Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 27(4): 1254-1264.e7, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31018138

ABSTRACT

In vivo delivery of genome-modifying enzymes holds significant promise for therapeutic applications and functional genetic screening. Delivery to endogenous tissue stem cells, which provide an enduring source of cell replacement during homeostasis and regeneration, is of particular interest. Here, we use a sensitive Cre/lox fluorescent reporter system to test the efficiency of genome modification following in vivo transduction by adeno-associated viruses (AAVs) in tissue stem and progenitor cells. We combine immunophenotypic analyses with in vitro and in vivo assays of stem cell function to reveal effective targeting of skeletal muscle satellite cells, mesenchymal progenitors, hematopoietic stem cells, and dermal cell subsets using multiple AAV serotypes. Genome modification rates achieved through this system reached >60%, and modified cells retained key functional properties. This study establishes a powerful platform to genetically alter tissue progenitors within their physiological niche while preserving their native stem cell properties and regulatory interactions.


Subject(s)
Cell Differentiation , Dependovirus/genetics , Genome , Hematopoietic Stem Cells/cytology , Satellite Cells, Skeletal Muscle/cytology , Skin/cytology , Animals , Cell Movement , Dependovirus/classification , Female , Gene Transfer Techniques , Genetic Therapy , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Satellite Cells, Skeletal Muscle/metabolism , Skin/metabolism
2.
Nat Methods ; 13(10): 868-74, 2016 10.
Article in English | MEDLINE | ID: mdl-27595405

ABSTRACT

CRISPR-Cas9 delivery by adeno-associated virus (AAV) holds promise for gene therapy but faces critical barriers on account of its potential immunogenicity and limited payload capacity. Here, we demonstrate genome engineering in postnatal mice using AAV-split-Cas9, a multifunctional platform customizable for genome editing, transcriptional regulation, and other previously impracticable applications of AAV-CRISPR-Cas9. We identify crucial parameters that impact efficacy and clinical translation of our platform, including viral biodistribution, editing efficiencies in various organs, antigenicity, immunological reactions, and physiological outcomes. These results reveal that AAV-CRISPR-Cas9 evokes host responses with distinct cellular and molecular signatures, but unlike alternative delivery methods, does not induce extensive cellular damage in vivo. Our study provides a foundation for developing effective genome therapeutics.


Subject(s)
CRISPR-Cas Systems/genetics , Dependovirus/genetics , Gene Transfer Techniques , Genetic Engineering/methods , Genetic Vectors/genetics , Animals , Enzyme-Linked Immunosorbent Assay , Gene Editing , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
3.
Science ; 351(6271): 407-411, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26721686

ABSTRACT

Frame-disrupting mutations in the DMD gene, encoding dystrophin, compromise myofiber integrity and drive muscle deterioration in Duchenne muscular dystrophy (DMD). Removing one or more exons from the mutated transcript can produce an in-frame mRNA and a truncated, but still functional, protein. In this study, we developed and tested a direct gene-editing approach to induce exon deletion and recover dystrophin expression in the mdx mouse model of DMD. Delivery by adeno-associated virus (AAV) of clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 endonucleases coupled with paired guide RNAs flanking the mutated Dmd exon23 resulted in excision of intervening DNA and restored the Dmd reading frame in myofibers, cardiomyocytes, and muscle stem cells after local or systemic delivery. AAV-Dmd CRISPR treatment partially recovered muscle functional deficiencies and generated a pool of endogenously corrected myogenic precursors in mdx mouse muscle.


Subject(s)
Genetic Therapy/methods , Muscular Dystrophy, Duchenne/therapy , Satellite Cells, Skeletal Muscle/metabolism , Transduction, Genetic/methods , Animals , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Dependovirus , Disease Models, Animal , Exons , Frameshift Mutation , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Myocardium/metabolism , RNA, Messenger/genetics , Sequence Deletion
4.
PLoS One ; 8(3): e60016, 2013.
Article in English | MEDLINE | ID: mdl-23527294

ABSTRACT

Human pluripotent stem cells (hPSCs) provide unprecedented opportunities to study the earliest stages of human development in vitro and have the potential to provide unlimited new sources of cells for regenerative medicine. Although previous studies have reported cytokeratin 14+/p63+ keratinocyte generation from hPSCs, the multipotent progenitors of epithelial lineages have not been described and the developmental pathways regulating epithelial commitment remain largely unknown. Here we report membrane localization of ß-catenin during retinoic acid (RA)--induced epithelial differentiation. In addition hPSC treatment with the Src family kinase inhibitor SU6656 modulated ß-catenin localization and produced an enriched population of simple epithelial cells under defined culture conditions. SU6656 strongly upregulated expression of cytokeratins 18 and 8 (K18/K8), which are expressed in simple epithelial cells, while repressing expression of the pluripotency gene Oct4. This homogeneous population of K18+K8+Oct4- simple epithelial precursor cells can further differentiate into cells expressing keratinocyte or corneal-specific markers. These enriched hPSC-derived simple epithelial cells may provide a ready source for development and toxicology cell models and may serve as a progenitor for epithelial cell transplantation applications.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/cytology , Gene Expression Regulation/drug effects , Indoles/pharmacology , Pluripotent Stem Cells/cytology , Sulfonamides/pharmacology , src-Family Kinases/antagonists & inhibitors , Blotting, Western , Cadherins/metabolism , Cell Differentiation/drug effects , Cells, Cultured , DNA Primers/genetics , Flow Cytometry , Fluorescent Antibody Technique , Humans , Keratin-18/metabolism , Keratin-8/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/physiology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tretinoin/pharmacology , beta Catenin/metabolism
5.
Nat Protoc ; 8(1): 162-75, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23257984

ABSTRACT

The protocol described here efficiently directs human pluripotent stem cells (hPSCs) to functional cardiomyocytes in a completely defined, growth factor- and serum-free system by temporal modulation of regulators of canonical Wnt signaling. Appropriate temporal application of a glycogen synthase kinase 3 (GSK3) inhibitor combined with the expression of ß-catenin shRNA or a chemical Wnt inhibitor is sufficient to produce a high yield (0.8-1.3 million cardiomyocytes per cm(2)) of virtually pure (80-98%) functional cardiomyocytes in 14 d from multiple hPSC lines without cell sorting or selection. Qualitative (immunostaining) and quantitative (flow cytometry) characterization of differentiated cells is described to assess the expression of cardiac transcription factors and myofilament proteins. Flow cytometry of BrdU incorporation or Ki67 expression in conjunction with cardiac sarcomere myosin protein expression can be used to determine the proliferative capacity of hPSC-derived cardiomyocytes. Functional human cardiomyocytes differentiated via these protocols may constitute a potential cell source for heart disease modeling, drug screening and cell-based therapeutic applications.


Subject(s)
Cell Culture Techniques , Cell Differentiation , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Wnt Signaling Pathway , Flow Cytometry , Humans , Lentivirus/metabolism , RNA, Small Interfering/metabolism , beta Catenin/metabolism
6.
Stem Cells ; 31(3): 447-57, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23193013

ABSTRACT

The study of the regulatory signaling hierarchies of human heart development is limited by a lack of model systems that can reproduce the precise developmental events that occur during human embryogenesis. The advent of human pluripotent stem cell (hPSC) technology and robust cardiac differentiation methods affords a unique opportunity to monitor the full course of cardiac induction in vitro. Here, we show that stage-specific activation of insulin signaling strongly inhibited cardiac differentiation during a monolayer-based differentiation protocol that used transforming growth factor ß superfamily ligands to generate cardiomyocytes. However, insulin did not repress cardiomyocyte differentiation in a defined protocol that used small molecule regulators of canonical Wnt signaling. By examining the context of insulin inhibition of cardiomyocyte differentiation, we determined that the inhibitory effects by insulin required Wnt/ß-catenin signaling and that the cardiomyocyte differentiation defect resulting from insulin exposure was rescued by inhibition of Wnt/ß-catenin during the cardiac mesoderm (Nkx2.5+) stage. Thus, insulin and Wnt/ß-catenin signaling pathways, as a network, coordinate to influence hPSC differentiation to cardiomyocytes, with the Wnt/ß-catenin pathway dominant to the insulin pathway. Our study contributes to the understanding of the regulatory hierarchies of human cardiomyocyte differentiation and has implications for modeling human heart development.


Subject(s)
Insulin/pharmacology , Myocardium/cytology , Myocytes, Cardiac/drug effects , Wnt Signaling Pathway/drug effects , Animals , Cell Differentiation/drug effects , Humans , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Myocytes, Cardiac/metabolism , Signal Transduction/drug effects
7.
Proc Natl Acad Sci U S A ; 109(27): E1848-57, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22645348

ABSTRACT

Human pluripotent stem cells (hPSCs) offer the potential to generate large numbers of functional cardiomyocytes from clonal and patient-specific cell sources. Here we show that temporal modulation of Wnt signaling is both essential and sufficient for efficient cardiac induction in hPSCs under defined, growth factor-free conditions. shRNA knockdown of ß-catenin during the initial stage of hPSC differentiation fully blocked cardiomyocyte specification, whereas glycogen synthase kinase 3 inhibition at this point enhanced cardiomyocyte generation. Furthermore, sequential treatment of hPSCs with glycogen synthase kinase 3 inhibitors followed by inducible expression of ß-catenin shRNA or chemical inhibitors of Wnt signaling produced a high yield of virtually (up to 98%) pure functional human cardiomyocytes from multiple hPSC lines. The robust ability to generate functional cardiomyocytes under defined, growth factor-free conditions solely by genetic or chemically mediated manipulation of a single developmental pathway should facilitate scalable production of cardiac cells suitable for research and regenerative applications.


Subject(s)
Cell Culture Techniques/methods , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Wnt Signaling Pathway/physiology , Cell Differentiation/physiology , Culture Media/pharmacology , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Humans , RNA, Small Interfering/genetics , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...