26 results on '"Nicole M, Gaudelli"'
Search Results
2. Structure of a bound peptide phosphonate reveals the mechanism of nocardicin bifunctional thioesterase epimerase-hydrolase half-reactions
- Author
-
Ketan D. Patel, Felipe B. d’Andrea, Nicole M. Gaudelli, Andrew R. Buller, Craig A. Townsend, and Andrew M. Gulick
- Subjects
Science - Abstract
NocTE is a nonribosomal peptide synthetase thioesterase that completes the biosynthesis of pro-nocardicin G, the precursor for nocardicin β-lactam antibiotics. Here the authors provide mechanistic insights into NocTE by determining its crystal structures in the ligand-free form and covalently linked to a fluorophosphonate substrate mimic.
- Published
- 2019
- Full Text
- View/download PDF
3. Improved cytosine base editors generated from TadA variants
- Author
-
Dieter K. Lam, Patricia R. Feliciano, Amena Arif, Tanggis Bohnuud, Thomas P. Fernandez, Jason M. Gehrke, Phil Grayson, Kin D. Lee, Manuel A. Ortega, Courtney Sawyer, Noah D. Schwaegerle, Leila Peraro, Lauren Young, Seung-Joo Lee, Giuseppe Ciaramella, and Nicole M. Gaudelli
- Subjects
ddc:660 ,Biomedical Engineering ,Molecular Medicine ,Bioengineering ,Applied Microbiology and Biotechnology ,Biotechnology - Abstract
Nature biotechnology 41(5), 686 - 697 (2023). doi:10.1038/s41587-022-01611-9, Cytosine base editors (CBEs) enable programmable genomic C·G-to-T·A transition mutations and typically comprise a modified CRISPR–Cas enzyme, a naturally occurring cytidine deaminase, and an inhibitor of uracil repair. Previous studies have shown that CBEs utilizing naturally occurring cytidine deaminases may cause unguided, genome-wide cytosine deamination. While improved CBEs that decrease stochastic genome-wide off-targets have subsequently been reported, these editors can suffer from suboptimal on-target performance. Here, we report the generation and characterization of CBEs that use engineered variants of TadA (CBE-T) that enable high on-target C·G to T·A across a sequence-diverse set of genomic loci, demonstrate robust activity in primary cells and cause no detectable elevation in genome-wide mutation. Additionally, we report cytosine and adenine base editors (CABEs) catalyzing both A-to-I and C-to-U editing (CABE-Ts). Together with ABEs, CBE-Ts and CABE-Ts enable the programmable installation of all transition mutations using laboratory-evolved TadA variants with improved properties relative to previously reported CBEs., Published by Springer Nature, New York, NY
- Published
- 2023
- Full Text
- View/download PDF
4. CRISPR-derived genome editing therapies: Progress from bench to bedside
- Author
-
Cameron A. Burnett, Alex C. Minella, Alexis C. Komor, Nicole M. Gaudelli, and Holly A. Rees
- Subjects
Technology ,Enter keywords here ,Biomedical ,Computer science ,Review ,Computational biology ,Medical and Health Sciences ,DNA sequencing ,Translational Research, Biomedical ,Cellular mechanism ,Genome editing ,Models ,CRISPR-Associated Protein 9 ,Translational Research ,Drug Discovery ,Genetics ,Animals ,Humans ,CRISPR ,Clustered Regularly Interspaced Short Palindromic Repeats ,Kinetoplastida ,Molecular Biology ,Gene Editing ,Pharmacology ,Clinical Trials as Topic ,Animal ,Human Genome ,Gene Transfer Techniques ,Recombinational DNA Repair ,Genetic Therapy ,Biological Sciences ,Bench to bedside ,Models, Animal ,RNA ,Molecular Medicine ,Human genome ,Generic health relevance ,CRISPR-Cas Systems ,Genetic Engineering ,Guide ,RNA, Guide, Kinetoplastida ,Biotechnology - Abstract
The development of CRISPR-derived genome editing technologies has enabled the precise manipulation of DNA sequences within the human genome. In this review, we discuss the initial development and cellular mechanism of action of CRISPR nucleases and DNA base editors. We then describe factors that must be taken into consideration when developing these tools into therapeutic agents, including the potential for unintended and off-target edits when using these genome editing tools, and methods to characterize these types of edits. We finish by considering specific challenges associated with bringing a CRISPR-based therapy to the clinic, including manufacturing, regulatory oversight, and considerations for clinical trials that involve genome editing agents.
- Published
- 2021
- Full Text
- View/download PDF
5. In vivo CRISPR base editing of PCSK9 durably lowers cholesterol in primates
- Author
-
Kallanthottathil G. Rajeev, Nicole M. Gaudelli, Yusuf S. Nasrullah, Soumyashree A. Gangopadhyay, Andrew M. Bellinger, Sara P. Garcia, James Madsen, Jamie E. DeNizio, Athul Sanjeev, Lisa N. Kasiewicz, Giuseppe Ciaramella, Aaron Beach, Ellen Rohde, Anne Marie Mazzola, Thomas V. Colace, Kui Wang, Sowmya Iyer, Caroline W. Reiss, Padma Malyala, Steven H. Y. Fan, Victoria Clendaniel, Christopher J. Cheng, Jennifer Lavoie, Taiji Mizoguchi, Yuri Matsumoto, Madeleine Shay, Kiran Musunuru, Leslie E. Stolz, Maurine C. Braun, Chaitali Dutta, Ying K. Tam, Joseph Nneji, Ryan Garrity, Alexandra C. Chadwick, Souvik Biswas, Michael Amaonye, John D. Ganey, Mary R. Stahley, Huilan Ren, Huei-Mei Chen, Kathleen Berth, and Sekar Kathiresan
- Subjects
Male ,0301 basic medicine ,Time Factors ,Genetic enhancement ,030204 cardiovascular system & hematology ,Pharmacology ,Biology ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Genome editing ,Loss of Function Mutation ,In vivo ,Animals ,Humans ,CRISPR ,Gene ,Cells, Cultured ,Gene Editing ,Gene knockdown ,Multidisciplinary ,Cholesterol ,Adenine ,PCSK9 ,Cholesterol, LDL ,Mice, Inbred C57BL ,Macaca fascicularis ,030104 developmental biology ,Liver ,chemistry ,Models, Animal ,Hepatocytes ,Mutagenesis, Site-Directed ,Female ,CRISPR-Cas Systems ,Proprotein Convertase 9 - Abstract
Gene-editing technologies, which include the CRISPR–Cas nucleases1–3 and CRISPR base editors4,5, have the potential to permanently modify disease-causing genes in patients6. The demonstration of durable editing in target organs of nonhuman primates is a key step before in vivo administration of gene editors to patients in clinical trials. Here we demonstrate that CRISPR base editors that are delivered in vivo using lipid nanoparticles can efficiently and precisely modify disease-related genes in living cynomolgus monkeys (Macaca fascicularis). We observed a near-complete knockdown of PCSK9 in the liver after a single infusion of lipid nanoparticles, with concomitant reductions in blood levels of PCSK9 and low-density lipoprotein cholesterol of approximately 90% and about 60%, respectively; all of these changes remained stable for at least 8 months after a single-dose treatment. In addition to supporting a ‘once-and-done’ approach to the reduction of low-density lipoprotein cholesterol and the treatment of atherosclerotic cardiovascular disease (the leading cause of death worldwide7), our results provide a proof-of-concept for how CRISPR base editors can be productively applied to make precise single-nucleotide changes in therapeutic target genes in the liver, and potentially in other organs. In a cynomolgus macaque model, CRISPR base editors delivered in lipid nanoparticles are shown to efficiently and stably knock down PCSK9 in the liver to reduce levels of PCSK9 and low-density lipoprotein cholesterol in the blood.
