Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Cell Res ; 29(10): 804-819, 2019 Oct.
Article En | MEDLINE | ID: mdl-31444470

In vivo genome editing represents a powerful strategy for both understanding basic biology and treating inherited diseases. However, it remains a challenge to develop universal and efficient in vivo genome-editing tools for tissues that comprise diverse cell types in either a dividing or non-dividing state. Here, we describe a versatile in vivo gene knock-in methodology that enables the targeting of a broad range of mutations and cell types through the insertion of a minigene at an intron of the target gene locus using an intracellularly linearized single homology arm donor. As a proof-of-concept, we focused on a mouse model of premature-aging caused by a dominant point mutation, which is difficult to repair using existing in vivo genome-editing tools. Systemic treatment using our new method ameliorated aging-associated phenotypes and extended animal lifespan, thus highlighting the potential of this methodology for a broad range of in vivo genome-editing applications.


Gene Editing/methods , Animals , CRISPR-Cas Systems/genetics , DNA Repair , Dependovirus/genetics , GATA3 Transcription Factor/genetics , Gene Knock-In Techniques , Genetic Therapy/methods , Genetic Vectors/metabolism , Human Embryonic Stem Cells , Humans , Introns , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neurons/cytology , Neurons/metabolism , RNA, Guide, Kinetoplastida/metabolism , Rats , Tubulin/genetics
2.
Nature ; 540(7631): 144-149, 2016 12 01.
Article En | MEDLINE | ID: mdl-27851729

Targeted genome editing via engineered nucleases is an exciting area of biomedical research and holds potential for clinical applications. Despite rapid advances in the field, in vivo targeted transgene integration is still infeasible because current tools are inefficient, especially for non-dividing cells, which compose most adult tissues. This poses a barrier for uncovering fundamental biological principles and developing treatments for a broad range of genetic disorders. Based on clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) technology, here we devise a homology-independent targeted integration (HITI) strategy, which allows for robust DNA knock-in in both dividing and non-dividing cells in vitro and, more importantly, in vivo (for example, in neurons of postnatal mammals). As a proof of concept of its therapeutic potential, we demonstrate the efficacy of HITI in improving visual function using a rat model of the retinal degeneration condition retinitis pigmentosa. The HITI method presented here establishes new avenues for basic research and targeted gene therapies.


CRISPR-Cas Systems/genetics , Gene Editing/methods , Gene Targeting/methods , Genome/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Animals , Cell Division , Disease Models, Animal , Gene Knock-In Techniques , Genetic Therapy/methods , Neurons/cytology , Neurons/metabolism , Rats , Sequence Homology
3.
Science ; 348(6239): 1160-3, 2015 Jun 05.
Article En | MEDLINE | ID: mdl-25931448

Werner syndrome (WS) is a premature aging disorder caused by WRN protein deficiency. Here, we report on the generation of a human WS model in human embryonic stem cells (ESCs). Differentiation of WRN-null ESCs to mesenchymal stem cells (MSCs) recapitulates features of premature cellular aging, a global loss of H3K9me3, and changes in heterochromatin architecture. We show that WRN associates with heterochromatin proteins SUV39H1 and HP1α and nuclear lamina-heterochromatin anchoring protein LAP2ß. Targeted knock-in of catalytically inactive SUV39H1 in wild-type MSCs recapitulates accelerated cellular senescence, resembling WRN-deficient MSCs. Moreover, decrease in WRN and heterochromatin marks are detected in MSCs from older individuals. Our observations uncover a role for WRN in maintaining heterochromatin stability and highlight heterochromatin disorganization as a potential determinant of human aging.


Aging/metabolism , Cellular Senescence , Exodeoxyribonucleases/metabolism , Heterochromatin/metabolism , Mesenchymal Stem Cells/metabolism , RecQ Helicases/metabolism , Werner Syndrome/metabolism , Aging/genetics , Animals , Cell Differentiation , Centromere/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Exodeoxyribonucleases/genetics , Gene Knockout Techniques , HEK293 Cells , Heterochromatin/chemistry , Humans , Membrane Proteins/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Models, Biological , RecQ Helicases/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Werner Syndrome/genetics , Werner Syndrome Helicase
4.
Cell Stem Cell ; 15(1): 31-6, 2014 Jul 03.
Article En | MEDLINE | ID: mdl-24996168

The utility of genome editing technologies for disease modeling and developing cellular therapies has been extensively documented, but the impact of these technologies on mutational load at the whole-genome level remains unclear. We performed whole-genome sequencing to evaluate the mutational load at single-base resolution in individual gene-corrected human induced pluripotent stem cell (hiPSC) clones in three different disease models. In single-cell clones, gene correction by helper-dependent adenoviral vector (HDAdV) or Transcription Activator-Like Effector Nuclease (TALEN) exhibited few off-target effects and a low level of sequence variation, comparable to that accumulated in routine hiPSC culture. The sequence variants were randomly distributed and unique to individual clones. We also combined both technologies and developed a TALEN-HDAdV hybrid vector, which significantly increased gene-correction efficiency in hiPSCs. Therefore, with careful monitoring via whole-genome sequencing it is possible to apply genome editing to human pluripotent cells with minimal impact on genomic mutational load.


