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.
Mol Ther ; 24(9): 1570-80, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27203437

ABSTRACT

Many future therapeutic applications of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 and related RNA-guided nucleases are likely to require their use to promote gene targeting, thus necessitating development of methods that provide for delivery of three components-Cas9, guide RNAs and recombination templates-to primary cells rendered proficient for homology-directed repair. Here, we demonstrate an electroporation/transduction codelivery method that utilizes mRNA to express both Cas9 and mutant adenoviral E4orf6 and E1b55k helper proteins in association with adeno-associated virus (AAV) vectors expressing guide RNAs and recombination templates. By transiently enhancing target cell permissiveness to AAV transduction and gene editing efficiency, this novel approach promotes efficient gene disruption and/or gene targeting at multiple loci in primary human T-cells, illustrating its broad potential for application in translational gene editing.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Mutant Proteins , T-Lymphocytes/metabolism , Viral Proteins/metabolism , Dependovirus/genetics , Gene Expression , Gene Knock-In Techniques , Gene Knockout Techniques , Gene Order , Gene Targeting , Gene Transfer Techniques , Genetic Vectors/genetics , Homologous Recombination , Humans , RNA, Guide, Kinetoplastida/genetics , Transduction, Genetic , Viral Proteins/genetics
2.
Sci Transl Med ; 7(307): 307ra156, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26424571

ABSTRACT

Genetic mutations or engineered nucleases that disrupt the HIV co-receptor CCR5 block HIV infection of CD4(+) T cells. These findings have motivated the engineering of CCR5-specific nucleases for application as HIV therapies. The efficacy of this approach relies on efficient biallelic disruption of CCR5, and the ability to efficiently target sequences that confer HIV resistance to the CCR5 locus has the potential to further improve clinical outcomes. We used RNA-based nuclease expression paired with adeno-associated virus (AAV)-mediated delivery of a CCR5-targeting donor template to achieve highly efficient targeted recombination in primary human T cells. This method consistently achieved 8 to 60% rates of homology-directed recombination into the CCR5 locus in T cells, with over 80% of cells modified with an MND-GFP expression cassette exhibiting biallelic modification. MND-GFP-modified T cells maintained a diverse repertoire and engrafted in immune-deficient mice as efficiently as unmodified cells. Using this method, we integrated sequences coding chimeric antigen receptors (CARs) into the CCR5 locus, and the resulting targeted CAR T cells exhibited antitumor or anti-HIV activity. Alternatively, we introduced the C46 HIV fusion inhibitor, generating T cell populations with high rates of biallelic CCR5 disruption paired with potential protection from HIV with CXCR4 co-receptor tropism. Finally, this protocol was applied to adult human mobilized CD34(+) cells, resulting in 15 to 20% homologous gene targeting. Our results demonstrate that high-efficiency targeted integration is feasible in primary human hematopoietic cells and highlight the potential of gene editing to engineer T cell products with myriad functional properties.


Subject(s)
Deoxyribonucleases/metabolism , Dependovirus/metabolism , Hematopoietic Stem Cells/metabolism , Receptors, CCR5/metabolism , Adult , Antigens, CD34/metabolism , CD3 Complex/metabolism , Cells, Cultured , DNA Repair , Genetic Loci , Genetic Therapy , Green Fluorescent Proteins/metabolism , Humans , RNA Editing/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
3.
Nucleic Acids Res ; 42(1): e4, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24121685

ABSTRACT

The creation of a DNA break at a specific locus by a designer endonuclease can be harnessed to edit a genome. However, DNA breaks may engage one of several competing repair pathways that lead to distinct types of genomic alterations. Therefore, understanding the contribution of different repair pathways following the introduction of a targeted DNA break is essential to further advance the safety and efficiency of nuclease-induced genome modification. To gain insight into the role of different DNA repair pathways in resolving nuclease-induced DNA breaks into genome editing outcomes, we previously developed a fluorescent-based reporter system, designated the Traffic Light Reporter, which provides a readout of gene targeting and gene disruption downstream of a targeted DNA double-strand break. Here we describe two related but novel reporters that extend this technology: one that allows monitoring of the transcriptional activity at the reporter locus, and thus can be applied to interrogate break resolution at active and repressed loci; and a second that reads out single-strand annealing in addition to gene targeting and gene disruption. Application of these reporters to assess repair pathway usage in several common gene editing contexts confirms the importance that chromatin status and initiation of end resection have on the resolution of nuclease-induced breaks.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Endodeoxyribonucleases , Genes, Reporter , Flow Cytometry , Fluorescence , Gene Silencing , Genes , Genetic Loci , Genome , Genomics/methods , HEK293 Cells , Humans , Luminescent Proteins/genetics , Transcription, Genetic
4.
Nat Methods ; 9(10): 973-5, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22941364

ABSTRACT

Targeted DNA double-strand breaks introduced by rare-cleaving designer endonucleases can be harnessed for gene disruption applications by engaging mutagenic nonhomologous end-joining DNA repair pathways. However, endonuclease-mediated DNA breaks are often subject to precise repair, which limits the efficiency of targeted genome editing. To address this issue, we coupled designer endonucleases to DNA end-processing enzymes to drive mutagenic break resolution, achieving up to 25-fold enhancements in gene disruption rates.


Subject(s)
DNA Breaks, Double-Stranded , Endonucleases/physiology , Animals , DNA End-Joining Repair , DNA Repair , Exodeoxyribonucleases/physiology , HEK293 Cells , Humans , Mice , Phosphoproteins/physiology , Receptors, CCR5/physiology
5.
Endocrinology ; 151(2): 502-12, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20056832

ABSTRACT

Insulin enhances the proliferation and survival of pancreatic beta-cells, but its mechanisms remain unclear. We hypothesized that Raf-1, a kinase upstream of both ERK and Bad, might be a critical target of insulin in beta-cells. To test this hypothesis, we treated human and mouse islets as well as MIN6 beta-cells with multiple insulin concentrations and examined putative downstream targets using immunoblotting, immunoprecipitation, quantitative fluorescent imaging, and cell death assays. Low doses of insulin rapidly activated Raf-1 by dephosphorylating serine 259 and phosphorylating serine 338 in human islets, mouse islets, and MIN6 cells. The phosphorylation of ERK by insulin was eliminated by exposure to a Raf inhibitor (GW5074) or transfection with a dominant-negative Raf-1 mutant. Insulin also enhanced the interaction between mitochondrial Raf-1 and Bcl-2 agonist of cell death (Bad), promoting Bad inactivation via its phosphorylation on serine 112. Insulin-stimulated ERK phosphorylation was abrogated by calcium chelation, calcineurin and calmodulin-dependent protein kinase II inhibitors, and Ned-19, a nicotinic acid adenine dinucleotide phosphate receptor (NAADPR) antagonist. Blocking Raf-1 and Ca(2+) signaling resulted in nonadditive beta-cell death. Autocrine insulin signaling partly accounted for the effects of glucose on ERK phosphorylation. Our results demonstrate that Raf-1 is a critical target of insulin in primary beta-cells. Activation of Raf-1 leads to both an ERK-dependent pathway that involves nicotinic acid adenine dinucleotide phosphate-sensitive Ca(2+) stores and Ca(2+)-dependent phosphorylation events, and an ERK-independent pathway that involves Bad inactivation at the mitochondria. Together our findings identify a novel insulin signaling pathway in beta-cells and shed light on insulin's antiapoptotic and mitogenic mechanisms.


Subject(s)
Calcium/physiology , Insulin-Secreting Cells/physiology , Insulin/physiology , Proto-Oncogene Proteins c-raf/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Division/drug effects , Cell Line , Humans , Indoles/pharmacology , Insulin/genetics , Insulin/pharmacology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/physiology , Phenols/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/genetics
6.
Am J Physiol Endocrinol Metab ; 296(4): E690-701, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19141690

