Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ayal Hendel is active.

Publication


Featured researches published by Ayal Hendel.


Nature Biotechnology | 2015

Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells.

Ayal Hendel; Rasmus O. Bak; Joseph T. Clark; Andrew Kennedy; Daniel E. Ryan; Subhadeep Roy; Israel Steinfeld; Benjamin D. Lunstad; Robert Kaiser; Alec B. Wilkens; Rosa Bacchetta; Anya Tsalenko; Douglas J. Dellinger; Laurakay Bruhn; Matthew H. Porteus

CRISPR-Cas-mediated genome editing relies on guide RNAs that direct site-specific DNA cleavage facilitated by the Cas endonuclease. Here we report that chemical alterations to synthesized single guide RNAs (sgRNAs) enhance genome editing efficiency in human primary T cells and CD34+ hematopoietic stem and progenitor cells. Co-delivering chemically modified sgRNAs with Cas9 mRNA or protein is an efficient RNA- or ribonucleoprotein (RNP)-based delivery method for the CRISPR-Cas system, without the toxicity associated with DNA delivery. This approach is a simple and effective way to streamline the development of genome editing with the potential to accelerate a wide array of biotechnological and therapeutic applications of the CRISPR-Cas technology.


Nature | 2016

CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells

Daniel P. Dever; Rasmus O. Bak; Andreas Reinisch; Joab Camarena; Gabriel Washington; Carmencita Nicolas; Mara Pavel-Dinu; Nivi Saxena; Alec B. Wilkens; Sruthi Mantri; Nobuko Uchida; Ayal Hendel; Anupama Narla; Ravindra Majeti; Kenneth I. Weinberg; Matthew H. Porteus

The β-haemoglobinopathies, such as sickle cell disease and β-thalassaemia, are caused by mutations in the β-globin (HBB) gene and affect millions of people worldwide. Ex vivo gene correction in patient-derived haematopoietic stem cells followed by autologous transplantation could be used to cure β-haemoglobinopathies. Here we present a CRISPR/Cas9 gene-editing system that combines Cas9 ribonucleoproteins and adeno-associated viral vector delivery of a homologous donor to achieve homologous recombination at the HBB gene in haematopoietic stem cells. Notably, we devise an enrichment model to purify a population of haematopoietic stem and progenitor cells with more than 90% targeted integration. We also show efficient correction of the Glu6Val mutation responsible for sickle cell disease by using patient-derived stem and progenitor cells that, after differentiation into erythrocytes, express adult β-globin (HbA) messenger RNA, which confirms intact transcriptional regulation of edited HBB alleles. Collectively, these preclinical studies outline a CRISPR-based methodology for targeting haematopoietic stem cells by homologous recombination at the HBB locus to advance the development of next-generation therapies for β-haemoglobinopathies.


Trends in Biotechnology | 2015

Quantifying on- and off-target genome editing

Ayal Hendel; Eli J. Fine; Gang Bao; Matthew H. Porteus

Genome editing with engineered nucleases is a rapidly growing field thanks to transformative technologies that allow researchers to precisely alter genomes for numerous applications including basic research, biotechnology, and human gene therapy. While the ability to make precise and controlled changes at specified sites throughout the genome has grown tremendously in recent years, we still lack a comprehensive and standardized battery of assays for measuring the different genome editing outcomes created at endogenous genomic loci. Here we review the existing assays for quantifying on- and off-target genome editing and describe their utility in advancing the technology. We also highlight unmet assay needs for quantifying on- and off-target genome editing outcomes and discuss their importance for the genome editing field.


Cell Reports | 2014

Quantifying Genome-Editing Outcomes at Endogenous Loci with SMRT Sequencing

Ayal Hendel; Eric J Kildebeck; Eli J. Fine; Joseph T. Clark; Niraj Punjya; Vittorio Sebastiano; Gang Bao; Matthew H. Porteus

Targeted genome editing with engineered nucleases has transformed the ability to introduce precise sequence modifications at almost any site within the genome. A major obstacle to probing the efficiency and consequences of genome editing is that no existing method enables the frequency of different editing events to be simultaneously measured across a cell population at any endogenous genomic locus. We have developed a method for quantifying individual genome-editing outcomes at any site of interest with single-molecule real-time (SMRT) DNA sequencing. We show that this approach can be applied at various loci using multiple engineered nuclease platforms, including transcription-activator-like effector nucleases (TALENs), RNA-guided endonucleases (CRISPR/Cas9), and zinc finger nucleases (ZFNs), and in different cell lines to identify conditions and strategies in which the desired engineering outcome has occurred. This approach offers a technique for studying double-strand break repair, facilitates the evaluation of gene-editing technologies, and permits sensitive quantification of editing outcomes in almost every experimental system used.


