1
|
Qie B, Tuo J, Chen F, Ding H, Lyu L. Gene therapy for genetic diseases: challenges and future directions. MedComm (Beijing) 2025; 6:e70091. [PMID: 39949979 PMCID: PMC11822459 DOI: 10.1002/mco2.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Genetic diseases constitute the majority of rare human diseases, resulting from abnormalities in an individual's genetic composition. Traditional treatments offer limited relief for these challenging conditions. In contrast, the rapid advancement of gene therapy presents significant advantages by directly addressing the underlying causes of genetic diseases, thereby providing the potential for precision treatment and the possibility of curing these disorders. This review aims to delineate the mechanisms and outcomes of current gene therapy approaches in clinical applications across various genetic diseases affecting different body systems. Additionally, genetic muscular disorders will be examined as a case study to investigate innovative strategies of novel therapeutic approaches, including gene replacement, gene suppression, gene supplementation, and gene editing, along with their associated advantages and limitations at both clinical and preclinical levels. Finally, this review emphasizes the existing challenges of gene therapy, such as vector packaging limitations, immunotoxicity, therapy specificity, and the subcellular localization and immunogenicity of therapeutic cargos, while discussing potential optimization directions for future research. Achieving delivery specificity, as well as long-term effectiveness and safety, will be crucial for the future development of gene therapies targeting genetic diseases.
Collapse
Affiliation(s)
- Beibei Qie
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Jianghua Tuo
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Feilong Chen
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Haili Ding
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Lei Lyu
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| |
Collapse
|
2
|
Miyake T, Tanaka K, Inoue Y, Nagai Y, Nishimura R, Seta T, Nakagawa S, Inoue KI, Hasegawa E, Minamimoto T, Doi M. Size-reduced DREADD derivatives for AAV-assisted multimodal chemogenetic control of neuronal activity and behavior. CELL REPORTS METHODS 2024; 4:100881. [PMID: 39437713 PMCID: PMC11573748 DOI: 10.1016/j.crmeth.2024.100881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/24/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) are engineered G-protein-coupled receptors that afford reversible manipulation of neuronal activity in vivo. Here, we introduce size-reduced DREADD derivatives miniDq and miniDi, which inherit the basic receptor properties from the Gq-coupled excitatory receptor hM3Dq and the Gi-coupled inhibitory receptor hM4Di, respectively, while being approximately 30% smaller in size. Taking advantage of the compact size of the receptors, we generated an adeno-associated virus (AAV) vector carrying both miniDq and the other DREADD family receptor (κ-opioid receptor-based inhibitory DREADD [KORD]) within the maximum AAV capacity (4.7 kb), allowing us to modulate neuronal activity and animal behavior in both excitatory and inhibitory directions using a single viral vector. We confirmed that expressing miniDq, but not miniDi, allowed activation of striatum activity in the cynomolgus monkey (Macaca fascicularis). The compact DREADDs may thus widen the opportunity for multiplexed interrogation and/or intervention in neuronal regulation in mice and non-human primates.
Collapse
Affiliation(s)
- Takahito Miyake
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan.
| | - Kaho Tanaka
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Yutsuki Inoue
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Yuji Nagai
- Advanced Neuroimaging Center, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Reo Nishimura
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Takehito Seta
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Shumpei Nakagawa
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Ken-Ichi Inoue
- Systems Neuroscience Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama 484-8506, Japan
| | - Emi Hasegawa
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Takafumi Minamimoto
- Advanced Neuroimaging Center, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Masao Doi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
3
|
Lau CH, Liang QL, Zhu H. Next-generation CRISPR technology for genome, epigenome and mitochondrial editing. Transgenic Res 2024; 33:323-357. [PMID: 39158822 DOI: 10.1007/s11248-024-00404-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
The application of rapidly growing CRISPR toolboxes and methods has great potential to transform biomedical research. Here, we provide a snapshot of up-to-date CRISPR toolboxes, then critically discuss the promises and hurdles associated with CRISPR-based nuclear genome editing, epigenome editing, and mitochondrial editing. The technical challenges and key solutions to realize epigenome editing in vivo, in vivo base editing and prime editing, mitochondrial editing in complex tissues and animals, and CRISPR-associated transposases and integrases in targeted genomic integration of very large DNA payloads are discussed. Lastly, we discuss the latest situation of the CRISPR/Cas9 clinical trials and provide perspectives on CRISPR-based gene therapy. Apart from technical shortcomings, ethical and societal considerations for CRISPR applications in human therapeutics and research are extensively highlighted.
Collapse
Affiliation(s)
- Cia-Hin Lau
- Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, China
| | - Qing-Le Liang
- Department of Clinical Laboratory Medicine, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Haibao Zhu
- Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, China.
| |
Collapse
|
4
|
Yurchenco PD, Kulczyk AW. Polymerizing laminins in development, health, and disease. J Biol Chem 2024; 300:107429. [PMID: 38825010 PMCID: PMC11260871 DOI: 10.1016/j.jbc.2024.107429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/12/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Polymerizing laminins are multi-domain basement membrane (BM) glycoproteins that self-assemble into cell-anchored planar lattices to establish the initial BM scaffold. Nidogens, collagen-IV and proteoglycans then bind to the scaffold at different domain loci to create a mature BM. The LN domains of adjacent laminins bind to each other to form a polymer node, while the LG domains attach to cytoskeletal-anchoring integrins and dystroglycan, as well as to sulfatides and heparan sulfates. The polymer node, the repeating unit of the polymer scaffold, is organized into a near-symmetrical triskelion. The structure, recently solved by cryo-electron microscopy in combination with AlphaFold2 modeling and biochemical studies, reveals how the LN surface residues interact with each other and how mutations cause failures of self-assembly in an emerging group of diseases, the LN-lamininopathies, that include LAMA2-related dystrophy and Pierson syndrome.
Collapse
Affiliation(s)
- Peter D Yurchenco
- Department of Pathology & Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.
| | - Arkadiusz W Kulczyk
- Department of Biochemistry and Microbiology, Institute for Quantitative Biomedicine, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
5
|
Ling C, Yu C, Wang C, Yang M, Yang H, Yang K, He Y, Shen Y, Tang S, Yu X, Zhou Z, Zhou S, Zhou J, Zhu L, Li J. rAAV capsid mutants eliminate leaky expression from DNA donor template for homologous recombination. Nucleic Acids Res 2024; 52:6518-6531. [PMID: 38783157 PMCID: PMC11194064 DOI: 10.1093/nar/gkae401] [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: 05/26/2023] [Revised: 04/28/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024] Open
Abstract
Precise genomic editing through the combination of CRISPR/Cas systems and recombinant adeno-associated virus (rAAV)-delivered homology directed repair (HDR) donor templates represents a powerful approach. However, the challenge of effectively suppressing leaky transcription from the rAAV vector, a phenomenon associated to cytotoxicity, persists. In this study, we demonstrated substantial promoter activities of various homology arms and inverted terminal repeats (ITR). To address this issue, we identified a novel rAAV variant, Y704T, which not only yields high-vector quantities but also effectively suppresses in cis mRNA transcription driven by a robust promoter. The Y704T variant maintains normal functionality in receptor interaction, intracellular trafficking, nuclear entry, uncoating, and second-strand synthesis, while specifically exhibiting defects in transcription. Importantly, this inhibitory effect is found to be independent of ITR, promoter types, and RNA polymerases. Mechanistic studies unveiled the involvement of Valosin Containing Protein (VCP/p97) in capsid-mediated transcription repression. Remarkably, the Y704T variant delivers HDR donor templates without compromising DNA replication ability and homologous recombination efficiency. In summary, our findings enhance the understanding of capsid-regulated transcription and introduce novel avenues for the application of the rAAV-CRISPR/Cas9 system in human gene therapy.
Collapse
Affiliation(s)
- Chen Ling
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chenghui Yu
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Cong Wang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Ming Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Hengbin Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Keying Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Yun He
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Yajie Shen
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Huashan Hospital, Shanghai Engineering Research Center of Industrial Microorganisms, MOE Engineering Research Center of Gene Technology, Fudan University, Shanghai 200438, China
| | - Shiyi Tang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaomin Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zhengjun Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shaolai Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liqing Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Department of Clinical Laboratory, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jixi Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Huashan Hospital, Shanghai Engineering Research Center of Industrial Microorganisms, MOE Engineering Research Center of Gene Technology, Fudan University, Shanghai 200438, China
| |
Collapse
|
6
|
Russo V, Antonini G, Massa R, Casali C, Mauriello A, Martino AM, Marconi R, Garibaldi M, Franciosa P, Zecchin M, Gaudio C, D’Andrea A, Strano S. Comprehensive Cardiovascular Management of Myotonic Dystrophy Type 1 Patients: A Report from the Italian Neuro-Cardiology Network. J Cardiovasc Dev Dis 2024; 11:63. [PMID: 38392277 PMCID: PMC10889677 DOI: 10.3390/jcdd11020063] [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: 01/01/2024] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024] Open
Abstract
Myotonic dystrophy is a hereditary disorder with systemic involvement. The Italian Neuro-Cardiology Network-"Rete delle Neurocardiologie" (INCN-RNC) is a unique collaborative experience involving neurology units combined with cardio-arrhythmology units. The INCN facilitates the creation of integrated neuro-cardiac teams in Neuromuscular Disease Centers for the management of cardiovascular involvement in the treatment of myotonic dystrophy type 1 (MD1).
Collapse
Affiliation(s)
- Vincenzo Russo
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”—“Monaldi” Hospital, 80126 Naples, Italy;
| | - Giovanni Antonini
- Neuromuscular Disease Centre, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, “Sant’Andrea” Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (G.A.); (M.G.)
| | - Roberto Massa
- Neuromuscular Diseases Unit, Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Carlo Casali
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00196 Rome, Italy;
| | - Alfredo Mauriello
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”—“Monaldi” Hospital, 80126 Naples, Italy;
- Department of Cardiology, “Umberto I” Hospital, 84014 Nocera Inferiore, Italy;
| | | | - Roberto Marconi
- Unit of Neurology, Cardio-Thoracic-Neuro-Vascular Department, “Misericordia” Hospital, 58100 Grosseto, Italy;
| | - Matteo Garibaldi
- Neuromuscular Disease Centre, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, “Sant’Andrea” Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (G.A.); (M.G.)
| | - Pasquale Franciosa
- Department of Internal, Anesthesiological and Cardiovascular Clinical Sciences, Sapienza University of Rome, 00196 Rome, Italy; (P.F.); (C.G.); (S.S.)
| | - Massimo Zecchin
- Cardiothoracovascular Department, “Cattinara” Hospital, ASUGI and University of Trieste, 34149 Trieste, Italy;
| | - Carlo Gaudio
- Department of Internal, Anesthesiological and Cardiovascular Clinical Sciences, Sapienza University of Rome, 00196 Rome, Italy; (P.F.); (C.G.); (S.S.)
| | - Antonello D’Andrea
- Department of Cardiology, “Umberto I” Hospital, 84014 Nocera Inferiore, Italy;
| | - Stefano Strano
- Department of Internal, Anesthesiological and Cardiovascular Clinical Sciences, Sapienza University of Rome, 00196 Rome, Italy; (P.F.); (C.G.); (S.S.)
| |
Collapse
|
7
|
El-Sobky TA, Abdulhady H, Mahmoud S, Amen J. Orthopedic manifestations of congenital muscular dystrophy subtypes in children: Emerging signatures need consolidation: a scoping review. JOURNAL OF MUSCULOSKELETAL SURGERY AND RESEARCH 2024; 8:11-23. [DOI: 10.25259/jmsr_229_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Our objective was to screen the literature on congenital muscular dystrophy (CMD) children/adolescents regarding the extent/nature of reporting orthopedic manifestations/deformities and to assess its appropriateness in informing clinical practice/research. We searched PubMed for original research on orthopedic surgical/non-surgical manifestations of CMD. Eligible articles needed to focus on orthopedic manifestations/deformities as one of the study objectives with no restrictions on study designs/types or search period. Eight hundred and thirty articles were initially identified and screened. Following the exclusion of 501 articles for disagreeing titles/abstracts, 329 were available for eligibility assessment. Two hundred and fifty-five articles were excluded for reasons. Of one hundred articles, 15 were captured manually and 11 through pre-submission searches, with 1078 patients included in the final analysis. The most common subtype was Laminin alpha-2 (LAMA2)-relatedCMD. Orthopedic manifestations of CMD are generally underreported and insufficiently detailed. There is reliable evidence that accurate reporting of orthopedic manifestations can be a valuable clinical supplement to the complex differential diagnosis process in collagen VI-related CMD, LAMA2-related-CMD, LMNA-related-CMD, and SEPN1-related CMD (SELENON). For alpha dystroglycan-related CMD, there is insufficient information to delineate a subtype-specific pattern. There is emerging evidence that reporting spine surgery outcomes may facilitate orthopedic decision making. The greatest clinical/research utility was provided by articles with longitudinal, comprehensive, and correlative reporting of larger cohorts. Detailed reporting of the orthopedic phenotype of CMD in future research may further uncover its diagnostic potential.