- Published
- 2021
- Full Text
- View/download PDF
6. Rationally Designed Base Editors for Precise Editing of the Sickle Cell Disease Mutation
- Author
-
Alexander Liquori, Ian Slaymaker, Daisy Lam, Giuseppe Ciaramella, Dieter Lam, Adam J. Hartigan, David A. Born, Jeremy Decker, Fei Ann Ran, S. Haihua Chu, Lo-I Cheng, Holly A. Rees, Jeffrey Marshall, Michael S. Packer, Bob Gantzer, Nicole M. Gaudelli, Jenny Olins, Luis A. Barrera, and Yi Yu
- Subjects
Transition (genetics) ,Computer science ,Cas9 ,Cell ,RNA ,Computational biology ,Base (topology) ,chemistry.chemical_compound ,Protospacer adjacent motif ,medicine.anatomical_structure ,chemistry ,Genetics ,medicine ,DNA ,Biotechnology ,Ribonucleoprotein - Abstract
Base editors are fusions of a deaminase and CRISPR-Cas ribonucleoprotein that allow programmable installment of transition mutations without double-strand DNA break intermediates. The breadth of potential base editing targets is frequently limited by the requirement of a suitably positioned Cas9 protospacer adjacent motif. To address this, we used structures of Cas9 and TadA to design a set of inlaid base editors (IBEs), in which deaminase domains are internal to Cas9. Several of these IBEs exhibit shifted editing windows and greater editing efficiency, enabling editing of targets outside the canonical editing window with reduced DNA and RNA off-target editing frequency. Finally, we show that IBEs enable conversion of the pathogenic sickle cell hemoglobin allele to the naturally occurring HbG-Makassar variant in patient-derived hematopoietic stem cells.
- Published
- 2021
- Full Text
- View/download PDF
7. Engineered Stem Cell Antibody Paired Evasion-2 (ESCAPE-2): Paired HSC Epitope Engineering and Direct Editing of Sickle Allele for Antibody-Mediated Autologous Hematopoietic Stem Cell Therapy Conditioning for the Treatment of Sickle Cell Disease
- Author
-
S. Haihua Chu, Elizabeth Budak, Nandini Mondal, Kathy Zhang, Alexander Harmon, Jeffrey Wong, Dieter Lam, Corrina Lucini, Brent Coisman, Kangjian Qiao, Tao Bai, Jeremy Decker, Bob Gantzer, Kindo Lee, Maya Sen, Tanggis Bohnuud, Luis Barrera, Brian Cafferty, Seung-Joo Lee, Paul Kopesky, Nicole M. Gaudelli, Charlotte F McDonagh, Giuseppe Ciaramella, and Adam J. Hartigan
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
8. Engineered Stem Cell Antibody Paired Evasion 1 (ESCAPE-1): Paired HSC Epitope Engineering and Upregulation of Fetal Hemoglobin for Antibody-Mediated Autologous Hematopoietic Stem Cell Therapy Conditioning for the Treatment of Hemoglobinopathies
- Author
-
Nandini Mondal, Alexander Harmon, Elizabeth Budak, Kathy Zhang, Jeffrey Wong, Conrad Rinaldi, Jenny Olins, Kara Hoar, Archita Venugopal Menon, Moriah White, Faith Musenge, Adam Camblin, Adam Wolin, Brent Coisman, Tao Bai, Kangjian Qiao, Mudra Patel, Jeremy Decker, Bob Gantzer, Thomas Leete, Yi Yu, Tanggis Bohnuud, Seung-Joo Lee, Sarah Smith, Charlotte F McDonagh, Nicole M. Gaudelli, S. Haihua Chu, Adam J. Hartigan, and Giuseppe Ciaramella
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
9. Directed evolution of adenine base editors with increased activity and therapeutic application
- Author
-
Jonathan Yen, Giuseppe Ciaramella, Luis A. Barrera, Aaron Edwards, Alexander Liquori, Nicole M. Gaudelli, Dieter K. Lam, Holly A. Rees, Noris M. Solá-Esteves, Lauren Young, Jason Michael Gehrke, Conrad Rinaldi, Michael S. Packer, Seung-Joo Lee, Ian Slaymaker, Ryan Murray, and David A. Born
- Subjects
HBG1 ,Biomedical Engineering ,Deamination ,Bioengineering ,Biology ,Applied Microbiology and Biotechnology ,03 medical and health sciences ,Adenosine deaminase ,0302 clinical medicine ,Guide RNA ,Gene ,030304 developmental biology ,0303 health sciences ,Messenger RNA ,Chemistry ,Point mutation ,RNA ,Directed evolution ,Molecular biology ,Cell biology ,genomic DNA ,Protospacer adjacent motif ,biology.protein ,Molecular Medicine ,030217 neurology & neurosurgery ,Biotechnology - Abstract
The foundational adenine base editors (for example, ABE7.10) enable programmable A•T to G•C point mutations but editing efficiencies can be low at challenging loci in primary human cells. Here we further evolve ABE7.10 using a library of adenosine deaminase variants to create ABE8s. At NGG protospacer adjacent motif (PAM) sites, ABE8s result in ~1.5× higher editing at protospacer positions A5-A7 and ~3.2× higher editing at positions A3-A4 and A8-A10 compared with ABE7.10. Non-NGG PAM variants have a ~4.2-fold overall higher on-target editing efficiency than ABE7.10. In human CD34+ cells, ABE8 can recreate a natural allele at the promoter of the γ-globin genes HBG1 and HBG2 with up to 60% efficiency, causing persistence of fetal hemoglobin. In primary human T cells, ABE8s achieve 98-99% target modification, which is maintained when multiplexed across three loci. Delivered as messenger RNA, ABE8s induce no significant levels of single guide RNA (sgRNA)-independent off-target adenine deamination in genomic DNA and very low levels of adenine deamination in cellular mRNA.
- Published
- 2020
- Full Text
- View/download PDF
10. Cytosine base editors with minimized unguided DNA and RNA off-target events and high on-target activity
- Author
-
Seung-Joo Lee, David A. Born, Holly A. Rees, Giuseppe Ciaramella, Thomas Leete, Nicole M. Gaudelli, Lauren Young, Yi Yu, and Luis A. Barrera
- Subjects
0301 basic medicine ,DNA Replication ,CRISPR-Cas systems ,Transcription, Genetic ,Science ,APOBEC-1 Deaminase ,Deamination ,General Physics and Astronomy ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cytosine Deaminase ,03 medical and health sciences ,chemistry.chemical_compound ,Cytosine ,0302 clinical medicine ,Transcription (biology) ,Humans ,lcsh:Science ,Gene Editing ,Multidisciplinary ,Genome ,Chemistry ,Point mutation ,RNA ,General Chemistry ,Genomics ,DNA ,030104 developmental biology ,HEK293 Cells ,Biochemistry ,Mutagenesis ,lcsh:Q ,Human genome ,Transcriptome ,DNA deamination ,030217 neurology & neurosurgery - Abstract
Cytosine base editors (CBEs) enable efficient, programmable reversion of T•A to C•G point mutations in the human genome. Recently, cytosine base editors with rAPOBEC1 were reported to induce unguided cytosine deamination in genomic DNA and cellular RNA. Here we report eight next-generation CBEs (BE4 with either RrA3F [wt, F130L], AmAPOBEC1, SsAPOBEC3B [wt, R54Q], or PpAPOBEC1 [wt, H122A, R33A]) that display comparable DNA on-target editing frequencies, whilst eliciting a 12- to 69-fold reduction in C-to-U edits in the transcriptome, and up to a 45-fold overall reduction in unguided off-target DNA deamination relative to BE4 containing rAPOBEC1. Further, no enrichment of genome-wide C•G to T•A edits are observed in mammalian cells following transfection of mRNA encoding five of these next-generation editors. Taken together, these next-generation CBEs represent a collection of base editing tools for applications in which minimized off-target and high on-target activity are required., Cytosine base editors have been reported to induce off-target mutations in DNA and RNA. Here the authors identify next-generation CBEs with reduced guide-independent off-target editing profiles and retain high on-target editing activity.
- Published
- 2020
11. Next-generation cytosine base editors with minimized unguided DNA and RNA off-target events and high on-target activity
- Author
-
Giuseppe Ciaramella, Luis A. Barrera, Yi Yu, David A. Born, Holly A. Rees, Thomas Leete, Seung-Joo Lee, Nicole M. Gaudelli, and Lauren Young
- Subjects
chemistry.chemical_compound ,chemistry ,Point mutation ,Mutagenesis ,Deamination ,Human genome ,Computational biology ,Cytidine deaminase ,DNA deamination ,Cytosine ,DNA - Abstract
/introductory paragraphCytosine base editors (CBEs) are molecular machines which enable efficient and programmable reversion of T•A to C•G point mutations in the human genome without induction of DNA double strand breaks1, 2. Recently, the foundational cytosine base editor (CBE) ‘BE3’, containing rAPOBEC1, was reported to induce unguided, genomic DNA3, 4 and cellular RNA5 cytosine deamination when expressed in living cells. To mitigate spurious off-target events, we developed a sensitive, high-throughput cellular assay to select next-generation CBEs that display reduced spurious deamination profiles relative to rAPOBEC1-based CBEs, whilst maintaining equivalent or superior on-target editing frequencies. We screened 153 CBEs containing cytidine deaminase enzymes with diverse sequences and identified four novel CBEs with the most promising on/off target ratios. These spurious-deamination-minimized CBEs (BE4 with either RrA3F, AmAPOBEC1, SsAPOBEC3B, or PpAPOBEC1) were further optimized for superior on- and off-target DNA editing profiles through structure-guided mutagenesis of the deaminase domain. These next-generation CBEs display comparable overall DNA on-target editing frequencies, whilst eliciting a 10- to 49-fold reduction in C-to-U edits in the transcriptome of treated cells, and up to a 33-fold overall reduction in unguided off-target DNA deamination relative to BE4 containing rAPOBEC1. Taken together, these next-generation CBEs represent a new collection of base editing tools for applications in which minimization of spurious deamination is desirable and high on-target activity is required.
