Network


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

Hotspot


Dive into the research topics where Mark A. DeWitt is active.

Publication


Featured researches published by Mark A. DeWitt.


Nature Biotechnology | 2016

Enhancing homology-directed genome editing by catalytically active and inactive CRISPR-Cas9 using asymmetric donor DNA

Christopher D. Richardson; Graham J. Ray; Mark A. DeWitt; Gemma L. Curie; Jacob E. Corn

Targeted genomic manipulation by Cas9 can efficiently generate knockout cells and organisms via error-prone nonhomologous end joining (NHEJ), but the efficiency of precise sequence replacement by homology-directed repair (HDR) is substantially lower. Here we investigate the interaction of Cas9 with target DNA and use our findings to improve HDR efficiency. We show that dissociation of Cas9 from double-stranded DNA (dsDNA) substrates is slow (lifetime ∼6 h) but that, before complete dissociation, Cas9 asymmetrically releases the 3′ end of the cleaved DNA strand that is not complementary to the sgRNA (nontarget strand). By rationally designing single-stranded DNA (ssDNA) donors of the optimal length complementary to the strand that is released first, we increase the rate of HDR in human cells when using Cas9 or nickase variants to up to 60%. We also demonstrate HDR rates of up to 0.7% using a catalytically inactive Cas9 mutant (dCas9), which binds DNA without cleaving it.


Science | 2012

Cytoplasmic Dynein Moves Through Uncoordinated Stepping of the AAA+ Ring Domains

Mark A. DeWitt; Amy Y. Chang; Peter A. Combs; Ahmet Yildiz

Doing the Side Step The molecular motor, dynein, contains two ring domains responsible for its movement along the microtubule. However, how the rings move relative to each other during processive motility and whether dynein processivity requires interhead coordination are unclear. To directly observe how dynein “walks” along microtubules, DeWitt et al. (p. 221, published online 8 December) performed advanced fluorescence-imaging studies to follow both motor domains of a single dynein motor at nanometer resolution. The data suggest that the two heads do not cooperate during movement, which suggests a fundamentally different mechanism of motility from that observed for other microtubule-based motors. The molecular motor dynein moves each of its two heads independently along the microtubule. Cytoplasmic dynein is a homodimeric AAA+ motor that transports a multitude of cargos toward the microtubule minus end. How the two catalytic head domains interact and move relative to each other during processive movement is unclear. Here, we tracked the relative positions of both heads with nanometer precision and directly observed the heads moving independently along the microtubule. The heads remained widely separated, and their stepping behavior varied as a function of interhead separation. One active head was sufficient for processive movement, and an active head could drag an inactive partner head forward. Thus, dynein moves processively without interhead coordination, a mechanism fundamentally distinct from the hand-over-hand stepping of kinesin and myosin.


Science Translational Medicine | 2016

Selection-free genome editing of the sickle mutation in human adult hematopoietic stem/progenitor cells

Mark A. DeWitt; Wendy Magis; Nicolas Bray; Tianjiao Wang; Jennifer R. Berman; Fabrizia Urbinati; Seok Jin Heo; Therese Mitros; Denise P. Muñoz; Dario Boffelli; Donald B. Kohn; Mark C. Walters; Dana Carroll; David I. K. Martin; Jacob E. Corn

Hematopoietic stem cells from patients with sickle cell disease can be edited by CRISPR/Cas9 and maintain the edits in vivo. Hammering out the sickle cell mutation Sickle cell disease is a genetic disorder caused by a mutation in one of the hemoglobin genes, which causes deformation of red blood cells and results in occlusion of blood vessels, severe pain crises, and progressive organ injury. To correct the mutation that causes this disease, DeWitt et al. modified hematopoietic stem cells from sickle cell disease patients using a CRISPR/Cas9 gene editing approach. The authors showed that the corrected cells successfully engrafted in a mouse model and produced enough normal hemoglobin to have a potential clinical benefit in the setting of sickle cell disease. Genetic diseases of blood cells are prime candidates for treatment through ex vivo gene editing of CD34+ hematopoietic stem/progenitor cells (HSPCs), and a variety of technologies have been proposed to treat these disorders. Sickle cell disease (SCD) is a recessive genetic disorder caused by a single-nucleotide polymorphism in the β-globin gene (HBB). Sickle hemoglobin damages erythrocytes, causing vasoocclusion, severe pain, progressive organ damage, and premature death. We optimize design and delivery parameters of a ribonucleoprotein (RNP) complex comprising Cas9 protein and unmodified single guide RNA, together with a single-stranded DNA oligonucleotide donor (ssODN), to enable efficient replacement of the SCD mutation in human HSPCs. Corrected HSPCs from SCD patients produced less sickle hemoglobin RNA and protein and correspondingly increased wild-type hemoglobin when differentiated into erythroblasts. When engrafted into immunocompromised mice, ex vivo treated human HSPCs maintain SCD gene edits throughout 16 weeks at a level likely to have clinical benefit. These results demonstrate that an accessible approach combining Cas9 RNP with an ssODN can mediate efficient HSPC genome editing, enables investigator-led exploration of gene editing reagents in primary hematopoietic stem cells, and suggests a path toward the development of new gene editing treatments for SCD and other hematopoietic diseases.


Nature Biomedical Engineering | 2017

Nanoparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo induces homology-directed DNA repair

Kunwoo Lee; Michael J. Conboy; Hyo Min Park; Fuguo Jiang; Hyun Jin Kim; Mark A. DeWitt; Vanessa Mackley; Kevin Chang; Anirudh Rao; Colin Skinner; Tamanna Shobha; Melod Mehdipour; Hui Liu; Wen-chin Huang; Freeman Lan; Nicolas Bray; Song Li; Jacob E. Corn; Kazunori Kataoka; Jennifer A. Doudna; Irina M. Conboy; Niren Murthy

Clustered regularly interspaced short palindromic repeats (CRISPR)–CRISPR associated protein 9 (Cas9)-based therapeutics, especially those that can correct gene mutations via homology-directed repair, have the potential to revolutionize the treatment of genetic diseases. However, it is challenging to develop homology-directed repair-based therapeutics because they require the simultaneous in vivo delivery of Cas9 protein, guide RNA and donor DNA. Here, we demonstrate that a delivery vehicle composed of gold nanoparticles conjugated to DNA and complexed with cationic endosomal disruptive polymers can deliver Cas9 ribonucleoprotein and donor DNA into a wide variety of cell types and efficiently correct the DNA mutation that causes Duchenne muscular dystrophy in mice via local injection, with minimal off-target DNA damage.Gold nanoparticles carrying Cas9 ribonucleoprotein and donor DNA, and complexed with endosomal disruptive polymers, correct the DNA mutation that causes Duchenne muscular dystrophy in mice, with minimal off-target effects.


Nature Communications | 2014

Tension on the linker gates the ATP-dependent release of dynein from microtubules

Frank B. Cleary; Mark A. DeWitt; Thomas Bilyard; Zaw Min Htet; Vladislav Belyy; Danna D. Chan; Amy Y. Chang; Ahmet Yildiz

Cytoplasmic dynein is a dimeric motor that transports intracellular cargoes towards the minus-end of microtubules (MTs). In contrast to other processive motors, stepping of the dynein motor domains (heads) is not precisely coordinated. Therefore, the mechanism of dynein processivity remains unclear. Here, by engineering the mechanical and catalytic properties of the motor, we show that dynein processivity minimally requires a single active head and a second inert MT binding domain. Processivity arises from a high ratio of MT-bound to unbound time, and not from interhead communication. Additionally, nucleotide-dependent microtubule release is gated by tension on the linker domain. Intramolecular tension sensing is observed in dynein’s stepping motion at high interhead separations. We developed a quantitative model for the stepping characteristics of dynein and its response to chemical and mechanical perturbation.


eLife | 2014

Bidirectional Helical Motility of Cytoplasmic Dynein around Microtubules

Sinan Can; Mark A. DeWitt; Ahmet Yildiz

Cytoplasmic dynein is a molecular motor responsible for minus-end-directed cargo transport along microtubules (MTs). Dynein motility has previously been studied on surface-immobilized MTs in vitro, which constrains the motors to move in two dimensions. In this study, we explored dynein motility in three dimensions using an MT bridge assay. We found that dynein moves in a helical trajectory around the MT, demonstrating that it generates torque during cargo transport. Unlike other cytoskeletal motors that produce torque in a specific direction, dynein generates torque in either direction, resulting in bidirectional helical motility. Dynein has a net preference to move along a right-handed helical path, suggesting that the heads tend to bind to the closest tubulin binding site in the forward direction when taking sideways steps. This bidirectional helical motility may allow dynein to avoid roadblocks in dense cytoplasmic environments during cargo transport. DOI: http://dx.doi.org/10.7554/eLife.03205.001


Methods | 2017

Genome editing via delivery of Cas9 ribonucleoprotein

Mark A. DeWitt; Jacob E. Corn; Dana Carroll

The CRISPR-Cas genome editing system is very powerful. The format of the CRISPR reagents and the means of delivery are often important factors in targeting efficiency. Delivery of recombinant Cas9 protein and guide RNA (gRNA) as a preformed ribonucleoprotein (RNP) complex has recently emerged as a powerful and general approach to genome editing. Here we outline methods to produce and deliver Cas9 RNPs. A donor DNA carrying desired sequence changes can also be included to program precise sequence introduction or replacement. RNP delivery limits exposure to genome editing reagents, reduces off-target events, drives high rates of homology-dependent repair, and can be applied to embryos to rapidly generate animal models. RNP delivery thus minimizes some of the pitfalls of alternative editing modalities and is rapidly being adopted by the genome editing community.


eLife | 2017

Synthetically modified guide RNA and donor DNA are a versatile platform for CRISPR-Cas9 engineering

Kunwoo Lee; Vanessa Mackley; Anirudh Rao; Anthony Chong; Mark A. DeWitt; Jacob E. Corn; Niren Murthy

Chemical modification of the gRNA and donor DNA has great potential for improving the gene editing efficiency of Cas9 and Cpf1, but has not been investigated extensively. In this report, we demonstrate that the gRNAs of Cas9 and Cpf1, and donor DNA can be chemically modified at their terminal positions without losing activity. Moreover, we show that 5’ fluorescently labeled donor DNA can be used as a marker to enrich HDR edited cells by a factor of two through cell sorting. In addition, we demonstrate that the gRNA and donor DNA can be directly conjugated together into one molecule, and show that this gRNA-donor DNA conjugate is three times better at transfecting cells and inducing HDR, with cationic polymers, than unconjugated gRNA and donor DNA. The tolerance of the gRNA and donor DNA to chemical modifications has the potential to enable new strategies for genome engineering. DOI: http://dx.doi.org/10.7554/eLife.25312.001


ACS Nano | 2017

Covalent Protein Labeling and Improved Single Molecule Optical Properties of Aqueous CdSe/CdS Quantum Dots

Sara M. Wichner; Victor R. Mann; Alexander S. Powers; Maya A. Segal; Mustafa Mir; Jigar N. Bandaria; Mark A. DeWitt; Xavier Darzacq; Ahmet Yildiz; Bruce E. Cohen

Semiconductor quantum dots (QDs) have proven to be superior probes for single-molecule imaging compared to organic or genetically encoded fluorophores, but they are limited by difficulties in protein targeting, their larger size, and on-off blinking. Here, we report compact aqueous CdSe/CdS QDs with significantly improved bioconjugation efficiency and superior single-molecule optical properties. We have synthesized covalent protein labeling ligands (i.e., SNAP tags) that are optimized for nanoparticle use, and QDs functionalized with these ligands label SNAP-tagged proteins ∼10-fold more efficiently than existing SNAP ligands. Single-molecule analysis of these QDs shows 99% of time spent in the fluorescent on-state, ∼4-fold higher quantum efficiency than standard CdSe/ZnS QDs, and 350 million photons detected before photobleaching. Bright signals of these QDs enable us to track the stepping movement of a kinesin motor in vitro, and the improved labeling efficiency enables tracking of single kinesins in live cells.


bioRxiv | 2016

Efficient Correction of the Sickle Mutation in Human Hematopoietic Stem Cells Using a Cas9 Ribonucleoprotein Complex

Mark A. DeWitt; Wendy Magis; Nicolas Bray; Tianjiao Wang; Jennifer R. Berman; Fabrizia Urbinati; Denise P. Muñoz; Donald B. Kohn; Mark C. Walters; Dana Carroll; David K Martin; Jacob E. Corn

Sickle Cell Disease (SCD) is a serious recessive genetic disorder caused by a single nucleotide polymorphism (SNP) in the ß-globin gene (HBB). Sickle hemoglobin polymerizes within red blood cells (RBCs), causing them to adopt an elongated “sickle” shape. Sickle RBCs damage vasculature, leading to severe symptoms, ultimately diminishing patient quality of life and reducing lifespan. Here, we use codelivery of a pre-formed Cas9 ribonucleoprotein complex (RNP) and a singlestranded DNA (ssDNA) oligonucleotide donor to drive sequence replacement at the SCD SNP in human CD34+ hematopoietic stem/progenitor cells (HSPCs). Corrected HSPCs from SCD patients produce less sickle hemoglobin protein and correspondingly increased wild-type hemoglobin when differentiated into erythroblasts. When injected into immunocompromised mice, treated HSPCs maintain editing long-term at therapeutically relevant levels. These results demonstrate that the Cas9 RNP/ssDNA donor approach can mediate efficient HSPC gene editing and could form the basis for treatment of SCD by autologous hematopoietic cell transplantation.

Collaboration


Dive into the Mark A. DeWitt's collaboration.

Top Co-Authors

Avatar

Jacob E. Corn

University of California

View shared research outputs
Top Co-Authors

Avatar

Ahmet Yildiz

University of California

View shared research outputs
Top Co-Authors

Avatar

Mark C. Walters

Children's Hospital Oakland

View shared research outputs
Top Co-Authors

Avatar

Wendy Magis

Children's Hospital Oakland Research Institute

View shared research outputs
Top Co-Authors

Avatar

Bruce E. Cohen

Lawrence Berkeley National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dario Boffelli

Children's Hospital Oakland Research Institute

View shared research outputs
Top Co-Authors

Avatar

David I. K. Martin

Children's Hospital Oakland Research Institute

View shared research outputs
Top Co-Authors

Avatar

Donald B. Kohn

University of California

View shared research outputs
Top Co-Authors

Avatar

Jonathan Vu

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge