225 results on '"Philip D. Gregory"'
Search Results
2. Targeted Correction and Restored Function of the CFTR Gene in Cystic Fibrosis Induced Pluripotent Stem Cells
- Author
-
Ana M. Crane, Philipp Kramer, Jacquelin H. Bui, Wook Joon Chung, Xuan Shirley Li, Manuel L. Gonzalez-Garay, Finn Hawkins, Wei Liao, Daniela Mora, Sangbum Choi, Jianbin Wang, Helena C. Sun, David E. Paschon, Dmitry Y. Guschin, Philip D. Gregory, Darrell N. Kotton, Michael C. Holmes, Eric J. Sorscher, and Brian R. Davis
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Recently developed reprogramming and genome editing technologies make possible the derivation of corrected patient-specific pluripotent stem cell sources—potentially useful for the development of new therapeutic approaches. Starting with skin fibroblasts from patients diagnosed with cystic fibrosis, we derived and characterized induced pluripotent stem cell (iPSC) lines. We then utilized zinc-finger nucleases (ZFNs), designed to target the endogenous CFTR gene, to mediate correction of the inherited genetic mutation in these patient-derived lines via homology-directed repair (HDR). We observed an exquisitely sensitive, homology-dependent preference for targeting one CFTR allele versus the other. The corrected cystic fibrosis iPSCs, when induced to differentiate in vitro, expressed the corrected CFTR gene; importantly, CFTR correction resulted in restored expression of the mature CFTR glycoprotein and restoration of CFTR chloride channel function in iPSC-derived epithelial cells.
- Published
- 2015
- Full Text
- View/download PDF
3. Human Intestinal Tissue with Adult Stem Cell Properties Derived from Pluripotent Stem Cells
- Author
-
Ryan Forster, Kunitoshi Chiba, Lorian Schaeffer, Samuel G. Regalado, Christine S. Lai, Qing Gao, Samira Kiani, Henner F. Farin, Hans Clevers, Gregory J. Cost, Andy Chan, Edward J. Rebar, Fyodor D. Urnov, Philip D. Gregory, Lior Pachter, Rudolf Jaenisch, and Dirk Hockemeyer
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Genetically engineered human pluripotent stem cells (hPSCs) have been proposed as a source for transplantation therapies and are rapidly becoming valuable tools for human disease modeling. However, many applications are limited due to the lack of robust differentiation paradigms that allow for the isolation of defined functional tissues. Here, using an endogenous LGR5-GFP reporter, we derived adult stem cells from hPSCs that gave rise to functional human intestinal tissue comprising all major cell types of the intestine. Histological and functional analyses revealed that such human organoid cultures could be derived with high purity and with a composition and morphology similar to those of cultures obtained from human biopsies. Importantly, hPSC-derived organoids responded to the canonical signaling pathways that control self-renewal and differentiation in the adult human intestinal stem cell compartment. This adult stem cell system provides a platform for studying human intestinal disease in vitro using genetically engineered hPSCs.
- Published
- 2014
- Full Text
- View/download PDF
4. LRRK2 mutations cause mitochondrial DNA damage in iPSC-derived neural cells from Parkinson's disease patients: Reversal by gene correction
- Author
-
Laurie H. Sanders, Josée Laganière, Oliver Cooper, Sally K. Mak, B. Joseph Vu, Y. Anne Huang, David E. Paschon, Malini Vangipuram, Ramya Sundararajan, Fyodor D. Urnov, J. William Langston, Philip D. Gregory, H. Steve Zhang, J. Timothy Greenamyre, Ole Isacson, and Birgitt Schüle
- Subjects
Parkinson's disease ,LRRK2 ,Mitochondrial DNA damage ,Stem cells ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Parkinson's disease associated mutations in leucine rich repeat kinase 2 (LRRK2) impair mitochondrial function and increase the vulnerability of induced pluripotent stem cell (iPSC)-derived neural cells from patients to oxidative stress. Since mitochondrial DNA (mtDNA) damage can compromise mitochondrial function, we examined whether LRRK2 mutations can induce damage to the mitochondrial genome. We found greater levels of mtDNA damage in iPSC-derived neural cells from patients carrying homozygous or heterozygous LRRK2 G2019S mutations, or at-risk individuals carrying the heterozygous LRRK2 R1441C mutation, than in cells from unrelated healthy subjects who do not carry LRRK2 mutations. After zinc finger nuclease-mediated repair of the LRRK2 G2019S mutation in iPSCs, mtDNA damage was no longer detected in differentiated neuroprogenitor and neural cells. Our results unambiguously link LRRK2 mutations to mtDNA damage and validate a new cellular phenotype that can be used for examining pathogenic mechanisms and screening therapeutic strategies.
- Published
- 2014
- Full Text
- View/download PDF
5. Use of zinc-finger nucleases to knock out the WAS gene in K562 cells: a human cellular model for Wiskott-Aldrich syndrome
- Author
-
Miguel G. Toscano, Per Anderson, Pilar Muñoz, Gema Lucena, Marién Cobo, Karim Benabdellah, Philip D. Gregory, Michael C. Holmes, and Francisco Martin
- Subjects
Medicine ,Pathology ,RB1-214 - Abstract
SUMMARY Mutations in the WAS gene cause Wiskott-Aldrich syndrome (WAS), which is characterized by eczema, immunodeficiency and microthrombocytopenia. Although the role of WASP in lymphocytes and myeloid cells is well characterized, its role on megakaryocyte (MK) development is poorly understood. In order to develop a human cellular model that mimics the megakaryocytic-derived defects observed in WAS patients we used K562 cells, a well-known model for study of megakaryocytic development. We knocked out the WAS gene in K562 cells using a zinc-finger nuclease (ZFN) pair targeting the WAS intron 1 and a homologous donor DNA that disrupted WASP expression. Knockout of WASP on K562 cells (K562WASKO cells) resulted in several megakaryocytic-related defects such as morphological alterations, lower expression of CD41ɑ, lower increments in F-actin polymerization upon stimulation, reduced CD43 expression and increased phosphatidylserine exposure. All these defects have been previously described either in WAS-knockout mice or in WAS patients, validating K562WASKO as a cell model for WAS. However, K562WASPKO cells showed also increased basal F-actin and adhesion, increased expression of CD61 and reduced expression of TGFβ and Factor VIII, defects that have never been described before for WAS-deficient cells. Interestingly, these phenotypic alterations correlate with different roles for WASP in megakaryocytic differentiation. All phenotypic alterations observed in K562WASKO cells were alleviated upon expression of WAS following lentiviral transduction, confirming the role of WASP in these phenotypes. In summary, in this work we have validated a human cellular model, K562WASPKO, that mimics the megakaryocytic-related defects found in WAS-knockout mice and have found evidences for a role of WASP as regulator of megakaryocytic differentiation. We propose the use of K562WASPKO cells as a tool to study the molecular mechanisms involved in the megakaryocytic-related defects observed in WAS patients and as a cellular model to study new therapeutic strategies.
- Published
- 2013
- Full Text
- View/download PDF
6. Preclinical development and qualification of ZFN-mediated CCR5 disruption in human hematopoietic stem/progenitor cells
- Author
-
David L DiGiusto, Paula M Cannon, Michael C Holmes, Lijing Li, Anitha Rao, Jianbin Wang, Gary Lee, Philip D. Gregory, Kenneth A Kim, Samuel B Hayward, Kathleen Meyer, Colin Exline, Evan Lopez, Jill Henley, Nancy Gonzalez, Victoria Bedell, Rodica Stan, and John A Zaia
- Subjects
Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Gene therapy for HIV-1 infection is a promising alternative to lifelong combination antiviral drug treatment. Chemokine receptor 5 (CCR5) is the coreceptor required for R5-tropic HIV-1 infection of human cells. Deletion of CCR5 renders cells resistant to R5-tropic HIV-1 infection, and the potential for cure has been shown through allogeneic stem cell transplantation with naturally occurring homozygous deletion of CCR5 in donor hematopoietic stem/progenitor cells (HSPC). The requirement for HLA-matched HSPC bearing homozygous CCR5 deletions prohibits widespread application of this approach. Thus, a strategy to disrupt CCR5 genomic sequences in HSPC using zinc finger nucleases was developed. Following discussions with regulatory agencies, we conducted IND-enabling preclinical in vitro and in vivo testing to demonstrate the feasibility and (preclinical) safety of zinc finger nucleases-based CCR5 disruption in HSPC. We report here the clinical-scale manufacturing process necessary to deliver CCR5-specific zinc finger nucleases mRNA to HSPC using electroporation and the preclinical safety data. Our results demonstrate effective biallelic CCR5 disruption in up to 72.9% of modified colony forming units from adult mobilized HSPC with maintenance of hematopoietic potential in vitro and in vivo. Tumorigenicity studies demonstrated initial product safety; further safety and feasibility studies are ongoing in subjects infected with HIV-1 (NCT02500849@clinicaltrials.gov).
- Published
- 2016
- Full Text
- View/download PDF
7. An association sequence suitable for producing ground-state RbCs molecules in optical lattices
- Author
-
Arpita Das, Philip D. Gregory, Tetsu Takekoshi, Luke Fernley, Manuele Landini, Jeremy M. Hutson, Simon L. Cornish, Hanns-Christoph Nägerl
- Subjects
Physics ,QC1-999 - Abstract
We identify a route for the production of $^{87}$Rb$^{133}$Cs molecules in the $X^1\Sigma^+$ rovibronic ground state that is compatible with efficient mixing of the atoms in optical lattices. We first construct a model for the excited-state structure using constants found by fitting to spectroscopy of the relevant $a\,^3 \Sigma^+ → b\, ^3\Pi_1$ transitions at 181.5 G and 217.1 G. We then compare the predicted transition dipole moments from this model to those found for the transitions that have been successfully used for STIRAP at 181.5 G. We form molecules by magnetoassociation on a broad interspecies Feshbach resonance at 352.7 G and explore the pattern of Feshbach states near 305 G. This allows us to navigate to a suitable initial state for STIRAP by jumping across an avoided crossing with radiofrequency radiation. We identify suitable transitions for STIRAP at 305 G. We characterize these transitions experimentally and demonstrate STIRAP to a single hyperfine level of the ground state with a one-way efficiency of 85(4)%.
- Published
- 2023
- Full Text
- View/download PDF
8. Acute Myeloid Leukemia Case after Gene Therapy for Sickle Cell Disease
- Author
-
Sunita Goyal, John Tisdale, Manfred Schmidt, Julie Kanter, Jennifer Jaroscak, Dustin Whitney, Hans Bitter, Philip D. Gregory, Geoffrey Parsons, Marianna Foos, Ashish Yeri, Maple Gioia, Sarah B. Voytek, Alex Miller, Jessie Lynch, Richard A. Colvin, and Melissa Bonner
- Subjects
Adult ,Carcinogenesis ,Sequence Analysis, RNA ,Genetic Vectors ,Lentivirus ,Hematopoietic Stem Cell Transplantation ,Gene Expression ,Anemia, Sickle Cell ,Genetic Therapy ,beta-Globins ,General Medicine ,Transplantation, Autologous ,Leukemia, Myeloid, Acute ,Risk Factors ,Humans ,Female ,Transgenes - Abstract
Gene therapy with LentiGlobin for sickle cell disease (bb1111, lovotibeglogene autotemcel) consists of autologous transplantation of a patient's hematopoietic stem cells transduced with the BB305 lentiviral vector that encodes the β
- Published
- 2022
9. Diatomic-py: A Python module for calculating the rotational and hyperfine structure of 1Σ molecules
- Author
-
Jacob A. Blackmore, Philip D. Gregory, Jeremy M. Hutson, and Simon L. Cornish
- Subjects
Quantum Gases (cond-mat.quant-gas) ,Atomic Physics (physics.atom-ph) ,Hardware and Architecture ,FOS: Physical sciences ,General Physics and Astronomy ,Physics::Atomic Physics ,Computational Physics (physics.comp-ph) ,Condensed Matter - Quantum Gases ,Physics - Computational Physics ,Physics - Atomic Physics - Abstract
We present a computer program to calculate the quantised rotational and hyperfine energy levels of diatomic molecules in the presence of dc electric, dc magnetic, and off-resonant optical fields. Our program is applicable to the bialkali molecules used in ongoing state-of-the-art experiments with ultracold molecular gases. We include functions for the calculation of space-fixed electric dipole moments, magnetic moments and transition dipole moments. Program summary Program Title: Diatomic-Py CPC Library link to program files: https://doi.org/10.17632/3yfxnh5bn5.1 Developer's repository link: https://doi.org/10.5281/zenodo.6632148 Licensing provisions: BSD 3-clause Programming language: Python ≥ 3.7 Nature of problem: Calculation of the rotational and hyperfine structure of molecules in the presence of dc magnetic, dc electric, and off-resonant laser fields. Solution method: A matrix representation of the Hamiltonian is constructed in the uncoupled basis set. Eigenstates and eigenenergies are calculated by numerical diagonalization of the Hamiltonian. Additional comments including restrictions and unusual features: Restricted to calculating the Stark and Zeeman shifts with co-axial electric and magnetic fields.