Adenoviridae/genetics , Endonucleases/metabolism , Genetic Therapy , Genetic Vectors/metabolism , Induced Pluripotent Stem Cells/physiology , CRISPR-Cas Systems/genetics , Clone Cells , DNA Repair/genetics , Endonucleases/genetics , Genetic Vectors/genetics , Genome/genetics , HEK293 Cells , Humans , Mutation/genetics , Regenerative Medicine , Sequence Analysis, DNA
5.
Nat Commun ; 5: 4330, 2014 Jul 07.
Article En | MEDLINE | ID: mdl-24999918

Fanconi anaemia (FA) is a recessive disorder characterized by genomic instability, congenital abnormalities, cancer predisposition and bone marrow (BM) failure. However, the pathogenesis of FA is not fully understood partly due to the limitations of current disease models. Here, we derive integration free-induced pluripotent stem cells (iPSCs) from an FA patient without genetic complementation and report in situ gene correction in FA-iPSCs as well as the generation of isogenic FANCA-deficient human embryonic stem cell (ESC) lines. FA cellular phenotypes are recapitulated in iPSCs/ESCs and their adult stem/progenitor cell derivatives. By using isogenic pathogenic mutation-free controls as well as cellular and genomic tools, our model serves to facilitate the discovery of novel disease features. We validate our model as a drug-screening platform by identifying several compounds that improve hematopoietic differentiation of FA-iPSCs. These compounds are also able to rescue the hematopoietic phenotype of FA patient BM cells.


Drug Evaluation, Preclinical/methods , Fanconi Anemia/etiology , Fanconi Anemia/pathology , Models, Biological , Stem Cells/pathology , Cell Differentiation , Epigenesis, Genetic , Fanconi Anemia/drug therapy , Fanconi Anemia Complementation Group A Protein/genetics , Humans , Induced Pluripotent Stem Cells , Male , Young Adult
6.
Nature ; 491(7425): 603-7, 2012 Nov 22.
Article En | MEDLINE | ID: mdl-23075850

Nuclear-architecture defects have been shown to correlate with the manifestation of a number of human diseases as well as ageing. It is therefore plausible that diseases whose manifestations correlate with ageing might be connected to the appearance of nuclear aberrations over time. We decided to evaluate nuclear organization in the context of ageing-associated disorders by focusing on a leucine-rich repeat kinase 2 (LRRK2) dominant mutation (G2019S; glycine-to-serine substitution at amino acid 2019), which is associated with familial and sporadic Parkinson's disease as well as impairment of adult neurogenesis in mice. Here we report on the generation of induced pluripotent stem cells (iPSCs) derived from Parkinson's disease patients and the implications of LRRK2(G2019S) mutation in human neural-stem-cell (NSC) populations. Mutant NSCs showed increased susceptibility to proteasomal stress as well as passage-dependent deficiencies in nuclear-envelope organization, clonal expansion and neuronal differentiation. Disease phenotypes were rescued by targeted correction of the LRRK2(G2019S) mutation with its wild-type counterpart in Parkinson's disease iPSCs and were recapitulated after targeted knock-in of the LRRK2(G2019S) mutation in human embryonic stem cells. Analysis of human brain tissue showed nuclear-envelope impairment in clinically diagnosed Parkinson's disease patients. Together, our results identify the nucleus as a previously unknown cellular organelle in Parkinson's disease pathology and may help to open new avenues for Parkinson's disease diagnoses as well as for the potential development of therapeutics targeting this fundamental cell structure.


Mutant Proteins/metabolism , Neural Stem Cells/pathology , Parkinson Disease/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Apoptosis , Cell Differentiation , Cell Division , Cell Line , Clone Cells/metabolism , Clone Cells/pathology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Gene Knock-In Techniques , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mutant Proteins/genetics , Mutation , Neural Stem Cells/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/pathology , Proteasome Endopeptidase Complex/metabolism , Stress, Physiological
7.
Cell Stem Cell ; 8(6): 688-94, 2011 Jun 03.
Article En | MEDLINE | ID: mdl-21596650

Combination of stem cell-based approaches with gene-editing technologies represents an attractive strategy for studying human disease and developing therapies. However, gene-editing methodologies described to date for human cells suffer from technical limitations including limited target gene size, low targeting efficiency at transcriptionally inactive loci, and off-target genetic effects that could hamper broad clinical application. To address these limitations, and as a proof of principle, we focused on homologous recombination-based gene correction of multiple mutations on lamin A (LMNA), which are associated with various degenerative diseases. We show that helper-dependent adenoviral vectors (HDAdVs) provide a highly efficient and safe method for correcting mutations in large genomic regions in human induced pluripotent stem cells and can also be effective in adult human mesenchymal stem cells. This type of approach could be used to generate genotype-matched cell lines for disease modeling and drug discovery and potentially also in therapeutics.


Induced Pluripotent Stem Cells/metabolism , Lamin Type A/genetics , Mutation , Cell Line , Genotype , Humans , Induced Pluripotent Stem Cells/pathology
...