- Published
- 2020
- Full Text
- View/download PDF
12. Conversion of HbS to Hb G-Makassar By Adenine Base Editing Is Compatible with Normal Hemoglobin Function
- Author
-
Adam J. Hartigan, Daisy Lam, Jeffrey Marshall, Bob Gantzer, Kyle Rehberger, Nicole M. Gaudelli, Jenny Olins, Chavonna Xu, Bo Yan, Luis A. Barrera, Conrad Rinaldi, Paty Feliciano, Jeremy Decker, Daniel Haupt, Sarah Smith, S. Haihua Chu, Michael S. Packer, Valerie Winton, Matthew Lee, Tanggis Bohnuud, Ian Slaymaker, Teresa McDonald, Colin Lazarra, Salette Martinez, Manuel Ortega, David A. Born, Alexander Liquori, Giuseppe Ciaramella, and Carlo Zambonelli
- Subjects
Stereochemistry ,Chemistry ,Immunology ,Normal hemoglobin ,Cell Biology ,Hematology ,Base (exponentiation) ,Biochemistry ,Function (biology) ,Hb G-Makassar - Abstract
Conversion of the pathogenic sickle allele to a naturally occurring, non-pathogenic hemoglobin variant, Hb G-Makassar, represents a long-term and durable treatment strategy for sickle cell disease (SCD). Using our engineered adenine base editor, we achieved highly efficient base editing in mobilized sickle trait (HbAS) and non-mobilized homozygous sickle (HbSS) CD34 + cells that led to >70% conversion of sickle allele to Makassar allele in in vitro erythroid differentiated (IVED) cells derived from ex vivo edited CD34 + cells. At this level of editing, >70% bi-allelic Makassar editing could be achieved in HbSS IVED cells, with ~20% of cells being mono-allelically Makassar edited. These mono-allelically edited cells behaved similarly to sickle trait (HbAS) cells, when exposed to hypoxic conditions in vitro. In vivo proof of concept xenotransplantation studies demonstrated that Makassar edited HbAS CD34 + cells achieved long-term, multi-lineage hematopoietic engraftment as well as Makassar globin protein expression in human erythroid glycophorin A + cells in thebone marrow of immunocompromised mice. Although the Makassar variant is naturally occurring in human genetics and present in individuals in Southeast Asia with normal hematologic parameters in both heterozygous and homozygous states, we sought to further characterize Makassar hemoglobin and assess its biophysical and biochemical properties. Recombinant Makassar globin was co-expressed with alpha globin in E. coli and tetramers were purified to homogeneity. Recombinant tetramers were assessed for identity, purity, globin content, and heme content demonstrating comparability to hemoglobin tetramers isolated from primary sources (whole blood). Several characterization methods were employed, to assess size, molecular weight, oligomerization state, tetramer composition, and oxygen binding properties. These studies indicated Makassar globin could properly assemble into hemoglobin tetramers, displaying biochemical properties characteristic of hemoglobins. Furthermore, we assessed polymerization potential using a temperature jump method previously employed for kinetic measurements of sickle-fiber formation and found Makassar hemoglobin did not polymerize in vitro under conditions where sickle hemoglobin (HbS) readily polymerizes, consistent with behavior observed previously by others. Finally, a crystal structure of Hb G-Makassar has been determined at the 2.2 Å resolution and showed high similarity to the HbA (wildtype hemoglobin) structure with a RMSD of 0.385 Å for all the Cα atoms, which indicates that the glutamic acid to alanine (E6A) substitution in beta-hemoglobin does not seem to induce any significant conformational change in hemoglobin structures. Altogether, our biophysical and biochemical characterization shows that the Makassar variant behaves as a functional hemoglobin. By replacing the pathogenic sickle globin with a benign hemoglobin variant with normal function, our base editing approach provides a promising autologous investigational cell therapy for the treatment of SCD. Disclosures Chu: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Ortega: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Feliciano: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Winton: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Xu: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Haupt: Beam Therapeutics: Current Employment, Current holder of stock options in a privately-held company. McDonald: Beam Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Martinez: Beam Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Liquori: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Marshall: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Lam: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Olins: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Rinaldi: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Rehberger: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Lazarra: Beam Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Decker: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Gantzer: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Bohnuud: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Born: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Barrera: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Yan: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Slaymaker: Beam Therapeutics: Current Employment, Current holder of stock options in a privately-held company, Patents & Royalties. Packer: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Smith: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Zambonelli: Beam Therapeutics: Current Employment, Current holder of stock options in a privately-held company. Lee: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Gaudelli: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Hartigan: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of stock options in a privately-held company. Ciaramella: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company, Membership on an entity's Board of Directors or advisory committees.
- Published
- 2021
- Full Text
- View/download PDF
13. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage
- Author
-
Holly A. Rees, Ahmed H. Badran, David R. Liu, David I. Bryson, Nicole M. Gaudelli, Alexis C. Komor, and Michael S. Packer
- Subjects
Models, Molecular ,0301 basic medicine ,Adenosine Deaminase ,Base pair ,Guanine ,CRISPR-Associated Proteins ,Deamination ,Biology ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,chemistry.chemical_compound ,Cell Line, Tumor ,Humans ,DNA Cleavage ,Base Pairing ,Gene Editing ,Genetics ,Multidisciplinary ,Genome, Human ,Point mutation ,DNA ,genomic DNA ,HEK293 Cells ,030104 developmental biology ,chemistry ,Human genome ,Cytosine - Abstract
The spontaneous deamination of cytosine is a major source of transitions from C•G to T•A base pairs, which account for half of known pathogenic point mutations in humans. The ability to efficiently convert targeted A•T base pairs to G•C could therefore advance the study and treatment of genetic diseases. The deamination of adenine yields inosine, which is treated as guanine by polymerases, but no enzymes are known to deaminate adenine in DNA. Here we describe adenine base editors (ABEs) that mediate the conversion of A•T to G•C in genomic DNA. We evolved a transfer RNA adenosine deaminase to operate on DNA when fused to a catalytically impaired CRISPR-Cas9 mutant. Extensive directed evolution and protein engineering resulted in seventh-generation ABEs that convert targeted A•T base pairs efficiently to G•C (approximately 50% efficiency in human cells) with high product purity (typically at least 99.9%) and low rates of indels (typically no more than 0.1%). ABEs introduce point mutations more efficiently and cleanly, and with less off-target genome modification, than a current Cas9 nuclease-based method, and can install disease-correcting or disease-suppressing mutations in human cells. Together with previous base editors, ABEs enable the direct, programmable introduction of all four transition mutations without double-stranded DNA cleavage.
- Published
- 2017
- Full Text
- View/download PDF
14. Celebrating Rosalind Franklin’s Centennial with a Nobel Win for Doudna and Charpentier
- Author
-
Nicole M. Gaudelli and Alexis C. Komor
- Subjects
Gene Editing ,Pharmacology ,media_common.quotation_subject ,COVID-19 ,Art history ,Art ,Nobel Prize ,Chemistry ,Editorial ,Centennial ,CRISPR-Associated Protein 9 ,Drug Discovery ,Genetics ,Humans ,Molecular Medicine ,Clustered Regularly Interspaced Short Palindromic Repeats ,Female ,CRISPR-Cas Systems ,Pandemics ,Molecular Biology ,media_common - Published
- 2020
- Full Text
- View/download PDF
15. Directed evolution of adenine base editors with increased activity and therapeutic application
- Author
-
Nicole M, Gaudelli, Dieter K, Lam, Holly A, Rees, Noris M, Solá-Esteves, Luis A, Barrera, David A, Born, Aaron, Edwards, Jason M, Gehrke, Seung-Joo, Lee, Alexander J, Liquori, Ryan, Murray, Michael S, Packer, Conrad, Rinaldi, Ian M, Slaymaker, Jonathan, Yen, Lauren E, Young, and Giuseppe, Ciaramella
- Subjects
Gene Editing ,Cytosine ,HEK293 Cells ,Adenosine Deaminase ,Adenine ,Mutation ,Humans ,DNA ,CRISPR-Cas Systems ,RNA, Guide, Kinetoplastida - Abstract
The foundational adenine base editors (for example, ABE7.10) enable programmable A•T to G•C point mutations but editing efficiencies can be low at challenging loci in primary human cells. Here we further evolve ABE7.10 using a library of adenosine deaminase variants to create ABE8s. At NGG protospacer adjacent motif (PAM) sites, ABE8s result in ~1.5× higher editing at protospacer positions A5-A7 and ~3.2× higher editing at positions A3-A4 and A8-A10 compared with ABE7.10. Non-NGG PAM variants have a ~4.2-fold overall higher on-target editing efficiency than ABE7.10. In human CD34
- Published
- 2019
16. Structure of a bound peptide phosphonate reveals the mechanism of nocardicin bifunctional thioesterase epimerase-hydrolase half-reactions
- Author
-
Nicole M. Gaudelli, Andrew M. Gulick, Craig A. Townsend, Andrew R. Buller, Ketan D. Patel, and Felipe B. d’Andrea
- Subjects
0301 basic medicine ,Models, Molecular ,Lactams ,Stereochemistry ,Hydrolases ,Science ,Organophosphonates ,General Physics and Astronomy ,Peptide ,Peptide binding ,02 engineering and technology ,Biosynthesis ,Crystallography, X-Ray ,General Biochemistry, Genetics and Molecular Biology ,Article ,Nocardia ,Protein Structure, Secondary ,Substrate Specificity ,03 medical and health sciences ,chemistry.chemical_compound ,Protein structure ,Thioesterase ,Bacterial Proteins ,Nonribosomal peptide ,Hydrolase ,Peptide Synthases ,lcsh:Science ,X-ray crystallography ,Amino Acid Isomerases ,chemistry.chemical_classification ,Multidisciplinary ,Chemistry ,Stereoisomerism ,General Chemistry ,021001 nanoscience & nanotechnology ,3. Good health ,Amino acid ,030104 developmental biology ,Enzyme mechanisms ,lcsh:Q ,0210 nano-technology ,Peptides - Abstract
Nonribosomal peptide synthetases (NRPSs) underlie the biosynthesis of many natural products that have important medicinal utility. Protection of the NRPS peptide products from proteolysis is critical to these pathways and is often achieved by structural modification, principally the introduction of d-amino acid residues into the elongating peptide. These amino acids are generally formed in situ from their l-stereoisomers by epimerization domains or dual-function condensation/epimerization domains. In singular contrast, the thioesterase domain of nocardicin biosynthesis mediates both the effectively complete l- to d-epimerization of its C-terminal amino acid residue (≥100:1) and hydrolytic product release. We report herein high-resolution crystal structures of the nocardicin thioesterase domain in ligand-free form and reacted with a structurally precise fluorophosphonate substrate mimic that identify the complete peptide binding pocket to accommodate both stereoisomers. These structures combined with additional functional studies provide detailed mechanistic insight into this unique dual-function NRPS domain., NocTE is a nonribosomal peptide synthetase thioesterase that completes the biosynthesis of pro-nocardicin G, the precursor for nocardicin β-lactam antibiotics. Here the authors provide mechanistic insights into NocTE by determining its crystal structures in the ligand-free form and covalently linked to a fluorophosphonate substrate mimic.
- Published
- 2019
17. Adenine Base Editing of the Sickle Allele in CD34+ Hematopoietic Stem and Progenitor Cells Eliminates Hemoglobin S
- Author
-
Giuseppe Ciaramella, Adam J. Hartigan, Calvin Lee, Alexander Liquori, Bo Yan, David A. Born, Tanggis Bohnuud, S. Haihua Chu, Jeffrey Marshall, Jennifer Olins, Daisy Lam, Luis A. Barrera, Ian Slaymaker, Jeremy Decker, Bob Gantzer, Nicole M. Gaudelli, Michael S. Packer, and Scott Haskett
- Subjects
Cellular differentiation ,Genetic enhancement ,Immunology ,Hemoglobin variants ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Virology ,Haematopoiesis ,Erythropoiesis ,Globin ,Progenitor cell ,Stem cell - Abstract
While there are several small molecule, gene therapy, and gene editing approaches for treating sickle cell disease (SCD), these strategies do not result in the direct elimination of the causative sickle β-globin (HbS) variant itself. The reduction or complete removal of this pathologic globin variant and expression of normal β-hemoglobin (HbB) or other non-polymerizing β-globin variant may increase the likelihood of beneficial outcomes for SCD patients. Adenine base editors (ABEs) can precisely convert the mutant A-T base pair responsible for SCD to a G-C base pair, thus generating the hemoglobin variant, Hb G-Makassar, a naturally occurring β-globin variant that is not associated with human disease. Our studies have identified ABEs that can achieve highly efficient Makassar editing (>70%) of the sickle mutation in both sickle trait (HbAS) and homozygous sickle (HbSS) patient CD34+ cells with high cell viability and recovery and without perturbation of immunophenotypic hematopoietic stem and progenitor cell (HSPC) frequencies after ex vivo delivery of guide RNA and mRNA encoding the ABE. Furthermore, Makassar editing was retained throughout erythropoiesis in bulk in vitro erythroid differentiated cells (IVED) derived from edited CD34+ cells. To gain an understanding of allelic editing at a single clone resolution, we assessed editing frequencies of clones from both single cell expansion in erythroid differentiation media, as well as from single BFU-E colonies. We found that we could achieve >70% of colonies with bi-allelic Makassar editing and approximately 20% of colonies with mono-allelic Makassar editing, while Previously, conventional hemoglobin capillary electrophoresis and high-performance liquid chromatography (HPLC) were unable to distinguish between HbS and HbG-Makassar. Here, we developed an ultra-high-performance liquid chromatography (UPLC) method that resolves sickle globin (HbS) from Hb G-Makassar globin in IVED cells. The Makassar globin variant was further confirmed by liquid chromatography mass spectrometry (LC-MS). By developing this new method to resolve these two β-globin variants in edited HbSS cells, we were able to detect, in bulk IVED cultures, >80% abundance Hb G-Makassar of total β-globins, which corresponded to a concomitant reduction of HbS levels to 70% of cells that had bi-allelic Makassar editing. Moreover, in the approximately 20% of colonies that were found to be mono-allelically edited for the Makassar variant, there was a 60:40 ratio of Hb G-Makassar:HbS globin abundance in individual clones, at levels remarkably similar to the HbA(wildtype HbB):HbS levels found in HbAS individuals, with minimal observable in vitro sickling when exposed to hypoxia. Thus, with our ABEs, we were able to reduce HbS to 90% of IVED cells and found that in vitro sickling under hypoxia inversely correlated with the level of Hb G-Makassar globin variant installation and corresponding reduction in HbS levels. By converting HbS to Hb G-Makassar, our direct and precise editing strategy replaces a pathogenic β-globin with one that has been shown to have normal hematologic parameters. Coupled with autologous stem cell transplant, this next generation gene editing strategy presents a promising new modality for treating patients with SCD. Disclosures Chu: Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Lam:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Packer:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Olins:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Liquori:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Marshall:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Lee:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Yan:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Decker:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Gantzer:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Haskett:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Bohnuud:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Born:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Barrera:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Slaymaker:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Gaudelli:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Hartigan:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company. Ciaramella:Beam Therapeutics: Current Employment, Current equity holder in publicly-traded company.