- Published
- 2023
10. Robust storage qubits in ultracold polar molecules
- Author
-
Jacob A. Blackmore, Sarah L. Bromley, Philip D. Gregory, Jeremy M. Hutson, and Simon L. Cornish
- Subjects
Physics ,Quantum Physics ,Coherence time ,Quantum decoherence ,Atomic Physics (physics.atom-ph) ,FOS: Physical sciences ,General Physics and Astronomy ,Rotational–vibrational spectroscopy ,Physics - Atomic Physics ,Quantum Gases (cond-mat.quant-gas) ,Quantum state ,Qubit ,Quantum information ,Atomic physics ,Condensed Matter - Quantum Gases ,Quantum Physics (quant-ph) ,Hyperfine structure ,Quantum computer ,Coherence (physics) - Abstract
Quantum states with long-lived coherence are essential for quantum computation, simulation and metrology. The nuclear spin states of ultracold molecules prepared in the singlet rovibrational ground state are an excellent candidate for encoding and storing quantum information. However, it is important to understand all sources of decoherence for these qubits, and then eliminate them, in order to reach the longest possible coherence times. Here, we fully characterise the dominant mechanisms for decoherence of a storage qubit in an optically trapped ultracold gas of RbCs molecules using high-resolution Ramsey spectroscopy. Guided by a detailed understanding of the hyperfine structure of the molecule, we tune the magnetic field to where a pair of hyperfine states have the same magnetic moment. These states form a qubit, which is insensitive to variations in magnetic field. Our experiments reveal an unexpected differential tensor light shift between the states, caused by weak mixing of rotational states. We demonstrate how this light shift can be eliminated by setting the angle between the linearly polarised trap light and the applied magnetic field to a magic angle of $\arccos{(1/\sqrt{3})}\approx55^{\circ}$. This leads to a coherence time exceeding 6.9 s (90% confidence level). Our results unlock the potential of ultracold molecules as a platform for quantum computation., Comment: 19 pages, 12 figures
- Published
- 2021
11. Targeted gene therapy and cell reprogramming in Fanconi anemia
- Author
-
Paula Rio, Rocio Baños, Angelo Lombardo, Oscar Quintana‐Bustamante, Lara Alvarez, Zita Garate, Pietro Genovese, Elena Almarza, Antonio Valeri, Begoña Díez, Susana Navarro, Yaima Torres, Juan P Trujillo, Rodolfo Murillas, Jose C Segovia, Enrique Samper, Jordi Surralles, Philip D Gregory, Michael C Holmes, Luigi Naldini, and Juan A Bueren
- Subjects
cell reprogramming ,Fanconi anemia ,gene‐targeting ,iPSCs ,zinc finger nucleases ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Gene targeting is progressively becoming a realistic therapeutic alternative in clinics. It is unknown, however, whether this technology will be suitable for the treatment of DNA repair deficiency syndromes such as Fanconi anemia (FA), with defects in homology‐directed DNA repair. In this study, we used zinc finger nucleases and integrase‐defective lentiviral vectors to demonstrate for the first time that FANCA can be efficiently and specifically targeted into the AAVS1 safe harbor locus in fibroblasts from FA‐A patients. Strikingly, up to 40% of FA fibroblasts showed gene targeting 42 days after gene editing. Given the low number of hematopoietic precursors in the bone marrow of FA patients, gene‐edited FA fibroblasts were then reprogrammed and re‐differentiated toward the hematopoietic lineage. Analyses of gene‐edited FA‐iPSCs confirmed the specific integration of FANCA in the AAVS1 locus in all tested clones. Moreover, the hematopoietic differentiation of these iPSCs efficiently generated disease‐free hematopoietic progenitors. Taken together, our results demonstrate for the first time the feasibility of correcting the phenotype of a DNA repair deficiency syndrome using gene‐targeting and cell reprogramming strategies.
- Published
- 2014
- Full Text
- View/download PDF
12. Coherent manipulation of the internal state of ultracold 87Rb133Cs molecules with multiple microwave fields
- Author
-
Jacob A. Blackmore, Philip D. Gregory, Simon L. Cornish, and Sarah L. Bromley
- Subjects
Physics ,education.field_of_study ,Coherence time ,Field (physics) ,Atomic Physics (physics.atom-ph) ,Population ,FOS: Physical sciences ,General Physics and Astronomy ,Quantum simulator ,01 natural sciences ,Physics - Atomic Physics ,010305 fluids & plasmas ,Superposition principle ,Quantum Gases (cond-mat.quant-gas) ,0103 physical sciences ,Physical and Theoretical Chemistry ,Atomic physics ,Condensed Matter - Quantum Gases ,010306 general physics ,education ,Ground state ,Hyperfine structure ,Quantum computer - Abstract
We explore coherent multi-photon processes in $^{87}$Rb$^{133}$Cs molecules using 3-level lambda and ladder configurations of rotational and hyperfine states, and discuss their relevance to future applications in quantum computation and quantum simulation. In the lambda configuration, we demonstrate the driving of population between two hyperfine levels of the rotational ground state via a two-photon Raman transition. Such pairs of states may be used in the future as a quantum memory, and we measure a Ramsey coherence time for a superposition of these states of 58(9) ms. In the ladder configuration, we show that we can generate and coherently populate microwave dressed states via the observation of an Autler-Townes doublet. We demonstrate that we can control the strength of this dressing by varying the intensity of the microwave coupling field. Finally, we perform spectroscopy of the rotational states of $^{87}$Rb$^{133}$Cs up to $N=6$, highlighting the potential of ultracold molecules for quantum simulation in synthetic dimensions. By fitting the measured transition frequencies we determine a new value of the centrifugal distortion coefficient $D_v=h\times207.3(2)~$Hz.
- Published
- 2020
13. Allele-selective transcriptional repression of mutant HTT for the treatment of Huntington’s disease
- Author
-
Yasaman Ataei, Larry Park, D. James Surmeier, B. Joseph Vu, Seung Kwak, Richard T. Surosky, Anand Narayanan, David A. Shivak, Josee Laganiere, Christer Halldin, Andrea Varrone, Matthew C. Mendel, Karsten Tillack, Lei Zhang, Bryan Zeitler, Dmitry Guschin, Lexi Kopan, Sarah J. Hinkley, Kimberly Marlen, Jocelynn R. Pearl, Qi Yu, Taneli Heikkinen, Annette Gärtner, Yalda Sedaghat, Christina Thiede, Miklós Tóth, Jennifer M. Cherone, David Paschon, Jyothisri Kondapalli, Andrea E. Kudwa, Ladislav Mrzljak, Rainier Amora, Kimmo Lehtimäki, Edward J. Rebar, Lenke Tari, Ignacio Munoz-Sanjuan, Jeffrey C. Miller, Sylvie Ramboz, Marie Svedberg, Steven Froelich, Irina Ankoudinova, Philip D. Gregory, Stephen Lam, Michelle Day, Jonathan Bard, Hoang Oanh B. Nguyen, Fyodor D. Urnov, Davis Li, Jenny Haggkvist, H. Steve Zhang, and Guijuan Qiao
- Subjects
0301 basic medicine ,Zinc finger ,congenital, hereditary, and neonatal diseases and abnormalities ,Mutant ,General Medicine ,Biology ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,nervous system diseases ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Huntington's disease ,Transcription (biology) ,030220 oncology & carcinogenesis ,mental disorders ,medicine ,Gene silencing ,Allele ,Gene ,Transcription factor - Abstract
Huntington’s disease (HD) is a dominantly inherited neurodegenerative disorder caused by a CAG trinucleotide expansion in the huntingtin gene (HTT), which codes for the pathologic mutant HTT (mHTT) protein. Since normal HTT is thought to be important for brain function, we engineered zinc finger protein transcription factors (ZFP-TFs) to target the pathogenic CAG repeat and selectively lower mHTT as a therapeutic strategy. Using patient-derived fibroblasts and neurons, we demonstrate that ZFP-TFs selectively repress >99% of HD-causing alleles over a wide dose range while preserving expression of >86% of normal alleles. Other CAG-containing genes are minimally affected, and virally delivered ZFP-TFs are active and well tolerated in HD neurons beyond 100 days in culture and for at least nine months in the mouse brain. Using three HD mouse models, we demonstrate improvements in a range of molecular, histopathological, electrophysiological and functional endpoints. Our findings support the continued development of an allele-selective ZFP-TF for the treatment of HD. Zinc finger protein transcription factors are developed for the selective silencing of the mutant huntingtin gene in human neurons in vitro and multiple animal models of Huntington’s disease in vivo while preserving expression of the wild-type allele.
- Published
- 2019
14. Molecule-molecule and atom-molecule collisions with ultracold RbCs molecules
- Author
-
Luke M. Fernley, Jacob A. Blackmore, Sarah L. Bromley, Simon L. Cornish, Matthew D. Frye, Jeremy M. Hutson, and Philip D. Gregory
- Subjects
Physics ,Condensed Matter::Quantum Gases ,Atomic Physics (physics.atom-ph) ,Inelastic collision ,FOS: Physical sciences ,General Physics and Astronomy ,01 natural sciences ,Molecular physics ,Physics - Atomic Physics ,010305 fluids & plasmas ,Quantum Gases (cond-mat.quant-gas) ,0103 physical sciences ,Atom ,Molecule ,Physics::Atomic Physics ,Physics::Chemical Physics ,Condensed Matter - Quantum Gases ,010306 general physics ,Ground state ,Hyperfine structure ,Excitation - Abstract
Understanding ultracold collisions involving molecules is of fundamental importance for current experiments, where inelastic collisions typically limit the lifetime of molecular ensembles in optical traps. Here we present a broad study of optically trapped ultracold RbCs molecules in collisions with one another, in reactive collisions with Rb atoms, and in nonreactive collisions with Cs atoms. For experiments with RbCs alone, we show that by modulating the intensity of the optical trap, such that the molecules spend 75% of each modulation cycle in the dark, we partially suppress collisional loss of the molecules. This is evidence for optical excitation of molecule pairs mediated via sticky collisions. We find that the suppression is less effective for molecules not prepared in the spin-stretched hyperfine ground state. This may be due either to longer lifetimes for complexes or to laser-free decay pathways. For atom-molecule mixtures, RbCs+Rb and RbCs+Cs, we demonstrate that the rate of collisional loss of molecules scales linearly with the density of atoms. This indicates that, in both cases, the loss of molecules is rate-limited by two-body atom-molecule processes. For both mixtures, we measure loss rates that are below the thermally averaged universal limit., Comment: 21 pages, 9 figures
- Published
- 2021
- Full Text
- View/download PDF
15. Controlling the ac Stark effect of RbCs with dc electric and magnetic fields
- Author
-
Philip D. Gregory, Rahul Sawant, Sarah L. Bromley, Jesus Aldegunde, Simon L. Cornish, Jacob A. Blackmore, and Jeremy M. Hutson
- Subjects
Physics ,Condensed matter physics ,Atomic Physics (physics.atom-ph) ,FOS: Physical sciences ,Astrophysics::Cosmology and Extragalactic Astrophysics ,01 natural sciences ,010305 fluids & plasmas ,Magnetic field ,Physics - Atomic Physics ,symbols.namesake ,Stark effect ,Quantum Gases (cond-mat.quant-gas) ,Electric field ,0103 physical sciences ,symbols ,Physics::Atomic Physics ,Physics::Chemical Physics ,010306 general physics ,Condensed Matter - Quantum Gases ,Differential (mathematics) ,Microwave - Abstract
We investigate the effects of static electric and magnetic fields on the differential ac Stark shifts for microwave transitions in ultracold bosonic $^{87}$Rb$^{133}$Cs molecules, for light of wavelength $\lambda = 1064~\mathrm{nm}$. Near this wavelength we observe unexpected two-photon transitions that may cause trap loss. We measure the ac Stark effect in external magnetic and electric fields, using microwave spectroscopy of the first rotational transition. We quantify the isotropic and anisotropic parts of the molecular polarizability at this wavelength. We demonstrate that a modest electric field can decouple the nuclear spins from the rotational angular momentum, greatly simplifying the ac Stark effect. We use this simplification to control the ac Stark shift using the polarization angle of the trapping laser.
- Published
- 2020
16. Loss of Ultracold Rb87Cs133 Molecules via Optical Excitation of Long-Lived Two-Body Collision Complexes
- Author
-
Jacob A. Blackmore, Philip D. Gregory, Simon L. Cornish, and Sarah L. Bromley
- Subjects
Physics ,General Physics and Astronomy ,Collision ,01 natural sciences ,3. Good health ,Trap (computing) ,Modulation ,0103 physical sciences ,Molecule ,Atomic physics ,010306 general physics ,Frequency modulation ,Intensity (heat transfer) ,Excitation - Abstract
We show that the lifetime of ultracold ground-state ^{87}Rb^{133}Cs molecules in an optical trap is limited by fast optical excitation of long-lived two-body collision complexes. We partially suppress this loss mechanism by applying square-wave modulation to the trap intensity, such that the molecules spend 75% of each modulation cycle in the dark. By varying the modulation frequency, we show that the lifetime of the collision complex is 0.53±0.06 ms in the dark. We find that the rate of optical excitation of the collision complex is 3_{-2}^{+4}×10^{3} W^{-1} cm^{2} s^{-1} for λ=1550 nm, leading to a lifetime of
- Published
- 2020
17. Loss of Ultracold ^{87}Rb^{133}Cs Molecules via Optical Excitation of Long-Lived Two-Body Collision Complexes
- Author
-
Philip D, Gregory, Jacob A, Blackmore, Sarah L, Bromley, and Simon L, Cornish
- Abstract
We show that the lifetime of ultracold ground-state ^{87}Rb^{133}Cs molecules in an optical trap is limited by fast optical excitation of long-lived two-body collision complexes. We partially suppress this loss mechanism by applying square-wave modulation to the trap intensity, such that the molecules spend 75% of each modulation cycle in the dark. By varying the modulation frequency, we show that the lifetime of the collision complex is 0.53±0.06 ms in the dark. We find that the rate of optical excitation of the collision complex is 3_{-2}^{+4}×10^{3} W^{-1} cm^{2} s^{-1} for λ=1550 nm, leading to a lifetime of100 ns for typical trap intensities. These results explain the two-body loss observed in experiments on nonreactive bialkali molecules.
- Published
- 2020
18. Loss of Ultracold 87Rb133Cs Molecules via Optical Excitation of Long-Lived Two-Body Collison Complexes
- Author
-
Philip D. Gregory, Jacob A. Blackmore, Sarah L. Bromley, and Simon L. Cornish
- Published
- 2020
- Full Text
- View/download PDF
19. Genome Editing in Neuroepithelial Stem Cells to Generate Human Neurons with High Adenosine-Releasing Capacity
- Author
-
Philipp Koch, Christa E. Müller, Julius A. Steinbeck, Ruven Wilkens, Philip D. Gregory, Daniel Poppe, Marion Schneider, Julia Ladewig, Andreas Reik, David Paschon, Allison Tam, Jonas Doerr, and Oliver Brüstle
- Subjects
Gene‐editing ,0301 basic medicine ,Adenosine ,Neurodegeneration/Neurological Disorders ,Human Embryonic Stem Cells ,Cellular homeostasis ,Adenosine kinase ,Polymorphism, Single Nucleotide ,Mass Spectrometry ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Translational Research Articles and Reviews ,Human neurons ,Neural Stem Cells ,medicine ,Animals ,Humans ,Enabling Technologies for Cell-Based Clinical Translation ,Adenosine Kinase ,Chromatography, High Pressure Liquid ,Gene Editing ,Mice, Knockout ,Neurons ,Enabling Technologies for Cell‐Based Clinical Translation ,Adenosine secretion ,biology ,Cell Biology ,General Medicine ,Gene Delivery Systems/Gene Therapy/Gene Editing Technology ,Embryonic stem cell ,Zinc Finger Nucleases ,Neural stem cell ,Neural/Progenitor Stem Cells ,ADK ,Cell biology ,Mice, Inbred C57BL ,Neuroepithelial cell ,030104 developmental biology ,Karyotyping ,biology.protein ,Neuroepithelial stem cells ,Stem cell ,030217 neurology & neurosurgery ,Developmental Biology ,medicine.drug - Abstract
As a powerful regulator of cellular homeostasis and metabolism, adenosine is involved in diverse neurological processes including pain, cognition, and memory. Altered adenosine homeostasis has also been associated with several diseases such as depression, schizophrenia, or epilepsy. Based on its protective properties, adenosine has been considered as a potential therapeutic agent for various brain disorders. Since systemic application of adenosine is hampered by serious side effects such as vasodilatation and cardiac suppression, recent studies aim at improving local delivery by depots, pumps, or cell-based applications. Here, we report on the characterization of adenosine-releasing human embryonic stem cell-derived neuroepithelial stem cells (long-term self-renewing neuroepithelial stem [lt-NES] cells) generated by zinc finger nuclease (ZFN)-mediated knockout of the adenosine kinase (ADK) gene. ADK-deficient lt-NES cells and their differentiated neuronal and astroglial progeny exhibit substantially elevated release of adenosine compared to control cells. Importantly, extensive adenosine release could be triggered by excitation of differentiated neuronal cultures, suggesting a potential activity-dependent regulation of adenosine supply. Thus, ZFN-modified neural stem cells might serve as a useful vehicle for the activity-dependent local therapeutic delivery of adenosine into the central nervous system.
- Published
- 2018
20. Prostaglandin E2 Increases Lentiviral Vector Transduction Efficiency of Adult Human Hematopoietic Stem and Progenitor Cells
- Author
-
Holly Horton, Lauryn Christiansen, Amanda Hamel, Olivia Garijo, Bruce E. Torbett, Philip D. Gregory, Garrett C. Heffner, Gretchen Lewis, Yegor Smurnyy, Dakota Campbell, Melissa Bonner, Wenliang Zhang, Gabor Istvan Veres, Seema Shaw, Francis J. Pierciey, Kendrick A. Goss, and Mitchell Finer
- Subjects
0301 basic medicine ,Genetic enhancement ,CD34 ,Antigens, CD34 ,Transduction (genetics) ,Mice ,Transduction, Genetic ,Drug Discovery ,Transgenes ,Gene Transfer Techniques ,Hematopoietic stem cell ,transduction ,gene therapy ,Cell biology ,Globins ,Haematopoiesis ,medicine.anatomical_structure ,Molecular Medicine ,Original Article ,Genetic Vectors ,Transplantation, Heterologous ,Anemia, Sickle Cell ,Biology ,Dinoprostone ,Viral vector ,Cell Line ,03 medical and health sciences ,vector copy number ,Genetics ,medicine ,hemoglobinopathy ,Animals ,Humans ,Progenitor cell ,Molecular Biology ,Gene Library ,Pharmacology ,Severe combined immunodeficiency ,prostaglandin E2 ,lentiviral vector ,Lentivirus ,beta-Thalassemia ,Genetic Therapy ,Virus Internalization ,medicine.disease ,Hematopoietic Stem Cells ,030104 developmental biology ,Immunology ,Leukocyte Common Antigens ,hematopoietic stem cell - Abstract
Gene therapy currently in development for hemoglobinopathies utilizes ex vivo lentiviral transduction of CD34+ hematopoietic stem and progenitor cells (HSPCs). A small-molecule screen identified prostaglandin E2 (PGE2) as a positive mediator of lentiviral transduction of CD34+ cells. Supplementation with PGE2 increased lentiviral vector (LVV) transduction of CD34+ cells approximately 2-fold compared to control transduction methods with no effect on cell viability. Transduction efficiency was consistently increased in primary CD34+ cells from multiple normal human donors and from patients with β-thalassemia or sickle cell disease. Notably, PGE2 increased transduction of repopulating human HSPCs in an immune-deficient (nonobese diabetic/severe combined immunodeficiency/interleukin-2 gamma receptor null [NSG]) xenotransplantation mouse model without evidence of in vivo toxicity, lineage bias, or a de novo bias of lentiviral integration sites. These data suggest that PGE2 improves lentiviral transduction and increases vector copy number, therefore resulting in increased transgene expression. As a result, PGE2 may be useful in clinical gene therapy applications using lentivirally modified HSPCs., Heffner et al. performed a small-molecule screen and identified prostaglandin E2 (PGE2) as a positive mediator of lentiviral transduction of CD34+ cells. Their data suggest that PGE2-mediated improvements in transduction efficiency may aid in clinical gene therapy applications using lentivirally modified HSPCs.
- Published
- 2017
21. Sensitive and adaptable pharmacological control of CAR T cells through extracellular receptor dimerization
- Author
-
Wai-Hang Leung, Joel Gay, Unja Martin, Tracy E. Garrett, Holly M. Horton, Michael T. Certo, Bruce R. Blazar, Richard A. Morgan, Philip D. Gregory, Jordan Jarjour, and Alexander Astrakhan
- Subjects
0301 basic medicine ,Antigen Targeting ,medicine.medical_treatment ,T cell ,Recombinant Fusion Proteins ,T-Lymphocytes ,Antigens, CD19 ,Lymphocyte Activation ,Immunotherapy, Adoptive ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,Antigen ,Protein Domains ,Cell Line, Tumor ,Neoplasms ,medicine ,Extracellular ,Animals ,Humans ,Receptor ,Promoter Regions, Genetic ,Sirolimus ,Receptors, Chimeric Antigen ,Chemistry ,General Medicine ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Signal transduction ,Protein Multimerization ,Single-Chain Antibodies ,Research Article - Abstract
Chimeric antigen receptor (CAR) T cell therapies have achieved promising outcomes in several cancers, however more challenging oncology indications may necessitate advanced antigen receptor designs and functions. Here we describe a bipartite receptor system comprised of separate antigen targeting and signal transduction polypeptides, each containing an extracellular dimerization domain. We demonstrate that T cell activation remains antigen dependent but can only be achieved in the presence of a dimerizing drug, rapamycin. Studies performed in vitro and in xenograft mouse models illustrate equivalent to superior anti-tumor potency compared to currently used CAR designs, and at rapamycin concentrations well below immunosuppressive levels. We further show that the extracellular positioning of the dimerization domains enables the administration of recombinant re-targeting modules, potentially extending antigen targeting. Overall, this novel regulatable CAR design has exquisite drug sensitivity, provides robust anti-tumor responses, and is uniquely flexible for multiplex antigen targeting or retargeting, which may further assist the development of safe, potent and durable T cell therapeutics.
- Published
- 2019
22. Gene editing of the multi-copy H2A.B gene and its importance for fertility
- Author
-
Peter Koopman, David J. Tremethick, Tatiana A. Soboleva, Lei Zhang, Josephine Bowles, Matthew A. Field, Sebastian Kurscheid, Philip D. Gregory, Thierry Buchou, Nur Diana Anuar, and Edward J. Rebar
- Subjects
Male ,lcsh:QH426-470 ,Chromosomal Proteins, Non-Histone ,Gene Expression ,RNA polymerase II ,Histones ,03 medical and health sciences ,Pre-mRNA splicing ,0302 clinical medicine ,Transcription Activator-Like Effector Nucleases ,Gene expression ,Animals ,Gene family ,lcsh:QH301-705.5 ,Gene ,Infertility, Male ,030304 developmental biology ,Gene Editing ,Mice, Knockout ,Histone variants ,Genetics ,0303 health sciences ,Base Sequence ,biology ,Research ,Splicing speckles ,Spermatozoa ,Chromatin ,lcsh:Genetics ,TALENs ,Fertility ,Histone ,lcsh:Biology (General) ,Mutation ,RNA splicing ,Knockout mouse ,biology.protein ,Female ,H2A.B ,030217 neurology & neurosurgery ,Genome editing - Abstract
Background Altering the biochemical makeup of chromatin by the incorporation of histone variants during development represents a key mechanism in regulating gene expression. The histone variant H2A.B, H2A.B.3 in mice, appeared late in evolution and is most highly expressed in the testis. In the mouse, it is encoded by three different genes. H2A.B expression is spatially and temporally regulated during spermatogenesis being most highly expressed in the haploid round spermatid stage. Active genes gain H2A.B where it directly interacts with polymerase II and RNA processing factors within splicing speckles. However, the importance of H2A.B for gene expression and fertility are unknown. Results Here, we report the first mouse knockout of this histone variant and its effects on fertility, nuclear organization, and gene expression. In view of the controversy related to the generation of off-target mutations by gene editing approaches, we test the specificity of TALENs by disrupting the H2A.B multi-copy gene family using only one pair of TALENs. We show that TALENs do display a high level of specificity since no off-target mutations are detected by bioinformatics analyses of exome sequences obtained from three consecutive generations of knockout mice and by Sanger DNA sequencing. Male H2A.B.3 knockout mice are subfertile and display an increase in the proportion of abnormal sperm and clogged seminiferous tubules. Significantly, a loss of proper RNA Pol II targeting to distinct transcription–splicing territories and changes to pre-mRNA splicing are observed. Conclusion We have produced the first H2A.B knockout mouse using the TALEN approach. Electronic supplementary material The online version of this article (10.1186/s13059-019-1633-3) contains supplementary material, which is available to authorized users.
- Published
- 2019
23. Production of Ultracold 87 Rb 133 Cs in the Absolute Ground State: Complete Characterisation of the Stimulated Raman Adiabatic Passage Transfer
- Author
-
Simon L. Cornish, Jeremy M. Hutson, Peter K. Molony, Philip D. Gregory, C. Ruth Le Sueur, and Avinash Kumar
- Subjects
Monte Carlo method ,Stimulated Raman adiabatic passage ,chemistry.chemical_element ,Rotational–vibrational spectroscopy ,01 natural sciences ,Atomic and Molecular Physics, and Optics ,010305 fluids & plasmas ,Rubidium ,Laser linewidth ,chemistry ,Caesium ,0103 physical sciences ,Physics::Atomic Physics ,Physical and Theoretical Chemistry ,Atomic physics ,010306 general physics ,Ground state ,Hyperfine structure - Abstract
We present the production of ultracold 87 RbCs molecules in the electronic, rovibrational and hyperfine ground state, using stimulated Raman adiabatic passage to transfer the molecules from a weakly bound Feshbach state. We measure one-way transfer efficiencies of 92(1)% and fully characterise the strengths and linewidths of the transitions used. We model the transfer, including a Monte Carlo simulation of the laser noise, and find this matches well with both the transfer efficiency and our previous measurements of the laser linewidth and frequency drifts.
- Published
- 2016
24. Long-term multilineage engraftment of autologous genome-edited hematopoietic stem cells in nonhuman primates
- Author
-
Jenny Jiacheng Yan, Richard G. Trimble, Edward J. Rebar, Philip D. Gregory, Jianbin Wang, Christopher W. Peterson, Collette K. Tse, Krystin K. Norman, Hans-Peter Kiem, Michael C. Holmes, Olivier Humbert, David A. Shivak, and Zachary K. Norgaard
- Subjects
0301 basic medicine ,Transplantation Conditioning ,Receptors, CCR5 ,medicine.medical_treatment ,Molecular Sequence Data ,Immunology ,Hematopoietic stem cell transplantation ,Biology ,Polymerase Chain Reaction ,Transplantation, Autologous ,Biochemistry ,Cell Line ,03 medical and health sciences ,Genome editing ,medicine ,Animals ,Autologous transplantation ,Cell Lineage ,Amino Acid Sequence ,RNA, Messenger ,Progenitor cell ,Bone Marrow Transplantation ,Gene Editing ,fungi ,Graft Survival ,Hematopoietic Stem Cell Transplantation ,food and beverages ,Zinc Fingers ,Gene Therapy ,Sequence Analysis, DNA ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Transplantation ,Haematopoiesis ,Electroporation ,030104 developmental biology ,Gene Knockdown Techniques ,Mutation ,Cancer research ,Feasibility Studies ,Macaca nemestrina ,Stem cell ,Whole-Body Irradiation ,Ex vivo - Abstract
Genome editing in hematopoietic stem and progenitor cells (HSPCs) is a promising novel technology for the treatment of many human diseases. Here, we evaluated whether the disruption of the C-C chemokine receptor 5 (CCR5) locus in pigtailed macaque HSPCs by zinc finger nucleases (ZFNs) was feasible. We show that macaque-specific CCR5 ZFNs efficiently induce CCR5 disruption at levels of up to 64% ex vivo, 40% in vivo early posttransplant, and 3% to 5% in long-term repopulating cells over 6 months following HSPC transplant. These genome-edited HSPCs support multilineage engraftment and generate progeny capable of trafficking to secondary tissues including the gut. Using deep sequencing technology, we show that these ZFNs are highly specific for the CCR5 locus in primary cells. Further, we have adapted our clonal tracking methodology to follow individual CCR5 mutant cells over time in vivo, reinforcing that CCR5 gene-edited HSPCs are capable of long-term engraftment. Together, these data demonstrate that genome-edited HSPCs engraft, and contribute to multilineage repopulation after autologous transplantation in a clinically relevant large animal model, an important step toward the development of stem cell-based genome-editing therapies for HIV and potentially other diseases as well.
