Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Stem Cell ; 30(11): 1538-1548.e4, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37922880

ABSTRACT

Immune rejection of allogeneic cell therapeutics remains a major problem for immuno-oncology and regenerative medicine. Allogeneic cell products so far have inferior persistence and efficacy when compared with autologous alternatives. Engineering of hypoimmune cells may greatly improve their therapeutic benefit. We present a new class of agonistic immune checkpoint engagers that protect human leukocyte antigen (HLA)-depleted induced pluripotent stem cell-derived endothelial cells (iECs) from innate immune cells. Engagers with agonistic functionality to their inhibitory receptors TIM3 and SIRPα effectively protect engineered iECs from natural killer (NK) cell and macrophage killing. The SIRPα engager can be combined with truncated CD64 to generate fully immune evasive iECs capable of escaping allogeneic cellular and immunoglobulin G (IgG) antibody-mediated rejection. Synthetic immune checkpoint engagers have high target specificity and lack retrograde signaling in the engineered cells. This modular design allows for the exploitation of more inhibitory immune pathways for immune evasion and could contribute to the advancement of allogeneic cell therapeutics.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Endothelial Cells/metabolism , HLA Antigens , Killer Cells, Natural , Immunity, Innate
2.
Nat Commun ; 12(1): 7101, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34880218

ABSTRACT

Genome editing therapy for Duchenne muscular dystrophy (DMD) holds great promise, however, one major obstacle is delivery of the CRISPR-Cas9/sgRNA system to skeletal muscle tissues. In general, AAV vectors are used for in vivo delivery, but AAV injections cannot be repeated because of neutralization antibodies. Here we report a chemically defined lipid nanoparticle (LNP) system which is able to deliver Cas9 mRNA and sgRNA into skeletal muscle by repeated intramuscular injections. Although the expressions of Cas9 protein and sgRNA were transient, our LNP system could induce stable genomic exon skipping and restore dystrophin protein in a DMD mouse model that harbors a humanized exon sequence. Furthermore, administration of our LNP via limb perfusion method enables to target multiple muscle groups. The repeated administration and low immunogenicity of our LNP system are promising features for a delivery vehicle of CRISPR-Cas9 to treat skeletal muscle disorders.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Muscle, Skeletal/metabolism , RNA, Messenger , Animals , CRISPR-Associated Protein 9 , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Exons , Genetic Therapy , Humans , Liposomes , Mice , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Nanoparticles , Neuromuscular Diseases/genetics , Neuromuscular Diseases/therapy
3.
Nat Commun ; 11(1): 1334, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32170079

ABSTRACT

Prolonged expression of the CRISPR-Cas9 nuclease and gRNA from viral vectors may cause off-target mutagenesis and immunogenicity. Thus, a transient delivery system is needed for therapeutic genome editing applications. Here, we develop an extracellular nanovesicle-based ribonucleoprotein delivery system named NanoMEDIC by utilizing two distinct homing mechanisms. Chemical induced dimerization recruits Cas9 protein into extracellular nanovesicles, and then a viral RNA packaging signal and two self-cleaving riboswitches tether and release sgRNA into nanovesicles. We demonstrate efficient genome editing in various hard-to-transfect cell types, including human induced pluripotent stem (iPS) cells, neurons, and myoblasts. NanoMEDIC also achieves over 90% exon skipping efficiencies in skeletal muscle cells derived from Duchenne muscular dystrophy (DMD) patient iPS cells. Finally, single intramuscular injection of NanoMEDIC induces permanent genomic exon skipping in a luciferase reporter mouse and in mdx mice, indicating its utility for in vivo genome editing therapy of DMD and beyond.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Exons/genetics , Extracellular Vesicles/metabolism , Nanoparticles/chemistry , RNA, Guide, Kinetoplastida/metabolism , Base Sequence , Cell Survival , Dimerization , Gene Editing , Genetic Vectors/metabolism , HEK293 Cells , HIV Protease/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Ligands , Luciferases/metabolism , RNA Splicing/genetics , RNA, Catalytic/metabolism , Ribonucleoproteins/metabolism , Tissue Donors , tat Gene Products, Human Immunodeficiency Virus/metabolism
4.
Methods Mol Biol ; 1828: 191-217, 2018.
Article in English | MEDLINE | ID: mdl-30171543

ABSTRACT

Duchenne muscular dystrophy (DMD) is a congenital X-linked disease caused by mutations in the gene encoding the dystrophin protein, which is required for myofiber integrity. Exon skipping therapy is an emerging strategy for restoring the open reading frame of the dystrophin gene to produce functional protein in DMD patients by skipping single or multiple exons. Although antisense oligonucleotides are able to target pre-mRNA for exon skipping, their half-lives are short and any therapeutic benefit is transient. In contrast, genome editing by DNA nucleases, such as the CRISPR-Cas9 system, could offer permanent correction by targeting genomic DNA. Our laboratory previously reported that disrupting the splicing acceptor site in exon 45 by plasmid delivery of the CRISPR-Cas9 system in iPS cells, derived from a DMD patient lacking exon 44, successfully restored dystrophin protein expression in differentiated myoblasts. Herein, we describe an optimized methodology to prepare myoblasts differentiated from iPS cells by mRNA transfection of the CRISPR-Cas9 system to skip exon 45 in myoblasts, and evaluate the restored dystrophin by RT-PCR and Western blotting.


Subject(s)
CRISPR-Cas Systems , Dystrophin/genetics , Exons , Gene Editing , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Muscular Dystrophy, Duchenne/genetics , Myoblasts/metabolism , Alternative Splicing , Cell Culture Techniques , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Differentiation/genetics , Cells, Cultured , Computational Biology/methods , Dystrophin/metabolism , Gene Expression Regulation , Humans , Mitomycin/pharmacology , Muscle Development , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Mutation , MyoD Protein/genetics , Myoblasts/cytology , RNA, Guide, Kinetoplastida , RNA, Messenger/genetics , Transduction, Genetic
5.
Cell Stem Cell ; 23(3): 343-354.e5, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30033120

ABSTRACT

Animal cloning can be achieved through somatic cell nuclear transfer (SCNT), although the live birth rate is relatively low. Recent studies have identified H3K9me3 in donor cells and abnormal Xist activation as epigenetic barriers that impede SCNT. Here we overcome these barriers using a combination of Xist knockout donor cells and overexpression of Kdm4 to achieve more than 20% efficiency of mouse SCNT. However, post-implantation defects and abnormal placentas were still observed, indicating that additional epigenetic barriers impede SCNT cloning. Comparative DNA methylome analysis of IVF and SCNT blastocysts identified abnormally methylated regions in SCNT embryos despite successful global reprogramming of the methylome. Strikingly, allelic transcriptomic and ChIP-seq analyses of pre-implantation SCNT embryos revealed complete loss of H3K27me3 imprinting, which may account for the postnatal developmental defects observed in SCNT embryos. Together, these results provide an efficient method for mouse cloning while paving the way for further improving SCNT efficiency.


Subject(s)
Embryo Implantation/genetics , Embryo, Mammalian/metabolism , Genomic Imprinting , Histones/metabolism , Nuclear Transfer Techniques , Animals , Embryo, Mammalian/embryology , Female , Male , Mice , Mice, Inbred Strains , Mice, Knockout
6.
Cell ; 159(4): 884-95, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25417163

ABSTRACT

Mammalian oocytes can reprogram somatic cells into a totipotent state enabling animal cloning through somatic cell nuclear transfer (SCNT). However, the majority of SCNT embryos fail to develop to term due to undefined reprogramming defects. Here, we identify histone H3 lysine 9 trimethylation (H3K9me3) of donor cell genome as a major barrier for efficient reprogramming by SCNT. Comparative transcriptome analysis identified reprogramming resistant regions (RRRs) that are expressed normally at 2-cell mouse embryos generated by in vitro fertilization (IVF) but not SCNT. RRRs are enriched for H3K9me3 in donor somatic cells and its removal by ectopically expressed H3K9me3 demethylase Kdm4d not only reactivates the majority of RRRs, but also greatly improves SCNT efficiency. Furthermore, use of donor somatic nuclei depleted of H3K9 methyltransferases markedly improves SCNT efficiency. Our study thus identifies H3K9me3 as a critical epigenetic barrier in SCNT-mediated reprogramming and provides a promising approach for improving mammalian cloning efficiency.


Subject(s)
Embryonic Development , Histone Code , Histones/metabolism , Nuclear Transfer Techniques , Animals , Cloning, Organism/methods , Embryo, Mammalian/metabolism , Female , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Methylation , Methyltransferases/metabolism , Mice, Inbred C57BL , Mice, Inbred DBA , Repressor Proteins/metabolism , Zygote
7.
Nature ; 499(7456): 88-91, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23728301

ABSTRACT

The generation of induced pluripotent stem (iPS) cells presents a challenge to normal developmental processes. The low efficiency and heterogeneity of most methods have hindered understanding of the precise molecular mechanisms promoting, and roadblocks preventing, efficient reprogramming. Although several intermediate populations have been described, it has proved difficult to characterize the rare, asynchronous transition from these intermediate stages to iPS cells. The rapid expansion of minor reprogrammed cells in the heterogeneous population can also obscure investigation of relevant transition processes. Understanding the biological mechanisms essential for successful iPS cell generation requires both accurate capture of cells undergoing the reprogramming process and identification of the associated global gene expression changes. Here we demonstrate that in mouse embryonic fibroblasts, reprogramming follows an orderly sequence of stage transitions, marked by changes in the cell-surface markers CD44 and ICAM1, and a Nanog-enhanced green fluorescent protein (Nanog-eGFP) reporter. RNA-sequencing analysis of these populations demonstrates two waves of pluripotency gene upregulation, and unexpectedly, transient upregulation of several epidermis-related genes, demonstrating that reprogramming is not simply the reversal of the normal developmental processes. This novel high-resolution analysis enables the construction of a detailed reprogramming route map, and the improved understanding of the reprogramming process will lead to new reprogramming strategies.


Subject(s)
Cellular Reprogramming/physiology , Hyaluronan Receptors/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cellular Reprogramming/genetics , Epidermis/metabolism , Fibroblasts , Flow Cytometry , Gene Expression Profiling , Genes, Reporter , Hyaluronan Receptors/genetics , Intercellular Adhesion Molecule-1/genetics , Mice , Sequence Analysis, RNA , Single-Cell Analysis , Up-Regulation/genetics
8.
PLoS One ; 6(5): e20461, 2011.
Article in English | MEDLINE | ID: mdl-21637830

ABSTRACT

The principal factors that lead to proliferation and pluripotency in embryonic stem cells (ESCs) have been vigorously investigated. However, the global network of factors and their full signaling cascade is still unclear. In this study, we found that ECAT11 (L1td1) is one of the ESC-associated transcripts harboring a truncated fragment of ORF-1, a component of the L1 retrotransposable element. We generated an ECAT11 knock-in mouse by replacing its coding region with green fluorescent protein. In the early stage of development, the fluorescence was observed at the inner cell mass of blastocysts and epiblasts. Despite this specific expression, ECAT11-null mice grow normally and are fertile. In addition, ECAT11 was dispensable for both the proliferation and pluripotency of ESCs.We found rapid and robust activation of ECAT11 in fibroblasts after the forced expression of transcription factors that can give rise pluripotency in somatic cells. However, iPS cells could be established from ECAT11-null fibroblasts. Our data demonstrate the dispensability of ECAT11/L1td1 in pluripotency, despite its specific expression.


Subject(s)
Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Proteins/metabolism , Amino Acid Sequence , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Genes, Reporter , Green Fluorescent Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Transport , Proteins/chemistry , Proteins/genetics , RNA-Binding Proteins , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...