- Published
- 2020
- Full Text
- View/download PDF
18. Circularly permuted and PAM-modified Cas9 variants broaden the targeting scope of base editors
- Author
-
Shannon M. Miller, David F. Savage, Nicole M. Gaudelli, Kevin T. Zhao, Christof Fellmann, David R. Liu, Tony P. Huang, and Benjamin L. Oakes
- Subjects
Computer science ,Biomedical Engineering ,Bioengineering ,Computational biology ,Applied Microbiology and Biotechnology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Cytosine ,0302 clinical medicine ,CRISPR-Associated Protein 9 ,Humans ,Nucleotide ,030304 developmental biology ,chemistry.chemical_classification ,Gene Editing ,0303 health sciences ,Extramural ,Cas9 ,Nucleotides ,Adenine ,Base (topology) ,Protospacer adjacent motif ,chemistry ,Molecular Medicine ,CRISPR-Cas Systems ,030217 neurology & neurosurgery ,Scope (computer science) ,Biotechnology ,Plasmids - Abstract
Base editing requires that the target sequence satisfy the PAM requirement of the Cas9 domain and that the target nucleotide is located within the editing window of the base editor. To increase the targeting scope of base editors, we engineered six optimized adenine base editors (ABEmax variants) that use SpCas9 variants compatible with non-NGG PAMs. To increase the range of target bases that can be modified within the protospacer, we use circularly permuted Cas9 variants to produce four cytosine and four adenine base editors with an editing window expanded from ~4–5 nucleotides to up to ~8–9 nucleotides and reduced byproduct formation. This set of base editors improves the targeting scope of cytosine and adenine base editing., Ed sum: Wider editing windows and different PAM requirements enable a broad set of genomic positions to be targeted with A and C base editors.
- Published
- 2018
19. Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C:G-to-T:A base editors with higher efficiency and product purity
- Author
-
Luke W. Koblan, Amanda L. Waterbury, Ahmed H. Badran, Kevin T. Zhao, David R. Liu, Michael S. Packer, Nicole M. Gaudelli, Alexis C. Komor, and Y. Bill Kim
- Subjects
0301 basic medicine ,DNA Repair ,Base pair ,DNA repair ,Stereochemistry ,Cell Line ,Bacteriophage mu ,03 medical and health sciences ,chemistry.chemical_compound ,Viral Proteins ,Genome editing ,Gene Frequency ,INDEL Mutation ,Gene Order ,Humans ,Genome Editing ,Uracil-DNA Glycosidase ,Base Pairing ,Research Articles ,Genetics ,Multidisciplinary ,Chemistry ,SciAdv r-articles ,Life Sciences ,Cytidine ,Base excision repair ,Cytidine deaminase ,Base (topology) ,DNA-Binding Proteins ,Enzyme Activation ,030104 developmental biology ,Bacteriophage Mu ,Research Article - Abstract
Probing base editing outcomes leads to new C:G to T:A base editors with greater efficiency and product purity, and fewer indels., We recently developed base editing, the programmable conversion of target C:G base pairs to T:A without inducing double-stranded DNA breaks (DSBs) or requiring homology-directed repair using engineered fusions of Cas9 variants and cytidine deaminases. Over the past year, the third-generation base editor (BE3) and related technologies have been successfully used by many researchers in a wide range of organisms. The product distribution of base editing—the frequency with which the target C:G is converted to mixtures of undesired by-products, along with the desired T:A product—varies in a target site–dependent manner. We characterize determinants of base editing outcomes in human cells and establish that the formation of undesired products is dependent on uracil N-glycosylase (UNG) and is more likely to occur at target sites containing only a single C within the base editing activity window. We engineered CDA1-BE3 and AID-BE3, which use cytidine deaminase homologs that increase base editing efficiency for some sequences. On the basis of these observations, we engineered fourth-generation base editors (BE4 and SaBE4) that increase the efficiency of C:G to T:A base editing by approximately 50%, while halving the frequency of undesired by-products compared to BE3. Fusing BE3, BE4, SaBE3, or SaBE4 to Gam, a bacteriophage Mu protein that binds DSBs greatly reduces indel formation during base editing, in most cases to below 1.5%, and further improves product purity. BE4, SaBE4, BE4-Gam, and SaBE4-Gam represent the state of the art in C:G-to-T:A base editing, and we recommend their use in future efforts.
- Published
- 2017
20. Highly Efficient Multiplexed Base Editing with Minimized Off-Targets for the Development of Universal CAR-T Cells to Treat Pediatric T-ALL
- Author
-
Waseem Qasim, Ryan Murray, Aaron Edwards, Yi Yu, Giuseppe Ciaramella, Jason Michael Gehrke, Yeh Chuin Poh, Sarah E. Smith, Nicole M. Gaudelli, and Amy Shaw
- Subjects
education.field_of_study ,CD52 ,Cell growth ,T cell ,Immunology ,Population ,Cell ,Cell Biology ,Hematology ,Computational biology ,Biology ,Biochemistry ,Cell therapy ,medicine.anatomical_structure ,Genome editing ,medicine ,Chimeric Antigen Receptor T-Cell Therapy ,education - Abstract
Autologous CAR-T therapies have demonstrated remarkable efficacy in treating some hematologic cancers. While these therapies can have substantial clinical benefit for patients, generating bespoke cell therapies creates manufacturing challenges, resulting in inconsistent products and delays in treatment that are often incompatible with effective clinical management of patients. Strategies to create universally-compatible CAR-T therapies, generated from single donors for the treatment of many patients, have been developed as a solution to these challenges, thereby reducing cost of goods, lot-to-lot variability and enabling timely treatment. Mitigating the risks of graft-versus-host-disease (GvHD) and host rejection of CAR-Ts are important components of any strategy to generate these universal therapies. Most first generation approaches utilize DNA double strand break (DSB)-inducing nucleases to ablate the expression of relevant genes in donor T cells to overcome these barriers. However, simultaneous induction of multiple DSBs results in a cell population containing genomic rearrangements, and can lead to significantly reduced cell proliferation. Approaches to develop CAR-T therapies for T cell malignancies, such as T-ALL, encounter additional challenges, including extensive fratricide caused by targeting T cell surface markers such as CD3 and CD7, which are present on both the malignant and the CAR-T cells. Resolving this issue requires additional gene editing, leading to, in the case of nuclease-based strategies, an increased number of DSBs, further genomic rearrangements, and decreased cell expansion. Base editors (BEs) are a novel class of gene editing reagents that enable programmable, single-base changes in genomic DNA without creating DSBs. Work in the Qasim lab has demonstrated proof of concept for an alternative means of producing universal, fratricide-resistant CAR-T cells for treating T cell malignancies by using cytosine base editor (CBE) technology. Here, we demonstrate a previously-unpublished CBE that produces greatly diminished off-target effects while preserving on-target activity. Using multiplex base editing, we demonstrate simultaneous knockout of four genes (TRAC, CD7, CD52, and PDCD1) with between 80-95% efficiency, producing engineered CAR-T cells with greatly diminished risk of GvHD, graft cell rejection, fratricide, and exhaustion. We show that, in contrast to nuclease editing, concurrent modification of four genetic loci using our reduced off-target CBE produces highly efficient gene knockouts with no detectable genomic rearrangements and no observable change in cell expansion compared to control conditions. T-ALL is a heterogeneous disease with variable expression of CD3 and CD7 across tumor cells in the same patient. To reduce the risk of antigen escape by tumor cells during the course of treatment with base edited CAR-Ts, we envisioned creating two independent CAR-T populations targeting CD3 (3CAR-Ts) or CD7 (7CAR-Ts). Alone or in combination, base edited 3CAR-Ts and 7CAR-Ts demonstrate robust cytokine release, potent in vitro cytotoxicity, and in vivo tumor control with antigen-positive tumor cells, and display minimal antigen-independent activity. Taken together, our approach addresses existing limitations in CAR-T cell manufacturing and demonstrates that simultaneous base editing using an improved specificity CBE at four target genes is a feasible strategy for generating universal, fratricide-resistant CAR-T cells for the potential treatment of T cell malignancies such as T-ALL. More generally, this program demonstrates the potential for base editing to create highly-engineered cell therapies featuring at least four simultaneous edits, which can confer a wide range of desirable therapeutic attributes. Disclosures Qasim: CellMedica: Research Funding; Bellicum: Research Funding; Servier: Research Funding; Orchard Therapeutics: Equity Ownership; UCLB: Other: revenue share eligibility; Autolus: Equity Ownership.
- Published
- 2019
- Full Text
- View/download PDF
21. Complementary Base Editing Approaches for the Treatment of Sickle Cell Disease and Beta Thalassemia
- Author
-
Sarah E. Smith, Jonathan Yen, Manmohan Singh, Elsie Akrawi, Giuseppe Ciaramella, Ling Lin, Fu Yanfang, Michael S. Packer, Nicole M. Gaudelli, Yeh Chuin Poh, Dana N. Levasseur, Conrad Rinaldi, Scott J. Haskett, Minerva E. Sanchez, and Adrian P. Rybak
- Subjects
0301 basic medicine ,education.field_of_study ,HBG1 ,Hereditary persistence of fetal hemoglobin ,Immunology ,Population ,Beta thalassemia ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,HBG2 ,Transplantation ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,hemic and lymphatic diseases ,Fetal hemoglobin ,medicine ,Autologous transplantation ,education ,030215 immunology - Abstract
Sickle cell disease (SCD) and Beta thalassemia are disorders of beta globin production and function that lead to severe anemia and significant disease complications across a multitude of organ systems. Autologous transplantation of hematopoietic stem cells engineered through the upregulation of fetal hemoglobin (HbF) or correction of the beta globin gene have the potential to reduce disease burden in patients with beta hemoglobinopathies. Base editing is a recently developed technology that enables precise modification of the genome without the introduction of double strand DNA breaks. Gamma globin gene promoters were comprehensively screened with cytosine and adenine base editors (ABE) for the identification of alterations that would derepress HbF. Three regions were identified that significantly upregulated HbF, and the most effective nucleotide residue conversions are supported by natural variation seen in patients with hereditary persistence of fetal hemoglobin (HPFH). ABEs have been developed that significantly increase the level of HbF following nucleotide conversion at key regulatory motifs within the HBG1 and HBG2 promoters. CD34+ hematopoietic stem and progenitor cells (HSPC) were purified at clinical scale and edited using a process designed to preserve self-renewal capacity. Editing at two independent sites with different ABEs reached 94 percent and resulted in up to 63 percent gamma globin by UPLC. The levels of HbF observed should afford protection to the majority of SCD and Beta thalassemia patients based on clinical observations of HPFH and non-interventional therapy that links higher HbF dosage with milder disease (Ngo et al, 2011 Brit J Hem; Musallam et al, 2012 Blood). Directly correcting the Glu6Val mutation of SCD has been a recent goal of genetic therapies designed for the SCD population. Current base editing technology cannot yet convert mutations like those that result from the A-T transversion in sickle beta globin; however, ABE variants have been designed to recognize and edit the opposite stranded adenine residue of valine. This results in the conversion of valine to alanine and the production of a naturally occurring variant known as Hb G-Makassar. Beta globin with alanine at this position does not contribute to polymer formation, and patients with Hb G-Makassar present with normal hematological parameters and red blood cell morphology. SCD patient fibroblasts edited with these ABE variants achieve up to 70 percent conversion of the target adenine. CD34 cells from healthy donors were then edited with a lead ABE variant, targeting a synonymous mutation in an adjacent proline that resides within the editing window and serves as a proxy for editing the SCD mutation. The average editing frequency was 40 percent. Donor myeloid chimerism documented at these levels in the allogeneic transplant setting exceeds the 20 percent that is required for reversing the sickle phenotype (Fitzhugh et al, 2017 Blood). These next generation editing approaches provide a promising new modality for treating patients with Beta thalassemia and SCD. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
- Full Text
- View/download PDF
22. Author Correction: Circularly permuted and PAM-modified Cas9 variants broaden the targeting scope of base editors
- Author
-
Nicole M. Gaudelli, Tony P. Huang, Kevin T. Zhao, Christof Fellmann, David R. Liu, David F. Savage, Shannon M. Miller, and Benjamin L. Oakes
- Subjects
Information retrieval ,Scope (project management) ,Computer science ,Published Erratum ,Biomedical Engineering ,Molecular Medicine ,Bioengineering ,Base (topology) ,Applied Microbiology and Biotechnology ,Biotechnology - Published
- 2019
- Full Text
- View/download PDF
23. Stereocontrolled Syntheses of Peptide Thioesters Containing Modified Seryl Residues as Probes of Antibiotic Biosynthesis
- Author
-
Nicole M. Gaudelli and Craig A. Townsend
- Subjects
chemistry.chemical_classification ,Oligopeptide ,Lactams ,Molecular Structure ,Stereochemistry ,Organic Chemistry ,Esters ,Stereoisomerism ,Peptide ,Thioester ,Pentapeptide repeat ,Article ,Anti-Bacterial Agents ,Peptide Synthases ,Serine ,chemistry ,Intramolecular force ,Sulfhydryl Compounds ,Oligopeptides - Abstract
Methods have been developed to synthesize tri- and pentapeptide thioesters containing one or more p-(hydroxyphenyl)glycine (pHPG) residues and L-serine, some where the latter is O-phosphorylated, O-acetylated, or exists as a β-lactam. Selection of orthogonal protection strategies and development of conditions to achieve seryl O-phosphorylation without β-elimination and to maintain stereochemical control, especially simultaneously at exceptionally base-labile pHPG α-carbons, are described. Intramolecular closure of a seryl peptide to a β-lactam-containing peptide and the syntheses of corresponding thioester analogues are also reported. Modification of classical Mitsunobu conditions is described in the synthesis of the β-lactam-containing products, and in a broadly useful observation, it was found that simple exclusion of light from the P(OEt)3-mediated Mitsunobu ring closure afforded yields of95%, presumably owing to reduced photodegradation of the azodicarboxylate used. These sensitive potential substrates and products will be used in mechanistic studies of the two nonribosomal peptide synthetases NocA and NocB that lie at the heart of nocardicin biosynthesis, a family of monocyclic β-lactam antibiotics.
- Published
- 2013
- Full Text
- View/download PDF
24. Publisher Correction: Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage
- Author
-
David I. Bryson, Ahmed H. Badran, Michael S. Packer, Nicole M. Gaudelli, Alexis C. Komor, Holly A. Rees, and David R. Liu
- Subjects
0301 basic medicine ,DNA metabolism ,03 medical and health sciences ,genomic DNA ,030104 developmental biology ,Multidisciplinary ,Dna cleavage ,Chemistry ,Molecular evolution ,Targeted Gene Repair ,Computational biology ,Base (exponentiation) - Abstract
In this Article, owing to an error during the production process, in Fig. 1a, the dark blue and light blue wedges were incorrectly labelled as ‘G•C → T•A’ and ‘G•C → A•T’, instead of ‘C•G → T•A’ and ‘C•G → A•T’, respectively. Fig. 1 has been corrected online.
- Published
- 2018
- Full Text
- View/download PDF
25. β-Lactam formation by a non-ribosomal peptide synthetase during antibiotic biosynthesis
- Author
-
Darcie H. Long, Nicole M. Gaudelli, and Craig A. Townsend
- Subjects
Lactams ,Stereochemistry ,medicine.medical_treatment ,Peptide ,Tripeptide ,Biology ,beta-Lactams ,Article ,Peptide Synthases ,Condensation domain ,chemistry.chemical_compound ,Biosynthesis ,medicine ,polycyclic compounds ,Serine ,Peptide bond ,Histidine ,chemistry.chemical_classification ,Multidisciplinary ,Protease ,biochemical phenomena, metabolism, and nutrition ,Amino acid ,Anti-Bacterial Agents ,Biosynthetic Pathways ,chemistry ,Biochemistry ,Cyclization ,Biocatalysis - Abstract
Non-ribosomal peptide synthetases are giant enzymes composed of modules that house repeated sets of functional domains, which select, activate and couple amino acids drawn from a pool of nearly 500 potential building blocks. The structurally and stereochemically diverse peptides generated in this manner underlie the biosynthesis of a large sector of natural products. Many of their derived metabolites are bioactive such as the antibiotics vancomycin, bacitracin, daptomycin and the β-lactam-containing penicillins, cephalosporins and nocardicins. Penicillins and cephalosporins are synthesized from a classically derived non-ribosomal peptide synthetase tripeptide (from δ-(L-α-aminoadipyl)-L-cysteinyl-D-valine synthetase). Here we report an unprecedented non-ribosomal peptide synthetase activity that both assembles a serine-containing peptide and mediates its cyclization to the critical β-lactam ring of the nocardicin family of antibiotics. A histidine-rich condensation domain, which typically performs peptide bond formation during product assembly, also synthesizes the embedded four-membered ring. We propose a mechanism, and describe supporting experiments, that is distinct from the pathways that have evolved to the three other β-lactam antibiotic families: penicillin/cephalosporins, clavams and carbapenems. These findings raise the possibility that β-lactam rings can be regio- and stereospecifically integrated into engineered peptides for application as, for example, targeted protease inactivators.
- Published
- 2014
26. Correction to Stereocontrolled Syntheses of Peptide Thioesters Containing Modified Seryl Residues as Probes of Antibiotic Biosynthesis
- Author
-
Nicole M. Gaudelli and Craig A. Townsend
- Subjects
chemistry.chemical_classification ,Biochemistry ,Chemistry ,Organic Chemistry ,Peptide ,Antibiotic biosynthesis - Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.