Nucleic Acids Research | 2014

Nuclease-mediated gene editing by homologous recombination of the human globin locus

Richard A. Voit; Ayal Hendel; Shondra M. Pruett-Miller; Matthew H. Porteus

Tal-effector nucleases (TALENs) are engineered proteins that can stimulate precise genome editing through specific DNA double-strand breaks. Sickle cell disease and β-thalassemia are common genetic disorders caused by mutations in β-globin, and we engineered a pair of highly active TALENs that induce modification of 54% of human β-globin alleles near the site of the sickle mutation. These TALENS stimulate targeted integration of therapeutic, full-length beta-globin cDNA to the endogenous β-globin locus in 19% of cells prior to selection as quantified by single molecule real-time sequencing. We also developed highly active TALENs to human γ-globin, a pharmacologic target in sickle cell disease therapy. Using the β-globin and γ-globin TALENs, we generated cell lines that express GFP under the control of the endogenous β-globin promoter and tdTomato under the control of the endogenous γ-globin promoter. With these fluorescent reporter cell lines, we screened a library of small molecule compounds for their differential effect on the transcriptional activity of the endogenous β- and γ-globin genes and identified several that preferentially upregulate γ-globin expression.


Circulation Research | 2017

A Comprehensive TALEN-Based Knockout Library for Generating Human-Induced Pluripotent Stem Cell–Based Models for Cardiovascular Diseases

Ioannis Karakikes; Vittavat Termglinchan; Diana Cepeda; Jaecheol Lee; Sebastian Diecke; Ayal Hendel; Ilanit Itzhaki; Mohamed Ameen; Rajani Shrestha; Haodi Wu; Ning Ma; Ning-Yi Shao; Timon Seeger; Nicole A. Woo; Kitchener D. Wilson; Elena Matsa; Matthew H. Porteus; Vittorio Sebastiano; Joseph C. Wu

Rationale: Targeted genetic engineering using programmable nucleases such as transcription activator–like effector nucleases (TALENs) is a valuable tool for precise, site-specific genetic modification in the human genome. Objective: The emergence of novel technologies such as human induced pluripotent stem cells (iPSCs) and nuclease-mediated genome editing represent a unique opportunity for studying cardiovascular diseases in vitro. Methods and Results: By incorporating extensive literature and database searches, we designed a collection of TALEN constructs to knockout 88 human genes that are associated with cardiomyopathies and congenital heart diseases. The TALEN pairs were designed to induce double-strand DNA break near the starting codon of each gene that either disrupted the start codon or introduced a frameshift mutation in the early coding region, ensuring faithful gene knockout. We observed that all the constructs were active and disrupted the target locus at high frequencies. To illustrate the utility of the TALEN–mediated knockout technique, 6 individual genes (TNNT2, LMNA/C, TBX5, MYH7, ANKRD1, and NKX2.5) were knocked out with high efficiency and specificity in human iPSCs. By selectively targeting a pathogenic mutation (TNNT2 p.R173W) in patient-specific iPSC-derived cardiac myocytes, we demonstrated that the knockout strategy ameliorates the dilated cardiomyopathy phenotype in vitro. In addition, we modeled the Holt–Oram syndrome in iPSC-cardiac myocytes in vitro and uncovered novel pathways regulated by TBX5 in human cardiac myocyte development. Conclusions: Collectively, our study illustrates the powerful combination of iPSCs and genome editing technologies for understanding the biological function of genes, and the pathological significance of genetic variants in human cardiovascular diseases. The methods, strategies, constructs, and iPSC lines developed in this study provide a validated, readily available resource for cardiovascular research.


Molecular therapy. Nucleic acids | 2018

Uridine Depletion and Chemical Modification Increase Cas9 mRNA Activity and Reduce Immunogenicity without HPLC Purification

Sriram Vaidyanathan; Krist T. Azizian; A.K.M. Ashiqul Haque; Jordana M. Henderson; Ayal Hendel; Sabrina Shore; Justin S. Antony; Richard I. Hogrefe; Michael Kormann; Matthew H. Porteus; Anton P. McCaffrey

The Cas9/guide RNA (Cas9/gRNA) system is commonly used for genome editing. mRNA expressing Cas9 can induce innate immune responses, reducing Cas9 expression. First-generation Cas9 mRNAs were modified with pseudouridine and 5-methylcytosine to reduce innate immune responses. We combined four approaches to produce more active, less immunogenic second-generation Cas9 mRNAs. First, we developed a novel co-transcriptional capping method yielding natural Cap 1. Second, we screened modified nucleotides in Cas9 mRNA to identify novel modifications that increase Cas9 activity. Third, we depleted the mRNA of uridines to improve mRNA activity. Lastly, we tested high-performance liquid chromatography (HPLC) purification to remove double-stranded RNAs. The activity of these mRNAs was tested in cell lines and primary human CD34+ cells. Cytokines were measured in whole blood and mice. These approaches yielded more active and less immunogenic mRNA. Uridine depletion (UD) most impacted insertion or deletion (indel) activity. Specifically, 5-methoxyuridine UD induced indel frequencies as high as 88% (average ± SD = 79% ± 11%) and elicited minimal immune responses without needing HPLC purification. Our work suggests that uridine-depleted Cas9 mRNA modified with 5-methoxyuridine (without HPLC purification) or pseudouridine may be optimal for the broad use of Cas9 both in vitro and in vivo.


Molecular Therapy | 2018

Global Transcriptional Response to CRISPR/Cas9-AAV6-Based Genome Editing in CD34+ Hematopoietic Stem and Progenitor Cells

M. Kyle Cromer; Sriram Vaidyanathan; Daniel E. Ryan; Bo Curry; Joab Camarena; Milan Kaushik; Sarah R. Hay; Renata Martin; Israel Steinfeld; Rasmus O. Bak; Daniel P. Dever; Ayal Hendel; Laurakay Bruhn; Matthew H. Porteus

Genome-editing technologies are currently being translated to the clinic. However, cellular effects of the editing machinery have yet to be fully elucidated. Here, we performed global microarray-based gene expression measurements on human CD34+ hematopoietic stem and progenitor cells that underwent editing. We probed effects of the entire editing process as well as each component individually, including electroporation, Cas9 (mRNA or protein) with chemically modified sgRNA, and AAV6 transduction. We identified differentially expressed genes relative to control treatments, which displayed enrichment for particular biological processes. All editing machinery components elicited immune, stress, and apoptotic responses. Cas9 mRNA invoked the greatest amount of transcriptional change, eliciting a distinct viral response and global transcriptional downregulation, particularly of metabolic and cell cycle processes. Electroporation also induced significant transcriptional change, with notable downregulation of metabolic processes. Surprisingly, AAV6 evoked no detectable viral response. We also found Cas9/sgRNA ribonucleoprotein treatment to be well tolerated, in spite of eliciting a DNA damage signature. Overall, this data establishes a benchmark for cellular tolerance of CRISPR/Cas9-AAV6-based genome editing, ensuring that the clinical protocol is as safe and efficient as possible.


Molecular Therapy | 2016

39. FACS-Based Enrichment of a Highly Purified HBB-Targeted Hematopoietic Stem and Progenitor Cell Population Using rAAV6 and CRISPR/Cas9

Daniel P. Dever; Rasmus O. Bak; Joab Camarena; Nivi Saxena; Andreas Reinisch; Carmencita Nicolas; Alec B. Wilkens; Ayal Hendel; Nobuko Uchida; Ravindra Majeti; Kenneth I. Weinberg; Matthew H. Porteus

Precise engineered nuclease-mediated gene correction via homologous recombination (HR) in hematopoietic stem and progenitor cells (HSPCs) has the power to transform curative therapies for monogenic diseases of the immune system. Sickle cell disease (SCD) is one of the most common monogenic diseases, affecting millions of people worldwide. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only curative treatment for patients with SCD; however, immunocompatibility issues, graft-versus-host disease, and graft rejection are major roadblocks for efficacious therapy. In theory, the ideal curative strategy for SCD and most monogenic immune diseases is ex vivo gene correction of patient-derived HSPCs followed by autologous HSCT of a highly purified targeted population to avoid possible complications of competition between unedited and gene-targeted HSPCs in vivo. By supplying a homologous GFP-expressing HBB donor via recombinant adeno-associated virus serotype 6 (rAAV6) in combination with a double strand break created by the CRISPR/Cas9 system, we achieved HR frequencies of 15-49% of HSPCs, and more importantly, identified a population of HBB-targeted HSPCs (HSPCHBB) through a log-fold MFI increase in chromosomal transgene expression. Fluorescence activated cell sorting (FACS) of this population revealed consistent GFP expression in >95% of cells over two weeks in vitro in multiple CD34+ donors isolated from either bone marrow, cord blood or peripheral blood. Single-cell FACS of HSPCHBB into methylcellulose led to both myeloid and lymphoid colony formation, and on-target PCR analysis revealed >95% of these HSPCHBB clones had either a mono or biallelic-targeting event. Notably, a fraction of HSPCHBB displayed the CD34+/CD38-/CD90+/CD45RA-immunophenotype, suggesting successful targeting of long-term repopulating hematopoietic stem cells (LT-HSCs). Furthermore, HSPCHBB displayed long-term engraftment in the bone marrow of immunodeficient NSG mice at 12 weeks post-transplant, where we identified that ~70% of human cells were GFP+ and also produced both myeloid (CD33+) and lymphoid (CD19+) cell types, implying that the HSPCHBB population contains true LT-HSCs that can repopulate a functional immune system. Altogether, these proof-of-concept studies showed that by combining a homologous rAAV6 donor, the CRISPR/Cas9 system, and FACS, it is feasible to generate and enrich a highly purified population of gene-corrected HSPCs that include LT-HSC potentials.


