49
|
Neumayer G, Torkelson JL, Li S, McCarthy K, Zhen HH, Vangipuram M, Mader MM, Gebeyehu G, Jaouni TM, Jacków-Malinowska J, Rami A, Hansen C, Guo Z, Gaddam S, Tate KM, Pappalardo A, Li L, Chow GM, Roy KR, Nguyen TM, Tanabe K, McGrath PS, Cramer A, Bruckner A, Bilousova G, Roop D, Tang JY, Christiano A, Steinmetz LM, Wernig M, Oro AE. A scalable and cGMP-compatible autologous organotypic cell therapy for Dystrophic Epidermolysis Bullosa. Nat Commun 2024; 15:5834. [PMID: 38992003 PMCID: PMC11239819 DOI: 10.1038/s41467-024-49400-z] [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: 03/28/2023] [Accepted: 05/25/2024] [Indexed: 07/13/2024] Open
Abstract
We present Dystrophic Epidermolysis Bullosa Cell Therapy (DEBCT), a scalable platform producing autologous organotypic iPS cell-derived induced skin composite (iSC) grafts for definitive treatment. Clinical-grade manufacturing integrates CRISPR-mediated genetic correction with reprogramming into one step, accelerating derivation of COL7A1-edited iPS cells from patients. Differentiation into epidermal, dermal and melanocyte progenitors is followed by CD49f-enrichment, minimizing maturation heterogeneity. Mouse xenografting of iSCs from four patients with different mutations demonstrates disease modifying activity at 1 month. Next-generation sequencing, biodistribution and tumorigenicity assays establish a favorable safety profile at 1-9 months. Single cell transcriptomics reveals that iSCs are composed of the major skin cell lineages and include prominent holoclone stem cell-like signatures of keratinocytes, and the recently described Gibbin-dependent signature of fibroblasts. The latter correlates with enhanced graftability of iSCs. In conclusion, DEBCT overcomes manufacturing and safety roadblocks and establishes a reproducible, safe, and cGMP-compatible therapeutic approach to heal lesions of DEB patients.
Collapse
Affiliation(s)
- Gernot Neumayer
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Jessica L Torkelson
- Department of Dermatology-Program in Epithelial Biology, Stanford University, School of Medicine, Stanford, CA, USA
- Center for Definitive and Curative Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Shengdi Li
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Kelly McCarthy
- Department of Dermatology-Program in Epithelial Biology, Stanford University, School of Medicine, Stanford, CA, USA
- Center for Definitive and Curative Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Hanson H Zhen
- Department of Dermatology-Program in Epithelial Biology, Stanford University, School of Medicine, Stanford, CA, USA
- Center for Definitive and Curative Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Madhuri Vangipuram
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Marius M Mader
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Gulilat Gebeyehu
- Thermo Fisher Scientific, Life Sciences Solutions Group, Cell Biology, Research and Development, Frederick, MD, USA
| | - Taysir M Jaouni
- Thermo Fisher Scientific, Life Sciences Solutions Group, Cell Biology, Research and Development, Frederick, MD, USA
| | - Joanna Jacków-Malinowska
- Department of Dermatology, Columbia University, New York, NY, USA
- St. John's Institute of Dermatology, King's College London, London, UK
| | - Avina Rami
- Department of Dermatology, Columbia University, New York, NY, USA
| | - Corey Hansen
- Department of Dermatology, Columbia University, New York, NY, USA
| | - Zongyou Guo
- Department of Dermatology, Columbia University, New York, NY, USA
| | - Sadhana Gaddam
- Department of Dermatology-Program in Epithelial Biology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Keri M Tate
- Center for Definitive and Curative Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | | | - Lingjie Li
- Department of Dermatology-Program in Epithelial Biology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Grace M Chow
- Department of Dermatology-Program in Epithelial Biology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Kevin R Roy
- Department of Genetics, Stanford University, School of Medicine, Stanford, CA, USA
- Stanford Genome Technology Center, Stanford University, School of Medicine, Stanford, CA, USA
| | - Thuylinh Michelle Nguyen
- Department of Genetics, Stanford University, School of Medicine, Stanford, CA, USA
- Stanford Genome Technology Center, Stanford University, School of Medicine, Stanford, CA, USA
| | | | - Patrick S McGrath
- Department of Dermatology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Amber Cramer
- Department of Dermatology-Program in Epithelial Biology, Stanford University, School of Medicine, Stanford, CA, USA
- Center for Definitive and Curative Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Anna Bruckner
- Department of Dermatology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Ganna Bilousova
- Department of Dermatology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Dennis Roop
- Department of Dermatology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Jean Y Tang
- Department of Dermatology-Program in Epithelial Biology, Stanford University, School of Medicine, Stanford, CA, USA
- Center for Definitive and Curative Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | | | - Lars M Steinmetz
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
- Department of Genetics, Stanford University, School of Medicine, Stanford, CA, USA
- Stanford Genome Technology Center, Stanford University, School of Medicine, Stanford, CA, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University, School of Medicine, Stanford, CA, USA.
- Department of Chemical and Systems Biology, Stanford University, School of Medicine, Stanford, CA, USA.
| | - Anthony E Oro
- Department of Dermatology-Program in Epithelial Biology, Stanford University, School of Medicine, Stanford, CA, USA
- Center for Definitive and Curative Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| |
Collapse
|
50
|
Peterman EL, Ploessl DS, Galloway KE. Accelerating Diverse Cell-Based Therapies Through Scalable Design. Annu Rev Chem Biomol Eng 2024; 15:267-292. [PMID: 38594944 DOI: 10.1146/annurev-chembioeng-100722-121610] [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] [Indexed: 04/11/2024]
Abstract
Augmenting cells with novel, genetically encoded functions will support therapies that expand beyond natural capacity for immune surveillance and tissue regeneration. However, engineering cells at scale with transgenic cargoes remains a challenge in realizing the potential of cell-based therapies. In this review, we introduce a range of applications for engineering primary cells and stem cells for cell-based therapies. We highlight tools and advances that have launched mammalian cell engineering from bioproduction to precision editing of therapeutically relevant cells. Additionally, we examine how transgenesis methods and genetic cargo designs can be tailored for performance. Altogether, we offer a vision for accelerating the translation of innovative cell-based therapies by harnessing diverse cell types, integrating the expanding array of synthetic biology tools, and building cellular tools through advanced genome writing techniques.
Collapse
Affiliation(s)
- Emma L Peterman
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| | - Deon S Ploessl
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| | - Kate E Galloway
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| |
Collapse
|