ABSTRACT

There are strong links between obesity, elevated free fatty acids, and type 2 diabetes. Specifically, the saturated fatty acid palmitate has pleiotropic effects on beta-cell function and survival. In the present study, we sought to determine the mechanism by which palmitate affects intracellular Ca2+, and in particular the role of the endoplasmic reticulum (ER). In human beta-cells and MIN6 cells, palmitate rapidly increased cytosolic Ca2+ through a combination of Ca2+ store release and extracellular Ca2+ influx. Palmitate caused a reversible lowering of ER Ca2+, measured directly with the fluorescent protein-based ER Ca2+ sensor D1ER. Using another genetically encoded indicator, we observed long-lasting oscillations of cytosolic Ca2+ in palmitate-treated cells. In keeping with this observed ER Ca2+ depletion, palmitate induced rapid phosphorylation of the ER Ca2+ sensor protein kinase R-like ER kinase (PERK) and subsequently ER stress and beta-cell death. We detected little palmitate-induced insulin secretion, suggesting that these Ca2+ signals are poorly coupled to exocytosis. In summary, we have characterized Ca2+-dependent mechanisms involved in altered beta-cell function and survival induced by the free fatty acid palmitate. We present the first direct evidence that free fatty acids reduce ER Ca2+ and shed light on pathways involved in lipotoxicity and the pathogenesis of type 2 diabetes.


Subject(s)
Calcium/metabolism , Cytosol/drug effects , Endoplasmic Reticulum/drug effects , Insulin-Secreting Cells/drug effects , Palmitic Acid/pharmacology , Animals , Calcium/physiology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Death/drug effects , Cells, Cultured , Cytosol/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum/metabolism , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Nonesterified/physiology , Homeostasis/drug effects , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Oleic Acid/pharmacology
7.
Diabetes ; 58(2): 422-32, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19033399

ABSTRACT

OBJECTIVE: Endoplasmic reticulum (ER) stress has been implicated in the pathogenesis of diabetes, but the roles of specific ER Ca(2+) release channels in the ER stress-associated apoptosis pathway remain unknown. Here, we examined the effects of stimulating or inhibiting the ER-resident inositol trisphosphate receptors (IP(3)Rs) and the ryanodine receptors (RyRs) on the induction of beta-cell ER stress and apoptosis. RESEARCH DESIGN AND METHODS: Kinetics of beta-cell death were tracked by imaging propidium iodide incorporation and caspase-3 activity in real time. ER stress and apoptosis were assessed by Western blot. Mitochondrial membrane potential was monitored by flow cytometry. Cytosolic Ca(2+) was imaged using fura-2, and genetically encoded fluorescence resonance energy transfer (FRET)-based probes were used to measure Ca(2+) in ER and mitochondria. RESULTS: Neither RyR nor IP(3)R inhibition, alone or in combination, caused robust death within 24 h. In contrast, blocking sarco/endoplasmic reticulum ATPase (SERCA) pumps depleted ER Ca(2+) and induced marked phosphorylation of PKR-like ER kinase (PERK) and eukaryotic initiation factor-2alpha (eIF2alpha), C/EBP homologous protein (CHOP)-associated ER stress, caspase-3 activation, and death. Notably, ER stress following SERCA inhibition was attenuated by blocking IP(3)Rs and RyRs. Conversely, stimulation of ER Ca(2+) release channels accelerated thapsigargin-induced ER depletion and apoptosis. SERCA block also activated caspase-9 and induced perturbations of the mitochondrial membrane potential, resulting eventually in the loss of mitochondrial polarization. CONCLUSIONS: This study demonstrates that the activity of ER Ca(2+) channels regulates the susceptibility of beta-cells to ER stress resulting from impaired SERCA function. Our results also suggest the involvement of mitochondria in beta-cell apoptosis associated with dysfunctional beta-cell ER Ca(2+) homeostasis and ER stress.


Subject(s)
Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/physiology , Insulin-Secreting Cells/metabolism , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Calcium/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Caspase 3/metabolism , Cell Death/drug effects , Cell Line , Cells, Cultured , Endoplasmic Reticulum/drug effects , Flow Cytometry , Fluorescence Resonance Energy Transfer , Immunoblotting , Inositol 1,4,5-Trisphosphate Receptors/agonists , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Kinetics , Macrocyclic Compounds/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Organometallic Compounds/pharmacology , Oxazoles/pharmacology , Propidium/metabolism , Ryanodine/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Thapsigargin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...