- Published
- 2016
25. Absence of WASp Enhances Hematopoietic and Megakaryocytic Differentiation in a Human Embryonic Stem Cell Model
- Author
-
Miguel G. Toscano, Philip D. Gregory, Olaf Neth, Francisco Martin, Almudena Sánchez-Gilabert, Karim Benabdellah, Marién Cobo, Pilar Muñoz, Per Anderson, Pedro J. Real, Verónica Ramos-Mejía, Michael C. Holmes, and Agueda Molinos-Quintana
- Subjects
Platelet Membrane Glycoprotein IIb ,0301 basic medicine ,Cellular differentiation ,CD34 ,Antigens, CD34 ,Stem cell factor ,Models, Biological ,Cell Line ,Gene Knockout Techniques ,03 medical and health sciences ,Drug Discovery ,Genetics ,Humans ,Progenitor cell ,Molecular Biology ,Embryonic Stem Cells ,Pharmacology ,biology ,Wiskott–Aldrich syndrome protein ,Cell Differentiation ,Hematopoietic Stem Cells ,Embryonic stem cell ,Cell biology ,Haematopoiesis ,030104 developmental biology ,Cell culture ,Immunology ,biology.protein ,Leukocyte Common Antigens ,Molecular Medicine ,Original Article ,Megakaryocytes ,Wiskott-Aldrich Syndrome Protein - Abstract
The Wiskott-Aldrich syndrome (WAS) is an X-linked primary immunodeficiency caused by mutations in the WAS gene and characterized by severe thrombocytopenia. Although the role of WASp in terminally differentiated lymphocytes and myeloid cells is well characterized, its role in early hematopoietic differentiation and in platelets (Plts) biology is poorly understood. In the present manuscript, we have used zinc finger nucleases targeted to the WAS locus for the development of two isogenic WAS knockout (WASKO) human embryonic stem cell lines (hESCs). Upon hematopoietic differentiation, hESCs-WASKO generated increased ratios of CD34(+)CD45(+) progenitors with altered responses to stem cell factor compared to hESCs-WT. When differentiated toward the megakaryocytic linage, hESCs-WASKO produced increased numbers of CD34(+)CD41(+) progenitors, megakaryocytes (MKs), and Plts. hESCs-WASKO-derived MKs and Plts showed altered phenotype as well as defective responses to agonist, mimicking WAS patients MKs and Plts defects. Interestingly, the defects were more evident in WASp-deficient MKs than in WASp-deficient Plts. Importantly, ectopic WAS expression using lentiviral vectors restored normal Plts development and MKs responses. These data validate the AND-1_WASKO cell lines as a human cellular model for basic research and for preclinical studies for WAS.
- Published
- 2016
26. Molecular Evidence of Genome Editing in a Mouse Model of Immunodeficiency
- Author
-
Jianbin Wang, Chv Gan, Cynthia C. Bartholomae, F. J. Molina-Estevez, Christine Kinnon, Richard Gabriel, HH Abdul-Razak, Michael C. Holmes, V. Prakash, Celine J. Rocca, Rafael J. Yáñez-Muñoz, Philip D. Gregory, J Pantoglou, JA Bueren, M. E. Alonso-Ferrero, Adam Roberts, C von Kalle, Guillermo Guenechea, Steven J. Howe, Marina I. Garin, Adrian J. Thrasher, Michael P. Blundell, and Manfred Schmidt
- Subjects
0301 basic medicine ,lcsh:Medicine ,DNA-Activated Protein Kinase ,Mice, SCID ,Biology ,Genome ,Article ,Mice ,03 medical and health sciences ,Genome editing ,medicine ,Animals ,Humans ,lcsh:Science ,Immunodeficiency ,Gene Editing ,Severe combined immunodeficiency ,Multidisciplinary ,lcsh:R ,Nuclear Proteins ,medicine.disease ,Zinc finger nuclease ,3. Good health ,Transplantation ,Disease Models, Animal ,030104 developmental biology ,Cancer research ,Primary immunodeficiency ,Severe Combined Immunodeficiency ,lcsh:Q ,Ex vivo - Abstract
Genome editing is the introduction of directed modifications in the genome, a process boosted to therapeutic levels by designer nucleases. Building on the experience of ex vivo gene therapy for severe combined immunodeficiencies, it is likely that genome editing of haematopoietic stem/progenitor cells (HSPC) for correction of inherited blood diseases will be an early clinical application. We show molecular evidence of gene correction in a mouse model of primary immunodeficiency. In vitro experiments in DNA-dependent protein kinase catalytic subunit severe combined immunodeficiency (Prkdc scid) fibroblasts using designed zinc finger nucleases (ZFN) and a repair template demonstrated molecular and functional correction of the defect. Following transplantation of ex vivo gene-edited Prkdc scid HSPC, some of the recipient animals carried the expected genomic signature of ZFN-driven gene correction. In some primary and secondary transplant recipients we detected double-positive CD4/CD8 T-cells in thymus and single-positive T-cells in blood, but no other evidence of immune reconstitution. However, the leakiness of this model is a confounding factor for the interpretation of the possible T-cell reconstitution. Our results provide support for the feasibility of rescuing inherited blood disease by ex vivo genome editing followed by transplantation, and highlight some of the challenges.
- Published
- 2018
27. In vivo genome editing of the albumin locus as a platform for protein replacement therapy
- Author
-
Katherine A. High, Thomas Wechsler, Jeffrey C. Miller, Philip D. Gregory, Edward J. Rebar, Robert J. Davidson, Russell Dekelver, Julianne M. Rieders, Rajiv Sharma, David Paschon, Michael C. Holmes, Yannick Doyon, Scott Sproul, Xavier M. Anguela, David A. Shivak, and Shangzhen Zhou
- Subjects
Mucopolysaccharidosis I ,Transgene ,Genetic Vectors ,Immunology ,Computational biology ,Biology ,Hemophilia A ,Real-Time Polymerase Chain Reaction ,Hemophilia B ,Biochemistry ,Genome ,Factor IX ,Mice ,Protein replacement therapy ,Genome editing ,Albumins ,Animals ,Humans ,Enzyme Replacement Therapy ,RNA, Messenger ,Transgenes ,Promoter Regions, Genetic ,Gene ,Mucopolysaccharidosis II ,Zinc finger ,Genetics ,Factor VIII ,Gaucher Disease ,Reverse Transcriptase Polymerase Chain Reaction ,High-Throughput Nucleotide Sequencing ,Zinc Fingers ,Genetic Therapy ,Gene Therapy ,Cell Biology ,Hematology ,Dependovirus ,Endonucleases ,Zinc finger nuclease ,Mice, Inbred C57BL ,Liver ,RNA editing ,Fabry Disease ,RNA Editing ,Lysosomes - Abstract
Site-specific genome editing provides a promising approach for achieving long-term, stable therapeutic gene expression. Genome editing has been successfully applied in a variety of preclinical models, generally focused on targeting the diseased locus itself; however, limited targeting efficiency or insufficient expression from the endogenous promoter may impede the translation of these approaches, particularly if the desired editing event does not confer a selective growth advantage. Here we report a general strategy for liver-directed protein replacement therapies that addresses these issues: zinc finger nuclease (ZFN) -mediated site-specific integration of therapeutic transgenes within the albumin gene. By using adeno-associated viral (AAV) vector delivery in vivo, we achieved long-term expression of human factors VIII and IX (hFVIII and hFIX) in mouse models of hemophilia A and B at therapeutic levels. By using the same targeting reagents in wild-type mice, lysosomal enzymes were expressed that are deficient in Fabry and Gaucher diseases and in Hurler and Hunter syndromes. The establishment of a universal nuclease-based platform for secreted protein production would represent a critical advance in the development of safe, permanent, and functional cures for diverse genetic and nongenetic diseases.
- Published
- 2015
28. Clinical Scale Zinc Finger Nuclease-mediated Gene Editing of PD-1 in Tumor Infiltrating Lymphocytes for the Treatment of Metastatic Melanoma
- Author
-
Joal D. Beane, David Paschon, Richard A. Morgan, Mary A. Black, Martin A. Giedlin, Andreas Reik, Philip D. Gregory, Zhili Zheng, Jeffrey C. Miller, Gary Lee, Daniel Abate-Daga, Michael C. Holmes, Matthew C. Mendel, Edward J. Rebar, Dmitry Guschin, Zhiya Yu, Smita S. Chandran, Mini Bharathan, Steven A. Feldman, Nicholas P. Restifo, Nimisha Gandhi, Dale Ando, and Steven A. Rosenberg
- Subjects
Adoptive cell transfer ,medicine.medical_treatment ,Green Fluorescent Proteins ,Programmed Cell Death 1 Receptor ,Population ,Cell Separation ,Biology ,Lymphocyte Activation ,Immunotherapy, Adoptive ,Interferon-gamma ,Mice ,Lymphocytes, Tumor-Infiltrating ,Endoribonucleases ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Interferon gamma ,Neoplasm Metastasis ,education ,Melanoma ,Molecular Biology ,Alleles ,Pharmacology ,Zinc finger ,education.field_of_study ,Tumor Necrosis Factor-alpha ,Tumor-infiltrating lymphocytes ,Granulocyte-Macrophage Colony-Stimulating Factor ,Zinc Fingers ,Immunotherapy ,Flow Cytometry ,medicine.disease ,Zinc finger nuclease ,Gene Expression Regulation, Neoplastic ,Phenotype ,Immunology ,Cancer research ,Cytokines ,Molecular Medicine ,Original Article ,Female ,Immunologic Memory ,Neoplasm Transplantation ,medicine.drug - Abstract
Programmed cell death-1 (PD-1) is expressed on activated T cells and represents an attractive target for gene-editing of tumor targeted T cells prior to adoptive cell transfer (ACT). We used zinc finger nucleases (ZFNs) directed against the gene encoding human PD-1 (PDCD-1) to gene-edit melanoma tumor infiltrating lymphocytes (TIL). We show that our clinical scale TIL production process yielded efficient modification of the PD-1 gene locus, with an average modification frequency of 74.8% (n = 3, range 69.9-84.1%) of the alleles in a bulk TIL population, which resulted in a 76% reduction in PD-1 surface-expression. Forty to 48% of PD-1 gene-edited cells had biallelic PD-1 modification. Importantly, the PD-1 gene-edited TIL product showed improved in vitro effector function and a significantly increased polyfunctional cytokine profile (TNFα, GM-CSF, and IFNγ) compared to unmodified TIL in two of the three donors tested. In addition, all donor cells displayed an effector memory phenotype and expanded approximately 500-2,000-fold in vitro. Thus, further study to determine the efficiency and safety of adoptive cell transfer using PD-1 gene-edited TIL for the treatment of metastatic melanoma is warranted.
- Published
- 2015
29. Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells
- Author
-
Zulema Romero, Edward J. Rebar, Andrew Wilber, Roger P. Hollis, Matthew C. Mendel, Lei Zhang, Fabrizia Urbinati, David Gray, Alok V. Joglekar, Michael L. Kaufman, Allen Zhu, Shantha Senadheera, Michael C. Holmes, Aaron R. Cooper, Philip D. Gregory, David Paschon, Michelle Ho, Megan D. Hoban, Dianne Lumaquin, Donald B. Kohn, Gregory J. Cost, Andreas Reik, Georgia R. Lill, Xiaoyan Wang, and Pei-Qi Liu
- Subjects
medicine.medical_treatment ,Molecular Sequence Data ,Immunology ,CD34 ,Antigens, CD34 ,Bone Marrow Cells ,Anemia, Sickle Cell ,Mice, SCID ,beta-Globins ,Hematopoietic stem cell transplantation ,Biology ,Biochemistry ,Mice ,Mice, Inbred NOD ,medicine ,Animals ,Humans ,Progenitor cell ,Cells, Cultured ,Severe combined immunodeficiency ,Endodeoxyribonucleases ,Base Sequence ,Point mutation ,Hematopoietic Stem Cell Transplantation ,Zinc Fingers ,Genetic Therapy ,Cell Biology ,Hematology ,Fetal Blood ,Hematopoietic Stem Cells ,medicine.disease ,Molecular biology ,Haematopoiesis ,medicine.anatomical_structure ,Genetic Loci ,Mutation ,Bone marrow ,Stem cell - Abstract
Sickle cell disease (SCD) is characterized by a single point mutation in the seventh codon of the β-globin gene. Site-specific correction of the sickle mutation in hematopoietic stem cells would allow for permanent production of normal red blood cells. Using zinc-finger nucleases (ZFNs) designed to flank the sickle mutation, we demonstrate efficient targeted cleavage at the β-globin locus with minimal off-target modification. By co-delivering a homologous donor template (either an integrase-defective lentiviral vector or a DNA oligonucleotide), high levels of gene modification were achieved in CD34(+) hematopoietic stem and progenitor cells. Modified cells maintained their ability to engraft NOD/SCID/IL2rγ(null) mice and to produce cells from multiple lineages, although with a reduction in the modification levels relative to the in vitro samples. Importantly, ZFN-driven gene correction in CD34(+) cells from the bone marrow of patients with SCD resulted in the production of wild-type hemoglobin tetramers.