Collapse
Affiliation(s)
- Tamer A. El-Sobky
- Department of Orthopaedic Surgery, Division of Paediatric Orthopaedics, Faculty of Medicine, Ain Shams University, Cairo, Egypt,
| | - Hala Abdulhady
- Department of Physical Medicine and Rheumatology and Rehabilitation, Faculty of Medicine, Ain Shams University, Cairo, Egypt,
| | - Shady Mahmoud
- Department of Orthopaedic Surgery, Division of Paediatric Orthopaedics, Faculty of Medicine, Ain Shams University, Cairo, Egypt,
| | - John Amen
- Department of Orthopaedic Surgery, Division of Paediatric Orthopaedics, Faculty of Medicine, Ain Shams University, Cairo, Egypt,
| |
Collapse
|
8
|
Thürkauf M, Lin S, Oliveri F, Grimm D, Platt RJ, Rüegg MA. Fast, multiplexable and efficient somatic gene deletions in adult mouse skeletal muscle fibers using AAV-CRISPR/Cas9. Nat Commun 2023; 14:6116. [PMID: 37777530 PMCID: PMC10542775 DOI: 10.1038/s41467-023-41769-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/19/2023] [Indexed: 10/02/2023] Open
Abstract
Molecular screens comparing different disease states to identify candidate genes rely on the availability of fast, reliable and multiplexable systems to interrogate genes of interest. CRISPR/Cas9-based reverse genetics is a promising method to eventually achieve this. However, such methods are sorely lacking for multi-nucleated muscle fibers, since highly efficient nuclei editing is a requisite to robustly inactive candidate genes. Here, we couple Cre-mediated skeletal muscle fiber-specific Cas9 expression with myotropic adeno-associated virus-mediated sgRNA delivery to establish a system for highly effective somatic gene deletions in mice. Using well-characterized genes, we show that local or systemic inactivation of these genes copy the phenotype of traditional gene-knockout mouse models. Thus, this proof-of-principle study establishes a method to unravel the function of individual genes or entire signaling pathways in adult skeletal muscle fibers without the cumbersome requirement of generating knockout mice.
Collapse
Affiliation(s)
| | - Shuo Lin
- Biozentrum, University of Basel, Basel, Switzerland
| | | | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, Heidelberg University, Heidelberg, Germany
- BioQuant, University of Heidelberg, Heidelberg, Germany
- German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), Heidelberg, Germany
| | - Randall J Platt
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel, Switzerland
- Department of Chemistry, University of Basel, Basel, Switzerland
| | | |
Collapse
|
9
|
Nikom D, Zheng S. Alternative splicing in neurodegenerative disease and the promise of RNA therapies. Nat Rev Neurosci 2023; 24:457-473. [PMID: 37336982 DOI: 10.1038/s41583-023-00717-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2023] [Indexed: 06/21/2023]
Abstract
Alternative splicing generates a myriad of RNA products and protein isoforms of different functions from a single gene. Dysregulated alternative splicing has emerged as a new mechanism broadly implicated in the pathogenesis of neurodegenerative diseases such as Alzheimer disease, amyotrophic lateral sclerosis, frontotemporal dementia, Parkinson disease and repeat expansion diseases. Understanding the mechanisms and functional outcomes of abnormal splicing in neurological disorders is vital in developing effective therapies to treat mis-splicing pathology. In this Review, we discuss emerging research and evidence of the roles of alternative splicing defects in major neurodegenerative diseases and summarize the latest advances in RNA-based therapeutic strategies to target these disorders.
Collapse
Affiliation(s)
- David Nikom
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, USA
- Center for RNA Biology and Medicine, University of California, Riverside, Riverside, CA, USA
| | - Sika Zheng
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, USA.
- Center for RNA Biology and Medicine, University of California, Riverside, Riverside, CA, USA.
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA, USA.
| |
Collapse
|
10
|
Tran VK, Nguyen NL, Tran LNT, Le PT, Tran AH, Pham TLA, Lien NTK, Xuan NT, Thanh LT, Ta TV, Tran TH, Nguyen HH. Merosin-deficient congenital muscular dystrophy type 1a: detection of LAMA2 variants in Vietnamese patients. Front Genet 2023; 14:1183663. [PMID: 37388928 PMCID: PMC10301838 DOI: 10.3389/fgene.2023.1183663] [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: 03/10/2023] [Accepted: 06/06/2023] [Indexed: 07/01/2023] Open
Abstract
Background: Merosin-deficient congenital muscular dystrophy type 1A (MDC1A), also known as laminin-α2 chain-deficient congenital muscular dystrophy (LAMA2-MD), is an autosomal recessive disease caused by biallelic variants in the LAMA2 gene. In MDC1A, laminin- α2 chain expression is absent or significantly reduced, leading to some early-onset clinical symptoms including severe hypotonia, muscle weakness, skeletal deformity, non-ambulation, and respiratory insufficiency. Methods: Six patients from five unrelated Vietnamese families presenting with congenital muscular dystrophy were investigated. Targeted sequencing was performed in the five probands. Sanger sequencing was carried out in their families. Multiplex ligation-dependent probe amplification was performed in one family to examine an exon deletion. Results: Seven variants of the LAMA2 (NM_000426) gene were identified and classified as pathogenic/likely pathogenic variants using American College of Medical Genetics and Genomics criteria. Two of these variants were not reported in the literature, including c.7156-5_7157delinsT and c.8974_8975insTGAT. Sanger sequencing indicated their parents as carriers. The mothers of family 4 and family 5 were pregnant and a prenatal testing was performed. The results showed that the fetus of the family 4 only carries c.4717 + 5G>A in the heterozygous form, while the fetus of the family 5 carries compound heterozygous variants, including a deletion of exon 3 and c.4644C>A. Conclusion: Our findings not only identified the underlying genetic etiology for the patients, but also provided genetic counseling for the parents whenever they have an offspring.
Collapse
Affiliation(s)
- Van Khanh Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Ngoc-Lan Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Lan Ngoc Thi Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Phuong Thi Le
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Anh Hai Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Tuan L. A. Pham
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Nguyen Thi Kim Lien
- Institute of Genome Research, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Nguyen Thi Xuan
- Institute of Genome Research, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Le Tat Thanh
- Institute of Genome Research, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Thanh Van Ta
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
- Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | - Thinh Huy Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
- Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | - Huy-Hoang Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| |
Collapse
|
11
|
Oliveira-Santos A, Dagda M, Wittmann J, Smalley R, Burkin DJ. Vemurafenib improves muscle histopathology in a mouse model of LAMA2-related congenital muscular dystrophy. Dis Model Mech 2023; 16:dmm049916. [PMID: 37021539 PMCID: PMC10184677 DOI: 10.1242/dmm.049916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
Laminin-α2-related congenital muscular dystrophy (LAMA2-CMD) is a neuromuscular disease affecting around 1-9 in 1,000,000 children. LAMA2-CMD is caused by mutations in the LAMA2 gene resulting in the loss of laminin-211/221 heterotrimers in skeletal muscle. LAMA2-CMD patients exhibit severe hypotonia and progressive muscle weakness. Currently, there is no effective treatment for LAMA2-CMD and patients die prematurely. The loss of laminin-α2 results in muscle degeneration, defective muscle repair and dysregulation of multiple signaling pathways. Signaling pathways that regulate muscle metabolism, survival and fibrosis have been shown to be dysregulated in LAMA2-CMD. As vemurafenib is a US Food and Drug Administration (FDA)-approved serine/threonine kinase inhibitor, we investigated whether vemurafenib could restore some of the serine/threonine kinase-related signaling pathways and prevent disease progression in the dyW-/- mouse model of LAMA2-CMD. Our results show that vemurafenib reduced muscle fibrosis, increased myofiber size and reduced the percentage of fibers with centrally located nuclei in dyW-/- mouse hindlimbs. These studies show that treatment with vemurafenib restored the TGF-β/SMAD3 and mTORC1/p70S6K signaling pathways in skeletal muscle. Together, our results indicate that vemurafenib partially improves histopathology but does not improve muscle function in a mouse model of LAMA2-CMD.
Collapse
Affiliation(s)
- Ariany Oliveira-Santos
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Marisela Dagda
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Jennifer Wittmann
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Robert Smalley
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Dean J. Burkin
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| |
Collapse
|
12
|
McKee KK, Yurchenco PD. Dual transgene amelioration of Lama2-null muscular dystrophy. Matrix Biol 2023; 118:1-15. [PMID: 36878377 PMCID: PMC10771811 DOI: 10.1016/j.matbio.2023.03.001] [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: 12/29/2022] [Revised: 02/14/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Null mutations of the Lama2-gene cause a severe congenital muscular dystrophy and associated neuropathy. In the absence of laminin-α2 (Lmα2) there is a compensatory replacement by Lmα4, a subunit that lacks the polymerization and α-dystroglycan (αDG)-binding properties of Lmα2. The dystrophic phenotype in the dy3K/dy3K Lama2-/- mouse were evaluated with transgenes driving expression of two synthetic laminin-binding linker proteins. Transgenic muscle-specific expression of αLNNd, a chimeric protein that enables α4-laminin polymerization, and miniagrin (mag), a protein that increases laminin binding to the receptor αDG, separately improved median mouse survival two-fold. The double transgenes (DT) improved mean survival three-fold with increases in overall body weight, muscle size, and grip strength, but, given absence of neuronal expression, did not prevent hindlimb paresis. Muscle improvements included increased myofiber size and number and reduced fibrosis. Myofiber hypertrophy with increased mTOR and Akt phosphorylation were characteristics of mag-dy3K/dy3K and DT-dy3K/dy3K muscle. Elevations of matrix-bound α4-, β1 and γ1 laminin subunits were detected in muscle extracts and immunostained sections in response to DT expression. Collectively, these findings reveal a complimentary polymerization and αDG-binding benefit to Lama2-/- mouse muscle largely mediated through modified laminin-411.