Molecular Therapy | 2016

43. CRISPR/Cas9 and rAAV6-Mediated Targeted Integration at the CCR5 Locus in Hematopoietic Stem and Progenitor Cells

Rasmus O. Bak; Daniel P. Dever; Nivedita Shankar Saxena; Joab Camarena; Andreas Reinisch; Alec B. Wilkens; Carmencita Nicolas; Ayal Hendel; Ravindra Majeti; Kenneth I. Weinberg; Matthew H. Porteus

CRISPR/Cas9-mediated genome editing relies on guide RNAs to direct site-specific DNA cleavage mediated by the Cas9 endonuclease. In this study, we identified a highly potent single guide RNA (sgRNA) targeting exon 3 of CCR5. This sgRNA was chemically synthesized with three modified nucleotides at each terminus with 2′O-Methyl and phosphorothioate modifications, and electroporated into cells either with Cas9 mRNA or complexed with Cas9 protein (RNP). Using Tracking of Indels by DEcomposition (TIDE) to quantify insertions and deletions (INDELs), we observe up to 80% INDELs in CD34+ hematopoietic stem and progenitor cells (HSPCs).To achieve targeted integration by homology-directed repair (HDR), we produced rAAV6 vectors carrying a GFP expression cassette flanked by CCR5 homology arms. Electroporation with Cas9 RNP followed by rAAV6 transduction led to targeted integration in up to 30% of the cells. Interestingly, we observed that cells with targeted integration expressed GFP at fluorescence intensities more than 10-fold higher than from episomal AAV vectors. This allowed us to sort targeted cells as early as four days after nucleofection and transduction. Upon fluorescence-activated cell sorting and culture of this population, >99% of cells remained GFP+ 20 days post sort. In a methylcellulose-based colony-forming unit (CFU) assay, we identified multipotent and lineage-committed progenitor cells in this population, and PCR of gDNA extracted from colonies confirmed targeted integration at CCR5 in at least 98% of cells. Phenotypic characterization of this targeted population confirmed the presence of CD34+ CD38− CD90+ CD45RA− cells, indicating genome editing of hematopoietic stem cells. We transplanted edited cells into immunodeficient NSG mice and analyzed the bone marrow 8 weeks post-transplant. In mice transplanted with cells that were not enriched for targeted integration, we found 0.1-1.9% GFP+ cells among the engrafted human cells. This was a significant decline compared to the 12-13% GFP+ cells in the input cell population following culture, which is a phenomenon consistent with findings reported by other groups using different nuclease platforms. In contrast, when transplanting cells enriched for targeted integration, we found that 75% of the engrafted human cells were GFP+, confirming the presence of cells with long-term engraftment potential in the enriched population.In conclusion, we have found that the combination of CRISPR/Cas9 and rAAV6 is an effective platform for HDR-mediated targeted integration of a transgene into the CCR5 locus. Furthermore, the GFP MFI shift observed when episomal rAAV6 vectors are integrated into the chromosome following HDR enables early isolation of a population highly enriched for targeted integration at this locus. Since CCR5 is considered a ‘safe harbor’ for targeted insertion of a gene, this approach might find general use in therapeutic genome editing. Additionally, since CCR5 is an important co-receptor during HIV infection, our findings may be used to generate immune cells resistant to HIV infection.

Collaboration


Dive into the Ayal Hendel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eli J. Fine

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge