Try a new search

Format these results:

Searched for:

person:longc08

in-biosketch:yes

Total Results:

21


PINK1 gene mutation by pair truncated sgRNA/Cas9-D10A in cynomolgus monkeys

Chen, Zhen-Zhen; Wang, Jian-Ying; Kang, Yu; Yang, Qiao-Yan; Gu, Xue-Ying; Zhi, Da-Long; Yan, Li; Long, Cheng-Zu; Shen, Bin; Niu, Yu-Yu
Mutations of PTEN-induced kinase I (PINK1) cause early-onset Parkinson's disease (PD) with selective neurodegeneration in humans. However, current PINK1 knockout mouse and pig models are unable to recapitulate the typical neurodegenerative phenotypes observed in PD patients. This suggests that generating PINK1 disease models in non-human primates (NHPs) that are close to humans is essential to investigate the unique function of PINK1 in primate brains. Paired single guide RNA (sgRNA)/Cas9-D10A nickases and truncated sgRNA/Cas9, both of which can reduce off-target effects without compromising on-target editing, are two optimized strategies in the CRISPR/Cas9 system for establishing disease animal models. Here, we combined the two strategies and injected Cas9-D10A mRNA and two truncated sgRNAs into one-cell-stage cynomolgus zygotes to target the PINK1 gene. We achieved precise and efficient gene editing of the target site in three newborn cynomolgus monkeys. The frame shift mutations of PINK1 in mutant fibroblasts led to a reduction in mRNA. However, western blotting and immunofluorescence staining confirmed the PINK1 protein levels were comparable to that in wild-type fibroblasts. We further reprogramed mutant fibroblasts into induced pluripotent stem cells (iPSCs), which showed similar ability to differentiate into dopamine (DA) neurons. Taken together, our results showed that co-injection of Cas9-D10A nickase mRNA and sgRNA into one-cell-stage cynomolgus embryos enabled the generation of human disease models in NHPs and target editing by pair truncated sgRNA/Cas9-D10A in PINK1 gene exon 2 did not impact protein expression.
PMCID:8317192
PMID: 34213093
ISSN: 2095-8137
CID: 4965112

Progress and challenges in CRISPR-mediated therapeutic genome editing for monogenic diseases

Konishi, Colin T; Long, Chengzu
There are an estimated 10 000 monogenic diseases affecting tens of millions of individuals worldwide. The application of CRISPR/Cas genome editing tools to treat monogenic diseases is an emerging strategy with the potential to generate personalized treatment approaches for these patients. CRISPR/Cas-based systems are programmable and sequence-specific genome editing tools with the capacity to generate base pair resolution manipulations to DNA or RNA. The complexity of genomic insults resulting in heritable disease requires patient-specific genome editing strategies with consideration of DNA repair pathways, and CRISPR/Cas systems of different types, species, and those with additional enzymatic capacity and/or delivery methods. In this review we aim to discuss broad and multifaceted therapeutic applications of CRISPR/Cas gene editing systems including in harnessing of homology directed repair, non-homologous end joining, microhomology-mediated end joining, and base editing to permanently correct diverse monogenic diseases.
PMCID:8038532
PMID: 33402545
ISSN: 1674-8301
CID: 4897882

Generation of a Hutchinson-Gilford progeria syndrome monkey model by base editing

Wang, Fang; Zhang, Weiqi; Yang, Qiaoyan; Kang, Yu; Fan, Yanling; Wei, Jingkuan; Liu, Zunpeng; Dai, Shaoxing; Li, Hao; Li, Zifan; Xu, Lizhu; Chu, Chu; Qu, Jing; Si, Chenyang; Ji, Weizhi; Liu, Guang-Hui; Long, Chengzu; Niu, Yuyu
Many human genetic diseases, including Hutchinson-Gilford progeria syndrome (HGPS), are caused by single point mutations. HGPS is a rare disorder that causes premature aging and is usually caused by a de novo point mutation in the LMNA gene. Base editors (BEs) composed of a cytidine deaminase fused to CRISPR/Cas9 nickase are highly efficient at inducing C to T base conversions in a programmable manner and can be used to generate animal disease models with single amino-acid substitutions. Here, we generated the first HGPS monkey model by delivering a BE mRNA and guide RNA (gRNA) targeting the LMNA gene via microinjection into monkey zygotes. Five out of six newborn monkeys carried the mutation specifically at the target site. HGPS monkeys expressed the toxic form of lamin A, progerin, and recapitulated the typical HGPS phenotypes including growth retardation, bone alterations, and vascular abnormalities. Thus, this monkey model genetically and clinically mimics HGPS in humans, demonstrating that the BE system can efficiently and accurately generate patient-specific disease models in non-human primates.
PMID: 32729022
ISSN: 1674-8018
CID: 4614972

In vivo non-invasive monitoring of dystrophin correction in a new Duchenne muscular dystrophy reporter mouse

Amoasii, Leonela; Li, Hui; Zhang, Yu; Min, Yi-Li; Sanchez-Ortiz, Efrain; Shelton, John M; Long, Chengzu; Mireault, Alex A; Bhattacharyya, Samadrita; McAnally, John R; Bassel-Duby, Rhonda; Olson, Eric N
Duchenne muscular dystrophy (DMD) is a fatal genetic disorder caused by mutations in the dystrophin gene. To enable the non-invasive analysis of DMD gene correction strategies in vivo, we introduced a luciferase reporter in-frame with the C-terminus of the dystrophin gene in mice. Expression of this reporter mimics endogenous dystrophin expression and DMD mutations that disrupt the dystrophin open reading frame extinguish luciferase expression. We evaluated the correction of the dystrophin reading frame coupled to luciferase in mice lacking exon 50, a common mutational hotspot, after delivery of CRISPR/Cas9 gene editing machinery with adeno-associated virus. Bioluminescence monitoring revealed efficient and rapid restoration of dystrophin protein expression in affected skeletal muscles and the heart. Our results provide a sensitive non-invasive means of monitoring dystrophin correction in mouse models of DMD and offer a platform for testing different strategies for amelioration of DMD pathogenesis.
PMID: 31586095
ISSN: 2041-1723
CID: 4116562

Modeling heart failure for therapeutic screens in engineered human myocardium [Meeting Abstract]

Tiburcy, Malte; Meyer, Tim; Long, Chengzu; Olson, Eric N.; Zimmermann, Wolfram H.
ISI:000444523000189
ISSN: 1056-8719
CID: 3319012

Myoediting: Toward Prevention of Muscular Dystrophy by Therapeutic Genome Editing

Zhang, Yu; Long, Chengzu; Bassel-Duby, Rhonda; Olson, Eric N
Muscular dystrophies represent a large group of genetic disorders that significantly impair quality of life and often progress to premature death. There is no effective treatment for these debilitating diseases. Most therapies, developed to date, focus on alleviating the symptoms or targeting the secondary effects, while the underlying gene mutation is still present in the human genome. The discovery and application of programmable nucleases for site-specific DNA double-stranded breaks provides a powerful tool for precise genome engineering. In particular, the CRISPR/Cas system has revolutionized the genome editing field and is providing a new path for disease treatment by targeting the disease-causing genetic mutations. In this review, we provide a historical overview of genome-editing technologies, summarize the most recent advances, and discuss potential strategies and challenges for permanently correcting genetic mutations that cause muscular dystrophies.
PMID: 29717930
ISSN: 1522-1210
CID: 3123962

Correction of diverse muscular dystrophy mutations in human engineered heart muscle by single-site genome editing

Long, Chengzu; Li, Hui; Tiburcy, Malte; Rodriguez-Caycedo, Cristina; Kyrychenko, Viktoriia; Zhou, Huanyu; Zhang, Yu; Min, Yi-Li; Shelton, John M; Mammen, Pradeep P A; Liaw, Norman Y; Zimmermann, Wolfram-Hubertus; Bassel-Duby, Rhonda; Schneider, Jay W; Olson, Eric N
Genome editing with CRISPR/Cas9 is a promising new approach for correcting or mitigating disease-causing mutations. Duchenne muscular dystrophy (DMD) is associated with lethal degeneration of cardiac and skeletal muscle caused by more than 3000 different mutations in the X-linked dystrophin gene (DMD). Most of these mutations are clustered in "hotspots." There is a fortuitous correspondence between the eukaryotic splice acceptor and splice donor sequences and the protospacer adjacent motif sequences that govern prokaryotic CRISPR/Cas9 target gene recognition and cleavage. Taking advantage of this correspondence, we screened for optimal guide RNAs capable of introducing insertion/deletion (indel) mutations by nonhomologous end joining that abolish conserved RNA splice sites in 12 exons that potentially allow skipping of the most common mutant or out-of-frame DMD exons within or nearby mutational hotspots. We refer to the correction of DMD mutations by exon skipping as myoediting. In proof-of-concept studies, we performed myoediting in representative induced pluripotent stem cells from multiple patients with large deletions, point mutations, or duplications within the DMD gene and efficiently restored dystrophin protein expression in derivative cardiomyocytes. In three-dimensional engineered heart muscle (EHM), myoediting of DMD mutations restored dystrophin expression and the corresponding mechanical force of contraction. Correcting only a subset of cardiomyocytes (30 to 50%) was sufficient to rescue the mutant EHM phenotype to near-normal control levels. We conclude that abolishing conserved RNA splicing acceptor/donor sites and directing the splicing machinery to skip mutant or out-of-frame exons through myoediting allow correction of the cardiac abnormalities associated with DMD by eliminating the underlying genetic basis of the disease.
PMCID:5796795
PMID: 29404407
ISSN: 2375-2548
CID: 2947522

Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy

Long, Chengzu; Amoasii, Leonela; Mireault, Alex A; McAnally, John R; Li, Hui; Sanchez-Ortiz, Efrain; Bhattacharyya, Samadrita; Shelton, John M; Bassel-Duby, Rhonda; Olson, Eric N
CRISPR/Cas9-mediated genome editing holds clinical potential for treating genetic diseases, such as Duchenne muscular dystrophy (DMD), which is caused by mutations in the dystrophin gene. To correct DMD by skipping mutant dystrophin exons in postnatal muscle tissue in vivo, we used adeno-associated virus-9 (AAV9) to deliver gene-editing components to postnatal mdx mice, a model of DMD. Different modes of AAV9 delivery were systematically tested, including intraperitoneal at postnatal day 1 (P1), intramuscular at P12, and retro-orbital at P18. Each of these methods restored dystrophin protein expression in cardiac and skeletal muscle to varying degrees, and expression increased from 3 to 12 weeks after injection. Postnatal gene editing also enhanced skeletal muscle function, as measured by grip strength tests 4 weeks after injection. This method provides a potential means of correcting mutations responsible for DMD and other monogenic disorders after birth.
PMCID:4760628
PMID: 26721683
ISSN: 1095-9203
CID: 2271092

Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline DNA

Long, Chengzu; McAnally, John R; Shelton, John M; Mireault, Alex A; Bassel-Duby, Rhonda; Olson, Eric N
Duchenne muscular dystrophy (DMD) is an inherited X-linked disease caused by mutations in the gene encoding dystrophin, a protein required for muscle fiber integrity. DMD is characterized by progressive muscle weakness and a shortened life span, and there is no effective treatment. We used clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9)-mediated genome editing to correct the dystrophin gene (Dmd) mutation in the germ line of mdx mice, a model for DMD, and then monitored muscle structure and function. Genome editing produced genetically mosaic animals containing 2 to 100% correction of the Dmd gene. The degree of muscle phenotypic rescue in mosaic mice exceeded the efficiency of gene correction, likely reflecting an advantage of the corrected cells and their contribution to regenerating muscle. With the anticipated technological advances that will facilitate genome editing of postnatal somatic cells, this strategy may one day allow correction of disease-causing mutations in the muscle tissue of patients with DMD.
PMCID:4398027
PMID: 25123483
ISSN: 1095-9203
CID: 2271082

Ataxia and Purkinje cell degeneration in mice lacking the CAMTA1 transcription factor

Long, Chengzu; Grueter, Chad E; Song, Kunhua; Qin, Song; Qi, Xiaoxia; Kong, Y Megan; Shelton, John M; Richardson, James A; Zhang, Chun-Li; Bassel-Duby, Rhonda; Olson, Eric N
Members of the calmodulin-binding transcription activator (CAMTA) family of proteins function as calcium-sensitive regulators of gene expression in multicellular organisms ranging from plants to humans. Here, we show that global or nervous system deletion of CAMTA1 in mice causes severe ataxia with Purkinje cell degeneration and cerebellar atrophy, partially resembling the consequences of haploinsufficiency of the human CAMTA1 locus. Gene-expression analysis identified a large collection of neuronal genes that were dysregulated in the brains of CAMTA1-mutant mice, and elucidation of a consensus sequence for binding of CAMTA proteins to DNA revealed the association of CAMTA-binding sites with many of these genes. We conclude that CAMTA1 plays an essential role in the control of Purkinje cell function and survival. CAMTA1-mutant mice provide a model to study the molecular mechanisms of neurodegenerative diseases and for screening potential therapeutic interventions for such disorders.
PMCID:4128133
PMID: 25049392
ISSN: 1091-6490
CID: 2271072