Collapse
Affiliation(s)
- Karen K McKee
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Peter D Yurchenco
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
13
|
Teng ACT, Tavassoli M, Shrestha S, Marks RM, McFadden MJ, Evagelou SL, Lindsay K, Vandenbelt A, Li W, Ivakine E, Cohn R, Santerre JP, Gramolini AO. An efficient and cost-effective purification protocol for Staphylococcus aureus Cas9 nuclease. STAR Protoc 2023; 4:101933. [PMID: 36574341 PMCID: PMC9813775 DOI: 10.1016/j.xpro.2022.101933] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/10/2022] [Accepted: 11/23/2022] [Indexed: 12/27/2022] Open
Abstract
Here, we describe a protocol for purifying functional clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) from Staphylococcus aureus within 24 h and over 90% purity. SaCas9 purification begins with immobilized metal affinity chromatography, followed by cation exchange chromatography, and ended with centrifugal concentrators. The simplicity, cost-effectiveness, and reproducibility of such protocols will enable general labs to produce a sizable amount of Cas9 proteins, further accelerating CRISPR research.
Collapse
Affiliation(s)
- Allen C T Teng
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th Floor, Toronto, ON M5G 1M1, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Marjan Tavassoli
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th Floor, Toronto, ON M5G 1M1, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Suja Shrestha
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th Floor, Toronto, ON M5G 1M1, Canada; Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1X3, Canada
| | - Ryan M Marks
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Meghan J McFadden
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th Floor, Toronto, ON M5G 1M1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Sonia L Evagelou
- Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Kyle Lindsay
- Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Ava Vandenbelt
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th Floor, Toronto, ON M5G 1M1, Canada; Faculty of Kinesiology & Physical Education, University of Toronto, Toronto, ON M5S 2W6, Canada
| | - Wenping Li
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th Floor, Toronto, ON M5G 1M1, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Evgueni Ivakine
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Ronald Cohn
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - J Paul Santerre
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th Floor, Toronto, ON M5G 1M1, Canada; Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1X3, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Anthony O Gramolini
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th Floor, Toronto, ON M5G 1M1, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
14
|
Poyatos-García J, Blázquez-Bernal Á, Selva-Giménez M, Bargiela A, Espinosa-Espinosa J, Vázquez-Manrique RP, Bigot A, Artero R, Vilchez JJ. CRISPR-Cas9 editing of a TNPO3 mutation in a muscle cell model of limb-girdle muscular dystrophy type D2. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:324-338. [PMID: 36789274 PMCID: PMC9898580 DOI: 10.1016/j.omtn.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 01/06/2023] [Indexed: 01/13/2023]
Abstract
A single-nucleotide deletion in the stop codon of the nuclear import receptor transportin-3 (TNPO3), also involved in human immunodeficiency virus type 1 (HIV-1) infection, causes the ultrarare autosomal dominant disease limb-girdle muscular dystrophy D2 (LGMDD2) by extending the wild-type protein. Here, we generated a patient-derived in vitro model of LGMDD2 as an immortalized myoblast cell line carrying the TNP O 3 mutation. The cell model reproduced critical molecular alterations seen in patients, such as TNP O 3 overexpression, defects in terminal muscle markers, and autophagy overactivation. Correction of the TNP O 3 mutation via CRISPR-Cas9 editing caused a significant reversion of the pathological phenotypes in edited cells, including a complete absence of the mutant TNPO3 protein, as detected with a polyclonal antibody specific against the abnormal 15-aa peptide. Transcriptomic analyses found that 15% of the transcriptome was differentially expressed in model myotubes. CRISPR-Cas9-corrected cells showed that 44% of the alterations were rescued toward normal levels. MicroRNAs (miRNAs) analyses showed that around 50% of miRNAs with impaired expression because of the disease were recovered on the mutation edition. In summary, this work provides proof of concept of the potential of CRISPR-Cas9-mediated gene editing of TNP O 3 as a therapeutic approach and describes critical reagents in LGMDD2 research.
Collapse
Affiliation(s)
- Javier Poyatos-García
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), U763, CB06/05/0091, 46026 Valencia, Spain
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain
| | - Águeda Blázquez-Bernal
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Burjasot, 46100 Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Marta Selva-Giménez
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), U763, CB06/05/0091, 46026 Valencia, Spain
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain
| | - Ariadna Bargiela
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain
| | - Jorge Espinosa-Espinosa
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Burjasot, 46100 Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Rafael P. Vázquez-Manrique
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), U763, CB06/05/0091, 46026 Valencia, Spain
- Grupo de Investigación en Biomedicina Molecular, Celular y Genómica, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain
- Joint Unit for Rare Diseases IIS La Fe-CIPF, 46012 Valencia, Spain
| | - Anne Bigot
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Ruben Artero
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Burjasot, 46100 Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Juan Jesús Vilchez
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), U763, CB06/05/0091, 46026 Valencia, Spain
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital La Fe (IIS La Fe), 46026 Valencia, Spain
| |
Collapse
|
15
|
Arockiaraj AI, Johnson MA, Munir A, Ekambaram P, Lucas PC, McAllister-Lucas LM, Kemaladewi DU. CRISPRa-induced upregulation of human LAMA1 compensates for LAMA2-deficiency in Merosin-deficient congenital muscular dystrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531347. [PMID: 36945402 PMCID: PMC10028808 DOI: 10.1101/2023.03.06.531347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Merosin-deficient congenital muscular dystrophy (MDC1A) is an autosomal recessive disorder caused by mutations in the LAMA2 gene, resulting in a defective form of the extracellular matrix protein laminin-α2 (LAMA2). Individuals diagnosed with MDC1A exhibit progressive muscle wasting and declining neuromuscular functions. No treatments for this disorder are currently available. We previously showed that postnatal Lama1 upregulation, achieved through CRISPR activation (CRISPRa), compensates for Lama2 deficiency and prevents neuromuscular pathophysiology in a mouse model of MDC1A. In this study, we assessed the feasibility of upregulating human LAMA1 as a potential therapeutic strategy for individuals with MDC1A, regardless of their mutations. We hypothesized that CRISPRa-mediated upregulation of human LAMA1 would compensate for the lack of LAMA2 and rescue cellular abnormalities in MDC1A fibroblasts. Global transcriptomic and pathway enrichment analyses of fibroblasts collected from individuals carrying pathogenic LAMA2 mutations, compared with healthy controls, indicated higher expression of transcripts encoding proteins that contribute to wound healing, including Transforming Growth Factor-β (TGF-β) and Fibroblast Growth Factor (FGF). These findings were supported by wound-healing assays indicating that MDC1A fibroblasts migrated significantly more rapidly than the controls. Subsequently, we treated the MDC1A fibroblasts with SadCas9-2XVP64 and sgRNAs targeting the LAMA1 promoter. We observed robust LAMA1 expression, which was accompanied by significant decreases in cell migration and expression of FGFR2, TGF-β2, and ACTA2, which are involved in the wound-healing mechanism in MDC1A fibroblasts. Collectively, our data suggest that CRISPRa-mediated LAMA1 upregulation may be a feasible mutation-independent therapeutic approach for MDC1A. This strategy might be adapted to address other neuromuscular diseases and inherited conditions in which strong compensatory mechanisms have been identified.
Collapse
Affiliation(s)
- Annie I. Arockiaraj
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Marie A. Johnson
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Anushe Munir
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Prasanna Ekambaram
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Peter C. Lucas
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | | | - Dwi U. Kemaladewi
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| |
Collapse
|
16
|
Genome Editing to Abrogate Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:157-176. [DOI: 10.1007/978-981-19-5642-3_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
17
|
Apolónio JD, Dias JS, Fernandes MT, Komosa M, Lipman T, Zhang CH, Leão R, Lee D, Nunes NM, Maia AT, Morera JL, Vicioso L, Tabori U, Castelo-Branco P. THOR is a targetable epigenetic biomarker with clinical implications in breast cancer. Clin Epigenetics 2022; 14:178. [PMID: 36529814 PMCID: PMC9759897 DOI: 10.1186/s13148-022-01396-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is the most frequently diagnosed cancer and a leading cause of death among women worldwide. Early BC is potentially curable, but the mortality rates still observed among BC patients demonstrate the urgent need of novel and more effective diagnostic and therapeutic options. Limitless self-renewal is a hallmark of cancer, governed by telomere maintenance. In around 95% of BC cases, this process is achieved by telomerase reactivation through upregulation of the human telomerase reverse transcriptase (hTERT). The hypermethylation of a specific region within the hTERT promoter, termed TERT hypermethylated oncological region (THOR) has been associated with increased hTERT expression in cancer. However, its biological role and clinical potential in BC have never been studied to the best of our knowledge. Therefore, we aimed to investigate the role of THOR as a biomarker and explore the functional impact of THOR methylation status in hTERT upregulation in BC. RESULTS THOR methylation status in BC was assessed by pyrosequencing on discovery and validation cohorts. We found that THOR is significantly hypermethylated in malignant breast tissue when compared to benign tissue (40.23% vs. 12.81%, P < 0.0001), differentiating malignant tumor from normal tissue from the earliest stage of disease. Using a reporter assay, the addition of unmethylated THOR significantly reduced luciferase activity by an average 1.8-fold when compared to the hTERT core promoter alone (P < 0.01). To further investigate its biological impact on hTERT transcription, targeted THOR demethylation was performed using novel technology based on CRISPR-dCas9 system and significant THOR demethylation was achieved. Cells previously demethylated on THOR region did not develop a histologic cancer phenotype in in vivo assays. Additional studies are required to validate these observations and to unravel the causality between THOR hypermethylation and hTERT upregulation in BC. CONCLUSIONS THOR hypermethylation is an important epigenetic mark in breast tumorigenesis, representing a promising biomarker and therapeutic target in BC. We revealed that THOR acts as a repressive regulatory element of hTERT and that its hypermethylation is a relevant mechanism for hTERT upregulation in BC.
Collapse
Affiliation(s)
- Joana Dias Apolónio
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Gambelas Campus, Bld. 2, 8005-139, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - João S Dias
- University Hospital Center of Algarve, Faro, Portugal
| | - Mónica Teotónio Fernandes
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
- Escola Superior de Saúde (ESSUAlg), Universidade Do Algarve, Faro, Portugal
| | - Martin Komosa
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Tatiana Lipman
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Cindy H Zhang
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Ricardo Leão
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Donghyun Lee
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Nuno Miguel Nunes
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Ana-Teresa Maia
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Gambelas Campus, Bld. 2, 8005-139, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
- Center for Research in Health Technologies and Information Systems (CINTESIS@RISE), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
| | - José L Morera
- University Hospital Center of Algarve, Faro, Portugal
| | - Luis Vicioso
- Faculty of Medicine, Department of Histology and Pathological Anatomy, University of Malaga, Malaga, Spain
| | - Uri Tabori
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Pedro Castelo-Branco
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Gambelas Campus, Bld. 2, 8005-139, Faro, Portugal.
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal.
- Algarve Biomedical Center (ABC), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal.
- Champalimaud Research Program, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
18
|
Abstract
Muscular dystrophies are a group of genetic disorders characterized by varying degrees of progressive muscle weakness and degeneration. They are clinically and genetically heterogeneous but share the common histological features of dystrophic muscle. There is currently no cure for muscular dystrophies, which is of particular concern for the more disabling and/or lethal forms of the disease. Through the years, several therapies have encouragingly been developed for muscular dystrophies and include genetic, cellular, and pharmacological approaches. In this chapter, we undertake a comprehensive exploration of muscular dystrophy therapeutics under current development. Our review includes antisense therapy, CRISPR, gene replacement, cell therapy, nonsense suppression, and disease-modifying small molecule compounds.