- Published
- 2015
30. Comparison of Zinc Finger Nucleases Versus CRISPR-Specific Nucleases for Genome Editing of the Wiskott-Aldrich Syndrome Locus
- Author
-
Marién Cobo, Francisco Martin, Alejandra Gutierrez-Guerrero, Giuseppe Galvani, Sabina Sánchez-Hernández, Philip D. Gregory, Javier Pinedo-Gomez, Araceli Aguilar-González, Almudena Sánchez-Gilabert, Karim Benabdellah, Michael C. Holmes, and Rocio Martin-Guerra
- Subjects
0301 basic medicine ,Wiskott–Aldrich syndrome ,Nucleofection ,Biology ,03 medical and health sciences ,Plasmid ,Genome editing ,Transduction, Genetic ,Genetics ,medicine ,CRISPR ,Humans ,Guide RNA ,Molecular Biology ,Gene Editing ,Cas9 ,Lentivirus ,medicine.disease ,Zinc finger nuclease ,Zinc Finger Nucleases ,Wiskott-Aldrich Syndrome ,030104 developmental biology ,Genetic Loci ,Molecular Medicine ,CRISPR-Cas Systems ,K562 Cells - Abstract
Primary immunodeficiencies, including Wiskott-Aldrich syndrome (WAS), are a main target for genome-editing strategies using specific nucleases (SNs) because a small number of corrected hematopoietic stem cells could cure patients. In this work, we have designed various WAS gene-specific CRISPR/Cas9 systems and compared their efficiency and specificity with homodimeric and heterodimeric WAS-specific zinc finger nucleases (ZFNs), using K-562 cells as a cellular model and plasmid nucleofection or integration-deficient lentiviral vectors (IDLVs) for delivery. The various CRISPR/Cas9 and ZFN SNs showed similar efficiency when using plasmid nucleofection for delivery. However, dual IDLVs expressing ZFNs were more efficient than dual IDLVs expressing Cas9 and guide RNA or all-in-one IDLVs, expressing Cas9 and guide RNA in the same vector. The specificity of heterodimeric ZFNs and CRISPR/Cas9, measured by increments in γ-H2AX focus formation in WAS-edited cells, was similar for both, and both outperformed homodimeric ZFNs independently of the delivery system used. Interestingly, we show that delivery of SNs, using IDLVs, is more efficient and less genotoxic than plasmid nucleofection. We also show the similar behavior of heterodimeric ZFNs and CRISPR/Cas9 for homology-directed gene knock-in strategies, with 88 and 83% of the donors inserted in the WAS locus, respectively, whereas when using homodimeric ZFNs only 45% of the insertions were on target. In summary, our data indicate that CRISPR/Cas9 and heterodimeric ZFNs are both good alternatives to further develop SN-based gene therapy strategies for WAS. However, IDLV delivery of WAS-specific heterodimeric ZFNs was the best option of all systems compared in this study.
- Published
- 2017
31. ac Stark effect in ultracold polar Rb87Cs133 molecules
- Author
-
Jesus Aldegunde, Jeremy M. Hutson, Jacob A. Blackmore, Simon L. Cornish, and Philip D. Gregory
- Subjects
Physics ,Linear polarization ,Polarization (waves) ,01 natural sciences ,010305 fluids & plasmas ,symbols.namesake ,Wavelength ,Stark effect ,Polarizability ,Excited state ,0103 physical sciences ,symbols ,Physics::Atomic Physics ,Rotational spectroscopy ,Atomic physics ,010306 general physics ,Hyperfine structure - Abstract
We investigate the effect of far-off-resonant trapping light on ultracold bosonic 87Rb133Cs molecules. We use kHz-precision microwave spectroscopy to measure the differential AC~Stark shifts between the ground and first excited rotational levels of the molecule with hyperfine-state resolution. We demonstrate through both experiment and theory that coupling between neighboring hyperfine states manifests in rich structure with many avoided crossings. This coupling may be tuned by rotating the polarization of the linearly polarized trapping light. A combination of spectroscopic and parametric heating measurements allows complete characterization of the molecule polarizability at a wavelength of 1550~nm in both the ground and first excited rotational states.
- Published
- 2017
32. Allele-selective transcriptional repression of mutant HTT for the treatment of Huntington's disease
- Author
-
Bryan, Zeitler, Steven, Froelich, Kimberly, Marlen, David A, Shivak, Qi, Yu, Davis, Li, Jocelynn R, Pearl, Jeffrey C, Miller, Lei, Zhang, David E, Paschon, Sarah J, Hinkley, Irina, Ankoudinova, Stephen, Lam, Dmitry, Guschin, Lexi, Kopan, Jennifer M, Cherone, Hoang-Oanh B, Nguyen, Guijuan, Qiao, Yasaman, Ataei, Matthew C, Mendel, Rainier, Amora, Richard, Surosky, Josee, Laganiere, B Joseph, Vu, Anand, Narayanan, Yalda, Sedaghat, Karsten, Tillack, Christina, Thiede, Annette, Gärtner, Seung, Kwak, Jonathan, Bard, Ladislav, Mrzljak, Larry, Park, Taneli, Heikkinen, Kimmo K, Lehtimäki, Marie M, Svedberg, Jenny, Häggkvist, Lenke, Tari, Miklós, Tóth, Andrea, Varrone, Christer, Halldin, Andrea E, Kudwa, Sylvie, Ramboz, Michelle, Day, Jyothisri, Kondapalli, D James, Surmeier, Fyodor D, Urnov, Philip D, Gregory, Edward J, Rebar, Ignacio, Muñoz-Sanjuán, and H Steve, Zhang
- Subjects
Male ,Huntingtin Protein ,Transcription, Genetic ,Zinc Fingers ,Neuroprotection ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Huntington Disease ,Trinucleotide Repeats ,Mutation ,Mice, Inbred CBA ,Animals ,Humans ,Female ,Alleles ,Cells, Cultured - Abstract
Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by a CAG trinucleotide expansion in the huntingtin gene (HTT), which codes for the pathologic mutant HTT (mHTT) protein. Since normal HTT is thought to be important for brain function, we engineered zinc finger protein transcription factors (ZFP-TFs) to target the pathogenic CAG repeat and selectively lower mHTT as a therapeutic strategy. Using patient-derived fibroblasts and neurons, we demonstrate that ZFP-TFs selectively repress 99% of HD-causing alleles over a wide dose range while preserving expression of 86% of normal alleles. Other CAG-containing genes are minimally affected, and virally delivered ZFP-TFs are active and well tolerated in HD neurons beyond 100 days in culture and for at least nine months in the mouse brain. Using three HD mouse models, we demonstrate improvements in a range of molecular, histopathological, electrophysiological and functional endpoints. Our findings support the continued development of an allele-selective ZFP-TF for the treatment of HD.
- Published
- 2017
33. Genetic and molecular identification of three human TPP1 functions in telomerase action: recruitment, activation, and homeostasis set point regulation
- Author
-
Samuel G. Regalado, Dirk Hockemeyer, Fyodor D. Urnov, Philip D. Gregory, Colleen M. O'Neil, Rudolf Jaenisch, Kathleen Collins, Gregory J. Cost, Christine S. Lai, Alec N. Sexton, Massachusetts Institute of Technology. Department of Biology, Lai, Christine S., and Jaenisch, Rudolf
- Subjects
Telomerase ,1.1 Normal biological development and functioning ,Cellular differentiation ,Telomere-Binding Proteins ,Stem Cell Research - Embryonic - Non-Human ,Biology ,telomerase ,Regenerative Medicine ,Medical and Health Sciences ,Shelterin Complex ,Gene Knockout Techniques ,Telomerase RNA component ,Underpinning research ,Genetics ,Humans ,Telomerase reverse transcriptase ,telomere maintenance ,Embryonic Stem Cells ,Cancer ,Telomere-binding protein ,Proto-Oncogene Proteins c-ets ,Genetic Complementation Test ,Psychology and Cognitive Sciences ,human genome engineering ,Telomere Homeostasis ,Telomere ,Biological Sciences ,Stem Cell Research ,Shelterin ,Cell biology ,Repressor Proteins ,Enzyme Activation ,Stem Cell Research - Nonembryonic - Non-Human ,Generic health relevance ,Stem cell ,shelterin ,Developmental Biology - Abstract
Telomere length homeostasis is essential for the long-term survival of stem cells, and its set point determines the proliferative capacity of differentiated cell lineages by restricting the reservoir of telomeric repeats. Knockdown and overexpression studies in human tumor cells showed that the shelterin subunit TPP1 recruits telomerase to telomeres through a region termed the TEL patch. However, these studies do not resolve whether the TPP1 TEL patch is the only mechanism for telomerase recruitment and whether telomerase regulation studied in tumor cells is representative of nontransformed cells such as stem cells. Using genome engineering of human embryonic stem cells, which have physiological telomere length homeostasis, we establish that the TPP1 TEL patch is genetically essential for telomere elongation and thus long-term cell viability. Furthermore, genetic bypass, protein fusion, and intragenic complementation assays define two distinct additional mechanisms of TPP1 involvement in telomerase action at telomeres. We demonstrate that TPP1 provides an essential step of telomerase activation as well as feedback regulation of telomerase by telomere length, which is necessary to determine the appropriate telomere length set point in human embryonic stem cells. These studies reveal and resolve multiple TPP1 roles in telomere elongation and stem cell telomere length homeostasis. Keywords: embryonic stem cells; human genome engineering; shelterin; telomerase telomere maintenance, National Institutes of Health (U.S.) (Grant R37-CA084198), National Institutes of Health (U.S.) (Grant RO1-CA087869), National Institutes of Health (U.S.) (Grant RO1-HD045022)
- Published
- 2014
34. Reactivation of Developmentally Silenced Globin Genes by Forced Chromatin Looping
- Author
-
Ann Dean, Gerd A. Blobel, Andreas Reik, Laura Breda, Wulan Deng, Ivan Krivega, Stefano Rivella, Jeremy Rupon, Kristen S. Jahn, Irene Motta, and Philip D. Gregory
- Subjects
Transcriptional Activation ,Erythroblasts ,Primary Cell Culture ,Antigens, CD34 ,beta-Globins ,Editorials: Cell Cycle Features ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Transcription (biology) ,hemic and lymphatic diseases ,Gene expression ,Animals ,Humans ,Enhancer ,Fetal Hemoglobin ,Locus control region ,Zinc finger ,Biochemistry, Genetics and Molecular Biology(all) ,Promoter ,Embryo, Mammalian ,Locus Control Region ,Molecular biology ,Embryonic stem cell ,Chromatin ,Hemoglobinopathies ,Genetic Techniques - Abstract
SummaryDistal enhancers commonly contact target promoters via chromatin looping. In erythroid cells, the locus control region (LCR) contacts β-type globin genes in a developmental stage-specific manner to stimulate transcription. Previously, we induced LCR-promoter looping by tethering the self-association domain (SA) of Ldb1 to the β-globin promoter via artificial zinc fingers. Here, we show that targeting the SA to a developmentally silenced embryonic globin gene in adult murine erythroblasts triggers its transcriptional reactivation. This activity depends on the LCR, consistent with an LCR-promoter looping mechanism. Strikingly, targeting the SA to the fetal γ-globin promoter in primary adult human erythroblasts increases γ-globin promoter-LCR contacts, stimulating transcription to approximately 85% of total β-globin synthesis, with a reciprocal reduction in adult β-globin expression. Our findings demonstrate that forced chromatin looping can override a stringent developmental gene expression program and suggest a novel approach to control the balance of globin gene transcription for therapeutic applications.
- Published
- 2014
35. Targeted Genome Editing in Human Repopulating Hematopoietic Stem Cells
- Author
-
Mirjam van der Burg, Giulia Schiroli, Bernhard Gentner, Angelo Lombardo, Giulia Escobar, Luigi Naldini, Michael C. Holmes, Roberta Mazzieri, Davide Moi, Chiara Bonini, Pietro Genovese, Philip D. Gregory, Claudia Firrito, Eugenio Montini, Andrea Calabria, Tiziano Di Tomaso, Immunology, Genovese, P, Schiroli, G, Escobar, G, Di Tomaso, T, Firrito, C, Calabria, A, Moi, D, Mazzieri, R, Bonini, MARIA CHIARA, Holmes, Mc, Gregory, Pd, van der Burg, M, Gentner, B, Montini, E, Lombardo, ANGELO LEONE, and Naldini, Luigi
- Subjects
Male ,DNA, Complementary ,Genetic enhancement ,Transgene ,Antigens, CD34 ,Biology ,Gene delivery ,X-Linked Combined Immunodeficiency Diseases ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Genome editing ,Animals ,Humans ,030304 developmental biology ,Genetics ,0303 health sciences ,Transcription activator-like effector nuclease ,Multidisciplinary ,Genome, Human ,Hematopoietic Stem Cell Transplantation ,Gene targeting ,Endonucleases ,Fetal Blood ,Hematopoietic Stem Cells ,Hematopoiesis ,Cell biology ,Haematopoiesis ,030220 oncology & carcinogenesis ,Mutation ,Gene Targeting ,Stem cell ,Interleukin Receptor Common gamma Subunit ,Targeted Gene Repair - Abstract
Targeted genome editing by artificial nucleases has brought the goal of site-specific transgene integration and gene correction within the reach of gene therapy. However, its application to long-term repopulating haematopoietic stem cells (HSCs) has remained elusive. Here we show that poor permissiveness to gene transfer and limited proficiency of the homology-directed DNA repair pathway constrain gene targeting in human HSCs. By tailoring delivery platforms and culture conditions we overcame these barriers and provide stringent evidence of targeted integration in human HSCs by long-term multilineage repopulation of transplanted mice. We demonstrate the therapeutic potential of our strategy by targeting a corrective complementary DNA into the IL2RG gene of HSCs from healthy donors and a subject with X-linked severe combined immunodeficiency (SCID-X1). Gene-edited HSCs sustained normal haematopoiesis and gave rise to functional lymphoid cells that possess a selective growth advantage over those carrying disruptive IL2RG mutations. These results open up new avenues for treating SCID-X1 and other diseases.
- Published
- 2014
36. Genomic Editing of the HIV-1 Coreceptor CCR5 in Adult Hematopoietic Stem and Progenitor Cells Using Zinc Finger Nucleases
- Author
-
David DiGiusto, Evan Lopez, Jianbin Wang, Pei-Qi Liu, Agnes M. Gardner, Jill Henley, Nancy Gonzalez, Michael C. Holmes, Ludmila Krymskaya, Paula M. Cannon, Qing Liu, Anitha Rao, John A. Zaia, Lijing Li, Chy-Anh Tran, Ursula Hofer, Lan-Feng Cao, Kenneth Kim, Monica Torres-Coronado, and Philip D. Gregory
- Subjects
Receptors, CCR5 ,Cell Survival ,Cellular differentiation ,Genetic Vectors ,Antigens, CD34 ,Apoptosis ,Biology ,Adenoviridae ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Granulocyte Colony-Stimulating Factor ,Drug Discovery ,Genetics ,Animals ,Humans ,Progenitor cell ,Extracellular Signal-Regulated MAP Kinases ,Molecular Biology ,Cells, Cultured ,Protein kinase C ,030304 developmental biology ,Pharmacology ,Zinc finger ,Acquired Immunodeficiency Syndrome ,0303 health sciences ,Hematopoietic Stem Cell Transplantation ,virus diseases ,Gene targeting ,Cell Differentiation ,Zinc Fingers ,Genomics ,Endonucleases ,Hematopoietic Stem Cells ,Molecular biology ,Zinc finger nuclease ,3. Good health ,Cell biology ,Disease Models, Animal ,Haematopoiesis ,030220 oncology & carcinogenesis ,Gene Targeting ,HIV-1 ,Molecular Medicine ,Original Article ,Stem cell ,Gene Deletion - Abstract
The HIV-1 coreceptor CCR5 is a validated target for HIV/AIDS therapy. The apparent elimination of HIV-1 in a patient treated with an allogeneic stem cell transplant homozygous for a naturally occurring CCR5 deletion mutation (CCR5(Δ32/Δ32)) supports the concept that a single dose of HIV-resistant hematopoietic stem cells can provide disease protection. Given the low frequency of naturally occurring CCR5(Δ32/Δ32) donors, we reasoned that engineered autologous CD34(+) hematopoietic stem/progenitor cells (HSPCs) could be used for AIDS therapy. We evaluated disruption of CCR5 gene expression in HSPCs isolated from granulocyte colony-stimulating factor (CSF)-mobilized adult blood using a recombinant adenoviral vector encoding a CCR5-specific pair of zinc finger nucleases (CCR5-ZFN). Our results demonstrate that CCR5-ZFN RNA and protein expression from the adenoviral vector is enhanced by pretreatment of HSPC with protein kinase C (PKC) activators resulting in25% CCR5 gene disruption and that activation of the mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling pathway is responsible for this activity. Importantly, using an optimized dose of PKC activator and adenoviral vector we could generate CCR5-modified HSPCs which engraft in a humanized mouse model (albeit at a reduced level) and support multilineage differentiation in vitro and in vivo. Together, these data establish the basis for improved approaches exploiting adenoviral vector delivery in the modification of HSPCs.
- Published
- 2013
37. Activation domains for controlling plant gene expression using designed transcription factors
- Author
-
Jocelynn R. Pearl, Philip D. Gregory, Joseph F. Petolino, Tonya L. Strange, Bryan Zeitler, Fyodor D. Urnov, Steve Evans, Ryan C. Blue, David Huizinga, and Jianquan Li
- Subjects
Transcriptional Activation ,Proteome ,Transcription, Genetic ,Amino Acid Motifs ,Molecular Sequence Data ,RNA polymerase II ,GUS reporter system ,Saccharomyces cerevisiae ,Plant Science ,Biology ,Protein Engineering ,Chromosomes, Plant ,Evolution, Molecular ,Gene Expression Regulation, Plant ,Genes, Reporter ,Tobacco ,Gene expression ,Amino Acid Sequence ,Plant Proteins ,Regulation of gene expression ,Genetics ,Reporter gene ,YY1 ,Herpes Simplex Virus Protein Vmw65 ,Promoter ,Protein Structure, Tertiary ,Cell biology ,Terminator (genetics) ,biology.protein ,Sequence Alignment ,Agronomy and Crop Science ,Transcription Factors ,Biotechnology - Abstract
Targeted gene regulation via designed transcription factors has great potential for precise phenotypic modification and acceleration of novel crop trait development. To this end, designed transcriptional activators have been constructed by fusing transcriptional activation domains to DNA-binding proteins. In this study, a transcriptional activator from the herpes simplex virus, VP16, was used to identify plant regulatory proteins. Transcriptional activation domains were identified from each protein and fused with zinc finger DNA-binding proteins (ZFPs) to generate designed transcriptional activators. In addition, specific sequences within each transcriptional activation domain were modified to mimic the VP16 contact motif that interacts directly with RNA polymerase II core transcription factors. To evaluate these designed transcriptional activators, test systems were built in yeast and tobacco comprising reporter genes driven by promoters containing ZFP-binding sites upstream of the transcriptional start site. In yeast, transcriptional domains from the plant proteins ERF2 and PTI4 activated MEL1 reporter gene expression to levels similar to VP16 and the modified sequences displayed even greater levels of activation. Following stable transformation of the tobacco reporter system with transcriptional activators derived from ERF2, GUS reporter gene transcript accumulation was equal to or greater than those derived from VP16. Moreover, a modified ERF2 domain displayed significantly enhanced transcriptional activation compared with VP16 and with the unmodified ERF2 sequence. These results demonstrate that plant sequences capable of facilitating transcriptional activation can be found and, when fused to DNA-binding proteins, can enhance gene expression.
- Published
- 2013
38. Efficient Clinical Scale Gene Modification via Zinc Finger Nuclease–Targeted Disruption of the HIV Co-receptor CCR5
- Author
-
Travis Wood, Dawn A. Maier, Andrea L. Brennan, Gabriela Plesa, Carl H. June, Gary Lee, Julio Cotte, Elena E. Perez, Bruce L. Levine, S. Kaye Spratt, Carmine Carpenito, Richard G. Carroll, Zhaohui Zheng, Philip D. Gregory, Michael C. Holmes, Gwendolyn Binder-Scholl, James L. Riley, Dale Ando, and Shuguang Jiang
- Subjects
Male ,Co-receptor ,CD3 Complex ,Receptors, CCR5 ,Chemokine receptor CCR5 ,T-Lymphocytes ,CD3 ,Genetic Vectors ,Transplantation, Heterologous ,HIV Infections ,Lymphocyte Activation ,Viral vector ,Mice ,Chemokine receptor ,CD28 Antigens ,Transduction, Genetic ,Genetics ,Animals ,Humans ,Molecular Biology ,Research Articles ,Zinc finger ,biology ,Adenoviruses, Human ,virus diseases ,CD28 ,Zinc Fingers ,DNA Restriction Enzymes ,Adoptive Transfer ,Molecular biology ,Zinc finger nuclease ,Cell biology ,Phenotype ,biology.protein ,Molecular Medicine ,Female - Abstract
Since HIV requires CD4 and a co-receptor, most commonly C-C chemokine receptor 5 (CCR5), for cellular entry, targeting CCR5 expression is an attractive approach for therapy of HIV infection. Treatment of CD4(+) T cells with zinc-finger protein nucleases (ZFNs) specifically disrupting chemokine receptor CCR5 coding sequences induces resistance to HIV infection in vitro and in vivo. A chimeric Ad5/F35 adenoviral vector encoding CCR5-ZFNs permitted efficient delivery and transient expression following anti-CD3/anti-CD28 costimulation of T lymphocytes. We present data showing CD3/CD28 costimulation substantially improved transduction efficiency over reported methods for Ad5/F35 transduction of T lymphocytes. Modifications to the laboratory scale process, incorporating clinically compatible reagents and methods, resulted in a robust ex vivo manufacturing process capable of generating10(10) CCR5 gene-edited CD4+ T cells from healthy and HIV+ donors. CD4+ T-cell phenotype, cytokine production, and repertoire were comparable between ZFN-modified and control cells. Following consultation with regulatory authorities, we conducted in vivo toxicity studies that showed no detectable ZFN-specific toxicity or T-cell transformation. Based on these findings, we initiated a clinical trial testing the safety and feasibility of CCR5 gene-edited CD4+ T-cell transfer in study subjects with HIV-1 infection.
- Published
- 2013
39. Potent and Broad Inhibition of HIV-1 by a Peptide from the gp41 Heptad Repeat-2 Domain Conjugated to the CXCR4 Amino Terminus
- Author
-
Jianbin Wang, Josephine Romano, Beth S. Haggarty, Michael C. Holmes, James A. Hoxie, James L. Riley, Kevin Hua, Kritika E. Kumar, Andrea P. O. Jordon, George J. Leslie, Max W. Richardson, Philip D. Gregory, Anthony Secreto, Carl H. June, Jennifer Duong, Joshua J. DeClercq, and Michael J. Root
- Subjects
0301 basic medicine ,RNA viruses ,CD4-Positive T-Lymphocytes ,Cell Membranes ,Peptide ,HIV Infections ,Pathology and Laboratory Medicine ,Membrane Fusion ,Cell Fusion ,White Blood Cells ,Mice ,0302 clinical medicine ,Immunodeficiency Viruses ,Animal Cells ,Mice, Inbred NOD ,Medicine and Health Sciences ,Biology (General) ,chemistry.chemical_classification ,Cell fusion ,T Cells ,virus diseases ,Animal Models ,Flow Cytometry ,HIV Envelope Protein gp41 ,3. Good health ,medicine.anatomical_structure ,Biochemistry ,Medical Microbiology ,030220 oncology & carcinogenesis ,Viral Pathogens ,Viruses ,Pathogens ,Cellular Types ,Cellular Structures and Organelles ,Coreceptors ,Research Article ,Signal Transduction ,Cell Physiology ,Receptors, CXCR4 ,QH301-705.5 ,T cell ,Immune Cells ,Immunology ,Mouse Models ,Biology ,Gp41 ,Research and Analysis Methods ,Microbiology ,03 medical and health sciences ,Model Organisms ,Viral entry ,Virology ,Retroviruses ,Genetics ,medicine ,Animals ,Humans ,T Helper Cells ,CCR5 coreceptor ,Molecular Biology ,Microbial Pathogens ,Blood Cells ,HEK 293 cells ,Lentivirus ,Organisms ,Biology and Life Sciences ,HIV ,Chemotaxis ,Cell Biology ,RC581-607 ,Virus Internalization ,Peptide Fragments ,Heptad repeat ,030104 developmental biology ,HEK293 Cells ,chemistry ,HIV-1 ,Parasitology ,Immunologic diseases. Allergy - Abstract
HIV-1 entry can be inhibited by soluble peptides from the gp41 heptad repeat-2 (HR2) domain that interfere with formation of the 6-helix bundle during fusion. Inhibition has also been seen when these peptides are conjugated to anchoring molecules and over-expressed on the cell surface. We hypothesized that potent anti-HIV activity could be achieved if a 34 amino acid peptide from HR2 (C34) were brought to the site of virus-cell interactions by conjugation to the amino termini of HIV-1 coreceptors CCR5 or CXCR4. C34-conjugated coreceptors were expressed on the surface of T cell lines and primary CD4 T cells, retained the ability to mediate chemotaxis in response to cognate chemokines, and were highly resistant to HIV-1 utilization for entry. Notably, C34-conjugated CCR5 and CXCR4 each exhibited potent and broad inhibition of HIV-1 isolates from diverse clades irrespective of tropism (i.e., each could inhibit R5, X4 and dual-tropic isolates). This inhibition was highly specific and dependent on positioning of the peptide, as HIV-1 infection was poorly inhibited when C34 was conjugated to the amino terminus of CD4. C34-conjugated coreceptors could also inhibit HIV-1 isolates that were resistant to the soluble HR2 peptide inhibitor, enfuvirtide. When introduced into primary cells, CD4 T cells expressing C34-conjugated coreceptors exhibited physiologic responses to T cell activation while inhibiting diverse HIV-1 isolates, and cells containing C34-conjugated CXCR4 expanded during HIV-1 infection in vitro and in a humanized mouse model. Notably, the C34-conjugated peptide exerted greater HIV-1 inhibition when conjugated to CXCR4 than to CCR5. Thus, antiviral effects of HR2 peptides can be specifically directed to the site of viral entry where they provide potent and broad inhibition of HIV-1. This approach to engineer HIV-1 resistance in functional CD4 T cells may provide a novel cell-based therapeutic for controlling HIV infection in humans., Author Summary HIV-1 infection persists and requires life-long therapy. Approaches to prevent viral replication in the absence of treatment will likely require effective antiviral immune responses, but this goal has been confounded by HIV-1’s ability to target CD4 T cells that coordinate adaptive immunity. We describe a novel approach to confer HIV-resistance to CD4 T cells using peptides from the HIV-1 gp41 heptad repeat-2 (HR2) domain to inhibit infection. By linking a 34 amino acid HR2 peptide to the amino terminus of CCR5 or CXCR4 we were able to use the physiologic trafficking of these coreceptors to deliver the inhibitory peptide to sites of viral fusion where they exerted potent, specific and broad resistance irrespective of viral clade or tropism. This effect was highly dependent on the positioning of the peptide and most effective when conjugated to CXCR4. In vitro and in humanized mice, primary CD4 T cells were protected and expanded following HIV-1 infection. This work represents a proof of concept that T cells can be genetically engineered to resist infection in vivo and provides a rationale to explore this approach as a novel cell-based therapeutic in strategies to augment antiviral immune responses that target viral reservoirs and for long-term control of HIV-1.
- Published
- 2016
40. Controlling the rotational and hyperfine state of ultracoldRb87Cs133molecules
- Author
-
Jesus Aldegunde, Philip D. Gregory, Simon L. Cornish, and Jeremy M. Hutson
- Subjects
Physics ,Rabi cycle ,01 natural sciences ,Measure (mathematics) ,010305 fluids & plasmas ,Magnetic field ,Coherent control ,0103 physical sciences ,Molecule ,Physics::Atomic Physics ,Atomic physics ,010306 general physics ,Ground state ,Hyperfine structure ,Microwave - Abstract
We demonstrate coherent control of the rotational and hyperfine state of ultracold, chemically stable $^{87}\mathrm{Rb}^{133}\mathrm{Cs}$ molecules with external microwave fields. We create a sample of $\ensuremath{\sim}2000$ molecules in the lowest hyperfine level of the rovibronic ground state $N=0$. We measure the transition frequencies to eight different hyperfine levels of the $N=1$ state at two magnetic fields $\ensuremath{\sim}23$ G apart. We determine accurate values of rotational and hyperfine coupling constants that agree well with previous calculations. We observe Rabi oscillations on each transition, allowing complete population transfer to a selected hyperfine level of $N=1$. Subsequent application of a second microwave pulse allows the transfer of molecules back to a different hyperfine level of $N=0$.
- Published
- 2016
41. Measurement of the binding energy of ultracoldRb87Cs133molecules using an offset-free optical frequency comb
- Author
-
Philip D. Gregory, Russell Kliese, Avinash Kumar, Simon L. Cornish, Jeremy M. Hutson, C. Ruth Le Sueur, Thomas Puppe, Peter K. Molony, and Jesus Aldegunde
- Subjects
Physics ,Binding energy ,Stimulated Raman adiabatic passage ,02 engineering and technology ,Rotational–vibrational spectroscopy ,021001 nanoscience & nanotechnology ,Laser ,01 natural sciences ,Bond-dissociation energy ,law.invention ,law ,0103 physical sciences ,Molecule ,Atomic physics ,010306 general physics ,0210 nano-technology ,Ground state ,Hyperfine structure - Abstract
We report the binding energy of $^{87}\mathrm{Rb}^{133}\mathrm{Cs}$ molecules in their rovibrational ground state measured using an offset-free optical frequency comb based on difference frequency generation technology. We create molecules in the absolute ground state using stimulated Raman adiabatic passage (STIRAP) with a transfer efficiency of 88%. By measuring the absolute frequencies of our STIRAP lasers, we find the energy-level difference from an initial weakly bound Feshbach state to the rovibrational ground state with a resolution of $\ensuremath{\sim}5$ kHz over an energy-level difference of more than $114\phantom{\rule{0.28em}{0ex}}\mathrm{T}\mathrm{Hz}$; this lets us discern the hyperfine splitting of the ground state. Combined with theoretical models of the Feshbach-state binding energies and ground-state hyperfine structure, we determine a zero-field binding energy of $h\ifmmode\times\else\texttimes\fi{}114\phantom{\rule{0.16em}{0ex}}268\phantom{\rule{0.16em}{0ex}}135.24(4)(3)\phantom{\rule{0.28em}{0ex}}\mathrm{M}\mathrm{Hz}$. To our knowledge, this is the most accurate determination to date of the dissociation energy of a molecule.
- Published
- 2016
42. Production of Ultracold
- Author
-
Peter K, Molony, Philip D, Gregory, Avinash, Kumar, C Ruth, Le Sueur, Jeremy M, Hutson, and Simon L, Cornish
- Abstract
We present the production of ultracold
- Published
- 2016
43. Site-specific genome editing in Plasmodium falciparum using engineered zinc-finger nucleases
- Author
-
Philip D. Gregory, Jocelynn R. Pearl, Edward J. Rebar, Lei Zhang, Manuel Llinás, David A. Fidock, Marcus C. S. Lee, Fyodor D. Urnov, Judith Straimer, Andrew H. Lee, April E. Williams, and Bryan Zeitler
- Subjects
Molecular Sequence Data ,Plasmodium falciparum ,Drug Resistance ,Locus (genetics) ,Biology ,Protein Engineering ,Biochemistry ,Genome ,Article ,Genome editing ,parasitic diseases ,Molecular Biology ,Gene ,Alleles ,Genetics ,Zinc finger ,Base Sequence ,Point mutation ,Chloroquine ,Zinc Fingers ,Cell Biology ,Endonucleases ,biology.organism_classification ,Zinc finger nuclease ,Genome, Protozoan ,Biotechnology - Abstract
Malaria afflicts over 200 million people worldwide and its most lethal etiologic agent, Plasmodium falciparum, is evolving to resist even the latest-generation therapeutics. Efficient tools for genome-directed investigations of P. falciparum pathogenesis, including drug resistance mechanisms, are clearly required. Here we report rapid and targeted genetic engineering of this parasite, using zinc-finger nucleases (ZFNs) that produce a double-strand break in a user-defined locus and trigger homology-directed repair. Targeting an integrated egfp locus, we obtained gene deletion parasites with unprecedented speed (two weeks), both with and without direct selection. ZFNs engineered against the endogenous parasite gene pfcrt, responsible for chloroquine treatment escape, rapidly produced parasites that carried either an allelic replacement or a panel of specified point mutations. The efficiency, versatility and precision of this method will enable a diverse array of genome editing approaches to interrogate this human pathogen.
- Published
- 2012
44. A Designed Zinc-finger Transcriptional Repressor of Phospholamban Improves Function of the Failing Heart
- Author
-
H Steve, Zhang, Dingang, Liu, Yan, Huang, Stefan, Schmidt, Reed, Hickey, Dmitry, Guschin, Haili, Su, Ion S, Jovin, Mike, Kunis, Sarah, Hinkley, Yuxin, Liang, Linda, Hinh, S Kaye, Spratt, Casey C, Case, Edward J, Rebar, Barbara E, Ehrlich, Barbara, Ehrlich, Philip D, Gregory, and Frank J, Giordano
- Subjects
Blotting, Western ,Druggability ,Repressor ,030204 cardiovascular system & hematology ,Biology ,Adenoviridae ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Calcium-binding protein ,Drug Discovery ,Genetics ,Animals ,Humans ,Myocytes, Cardiac ,Molecular Biology ,Gene ,Psychological repression ,Transcription factor ,030304 developmental biology ,Pharmacology ,Zinc finger ,Heart Failure ,0303 health sciences ,Reverse Transcriptase Polymerase Chain Reaction ,Calcium-Binding Proteins ,Zinc Fingers ,3. Good health ,Cell biology ,Phospholamban ,Rats ,Kinetics ,030220 oncology & carcinogenesis ,Molecular Medicine ,Original Article ,Corrigendum ,Transcription Factors - Abstract
Selective inhibition of disease-related proteins underpins the majority of successful drug–target interactions. However, development of effective antagonists is often hampered by targets that are not druggable using conventional approaches. Here, we apply engineered zinc-finger protein transcription factors (ZFP TFs) to the endogenous phospholamban (PLN) gene, which encodes a well validated but recalcitrant drug target in heart failure. We show that potent repression of PLN expression can be achieved with specificity that approaches single-gene regulation. Moreover, ZFP-driven repression of PLN increases calcium reuptake kinetics and improves contractile function of cardiac muscle both in vitro and in an animal model of heart failure. These results support the development of the PLN repressor as therapy for heart failure, and provide evidence that delivery of engineered ZFP TFs to native organs can drive therapeutically relevant levels of gene repression in vivo. Given the adaptability of designed ZFPs for binding diverse DNA sequences and the ubiquity of potential targets (promoter proximal DNA), our findings suggest that engineered ZFP repressors represent a powerful tool for the therapeutic inhibition of disease-related genes, therefore, offering the potential for therapeutic intervention in heart failure and other poorly treated human diseases.
- Published
- 2012
45. Controlling Long-Range Genomic Interactions at a Native Locus by Targeted Tethering of a Looping Factor
- Author
-
Philip D. Gregory, Ann Dean, Hongxin Wang, Gerd A. Blobel, Jongjoo Lee, Wulan Deng, Andreas Reik, and Jeffrey C. Miller
- Subjects
Male ,Erythroblasts ,Transcription, Genetic ,RNA polymerase II ,Cell Separation ,beta-Globins ,Biology ,DNA-binding protein ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,Animals ,GATA1 Transcription Factor ,Promoter Regions, Genetic ,Enhancer ,Locus control region ,030304 developmental biology ,Genetics ,0303 health sciences ,Biochemistry, Genetics and Molecular Biology(all) ,Zinc Fingers ,Promoter ,LIM Domain Proteins ,Embryo, Mammalian ,Chromatin ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Gene Expression Regulation ,biology.protein ,Female ,Chromatin Loop ,030217 neurology & neurosurgery - Abstract
SummaryChromatin loops juxtapose distal enhancers with active promoters, but their molecular architecture and relationship with transcription remain unclear. In erythroid cells, the locus control region (LCR) and β-globin promoter form a chromatin loop that requires transcription factor GATA1 and the associated molecule Ldb1. We employed artificial zinc fingers (ZF) to tether Ldb1 to the β-globin promoter in GATA1 null erythroblasts, in which the β-globin locus is relaxed and inactive. Remarkably, targeting Ldb1 or only its self-association domain to the β-globin promoter substantially activated β-globin transcription in the absence of GATA1. Promoter-tethered Ldb1 interacted with endogenous Ldb1 complexes at the LCR to form a chromatin loop, causing recruitment and phosphorylation of RNA polymerase II. ZF-Ldb1 proteins were inactive at alleles lacking the LCR, demonstrating that their activities depend on long-range interactions. Our findings establish Ldb1 as a critical effector of GATA1-mediated loop formation and indicate that chromatin looping causally underlies gene regulation.PaperFlick
- Published
- 2012
46. Transcriptional activation ofBrassica napusβ-ketoacyl-ACP synthase II with an engineered zinc finger protein transcription factor
- Author
-
Jeffrey C. Miller, Stephen Novak, Carla Clifford, Joseph F. Petolino, Manju Gupta, Thomas G. Patterson, Sunita Gopalan, Russell Dekelver, James Patrick Connell, Edward J. Rebar, Josh Flook, Philip D. Gregory, Daniel DeSloover, Kelly R. Robbins, Asha M. Palta, Daniel J Gachotte, and Fyodor D. Urnov
- Subjects
Zinc finger ,Regulation of gene expression ,Genetics ,Agrobacterium ,fungi ,food and beverages ,Plant Science ,Biology ,biology.organism_classification ,Transformation (genetics) ,Biochemistry ,Saturated fatty acid ,Agronomy and Crop Science ,Transcription factor ,Gene ,Selectable marker ,Biotechnology - Abstract
Targeted gene regulation via designed transcription factors has great potential for precise phenotypic modification and acceleration of novel crop trait development. Canola seed oil composition is dictated largely by the expression of genes encoding enzymes in the fatty acid biosynthetic pathway. In the present study, zinc finger proteins (ZFPs) were designed to bind DNA sequences common to two canola β-ketoacyl-ACP Synthase II (KASII) genes downstream of their transcription start site. Transcriptional activators (ZFP-TFs) were constructed by fusing these ZFP DNA-binding domains to the VP16 transcriptional activation domain. Following transformation using Agrobacterium, transgenic events expressing ZFP-TFs were generated and shown to have elevated KASII transcript levels in the leaves of transgenic T(0) plants when compared to 'selectable marker only' controls as well as of T(1) progeny plants when compared to null segregants. In addition, leaves of ZFP-TF-expressing T(1) plants contained statistically significant decreases in palmitic acid (consistent with increased KASII activity) and increased total C18. Similarly, T(2) seed displayed statistically significant decreases in palmitic acid, increased total C18 and reduced total saturated fatty acid contents. These results demonstrate that designed ZFP-TFs can be used to regulate the expression of endogenous genes to elicit specific phenotypic modifications of agronomically relevant traits in a crop species.
