Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 23(17)2022 Aug 28.
Article in English | MEDLINE | ID: mdl-36077152

ABSTRACT

Monocytes and their downstream effectors are critical components of the innate immune system. Monocytes are equipped with chemokine receptors, allowing them to migrate to various tissues, where they can differentiate into macrophage and dendritic cell subsets and participate in tissue homeostasis, infection, autoimmune disease, and cancer. Enabling genome engineering in monocytes and their effector cells will facilitate a myriad of applications for basic and translational research. Here, we demonstrate that CRISPR-Cas9 RNPs can be used for efficient gene knockout in primary human monocytes. In addition, we demonstrate that intracellular RNases are likely responsible for poor and heterogenous mRNA expression as incorporation of pan-RNase inhibitor allows efficient genome engineering following mRNA-based delivery of Cas9 and base editor enzymes. Moreover, we demonstrate that CRISPR-Cas9 combined with an rAAV vector DNA donor template mediates site-specific insertion and expression of a transgene in primary human monocytes. Finally, we demonstrate that SIRPa knock-out monocyte-derived macrophages have enhanced activity against cancer cells, highlighting the potential for application in cellular immunotherapies.


Subject(s)
CRISPR-Cas Systems , Ribonucleases , CRISPR-Cas Systems/genetics , Endoribonucleases/genetics , Gene Editing , Gene Knockout Techniques , Genetic Engineering , Humans , Monocytes , RNA, Messenger/genetics , Ribonucleases/genetics
2.
Nat Commun ; 12(1): 2437, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893286

ABSTRACT

CRISPR-Cas9 cytidine and adenosine base editors (CBEs and ABEs) can disrupt genes without introducing double-stranded breaks by inactivating splice sites (BE-splice) or by introducing premature stop (pmSTOP) codons. However, no in-depth comparison of these methods or a modular tool for designing BE-splice sgRNAs exists. To address these needs, we develop SpliceR ( http://z.umn.edu/spliceR ) to design and rank BE-splice sgRNAs for any Ensembl annotated genome, and compared disruption approaches in T cells using a screen against the TCR-CD3 MHC Class I immune synapse. Among the targeted genes, we find that targeting splice-donors is the most reliable disruption method, followed by targeting splice-acceptors, and introducing pmSTOPs. Further, the CBE BE4 is more effective for disruption than the ABE ABE7.10, however this disparity is eliminated by employing ABE8e. Collectively, we demonstrate a robust method for gene disruption, accompanied by a modular design tool that is of use to basic and translational researchers alike.


Subject(s)
Adenosine/metabolism , CRISPR-Cas Systems , Computational Biology/methods , Cytidine/metabolism , Gene Editing/methods , Adenosine/chemistry , Base Sequence , Cells, Cultured , Cytidine/chemistry , Humans , Internet , K562 Cells , Reproducibility of Results , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
3.
Mol Ther ; 28(1): 52-63, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31704085

ABSTRACT

Enhancing natural killer (NK) cell cytotoxicity by blocking inhibitory signaling could lead to improved NK-based cancer immunotherapy. Thus, we have developed a highly efficient method for editing the genome of human NK cells using CRISPR/Cas9 to knock out inhibitory signaling molecules. Our method efficiently edits up to 90% of primary peripheral blood NK cells. As a proof-of-principle we demonstrate highly efficient knockout of ADAM17 and PDCD1, genes that have a functional impact on NK cells, and demonstrate that these gene-edited NK cells have significantly improved activity, cytokine production, and cancer cell cytotoxicity. Furthermore, we were able to expand cells to clinically relevant numbers, without loss of activity. We also demonstrate that our CRISPR/Cas9 method can be used for efficient knockin of genes by delivering homologous recombination template DNA using recombinant adeno-associated virus serotype 6 (rAAV6). Our platform represents a feasible method for generating engineered primary NK cells as a universal therapeutic for cancer immunotherapy.


Subject(s)
Adoptive Transfer/methods , Cell Engineering/methods , Genetic Engineering/methods , Killer Cells, Natural/immunology , Ovarian Neoplasms/therapy , ADAM17 Protein/genetics , Animals , CRISPR-Cas Systems , Cytotoxicity, Immunologic/genetics , Dependovirus , Female , Gene Knockout Techniques , Healthy Volunteers , Humans , K562 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/pathology , Parvovirinae/genetics , Programmed Cell Death 1 Receptor/genetics , Treatment Outcome , Xenograft Model Antitumor Assays
5.
Nat Commun ; 10(1): 5222, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31745080

ABSTRACT

The fusion of genome engineering and adoptive cellular therapy holds immense promise for the treatment of genetic disease and cancer. Multiplex genome engineering using targeted nucleases can be used to increase the efficacy and broaden the application of such therapies but carries safety risks associated with unintended genomic alterations and genotoxicity. Here, we apply base editor technology for multiplex gene modification in primary human T cells in support of an allogeneic CAR-T platform and demonstrate that base editor can mediate highly efficient multiplex gene disruption with minimal double-strand break induction. Importantly, multiplex base edited T cells exhibit improved expansion and lack double strand break-induced translocations observed in T cells edited with Cas9 nuclease. Our findings highlight base editor as a powerful platform for genetic modification of therapeutically relevant primary cell types.


Subject(s)
CRISPR-Cas Systems , Cell Engineering/methods , DNA Breaks, Double-Stranded , Gene Editing/methods , T-Lymphocytes/metabolism , Cells, Cultured , High-Throughput Nucleotide Sequencing/methods , Humans , Immunotherapy, Adoptive/methods , Reproducibility of Results , T-Lymphocytes/cytology
6.
Stem Cells Dev ; 25(20): 1591-1603, 2016 10.
Article in English | MEDLINE | ID: mdl-27538887

ABSTRACT

The ability to rationally target disease-causing mutations has been made possible with programmable nucleases with the CRISPR/Cas9 system representing a facile platform for individualized gene-based medicine. In this study we employed footprint free reprogramming of fibroblasts from a patient with mutations to the Fanconi anemia I (FANCI) gene to generate induced pluripotent stem cells (iPSC). This process was accomplished without gene complementation and the resultant iPSC were able to be gene corrected in a robust manner using the Cas9 nickase. The self-renewing iPSC that were maintained under feeder free conditions were differentiated into cells with characteristics of definitive hematopoiesis. This defined and highly efficient procedure employed small molecule modulation of the hematopoietic differentiation pathway and a vascular induction technique to generate hematopoietic progenitors. In sum, our results demonstrate the ability to induce patient derived FA cells to pluripotency for patient specific therapeutic cell derivation.

7.
Mol Ther ; 24(3): 570-81, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26502778

ABSTRACT

Present adoptive immunotherapy strategies are based on the re-targeting of autologous T-cells to recognize tumor antigens. As T-cell properties may vary significantly between patients, this approach can result in significant variability in cell potency that may affect therapeutic outcome. More consistent results could be achieved by generating allogeneic cells from healthy donors. An impediment to such an approach is the endogenous T-cell receptors present on T-cells, which have the potential to direct dangerous off-tumor antihost reactivity. To address these limitations, we assessed the ability of three different TCR-α-targeted nucleases to disrupt T-cell receptor expression in primary human T-cells. We optimized the conditions for the delivery of each reagent and assessed off-target cleavage. The megaTAL and CRISPR/Cas9 reagents exhibited the highest disruption efficiency combined with low levels of toxicity and off-target cleavage, and we used them for a translatable manufacturing process to produce safe cellular substrates for next-generation immunotherapies.


Subject(s)
CRISPR-Cas Systems , Endonucleases , Gene Editing , Receptors, Antigen, T-Cell/genetics , Transcription Activator-Like Effector Nucleases , Binding Sites , Cell Culture Techniques , Cell Line , Gene Targeting , Gene Transfer Techniques , Genetic Loci , Genome , Humans , Immunophenotyping , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Phenotype , Protein Binding , Recombinant Fusion Proteins , T-Lymphocytes/metabolism , Transduction, Genetic
8.
Article in English | MEDLINE | ID: mdl-28250968

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) is a severe disorder caused by mutations to the COL7A1 gene that deactivate production of a structural protein essential for skin integrity. Haematopoietic cell transplantation can ameliorate some of the symptoms; however, significant side effects from the allogeneic transplant procedure can occur and unresponsive areas of blistering persist. Therefore, we employed genome editing in patient-derived cells to create an autologous platform for multilineage engineering of therapeutic cell types. The clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 system facilitated correction of an RDEB-causing COL7A1 mutation in primary fibroblasts that were then used to derive induced pluripotent stem cells (iPSCs). The resulting iPSCs were subsequently re-differentiated into keratinocytes, mesenchymal stem cells (MSCs) and haematopoietic progenitor cells using defined differentiation strategies. Gene-corrected keratinocytes exhibited characteristic epithelial morphology and expressed keratinocyte-specific genes and transcription factors. iPSC-derived MSCs exhibited a spindle morphology and expression of CD73, CD90 and CD105 with the ability to undergo adipogenic, chondrogenic and osteogenic differentiation in vitro in a manner indistinguishable from bone marrow-derived MSCs. Finally, we used a vascular induction strategy to generate potent definitive haematopoietic progenitors capable of multilineage differentiation in methylcellulose-based assays. In totality, we have shown that CRISPR/Cas9 is an adaptable gene-editing strategy that can be coupled with iPSC technology to produce multiple gene-corrected autologous cell types with therapeutic potential for RDEB.

9.
Stem Cells ; 31(8): 1611-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23606600

ABSTRACT

Transplantation of a myogenic cell population into an immunodeficient recipient is an excellent way of assessing the in vivo muscle-generating capacity of that cell population. To facilitate both allogeneic and xenogeneic transplantations of muscle-forming cells in mice, we have developed a novel immunodeficient muscular dystrophy model, the NSG-mdx(4Cv) mouse. The IL2Rg mutation, which is linked to the Dmd gene on the X chromosome, simultaneously depletes NK cells and suppresses thymic lymphomas, issues that limit the utility of the SCID/mdx model. The NSG-mdx(4Cv) mouse presents a muscular dystrophy of similar severity to the conventional mdx mouse. We show that this animal supports robust engraftment of both pig and dog muscle mononuclear cells. The question of whether satellite cells prospectively isolated by flow cytometry can confer a functional benefit upon transplantation has been controversial. Using allogeneic Pax7-ZsGreen donors and NSG-mdx(4Cv) recipients, we demonstrate definitively that as few as 900 FACS-isolated satellite cells can provide functional regeneration in vivo, in the form of an increased mean maximal force-generation capacity in cell-transplanted muscles, compared to a sham-injected control group. These studies highlight the potency of satellite cells to improve muscle function and the utility of the NSG-mdx(4Cv) model for studies on muscle regeneration and Duchenne muscular dystrophy therapy.


Subject(s)
Dystrophin/deficiency , Muscular Dystrophy, Duchenne/surgery , Satellite Cells, Skeletal Muscle/transplantation , Stem Cell Transplantation/methods , Animals , Disease Models, Animal , Dogs , Dystrophin/genetics , Dystrophin/metabolism , Female , Genotype , Heterografts , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Satellite Cells, Skeletal Muscle/cytology , Swine , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...