Collapse
|
19
|
Li R, Wang Q, She K, Lu F, Yang Y. CRISPR/Cas systems usher in a new era of disease treatment and diagnosis. MOLECULAR BIOMEDICINE 2022; 3:31. [PMID: 36239875 PMCID: PMC9560888 DOI: 10.1186/s43556-022-00095-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/27/2022] [Indexed: 11/21/2022] Open
Abstract
The discovery and development of the CRISPR/Cas system is a milestone in precise medicine. CRISPR/Cas nucleases, base-editing (BE) and prime-editing (PE) are three genome editing technologies derived from CRISPR/Cas. In recent years, CRISPR-based genome editing technologies have created immense therapeutic potential with safe and efficient viral or non-viral delivery systems. Significant progress has been made in applying genome editing strategies to modify T cells and hematopoietic stem cells (HSCs) ex vivo and to treat a wide variety of diseases and disorders in vivo. Nevertheless, the clinical translation of this unique technology still faces many challenges, especially targeting, safety and delivery issues, which require further improvement and optimization. In addition, with the outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), CRISPR-based molecular diagnosis has attracted extensive attention. Growing from the specific set of molecular biological discoveries to several active clinical trials, CRISPR/Cas systems offer the opportunity to create a cost-effective, portable and point-of-care diagnosis through nucleic acid screening of diseases. In this review, we describe the development, mechanisms and delivery systems of CRISPR-based genome editing and focus on clinical and preclinical studies of therapeutic CRISPR genome editing in disease treatment as well as its application prospects in therapeutics and molecular detection.
Collapse
Affiliation(s)
- Ruiting Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, China
| | - Qin Wang
- School of Pharmacy, Southwest Minzu University, Chengdu, 610225, Sichuan, China
| | - Kaiqin She
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, China
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fang Lu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, China.
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
20
|
López-Márquez A, Martínez-Pizarro A, Pérez B, Richard E, Desviat LR. Modeling Splicing Variants Amenable to Antisense Therapy by Use of CRISPR-Cas9-Based Gene Editing in HepG2 Cells. Methods Mol Biol 2022; 2434:167-184. [PMID: 35213016 PMCID: PMC9703257 DOI: 10.1007/978-1-0716-2010-6_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The field of splice modulating RNA therapy has gained new momentum with FDA approved antisense-based drugs for several rare diseases. In vitro splicing assays with minigenes or patient-derived cells are commonly employed for initial preclinical testing of antisense oligonucleotides aiming to modulate splicing. However, minigenes do not include the full genomic context of the exons under study and patients' samples are not always available, especially if the gene is expressed solely in certain tissues (e.g. liver or brain). This is the case for specific inherited metabolic diseases such as phenylketonuria (PKU) caused by mutations in the liver-expressed PAH gene.Herein we describe the generation of mutation-specific hepatic cellular models of PKU using CRISPR/Cas9 system, which is a versatile and easy-to-use gene editing tool. We describe in detail the selection of the appropriate cell line, guidelines for design of RNA guides and donor templates, transfection procedures and growth and selection of single-cell colonies with the desired variant , which should result in the accurate recapitulation of the splicing defect.
Collapse
Affiliation(s)
- Arístides López-Márquez
- Centro de Biología Molecular Severo Ochoa CSIC-UAM, CEDEM, CIBERER (ISCIII), IdiPaz (ISCIII), Universidad Autónoma de Madrid, Madrid, Spain
| | - Ainhoa Martínez-Pizarro
- Centro de Biología Molecular Severo Ochoa CSIC-UAM, CEDEM, CIBERER (ISCIII), IdiPaz (ISCIII), Universidad Autónoma de Madrid, Madrid, Spain
| | - Belén Pérez
- Centro de Biología Molecular Severo Ochoa CSIC-UAM, CEDEM, CIBERER (ISCIII), IdiPaz (ISCIII), Universidad Autónoma de Madrid, Madrid, Spain
| | - Eva Richard
- Centro de Biología Molecular Severo Ochoa CSIC-UAM, CEDEM, CIBERER (ISCIII), IdiPaz (ISCIII), Universidad Autónoma de Madrid, Madrid, Spain
| | - Lourdes R Desviat
- Centro de Biología Molecular Severo Ochoa CSIC-UAM, CEDEM, CIBERER (ISCIII), IdiPaz (ISCIII), Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
21
|
Tan D, Ge L, Fan Y, Chang X, Wang S, Wei C, Ding J, Liu A, Wang S, Li X, Gao K, Yang H, Que C, Huang Z, Li C, Zhu Y, Mao B, Jin B, Hua Y, Zhang X, Zhang B, Zhu W, Zhang C, Wang Y, Yuan Y, Jiang Y, Rutkowski A, Bönnemann CG, Wu X, Xiong H. Natural history and genetic study of LAMA2-related muscular dystrophy in a large Chinese cohort. Orphanet J Rare Dis 2021; 16:319. [PMID: 34281576 PMCID: PMC8287797 DOI: 10.1186/s13023-021-01950-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/11/2021] [Indexed: 11/12/2022] Open
Abstract
Background LAMA2-related muscular dystrophy including LAMA2-related congenital muscular dystrophy (LAMA2-CMD) and autosomal recessive limb-girdle muscular dystrophy-23 (LGMDR23) is caused by LAMA2 pathogenic variants. We aimed to describe the natural history and establish genotype–phenotype correlations in a large cohort of Chinese patients with LAMA2-related muscular dystrophy. Methods Clinical and genetic data of LAMA2-related muscular dystrophy patients enrolled from ten research centers between January 2003 and March 2021 were collected and analyzed. Results One hundred and thirty patients (116 LAMA2-CMD and 14 LGMDR23) were included. LAMA2-CMD group had earlier onset than LGMDR23 group. Head control, independent sitting and ambulation were achieved in 76.3%, 92.6% and 18.4% of LAMA2-CMD patients at median ages of 6.0 months (range 2.0–36.0 months), 11.0 months (range 6.0–36.0 months), and 27.0 months (range 18.0–84.0 months), respectively. All LGMDR23 patients achieved independent ambulation at median age of 18.0 months (range 13.0–20.0 months). Motor regression in LAMA2-CMD mainly occurred concurrently with rapid progression of contractures during 6–9 years old. Twenty-four LAMA2-related muscular dystrophy patients died, mostly due to severe pneumonia. Seizures occurred in 35.7% of LGMDR23 and 9.5% of LAMA2-CMD patients. Forty-six novel and 97 known LAMA2 disease-causing variants were identified. The top three high-frequency disease-causing variants in Han Chinese patients were c.7147C > T (p.R2383*), exon 4 deletion, and c.5156_5159del (p.K1719Rfs*5). In LAMA2-CMD, splicing variants tended to be associated with a relatively mild phenotype. Nonsense variants were more frequent in LAMA2-CMD (56.9%, 66/116) than in LGMDR23 (21.4%, 3/14), while missense disease-causing variants were more frequent in LGMDR23 (71.4%, 10/14) than in LAMA2-CMD (12.9%, 15/116). Copy number variations were identified in 26.4% of survivors and 50.0% of nonsurvivors, suggesting that copy number variations were associated with lower rate of survival (p = 0.029). Conclusions This study provides better understandings of natural history and genotype–phenotype correlations in LAMA2-related muscular dystrophy, and supports therapeutic targets for future researches. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01950-x.
Collapse
Affiliation(s)
- Dandan Tan
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Lin Ge
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Yanbin Fan
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Xingzhi Chang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Shuang Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Cuijie Wei
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Juan Ding
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Aijie Liu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Shuo Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Xueying Li
- Department of Statistics, Peking University First Hospital, Beijing, 100034, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Haipo Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Chengli Que
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Zhen Huang
- Department of Rehabilitation Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Chunde Li
- Department of Orthopedic/Spine Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Ying Zhu
- Department of Radiology, Peking University First Hospital, Beijing, 100034, China
| | - Bing Mao
- Department of Neurology, Wuhan Children's Hospital, Wuhan, 430015, Hubei Province, China
| | - Bo Jin
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, Jiangsu Province, China
| | - Ying Hua
- Department of Pediatrics, Wuxi Children's Hospital, Wuxi, 214000, Jiangsu Province, China
| | - Xiaoli Zhang
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Bingbing Zhang
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu Province, China
| | - Wenhua Zhu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Cheng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong Province, China
| | - Yanjuan Wang
- Department of Neurology, School of Medicine, Chengdu Women's & Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 610091, Sichuan Province, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | | | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Xiru Wu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Hui Xiong
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
22
|
Smeets HJM, Verbrugge B, Springuel P, Voermans NC. Merosin deficient congenital muscular dystrophy type 1A: An international workshop on the road to therapy 15-17 November 2019, Maastricht, the Netherlands. Neuromuscul Disord 2021; 31:673-680. [PMID: 34130888 PMCID: PMC8994498 DOI: 10.1016/j.nmd.2021.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Hubert J M Smeets
- Department of Toxicogenomics, Research Schools GROW and MHeNS, Maastricht University, Maastricht, The Netherlands.
| | - Bram Verbrugge
- MDC1A Foundation "Voor Sara", Dordrecht, The Netherlands
| | | | - Nicol C Voermans
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
23
|
Maino E, Wojtal D, Evagelou SL, Farheen A, Wong TWY, Lindsay K, Scott O, Rizvi SZ, Hyatt E, Rok M, Visuvanathan S, Chiodo A, Schneeweiss M, Ivakine EA, Cohn RD. Targeted genome editing in vivo corrects a Dmd duplication restoring wild-type dystrophin expression. EMBO Mol Med 2021; 13:e13228. [PMID: 33724658 PMCID: PMC8103086 DOI: 10.15252/emmm.202013228] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 02/06/2021] [Accepted: 02/10/2021] [Indexed: 12/26/2022] Open
Abstract
Tandem duplication mutations are increasingly found to be the direct cause of many rare heritable diseases, accounting for up to 10% of cases. Unfortunately, animal models recapitulating such mutations are scarce, limiting our ability to study them and develop genome editing therapies. Here, we describe the generation of a novel duplication mouse model, harboring a multi-exonic tandem duplication in the Dmd gene which recapitulates a human mutation. Duplication correction of this mouse was achieved by implementing a single-guide RNA (sgRNA) CRISPR/Cas9 approach. This strategy precisely removed a duplication mutation in vivo, restored full-length dystrophin expression, and was accompanied by improvements in both histopathological and clinical phenotypes. We conclude that CRISPR/Cas9 represents a powerful tool to accurately model and treat tandem duplication mutations. Our findings will open new avenues of research for exploring the study and therapeutics of duplication disorders.
Collapse
Affiliation(s)
- Eleonora Maino
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Daria Wojtal
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Sonia L Evagelou
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Aiman Farheen
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Tatianna W Y Wong
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Kyle Lindsay
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Ori Scott
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
- Department of Pediatricsthe Hospital for Sick ChildrenTorontoONCanada
| | - Samar Z Rizvi
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Elzbieta Hyatt
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Matthew Rok
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Shagana Visuvanathan
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Amanda Chiodo
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Michelle Schneeweiss
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
| | - Evgueni A Ivakine
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of PhysiologyUniversity of TorontoTorontoONCanada
| | - Ronald D Cohn
- Program in Genetics and Genome Biologythe Hospital for Sick Children Research InstituteTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
- Department of Pediatricsthe Hospital for Sick ChildrenTorontoONCanada
| |
Collapse
|
24
|
He X, Urip BA, Zhang Z, Ngan CC, Feng B. Evolving AAV-delivered therapeutics towards ultimate cures. J Mol Med (Berl) 2021; 99:593-617. [PMID: 33594520 PMCID: PMC7885987 DOI: 10.1007/s00109-020-02034-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022]
Abstract
Gene therapy has entered a new era after decades-long efforts, where the recombinant adeno-associated virus (AAV) has stood out as the most potent vector for in vivo gene transfer and demonstrated excellent efficacy and safety profiles in numerous preclinical and clinical studies. Since the first AAV-derived therapeutics Glybera was approved by the European Medicines Agency (EMA) in 2012, there is an increasing number of AAV-based gene augmentation therapies that have been developed and tested for treating incurable genetic diseases. In the subsequent years, the United States Food and Drug Administration (FDA) approved two additional AAV gene therapy products, Luxturna and Zolgensma, to be launched into the market. Recent breakthroughs in genome editing tools and the combined use with AAV vectors have introduced new therapeutic modalities using somatic gene editing strategies. The promising outcomes from preclinical studies have prompted the continuous evolution of AAV-delivered therapeutics and broadened the scope of treatment options for untreatable diseases. Here, we describe the clinical updates of AAV gene therapies and the latest development using AAV to deliver the CRISPR components as gene editing therapeutics. We also discuss the major challenges and safety concerns associated with AAV delivery and CRISPR therapeutics, and highlight the recent achievement and toxicity issues reported from clinical applications.
Collapse
Affiliation(s)
- Xiangjun He
- School of Biomedical Sciences, Faculty of Medicine; Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Brian Anugerah Urip
- School of Biomedical Sciences, Faculty of Medicine; Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Zhenjie Zhang
- School of Biomedical Sciences, Faculty of Medicine; Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
| | - Chun Christopher Ngan
- School of Biomedical Sciences, Faculty of Medicine; Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Shatin N.T., Hong Kong SAR, China
| | - Bo Feng
- School of Biomedical Sciences, Faculty of Medicine; Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Shatin N.T., Hong Kong SAR, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Shatin N.T., Hong Kong SAR, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510320, China.
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
25
|
Chuang YF, Phipps AJ, Lin FL, Hecht V, Hewitt AW, Wang PY, Liu GS. Approach for in vivo delivery of CRISPR/Cas system: a recent update and future prospect. Cell Mol Life Sci 2021; 78:2683-2708. [PMID: 33388855 PMCID: PMC11072787 DOI: 10.1007/s00018-020-03725-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022]
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) system provides a groundbreaking genetic technology that allows scientists to modify genes by targeting specific genomic sites. Due to the relative simplicity and versatility of the CRISPR/Cas system, it has been extensively applied in human genetic research as well as in agricultural applications, such as improving crops. Since the gene editing activity of the CRISPR/Cas system largely depends on the efficiency of introducing the system into cells or tissues, an efficient and specific delivery system is critical for applying CRISPR/Cas technology. However, there are still some hurdles remaining for the translatability of CRISPR/Cas system. In this review, we summarized the approaches used for the delivery of the CRISPR/Cas system in mammals, plants, and aquacultures. We further discussed the aspects of delivery that can be improved to elevate the potential for CRISPR/Cas translatability.
Collapse
Affiliation(s)
- Yu-Fan Chuang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, China
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
| | - Andrew J Phipps
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Fan-Li Lin
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, China
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
| | - Valerie Hecht
- School of Natural Sciences, University of Tasmania, Hobart, TAS, Australia
| | - Alex W Hewitt
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, Australia
| | - Peng-Yuan Wang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, China.
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, VIC, Australia.
| | - Guei-Sheung Liu
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia.
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, Australia.
- Aier Eye Institute, Changsha, Hunan, China.
| |
Collapse
|
26
|
Packer D, Martin PT. Micro-laminin gene therapy can function as an inhibitor of muscle disease in the dy W mouse model of MDC1A. Mol Ther Methods Clin Dev 2021; 21:274-287. [PMID: 33869655 PMCID: PMC8026908 DOI: 10.1016/j.omtm.2021.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/03/2021] [Indexed: 02/04/2023]
Abstract
Gene replacement for laminin-α2-deficient congenital muscular dystrophy 1A (MDC1A) is currently not possible using a single adeno-associated virus (AAV) vector due to the large size of the LAMA2 gene. LAMA2 encodes laminin-α2, a subunit of the trimeric laminin-211 extracellular matrix (ECM) protein that is the predominant laminin expressed in skeletal muscle. LAMA2 expression stabilizes skeletal muscle, in part by binding membrane receptors via its five globular (G) domains. We created a small, AAV-deliverable, micro-laminin gene therapy that expresses these G1-5 domains, LAMA2(G1-5), to test their therapeutic efficacy in the dyW mouse model for MDC1A. We also fused the heparin-binding (HB) domain from HB epidermal growth factor-like growth factor (HB-EGF) to LAMA2(G1-5) to test whether this would increase muscle ECM expression. dyW mice treated intravenously with rAAV9.CMV.HB-LAMA2(G1-5) showed increased muscle ECM expression of transgenic protein relative to mice treated with rAAV9.CMV.LAMA2(G1-5) and showed improved weight-normalized forelimb grip strength relative to untreated dyW mice. Additionally, dyW muscle fibers expressing either micro-laminin protein showed some measures of reduced pathology, although levels of muscle cell apoptosis and inflammation were not decreased. Although systemic expression of rAAV9.CMV.HB-LAMA2(G1-5) did not inhibit all disease phenotypes, these studies demonstrate the feasibility of using a micro-laminin gene therapy strategy to deliver gene replacement for MDC1A.
Collapse
Affiliation(s)
- Davin Packer
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
- Center for Gene Therapy, Abigail Wexner Research Institute, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Paul T. Martin
- Center for Gene Therapy, Abigail Wexner Research Institute, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Corresponding author Paul T. Martin, Center for Gene Therapy, Abigail Wexner Research Institute, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43209, USA.
| |
Collapse
|
27
|
Lau CH, Tin C, Suh Y. CRISPR-based strategies for targeted transgene knock-in and gene correction. Fac Rev 2020; 9:20. [PMID: 33659952 PMCID: PMC7886068 DOI: 10.12703/r/9-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The last few years have seen tremendous advances in CRISPR-mediated genome editing. Great efforts have been made to improve the efficiency, specificity, editing window, and targeting scope of CRISPR/Cas9-mediated transgene knock-in and gene correction. In this article, we comprehensively review recent progress in CRISPR-based strategies for targeted transgene knock-in and gene correction in both homology-dependent and homology-independent approaches. We cover homology-directed repair (HDR), synthesis-dependent strand annealing (SDSA), microhomology-mediated end joining (MMEJ), and homology-mediated end joining (HMEJ) pathways for a homology-dependent strategy and alternative DNA repair pathways such as non-homologous end joining (NHEJ), base excision repair (BER), and mismatch repair (MMR) for a homology-independent strategy. We also discuss base editing and prime editing that enable direct conversion of nucleotides in genomic DNA without damaging the DNA or requiring donor DNA. Notably, we illustrate the key mechanisms and design principles for each strategy, providing design guidelines for multiplex, flexible, scarless gene insertion and replacement at high efficiency and specificity. In addition, we highlight next-generation base editors that provide higher editing efficiency, fewer undesired by-products, and broader targeting scope.
Collapse
Affiliation(s)
- Cia-Hin Lau
- Department of Biomedical Engineering, Academic 1, 83 Tat Chee Avenue, City University of Hong Kong, Hong Kong
| | - Chung Tin
- Department of Biomedical Engineering, Academic 1, 83 Tat Chee Avenue, City University of Hong Kong, Hong Kong
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA
- Department of Genetics and Development, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA
| |
Collapse
|
28
|
Wong TWY, Ahmed A, Yang G, Maino E, Steiman S, Hyatt E, Chan P, Lindsay K, Wong N, Golebiowski D, Schneider J, Delgado-Olguín P, Ivakine EA, Cohn RD. A novel mouse model of Duchenne muscular dystrophy carrying a multi-exonic Dmd deletion exhibits progressive muscular dystrophy and early-onset cardiomyopathy. Dis Model Mech 2020; 13:dmm045369. [PMID: 32988972 PMCID: PMC7522028 DOI: 10.1242/dmm.045369] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a life-threatening neuromuscular disease caused by the lack of dystrophin, resulting in progressive muscle wasting and locomotor dysfunctions. By adulthood, almost all patients also develop cardiomyopathy, which is the primary cause of death in DMD. Although there has been extensive effort in creating animal models to study treatment strategies for DMD, most fail to recapitulate the complete skeletal and cardiac disease manifestations that are presented in affected patients. Here, we generated a mouse model mirroring a patient deletion mutation of exons 52-54 (Dmd Δ52-54). The Dmd Δ52-54 mutation led to the absence of dystrophin, resulting in progressive muscle deterioration with weakened muscle strength. Moreover, Dmd Δ52-54 mice present with early-onset hypertrophic cardiomyopathy, which is absent in current pre-clinical dystrophin-deficient mouse models. Therefore, Dmd Δ52-54 presents itself as an excellent pre-clinical model to evaluate the impact on skeletal and cardiac muscles for both mutation-dependent and -independent approaches.
Collapse
Affiliation(s)
- Tatianna Wai Ying Wong
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Abdalla Ahmed
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Program in Translational Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Grace Yang
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Eleonora Maino
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sydney Steiman
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Elzbieta Hyatt
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Parry Chan
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Kyle Lindsay
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Nicole Wong
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | | | | | - Paul Delgado-Olguín
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Program in Translational Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Evgueni A Ivakine
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Physiology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ronald D Cohn
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Pediatrics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
29
|
Suñé-Pou M, Limeres MJ, Moreno-Castro C, Hernández-Munain C, Suñé-Negre JM, Cuestas ML, Suñé C. Innovative Therapeutic and Delivery Approaches Using Nanotechnology to Correct Splicing Defects Underlying Disease. Front Genet 2020; 11:731. [PMID: 32760425 PMCID: PMC7373156 DOI: 10.3389/fgene.2020.00731] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
Alternative splicing of pre-mRNA contributes strongly to the diversity of cell- and tissue-specific protein expression patterns. Global transcriptome analyses have suggested that >90% of human multiexon genes are alternatively spliced. Alterations in the splicing process cause missplicing events that lead to genetic diseases and pathologies, including various neurological disorders, cancers, and muscular dystrophies. In recent decades, research has helped to elucidate the mechanisms regulating alternative splicing and, in some cases, to reveal how dysregulation of these mechanisms leads to disease. The resulting knowledge has enabled the design of novel therapeutic strategies for correction of splicing-derived pathologies. In this review, we focus primarily on therapeutic approaches targeting splicing, and we highlight nanotechnology-based gene delivery applications that address the challenges and barriers facing nucleic acid-based therapeutics.
Collapse
Affiliation(s)
- Marc Suñé-Pou
- Drug Development Service (SDM), Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - María J Limeres
- Institute of Research in Microbiology and Medical Parasitology (IMPaM), Faculty of Medicine, University of Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Cristina Moreno-Castro
- Department of Molecular Biology, Institute of Parasitology and Biomedicine "López-Neyra" (IPBLN-CSIC), Granada, Spain
| | - Cristina Hernández-Munain
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine "López-Neyra" (IPBLN-CSIC), Granada, Spain
| | - Josep M Suñé-Negre
- Drug Development Service (SDM), Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - María L Cuestas
- Institute of Research in Microbiology and Medical Parasitology (IMPaM), Faculty of Medicine, University of Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Carlos Suñé
- Department of Molecular Biology, Institute of Parasitology and Biomedicine "López-Neyra" (IPBLN-CSIC), Granada, Spain
| |
Collapse
|
30
|
van Haasteren J, Li J, Scheideler OJ, Murthy N, Schaffer DV. The delivery challenge: fulfilling the promise of therapeutic genome editing. Nat Biotechnol 2020; 38:845-855. [PMID: 32601435 DOI: 10.1038/s41587-020-0565-5] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/19/2020] [Indexed: 12/15/2022]
Abstract
Genome editing has the potential to treat an extensive range of incurable monogenic and complex diseases. In particular, advances in sequence-specific nuclease technologies have dramatically accelerated the development of therapeutic genome editing strategies that are based on either the knockout of disease-causing genes or the repair of endogenous mutated genes. These technologies are progressing into human clinical trials. However, challenges remain before the therapeutic potential of genome editing can be fully realized. Delivery technologies that have serendipitously been developed over the past couple decades in the protein and nucleic acid delivery fields have been crucial to genome editing success to date, including adeno-associated viral and lentiviral vectors for gene therapy and lipid nanoparticle and other non-viral vectors for nucleic acid and protein delivery. However, the efficiency and tissue targeting capabilities of these vehicles must be further improved. In addition, the genome editing enzymes themselves need to be optimized, and challenges regarding their editing efficiency, specificity and immunogenicity must be addressed. Emerging protein engineering and synthetic chemistry approaches can offer solutions and enable the development of safe and efficacious clinical genome editing.
Collapse
Affiliation(s)
- Joost van Haasteren
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, USA
| | - Jie Li
- Department of Bioengineering, University of California, Berkeley, CA, USA.,Innovative Genomics Institute (IGI), University of California, Berkeley, CA, USA
| | | | - Niren Murthy
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, USA. .,Department of Bioengineering, University of California, Berkeley, CA, USA. .,Innovative Genomics Institute (IGI), University of California, Berkeley, CA, USA.
| | - David V Schaffer
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, USA. .,Department of Bioengineering, University of California, Berkeley, CA, USA. .,Innovative Genomics Institute (IGI), University of California, Berkeley, CA, USA. .,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA. .,Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA. .,Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA.
| |
Collapse
|
31
|
In Vivo Genome Engineering for the Treatment of Muscular Dystrophies. CURRENT STEM CELL REPORTS 2020. [DOI: 10.1007/s40778-020-00173-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
32
|
Datta N, Ghosh PS. Update on Muscular Dystrophies with Focus on Novel Treatments and Biomarkers. Curr Neurol Neurosci Rep 2020; 20:14. [DOI: 10.1007/s11910-020-01034-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
33
|
Previtali SC, Zambon AA. LAMA2 Neuropathies: Human Findings and Pathomechanisms From Mouse Models. Front Mol Neurosci 2020; 13:60. [PMID: 32390798 PMCID: PMC7190814 DOI: 10.3389/fnmol.2020.00060] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/26/2020] [Indexed: 12/18/2022] Open
Abstract
Merosin deficient Congenital Muscular Dystrophy (MDC1A), or LAMA2-related muscular dystrophy (LAMA2-RD), is a recessive disorder resulting from mutations in the LAMA2 gene, encoding for the alpha-2 chain of laminin-211. The disease is predominantly characterized by progressive muscular dystrophy affecting patient motor function and reducing life expectancy. However, LAMA2-RD also comprises a developmentally-associated dysmyelinating neuropathy that contributes to the disease progression, in addition to brain abnormalities; the latter often underappreciated. In this brief review, we present data supporting the impact of peripheral neuropathy in the LAMA2-RD phenotype, including both mouse models and human studies. We discuss the molecular mechanisms underlying nerve abnormalities and involved in the laminin-211 pathway, which affects axon sorting, ensheathing and myelination. We conclude with some final considerations of consequences on nerve regeneration and potential therapeutic strategies.
Collapse
Affiliation(s)
- Stefano Carlo Previtali
- Neuromuscular Repair Unit, Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy.,Department of Neurology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | |
Collapse
|
34
|
Gawlik KI, Durbeej M. A Family of Laminin α2 Chain-Deficient Mouse Mutants: Advancing the Research on LAMA2-CMD. Front Mol Neurosci 2020; 13:59. [PMID: 32457577 PMCID: PMC7188397 DOI: 10.3389/fnmol.2020.00059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
The research on laminin α2 chain-deficient congenital muscular dystrophy (LAMA2-CMD) advanced rapidly in the last few decades, largely due to availability of good mouse models for the disease and a strong interest in preclinical studies from scientists all over the world. These mouse models continue to provide a solid platform for understanding the LAMA2-CMD pathology. In addition, they enable researchers to test laborious, necessary routines, but also the most creative scientific approaches in order to design therapy for this devastating disorder. In this review we present animals belonging to the laminin α2 chain-deficient “dy/dy” mouse family (dy/dy, dy2J/dy2J, dy3K/dy3K, dyW/dyW, et al.) and a summary of the scientific progress they facilitated. We also raise a few questions that need to be addressed in order to maximize the usefulness of laminin α2 murine mutants and to further advance the LAMA2-CMD studies. We believe that research opportunities offered by the mouse models for LAMA2-CMD will continuously support our efforts to find a treatment for the disease.
Collapse
Affiliation(s)
- Kinga I Gawlik
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Madeleine Durbeej
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
35
|
Wang C, Sun W, Ling S, Wang Y, Wang X, Meng H, Li Y, Yuan X, Li J, Liu R, Zhao D, Lu Q, Wang A, Guo Q, Lu S, Tian H, Li Y, Peng J. AAV-Anti-miR-214 Prevents Collapse of the Femoral Head in Osteonecrosis by Regulating Osteoblast and Osteoclast Activities. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:841-850. [PMID: 31739209 PMCID: PMC6861671 DOI: 10.1016/j.omtn.2019.09.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 09/10/2019] [Accepted: 09/15/2019] [Indexed: 02/07/2023]
Abstract
Osteonecrosis of the femoral head, an intractable but common disease that eventually triggers collapse of the femoral head, is characterized by increased osteoclast activity and markedly decreased osteoblast activity in the necrotic region of the femoral head. MicroRNA (miRNA)-214 (miR-214) may play important roles in vertebrate skeletal development by inhibiting osteoblast function by targeting activating transcription factor 4 (ATF4) and promoting osteoclast function via phosphatase and tensin homolog (PTEN). This study revealed significantly increased levels of miR-214 in necrotic regions, with commensurate changes in the numbers of its target cells (both osteoblasts and osteoclasts). To investigate whether targeting miR-214 could prevent femoral head collapse, we constructed an adeno-associated virus (AAV)-associated anti-miR-214 (AAV-anti-miR-214) and evaluated its function in vivo. AAV-anti-miR-214 promoted osteoblast activity and diminished osteoclast activity, effectively preventing collapse of the femoral head in a rat model of osteonecrosis.
Collapse
Affiliation(s)
- Cheng Wang
- Institute of Orthopedics, Peking Key Lab of Regenerative Medicine in Orthopaedics, Key Lab of Chinese PLA, Chinese PLA General Hospital, Beijing, China; Department of Orthopedics, Peking University Third Hospital, Beijing, People's Republic of China
| | - Weijia Sun
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Shukuan Ling
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yu Wang
- Institute of Orthopedics, Peking Key Lab of Regenerative Medicine in Orthopaedics, Key Lab of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Xin Wang
- Institute of Orthopedics, Peking Key Lab of Regenerative Medicine in Orthopaedics, Key Lab of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Haoye Meng
- Institute of Orthopedics, Peking Key Lab of Regenerative Medicine in Orthopaedics, Key Lab of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Yuheng Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China; The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, Xi'an, Shanxi, China
| | - Xueling Yuan
- Institute of Orthopedics, Peking Key Lab of Regenerative Medicine in Orthopaedics, Key Lab of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Jianwei Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Ruoxi Liu
- Institute of Orthopedics, Peking Key Lab of Regenerative Medicine in Orthopaedics, Key Lab of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Dingsheng Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Qiang Lu
- Institute of Orthopedics, Peking Key Lab of Regenerative Medicine in Orthopaedics, Key Lab of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Aiyuan Wang
- Institute of Orthopedics, Peking Key Lab of Regenerative Medicine in Orthopaedics, Key Lab of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Quanyi Guo
- Institute of Orthopedics, Peking Key Lab of Regenerative Medicine in Orthopaedics, Key Lab of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Shibi Lu
- Institute of Orthopedics, Peking Key Lab of Regenerative Medicine in Orthopaedics, Key Lab of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Hua Tian
- Department of Orthopedics, Peking University Third Hospital, Beijing, People's Republic of China.
| | - Yingxian Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China.
| | - Jiang Peng
- Institute of Orthopedics, Peking Key Lab of Regenerative Medicine in Orthopaedics, Key Lab of Chinese PLA, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
36
|
Young CS, Pyle AD, Spencer MJ. CRISPR for Neuromuscular Disorders: Gene Editing and Beyond. Physiology (Bethesda) 2019; 34:341-353. [PMID: 31389773 PMCID: PMC6863376 DOI: 10.1152/physiol.00012.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 12/18/2022] Open
Abstract
This is a review describing advances in CRISPR/Cas-mediated therapies for neuromuscular disorders (NMDs). We explore both CRISPR-mediated editing and dead Cas approaches as potential therapeutic strategies for multiple NMDs. Last, therapeutic considerations, including delivery and off-target effects, are also discussed.
Collapse
Affiliation(s)
- Courtney S Young
- Department of Neurology, University of California, Los Angeles, California
- Center for Duchenne Muscular Dystrophy at UCLA, University of California, Los Angeles, California
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, University of California, Los Angeles, California
| | - April D Pyle
- Center for Duchenne Muscular Dystrophy at UCLA, University of California, Los Angeles, California
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, University of California, Los Angeles, California
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California
| | - Melissa J Spencer
- Department of Neurology, University of California, Los Angeles, California
- Center for Duchenne Muscular Dystrophy at UCLA, University of California, Los Angeles, California
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, University of California, Los Angeles, California
| |
Collapse
|
37
|
Kemaladewi DU, Bassi PS, Erwood S, Al-Basha D, Gawlik KI, Lindsay K, Hyatt E, Kember R, Place KM, Marks RM, Durbeej M, Prescott SA, Ivakine EA, Cohn RD. A mutation-independent approach for muscular dystrophy via upregulation of a modifier gene. Nature 2019; 572:125-130. [PMID: 31341277 DOI: 10.1038/s41586-019-1430-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/21/2019] [Indexed: 01/18/2023]
Abstract
Neuromuscular disorders are often caused by heterogeneous mutations in large, structurally complex genes. Targeting compensatory modifier genes could be beneficial to improve disease phenotypes. Here we report a mutation-independent strategy to upregulate the expression of a disease-modifying gene associated with congenital muscular dystrophy type 1A (MDC1A) using the CRISPR activation system in mice. MDC1A is caused by mutations in LAMA2 that lead to nonfunctional laminin-α2, which compromises the stability of muscle fibres and the myelination of peripheral nerves. Transgenic overexpression of Lama1, which encodes a structurally similar protein called laminin-α1, ameliorates muscle wasting and paralysis in mouse models of MDC1A, demonstrating its importance as a compensatory modifier of the disease1. However, postnatal upregulation of Lama1 is hampered by its large size, which exceeds the packaging capacity of vehicles that are clinically relevant for gene therapy. We modulate expression of Lama1 in the dy2j/dy2j mouse model of MDC1A using an adeno-associated virus (AAV9) carrying a catalytically inactive Cas9 (dCas9), VP64 transactivators and single-guide RNAs that target the Lama1 promoter. When pre-symptomatic mice were treated, Lama1 was upregulated in skeletal muscles and peripheral nerves, which prevented muscle fibrosis and paralysis. However, for many disorders it is important to investigate the therapeutic window and reversibility of symptoms. In muscular dystrophies, it has been hypothesized that fibrotic changes in skeletal muscle are irreversible. However, we show that dystrophic features and disease progression were improved and reversed when the treatment was initiated in symptomatic dy2j/dy2j mice with apparent hindlimb paralysis and muscle fibrosis. Collectively, our data demonstrate the feasibility and therapeutic benefit of CRISPR-dCas9-mediated upregulation of Lama1, which may enable mutation-independent treatment for all patients with MDC1A. This approach has a broad applicability to a variety of disease-modifying genes and could serve as a therapeutic strategy for many inherited and acquired diseases.
Collapse
Affiliation(s)
- Dwi U Kemaladewi
- Program in Genetics and Genome Biology, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Prabhpreet S Bassi
- Program in Genetics and Genome Biology, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Steven Erwood
- Program in Genetics and Genome Biology, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Dhekra Al-Basha
- Program in Neurosciences and Mental Health, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Kinga I Gawlik
- Unit of Muscle Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Kyle Lindsay
- Program in Genetics and Genome Biology, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Elzbieta Hyatt
- Program in Genetics and Genome Biology, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Rebekah Kember
- Program in Genetics and Genome Biology, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Kara M Place
- Program in Genetics and Genome Biology, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Ryan M Marks
- Program in Genetics and Genome Biology, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Madeleine Durbeej
- Unit of Muscle Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Steven A Prescott
- Program in Neurosciences and Mental Health, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Evgueni A Ivakine
- Program in Genetics and Genome Biology, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Ronald D Cohn
- Program in Genetics and Genome Biology, the Hospital for Sick Children Research Institute, Toronto, Ontario, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada. .,Department of Pediatrics, the Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
38
|
Çiçek YA, Luther DC, Kretzmann JA, Rotello VM. Advances in CRISPR/Cas9 Technology for in Vivo Translation. Biol Pharm Bull 2019; 42:304-311. [PMID: 30828060 DOI: 10.1248/bpb.b18-00811] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology has revolutionized therapeutic gene editing by providing researchers with a new method to study and cure diseases previously considered untreatable. While the full range and power of CRISPR technology for therapeutics is being elucidated through in vitro studies, translation to in vivo studies is slow. To date there is no totally effective delivery strategy to carry CRISPR components to the target site in vivo. The complexity of in vivo delivery is furthered by the number of potential delivery methods, the different forms in which CRISPR can be delivered as a therapeutic, and the disease target and tissue type in question. There are major challenges and limitations to delivery strategies, and it is imperative that future directions are guided by well-conducted studies that consider the full effect these variables have on the eventual outcome. In this review we will discuss the advances of the latest in vivo CRISPR/Cas9 delivery strategies and highlight the challenges yet to be overcome.
Collapse
Affiliation(s)
- Yağız Anıl Çiçek
- Department of Chemistry, Middle East Technical University (METU)
| | | | - Jessica A Kretzmann
- Department of Chemistry, University of Massachusetts.,School of Molecular Sciences, The University of Western Australia
| | | |
Collapse
|
39
|
Nguyen Q, Lim KRQ, Yokota T. Current understanding and treatment of cardiac and skeletal muscle pathology in laminin-α2 chain-deficient congenital muscular dystrophy. APPLICATION OF CLINICAL GENETICS 2019; 12:113-130. [PMID: 31308722 PMCID: PMC6618038 DOI: 10.2147/tacg.s187481] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/08/2019] [Indexed: 01/04/2023]
Abstract
Congenital muscular dystrophy (CMD) is a class of severe early-onset muscular dystrophies affecting skeletal/cardiac muscles as well as the central nervous system (CNS). Laminin-α2 chain-deficient congenital muscular dystrophy (LAMA2 MD), also known as merosin-deficient congenital muscular dystrophy type 1A (MDC1A), is an autosomal recessive CMD characterized by severe muscle weakness and degeneration apparent at birth or in the first 6 months of life. LAMA2 MD is the most common congenital muscular dystrophy, affecting approximately 4 in 500,000 children. The most common cause of death in early-onset LAMA2 MD is respiratory tract infection, with 30% of them dying within the first decade of life. LAMA2 MD is caused by loss-of-function mutations in the LAMA2 gene encoding for the laminin-α2 chain, one of the subunits of laminin-211. Laminin-211 is an extracellular matrix protein that functions to stabilize the basement membrane and muscle fibers during contraction. Since laminin-α2 is expressed in many tissue types including skeletal muscle, cardiac muscle, Schwann cells, and trophoblasts, patients with LAMA2 MD experience a multi-systemic clinical presentation depending on the extent of laminin-α2 chain deficiency. Cardiac manifestations are typically associated with a complete absence of laminin-α2; however, recent case reports highlight cardiac involvement in partial laminin-α2 chain deficiency. Laminin-211 is also expressed in the brain, and many patients have abnormalities on brain imaging; however, mental retardation and/or seizures are rarely seen. Currently, there is no cure for LAMA2 MD, but various therapies are being investigated in an effort to lessen the severity of LAMA2 MD. For example, antisense oligonucleotide-mediated exon skipping and CRISPR-Cas9 genome editing have efficiently restored the laminin-α2 chain in mouse models in vivo. This review consolidates information on the clinical presentation, genetic basis, pathology, and current treatment approaches for LAMA2 MD.
Collapse
Affiliation(s)
- Quynh Nguyen
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Kenji Rowel Q Lim
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,The Friends of Garrett Cumming Research & Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, AB, Canada
| |
Collapse
|
40
|
Ideno N, Yamaguchi H, Okumura T, Huang J, Brun MJ, Ho ML, Suh J, Gupta S, Maitra A, Ghosh B. A pipeline for rapidly generating genetically engineered mouse models of pancreatic cancer using in vivo CRISPR-Cas9-mediated somatic recombination. J Transl Med 2019; 99:1233-1244. [PMID: 30728464 DOI: 10.1038/s41374-018-0171-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/13/2018] [Accepted: 12/02/2018] [Indexed: 12/20/2022] Open
Abstract
Genetically engineered mouse models (GEMMs) that recapitulate the major genetic drivers in pancreatic ductal adenocarcinoma (PDAC) have provided unprecedented insights into the pathogenesis of this lethal neoplasm. Nonetheless, generating an autochthonous model is an expensive, time consuming and labor intensive process, particularly when tissue specific expression or deletion of compound alleles are involved. In addition, many of the current PDAC GEMMs cause embryonic, pancreas-wide activation or loss of driver alleles, neither of which reflects the cognate human disease scenario. The advent of CRISPR/Cas9 based gene editing can potentially circumvent many of the aforementioned shortcomings of conventional breeding schema, but ensuring the efficiency of gene editing in vivo remains a challenge. Here we have developed a pipeline for generating PDAC GEMMs of complex genotypes with high efficiency using a single "workhorse" mouse strain expressing Cas9 in the adult pancreas under a p48 promoter. Using adeno-associated virus (AAV) mediated delivery of multiplexed guide RNAs (sgRNAs) to the adult murine pancreas of p48-Cre; LSL-Cas9 mice, we confirm our ability to express an oncogenic Kras G12D allele through homology-directed repair (HDR), in conjunction with CRISPR-induced disruption of cooperating alleles (Trp53, Lkb1 and Arid1A). The resulting GEMMs demonstrate a spectrum of precursor lesions (pancreatic intraepithelial neoplasia [PanIN] or Intraductal papillary mucinous neoplasm [IPMN] with eventual progression to PDAC. Next generation sequencing of the resulting murine PDAC confirms HDR of oncogenic KrasG12D allele at the endogenous locus, and insertion deletion ("indel") and frameshift mutations of targeted tumor suppressor alleles. By using a single "workhorse" mouse strain and optimal AAV serotype for in vivo gene editing with combination of driver alleles, we present a facile autochthonous platform for interrogation of the PDAC genome.
Collapse
Affiliation(s)
- Noboru Ideno
- Department of Translational Molecular Pathology and Sheikh Ahmed Pancreatic Cancer Research Center, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Hiroshi Yamaguchi
- Department of Translational Molecular Pathology and Sheikh Ahmed Pancreatic Cancer Research Center, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Takashi Okumura
- Department of Translational Molecular Pathology and Sheikh Ahmed Pancreatic Cancer Research Center, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Jonathon Huang
- Department of Translational Molecular Pathology and Sheikh Ahmed Pancreatic Cancer Research Center, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Mitchell J Brun
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, USA
| | - Michelle L Ho
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Junghae Suh
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Sonal Gupta
- Department of Translational Molecular Pathology and Sheikh Ahmed Pancreatic Cancer Research Center, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology and Sheikh Ahmed Pancreatic Cancer Research Center, UT MD Anderson Cancer Center, Houston, TX, USA.
| | - Bidyut Ghosh
- Department of Translational Molecular Pathology and Sheikh Ahmed Pancreatic Cancer Research Center, UT MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
41
|
Zhan H, Zhou Q, Gao Q, Li J, Huang W, Liu Y. Multiplexed promoterless gene expression with CRISPReader. Genome Biol 2019; 20:113. [PMID: 31159834 PMCID: PMC6545682 DOI: 10.1186/s13059-019-1712-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/08/2019] [Indexed: 02/06/2023] Open
Abstract
Background Genes are comprised of DNA codes and contain promoters and other control elements for reading these codes. The rapid development of clustered regularly interspaced short palindromic repeats (CRISPR) technology has made possible the construction of a novel code-reading system with low dependency on the native control elements. Results We develop CRISPReader, a technology for controlling promoterless gene expression in a robust fashion. We demonstrate that this tool is highly efficient in controlling transcription and translation initiation of a targeted transgene. A notable feature of CRISPReader is the ability to “read” the open reading frames of a cluster of gene without traditional regulatory elements or other cofactors. In particular, we use this strategy to construct an all-in-one AAV-CRISPR-Cas9 system by removing promoter-like elements from the expression cassette to resolve the existing AAV packaging size problem. The compact AAV-CRISPR-Cas9 is also more efficient in transactivation, DNA cleavage, and gene editing than the dual-AAV vector encoding two separate Cas9 elements, shown by targeting both reporter and endogenous genes in vitro and in vivo. Conclusions CRISPReader represents a novel approach for gene regulation that enables minimal gene constructs to be expressed and can be used in potential biomedical applications. Electronic supplementary material The online version of this article (10.1186/s13059-019-1712-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hengji Zhan
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Qun Zhou
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Qunjun Gao
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Jianfa Li
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Weiren Huang
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China. .,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| | - Yuchen Liu
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China. .,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| |
Collapse
|
42
|
Therapeutic application of the CRISPR system: current issues and new prospects. Hum Genet 2019; 138:563-590. [PMID: 31115652 DOI: 10.1007/s00439-019-02028-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 05/13/2019] [Indexed: 12/23/2022]
|
43
|
Kölbel H, Hathazi D, Jennings M, Horvath R, Roos A, Schara U. Identification of Candidate Protein Markers in Skeletal Muscle of Laminin-211-Deficient CMD Type 1A-Patients. Front Neurol 2019; 10:470. [PMID: 31133972 PMCID: PMC6514157 DOI: 10.3389/fneur.2019.00470] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/17/2019] [Indexed: 12/11/2022] Open
Abstract
Laminin-211 deficiency leads to the most common form of congenital muscular dystrophy in childhood, MDC1A. The clinical picture is characterized by severe muscle weakness, brain abnormalities and delayed motor milestones defining MDC1A as one of the most severe forms of congenital muscular diseases. Although the molecular genetic basis of this neurological disease is well-known and molecular studies of mouse muscle and human cultured muscle cells allowed first insights into the underlying pathophysiology, the definition of marker proteins in human vulnerable tissue such as skeletal muscle is still lacking. To systematically address this need, we analyzed the proteomic signature of laminin-211-deficient vastus muscle derived from four patients and identified 86 proteins (35 were increased and 51 decreased) as skeletal muscle markers and verified paradigmatic findings in a total of two further MDC1A muscle biopsies. Functions of proteins suggests fibrosis but also hints at altered synaptic transmission and accords with central nervous system alterations as part of the clinical spectrum of MDC1A. In addition, a profound mitochondrial vulnerability of the laminin-211-deficient muscle is indicated and also altered abundances of other proteins support the concept that metabolic alterations could be novel mechanisms that underline MDC1A and might constitute therapeutic targets. Intersection of our data with the proteomic signature of murine laminin-211-deficient gastrocnemius and diaphragm allowed the definition of nine common vulnerable proteins representing potential tissue markers.
Collapse
Affiliation(s)
- Heike Kölbel
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, University of Duisburg-Essen, Essen, Germany
| | - Denisa Hathazi
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany.,Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Matthew Jennings
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Rita Horvath
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Andreas Roos
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, University of Duisburg-Essen, Essen, Germany.,Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany
| | - Ulrike Schara
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
44
|
Development of therapeutic genome engineering in laminin-α2-deficient congenital muscular dystrophy. Emerg Top Life Sci 2019; 3:11-18. [PMID: 33523194 DOI: 10.1042/etls20180059] [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: 01/21/2019] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 11/17/2022]
Abstract
Muscular dystrophies are a heterogeneous group of genetic muscle diseases that are often characterized by pathological findings of muscle fiber degeneration and the replacement of muscle fibers with fibrotic/connective tissues. In spite of the genetic causes of many of these conditions having been identified, curative treatments are still lacking. Recently, genome engineering technologies, including targeted gene editing and gene regulation, have emerged as attractive therapeutic tools for a variety of muscular dystrophies. This review summarizes the genome engineering strategies that are currently under preclinical evaluation for the treatment of LAMA2-deficient congenital muscular dystrophy. In particular, we focus on the applications of CRISPR/Cas9 to correct a splice site mutation in LAMA2 and to up-regulate a disease-modifying gene LAMA1. Finally, the challenges faced in the clinical translation of these strategies are discussed.
Collapse
|
45
|
He X, Xie H, Liu X, Gu F. Basic and Clinical Application of Adeno-Associated Virus-Mediated Genome Editing. Hum Gene Ther 2019; 30:673-681. [PMID: 30588843 DOI: 10.1089/hum.2018.190] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Traditional gene therapy (gene replacement) has made a breakthrough in treating inherited diseases. Adeno-associated virus (AAV) has emerged as a highly promising vector with innate ability, boosting the development of gene replacement and gene targeting. With the recent advance of engineered nucleases that work efficiently in human cells, AAV mediated-genome editing with nucleases has raised hopes for in situ gene therapy of inherited and non-inherited diseases. Here, the applications of AAV-mediated genome editing are highlighted, and the prospect of AAV and nucleases that will render extension of such success in clinical gene therapy is discussed.
Collapse
Affiliation(s)
- Xiubin He
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, P.R. China
| | - Haihua Xie
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, P.R. China
| | - Xiexie Liu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, P.R. China
| | - Feng Gu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, P.R. China
| |
Collapse
|
46
|
Babačić H, Mehta A, Merkel O, Schoser B. CRISPR-cas gene-editing as plausible treatment of neuromuscular and nucleotide-repeat-expansion diseases: A systematic review. PLoS One 2019; 14:e0212198. [PMID: 30794581 PMCID: PMC6386526 DOI: 10.1371/journal.pone.0212198] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/29/2019] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION The system of clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated proteins (cas) is a new technology that allows easier manipulation of the genome. Its potential to edit genes opened a new door in treatment development for incurable neurological monogenic diseases (NMGDs). The aim of this systematic review was to summarise the findings on the current development of CRISPR-cas for therapeutic purposes in the most frequent NMGDs and provide critical assessment. METHODS AND DATA ACQUISITION We searched the MEDLINE and EMBASE databases, looking for original studies on the use of CRISPR-cas to edit pathogenic variants in models of the most frequent NMGDs, until end of 2017. We included all the studies that met the following criteria: 1. Peer-reviewed study report with explicitly described experimental designs; 2. In vitro, ex vivo, or in vivo study using human or other animal biological systems (including cells, tissues, organs, organisms); 3. focusing on CRISPR as the gene-editing method of choice; and 5. featured at least one NMGD. RESULTS We obtained 404 papers from MEDLINE and 513 from EMBASE. After removing the duplicates, we screened 490 papers by title and abstract and assessed them for eligibility. After reading 50 full-text papers, we finally selected 42 for the review. DISCUSSION Here we give a systematic summary on the preclinical development of CRISPR-cas for therapeutic purposes in NMGDs. Furthermore, we address the clinical interpretability of the findings, giving a comprehensive overview of the current state of the art. Duchenne's muscular dystrophy (DMD) paves the way forward, with 26 out of 42 studies reporting different strategies on DMD gene editing in different models of the disease. Most of the strategies aimed for permanent exon skipping by deletion with CRISPR-cas. Successful silencing of the mHTT gene with CRISPR-cas led to successful reversal of the neurotoxic effects in the striatum of mouse models of Huntington's disease. Many other strategies have been explored, including epigenetic regulation of gene expression, in cellular and animal models of: myotonic dystrophy, Fraxile X syndrome, ataxias, and other less frequent dystrophies. Still, before even considering the clinical application of CRISPR-cas, three major bottlenecks need to be addressed: efficacy, safety, and delivery of the systems. This requires a collaborative approach in the research community, while having ethical considerations in mind.
Collapse
Affiliation(s)
- Haris Babačić
- Friedrich Baur Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
- * E-mail: (BS); (HB)
| | - Aditi Mehta
- Faculty of Pharmacy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Olivia Merkel
- Faculty of Pharmacy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Benedikt Schoser
- Friedrich Baur Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
- * E-mail: (BS); (HB)
| |
Collapse
|
47
|
Montes M, Sanford BL, Comiskey DF, Chandler DS. RNA Splicing and Disease: Animal Models to Therapies. Trends Genet 2019; 35:68-87. [PMID: 30466729 PMCID: PMC6339821 DOI: 10.1016/j.tig.2018.10.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/01/2018] [Accepted: 10/16/2018] [Indexed: 02/07/2023]
Abstract
Alternative splicing of pre-mRNA increases genetic diversity, and recent studies estimate that most human multiexon genes are alternatively spliced. If this process is not highly regulated and accurate, it leads to mis-splicing events, which may result in proteins with altered function. A growing body of work has implicated mis-splicing events in a range of diseases, including cancer, neurodegenerative diseases, and muscular dystrophies. Understanding the mechanisms that cause aberrant splicing events and how this leads to disease is vital for designing effective therapeutic strategies. In this review, we focus on advances in therapies targeting splicing, and highlight the animal models developed to recapitulate disease phenotypes as a model for testing these therapies.
Collapse
Affiliation(s)
- Matías Montes
- Molecular, Cellular, and Developmental Biology Graduate Program and The Center for RNA Biology, The Ohio State University, Columbus, OH, USA; Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Brianne L Sanford
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Daniel F Comiskey
- Molecular, Cellular, and Developmental Biology Graduate Program and The Center for RNA Biology, The Ohio State University, Columbus, OH, USA; Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Dawn S Chandler
- Molecular, Cellular, and Developmental Biology Graduate Program and The Center for RNA Biology, The Ohio State University, Columbus, OH, USA; Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
48
|
Abstract
CRISPR gene editing is poised to transform the therapeutic landscape for diseases of genetic origin. The ease and agility by which CRISPR can make specific changes to DNA holds great promise not only for the treatment of heritable diseases, but also their prevention through germline editing. CRISPR-based therapeutic strategies are currently under development for numerous monogenic diseases. These strategies range from proof of concept studies demonstrating pre-fertilization gamete editing to recently initiated clinical trials for postnatal ex vivo therapies. The promise of CRISPR's human genome editing potential has captivated the public's attention. It is of paramount importance that medical professionals who work with patients who may have or carry a monogenic heritable disease understand CRISPR technology in order to have informed and compassionate discussions with their patients. Understanding CRISPR means understanding its evolving therapeutic applications' nuances, limitations, and barriers to access as well as the regulatory landscape they inhabit. In this piece we provide a review of the promises and pitfalls of CRISPR germline gene editing and their implications for patient decision-making throughout various stages of the reproductive process.
Collapse
Affiliation(s)
- Natalie Kofler
- Founder, Editing Nature and Associate Research Scientist, Yale Institute of Biospheric Studies, Yale Interdisciplinary Center for Bioethics, 21 Sachem St., New Haven, CT 06511, United States.
| | - Katherine L Kraschel
- Executive Director, Solomon Center for Health Law and Policy; Lecturer in Law; and Research Scholar, Yale Law School, 127 Wall Street, New Haven, CT 06511, United States
| |
Collapse
|
49
|
Xia E, Duan R, Shi F, Seigel KE, Grasemann H, Hu J. Overcoming the Undesirable CRISPR-Cas9 Expression in Gene Correction. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 13:699-709. [PMID: 30513454 PMCID: PMC6278715 DOI: 10.1016/j.omtn.2018.10.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 10/25/2018] [Accepted: 10/25/2018] [Indexed: 01/06/2023]
Abstract
The CRISPR-Cas9 system is attractive for gene therapy, as it allows for permanent genetic correction. However, as a new technology, Cas9 gene editing in clinical applications faces major challenges, such as safe delivery and gene targeting efficiency. Cas9 is a foreign protein to recipient cells; thus, its expression may prompt the immune system to eliminate gene-edited cells. To overcome these challenges, we have engineered a novel delivery system based on the helper-dependent adenoviral (HD-Ad) vector, which is capable of delivering genes to airway basal stem cells in vivo. Using this system, we demonstrate the successful co-delivery of both CRISPR-Cas9/single-guide RNA and the LacZ reporter or CFTR gene as donor DNA to cultured cells. HD-Ad vector genome integrity is compromised following donor DNA integration, and because the CRISPR-Cas9/single-guide RNA and donor DNA are carried on the same vector, CRISPR-Cas9 expression is concurrently eliminated. Thus, we show the feasibility of site-specific gene targeting with limited Cas9 expression. In addition, we achieved stable CFTR expression and functional correction in cultured cells following successful gene integration.
Collapse
Affiliation(s)
- Emily Xia
- Translational Medicine, Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Rongqi Duan
- Translational Medicine, Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Fushan Shi
- Translational Medicine, Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Kyle E Seigel
- Translational Medicine, Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Hartmut Grasemann
- Translational Medicine, Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Paediatrics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Jim Hu
- Translational Medicine, Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Paediatrics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
50
|
Abstract
Pompe disease is a rare and deadly muscle disorder. As a clinical entity, the disease has been known for over 75 years. While an optimist might be excited about the advances made during this time, a pessimist would note that we have yet to find a cure. However, both sides would agree that many findings in basic science-such as the Nobel prize-winning discoveries of glycogen metabolism, the lysosome, and autophagy-have become the foundation of our understanding of Pompe disease. The disease is a glycogen storage disorder, a lysosomal disorder, and an autophagic myopathy. In this review, we will discuss how these past discoveries have guided Pompe research and impacted recent therapeutic developments.
Collapse
Affiliation(s)
- Lara Kohler
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Nina Raben
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|