- Published
- 2012
47. Zinc-finger Nuclease Editing of Human cxcr4 Promotes HIV-1 CD4+ T Cell Resistance and Enrichment
- Author
-
Kenneth Kim, Colleen Fearns, Philip D. Gregory, Jinyun Yuan, Kevin Hua, Bruce E. Torbett, Karen Crain, Michael C. Holmes, and Jianbin Wang
- Subjects
CD4-Positive T-Lymphocytes ,Receptors, CXCR4 ,Biology ,Peripheral blood mononuclear cell ,CXCR4 ,Adenoviridae ,Mice ,Interleukin 21 ,Viral entry ,Drug Discovery ,Genetics ,Animals ,Humans ,Cytotoxic T cell ,IL-2 receptor ,RNA, Small Interfering ,Antigen-presenting cell ,Molecular Biology ,Cells, Cultured ,Pharmacology ,fungi ,Zinc Fingers ,Endonucleases ,Zinc finger nuclease ,Molecular biology ,HIV-1 ,Molecular Medicine ,Original Article - Abstract
HIV-1-infected individuals can harbor viral isolates that can use CCR5, as well as CXCR4, for viral entry. To genetically engineer HIV-1 resistance in CD4(+) T cells, we assessed whether transient, adenovirus delivered zinc-finger nuclease (ZFN) disruption of genomic cxcr4 or stable lentiviral expression of short hairpin RNAs (shRNAs) targeting CXCR4 mRNAs provides durable resistance to HIV-1 challenge. ZFN-modification of cxcr4 in CD4(+) T cells was found to be superior to cell integrated lentivirus-expressing CXCR4 targeting shRNAs when CD4(+) T cells were challenged with HIV-1s that utilizes CXCR4 for entry. Cxcr4 disruption in CD4(+) T cells was found to be stable, conferred resistance, and provided for continued cell enrichment during HIV-1 infection in tissue culture and, in vivo, in peripheral blood mononuclear cell transplanted NSG mice. Moreover, HIV-1-infected mice with engrafted cxcr4 ZFN-modified CD4(+) T cells demonstrated lower viral levels in contrast to mice engrafted with unmodified CD4(+) T cells. These findings provide evidence that ZFN-mediated disruption of cxcr4 provides a selective advantage to CD4(+) T cells during HIV-1 infection.
- Published
- 2012
48. Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer
- Author
-
Giulia Casorati, Pietro Genovese, Luigi Naldini, Philip D. Gregory, Angelo Lombardo, Maurilio Ponzoni, Chiara Bonini, Claudio Bordignon, Jürgen Kuball, Philip D. Greenberg, David Paschon, Victoria Chu, Andreas Reik, Michael C. Holmes, Elena Provasi, Zulma Magnani, Attilio Bondanza, Pei-Qi Liu, Lei Zhang, Fabio Ciceri, Barbara Camisa, Provasi, E, Genovese, P, Lombardo, ANGELO LEONE, Magnani, Z, Liu Pei, Q, Reik, A, Chu, V, Paschon, D. E., Zhang, L, Kuball, J, Camisa, B, Bondanza, Attilio, Casorati, G, Ponzoni, Maurilio, Ciceri, Fabio, Bordignon, Claudio, Greenberg, P. D., Holmes, Mc, Gregory, Pd, Naldini, Luigi, and Bonini, MARIA CHIARA
- Subjects
T-Lymphocytes ,medicine.medical_treatment ,CD3 ,Molecular Sequence Data ,Receptors, Antigen, T-Cell ,T-Cell Antigen Receptor Specificity ,chemical and pharmacologic phenomena ,Endogeny ,Gene transfer ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Jurkat Cells ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,medicine ,Humans ,Deoxyribonucleases, Type II Site-Specific ,WT1 Proteins ,030304 developmental biology ,0303 health sciences ,Leukemia ,Base Sequence ,Lentivirus ,T-cell receptor ,Gene Transfer Techniques ,food and beverages ,Zinc Fingers ,hemic and immune systems ,General Medicine ,medicine.disease ,Zinc finger nuclease ,Molecular biology ,Tumor antigen ,3. Good health ,030220 oncology & carcinogenesis ,biology.protein - Abstract
The transfer of high-avidity T cell receptor (TCR) genes isolated from rare tumor-specific lymphocytes into polyclonal T cells is an attractive cancer immunotherapy strategy. However, TCR gene transfer results in competition for surface expression and inappropriate pairing between the exogenous and endogenous TCR chains, resulting in suboptimal activity and potentially harmful unpredicted antigen specificities of the resultant TCRs. We designed zinc-finger nucleases (ZFNs) that promoted the disruption of endogenous TCR beta- and alpha-chain genes. Lymphocytes treated with ZFNs lacked surface expression of CD3-TCR and expanded with the addition of interleukin-7 (IL-7) and IL-15. After lentiviral transfer of a TCR specific for the Wilms tumor 1 (WT1) antigen, these TCR-edited cells expressed the new TCR at high levels, were easily expanded to near purity and were superior at specific antigen recognition compared to donor-matched, unedited TCR-transferred cells. In contrast to unedited TCR-transferred cells, the TCR-edited lymphocytes did not mediate off-target reactivity while maintaining their anti-tumor activity in vivo, thus showing that complete editing of T cell specificity generates tumor-specific lymphocytes with improved biosafety profiles.
- Published
- 2012
49. Targeted gene addition to a predetermined site in the human genome using a ZFN-based nicking enzyme
- Author
-
Michael C. Holmes, Yannick Doyon, Kevin Hua, Jianbin Wang, Jenny Jiacheng Yan, Geoffrey Friedman, Philip D. Gregory, Joshua E. Babiarz, Nathaniel Wang, Jeffrey C. Miller, and Carrie Jiaxin Li
- Subjects
Genetics ,DNA repair ,fungi ,Method ,Gene targeting ,DNA Repair Pathway ,Biology ,Zinc finger nuclease ,Non-homologous end joining ,DNA End-Joining Repair ,Genome editing ,Homologous recombination ,Genetics (clinical) - Abstract
Zinc-finger nucleases (ZFNs) are chimeric endonucleases created by fusing the cleavage domain from the type IIS restriction enzyme FokI to a designed zinc-finger protein (ZFP). In order to generate a DNA double strand break (DSB) at a predetermined genomic site, two engineered ZFNs must be designed to bind DNA with the appropriate orientation and spacing (i.e., on opposite sides of the DNA with 5 or 6 bp of sequence separating them) (Carroll 2008; Cathomen and Joung 2008). This architecture (analogous to that depicted in Fig. 1A) permits the two FokI cleavage domains to heterodimerize—a prerequisite for subsequent cleavage of the targeted DNA (Bitinaite et al. 1998). Figure 1. Mutation of the catalytic domain of a ZFN monomer generates a ZFN heterodimer with nicking activity. (A) Illustration of the ZFN heterodimer architecture for single-stranded break (SSB) generation. The FokI catalytic domain of ZFN (left, blue) is mutated ... Repair of the ZFN-induced site-specific DSB provides the molecular basis for gene disruption, gene correction, or gene addition—the different potential outcomes of ZFN-mediated genome editing (for review, see Urnov et al. 2010). Specifically, a given DSB may be repaired via either the nonhomologous end joining (NHEJ) or homology-directed repair (HDR) pathways (Kowalczykowski 2000; Sung and Klein 2006; Wyman and Kanaar 2006; Brugmans et al. 2007). HDR utilizes a homologous donor sequence as template for the conservative repair of the DNA break. Provision of a suitably designed donor DNA molecule can, therefore, specify gene addition or gene correction at the ZFN-induced DSB. NHEJ, on the other hand, simply catalyzes the rejoining of the two DNA ends—a process that can result in the deletion or insertion of nucleotides at the repair junction. DNA repair via NHEJ is, therefore, mutagenic. To date, ZFN-induced DNA repair via HDR or NHEJ has been utilized to target modifications to the genomes of numerous species (Le Provost et al. 2010) including Caenorhabditis elegans (Morton et al. 2006), Drosophila melanogaster (Bibikova et al. 2002), silkworms (Takasu et al. 2010), zebrafish (Doyon et al. 2008; Meng et al. 2008), sea urchins (Ochiai et al. 2010), Arabidopsis thaliana (Osakabe et al. 2010; Zhang et al. 2010), tobacco (Maeder et al. 2008; Cai et al. 2009; Townsend et al. 2009), corn (Shukla et al. 2009), mice (Carbery et al. 2010; Meyer et al. 2010), and rats (Geurts et al. 2009). ZFNs have also been used to efficiently engineer a diverse range of mammalian cell types (Urnov et al. 2010), including human stem cells, with subsequent retention of full potency and normal growth characteristics (Hockemeyer et al. 2009; Zou et al. 2009; Holt et al. 2010). Moreover, the therapeutic potential of ZFNs is currently under evaluation in clinical trials (clinicaltrials.gov identifiers NCT00842634, NCT01252641, NCT01044654). Given the markedly different genome editing outcomes mediated by NHEJ and HDR, it would be desirable to have control over the choice of pathway used and thus specify the outcome of repair. In practice, for gene disruption this can already be achieved, since NHEJ is the predominant pathway in mammalian cells (Lieber 2008) and codelivery of donor DNA is unnecessary. For gene correction and addition, however, NHEJ events represent competition to the desired homology-directed outcome at the target locus. One potential strategy for enforcing DNA repair pathway bias would be the targeted introduction of a DNA single strand break (SSB) or nick. Like the DSB, a DNA SSB/nick can theoretically stimulate homology-directed repair pathways (Holliday 1964; Meselson and Radding 1975; Radding 1982). For example, SSBs induced by the bacteriophage fd gene II protein (gIIp) stimulated HDR in yeast (Galli and Schiestl 1998), while nicks generated by specific mutants of the RAG proteins—critical components of the V(D)J recombination system—are repaired via initiation of homologous recombination (Lee et al. 2004). Moreover, experiments using a nick-inducing variant of the I-Anil LAGLIDADG homing endonuclease showed that the SSB/nicks induced by these engineered enzymes are also repaired via HDR (Smith et al. 2009). Importantly, and in contrast to a DSB, a DNA nick or SSB is not a bona fide substrate for repair via the NHEJ pathway. Thus, a targeted SSB has the potential to restrict repair to the homology-directed pathway. Here, we describe the engineering of a zinc finger nickase (ZFNickase) by mutation of a residue critical for FokI cleavage activity in one monomer of the ZFN heterodimer, thus allowing the ZFNs to heterodimerize on DNA but restricting cleavage to a single DNA strand. We show that targeting of these ZFNickases to the endogenous CCR5 locus stimulates gene correction and gene addition via HDR at frequencies of between ∼1%–8% in both transformed and primary human lines. Importantly, we show that the CCR5 ZFNickase-induced gene addition occurs without a marked increase in detectable mutagenesis by the error-prone NHEJ repair pathway. Specific outcomes of either gene addition/correction or gene disruption may, therefore, be modulated via the targeted introduction of a single- or double-strand break, respectively. Moreover, beyond the targeted event itself, elimination of DNA repair via NHEJ has the potential to eradicate mutagenesis at putative off-target cleavage sites, further improving the fidelity of genome editing.
- Published
- 2012
50. Chromatin structure of two genomic sites for targeted transgene integration in induced pluripotent stem cells and hematopoietic stem cells
- Author
-
R. Van Rensburg, Philip D. Gregory, O. Denisenko, Hongjie Wang, Ines Beyer, André Lieber, Karol Bomsztyk, Daniel G. Miller, Michael C. Holmes, David W. Russell, Zong Yi Li, X. Y. Yao, Faculty of Economic and Social Sciences and Solvay Business School, Faculty of Law and Criminology, and Faculty of Sciences and Bioengineering Sciences
- Subjects
Receptors, CCR5 ,Transcription, Genetic ,Genetic Vectors ,Induced Pluripotent Stem Cells ,Antigens, CD34 ,Article ,03 medical and health sciences ,Endonuclease ,Viral Proteins ,0302 clinical medicine ,Protein Phosphatase 1 ,Genetics ,hematopoïetic stem cells ,Humans ,Transgenes ,Induced pluripotent stem cell ,Endodeoxyribonucleases ,Molecular Biology ,ChIA-PET ,030304 developmental biology ,0303 health sciences ,biology ,Genome, Human ,Gene targeting ,Zinc Fingers ,Dependovirus ,Hematopoietic Stem Cells ,Molecular biology ,Chromatin ,DNA-Binding Proteins ,Genetic Loci ,030220 oncology & carcinogenesis ,Gene Targeting ,biology.protein ,Molecular Medicine ,Stem cell ,Chromatin immunoprecipitation - Abstract
Achieving transgene integration into preselected genomic sites is currently one of the central tasks in stem cell gene therapy. A strategy to mediate such targeted integration involves site-specific endonucleases. Two genomic sites within the MBS85 and chemokine (C-C motif) receptor 5 (CCR5) genes (AAVS1 and CCR5 zinc-finger nuclease (CCR5-ZFN) sites, respectively) have recently been suggested as potential target regions for integration as their disruption has no functional consequence. We hypothesized that efficient transgene integration maybe affected by DNA accessibility of endonucleases and therefore studied the transcriptional and chromatin status of the AAVS1 and CCR5 sites in eight human induced pluripotent stem (iPS) cell lines and pooled CD34+ hematopoietic stem cells (HSCs). Matrix chromatin immunoprecipitation (ChIP) assays demonstrated that the CCR5 site and surrounding regions possessed a predominantly closed chromatin configuration consistent with its transcriptional inactivity in these cell types. In contrast, the AAVS1 site was located within a transcriptionally active region and exhibited an open chromatin configuration in both iPS cells and HSCs. To show that the AAVS1 site is readily amendable to genome modification, we expressed Rep78, an AAV2-derived protein with AAVS1-specific endonuclease activity, in iPS cells after adenoviral gene transfer. We showed that Rep78 efficiently associated with the AAVS1 site and triggered genome modifications within this site. On the other hand, binding to and modification of the CCR5-ZFN site by a ZFN was relatively inefficient. Our data suggest a critical influence of chromatin structure on efficacy of site-specific endonucleases used for genome editing.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.