Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
iScience ; 25(6): 104374, 2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35633935

ABSTRACT

Background: A point mutation in sickle cell disease (SCD) alters one amino acid in the ß-globin subunit of hemoglobin, with resultant anemia and multiorgan damage that typically shortens lifespan by decades. Because SCD is caused by a single mutation, and hematopoietic stem cells (HSCs) can be harvested, manipulated, and returned to an individual, it is an attractive target for gene correction. Results: An optimized Cas9 ribonucleoprotein (RNP) with an ssDNA oligonucleotide donor together generated correction of at least one ß-globin allele in more than 30% of long-term engrafting human HSCs. After adopting a high-fidelity Cas9 variant, efficient correction with minimal off-target events also was observed. In vivo erythroid differentiation markedly enriches for corrected ß-globin alleles, indicating that erythroblasts carrying one or more corrected alleles have a survival advantage. Significance: These findings indicate that the sickle mutation can be corrected in autologous HSCs with an optimized protocol suitable for clinical translation.

2.
Elife ; 112022 02 11.
Article in English | MEDLINE | ID: mdl-35147495

ABSTRACT

Naturally occurring point mutations in the HBG promoter switch hemoglobin synthesis from defective adult beta-globin to fetal gamma-globin in sickle cell patients with hereditary persistence of fetal hemoglobin (HPFH) and ameliorate the clinical severity. Inspired by this natural phenomenon, we tiled the highly homologous HBG proximal promoters using adenine and cytosine base editors that avoid the generation of large deletions and identified novel regulatory regions including a cluster at the -123 region. Base editing at -123 and -124 bp of HBG promoter induced fetal hemoglobin (HbF) to a higher level than disruption of well-known BCL11A binding site in erythroblasts derived from human CD34+ hematopoietic stem and progenitor cells (HSPC). We further demonstrated in vitro that the introduction of -123T > C and -124T > C HPFH-like mutations drives gamma-globin expression by creating a de novo binding site for KLF1. Overall, our findings shed light on so far unknown regulatory elements within the HBG promoter and identified additional targets for therapeutic upregulation of fetal hemoglobin.


Subject(s)
Anemia, Sickle Cell/genetics , CRISPR-Cas Systems , Fetal Hemoglobin/genetics , Gene Editing/methods , Adenine/metabolism , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Cytosine/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Point Mutation , Promoter Regions, Genetic , beta-Globins/genetics , beta-Thalassemia/genetics , gamma-Globins/genetics
3.
Cell Rep ; 32(9): 108093, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32877675

ABSTRACT

Genome editing often takes the form of either error-prone sequence disruption by non-homologous end joining (NHEJ) or sequence replacement by homology-directed repair (HDR). Although NHEJ is generally effective, HDR is often difficult in primary cells. Here, we use a combination of immunophenotyping, next-generation sequencing, and single-cell RNA sequencing to investigate and reprogram genome editing outcomes in subpopulations of adult hematopoietic stem and progenitor cells. We find that although quiescent stem-enriched cells mostly use NHEJ, non-quiescent cells with the same immunophenotype use both NHEJ and HDR. Inducing quiescence before editing results in a loss of HDR in all cell subtypes. We develop a strategy of controlled cycling and quiescence that yields a 6-fold increase in the HDR/NHEJ ratio in quiescent stem cells ex vivo and in vivo. Our results highlight the tension between editing and cellular physiology and suggest strategies to manipulate quiescent cells for research and therapeutic genome editing.


Subject(s)
CRISPR-Cas Systems/genetics , GATA3 Transcription Factor/metabolism , Gene Editing/methods , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Recombinational DNA Repair/genetics , Stem Cells/metabolism , Humans
4.
Cell Rep ; 32(5): 107993, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32755585

ABSTRACT

ß-Hemoglobinopathies can trigger rapid production of red blood cells in a process known as stress erythropoiesis. Cellular stress prompts differentiating erythroid precursors to express high levels of fetal γ-globin. However, the mechanisms underlying γ-globin production during cellular stress are still poorly defined. Here, we use CRISPR-Cas genome editing to model the stress caused by reduced levels of adult ß-globin. We find that decreased ß-globin is sufficient to induce robust re-expression of γ-globin, and RNA sequencing (RNA-seq) of differentiating isogenic erythroid precursors implicates ATF4 as a causal regulator of this response. ATF4 binds within the HBS1L-MYB intergenic enhancer and regulates expression of MYB, a known γ-globin regulator. Overall, the reduction of ATF4 upon ß-globin knockout decreases the levels of MYB and BCL11A. Identification of ATF4 as a key regulator of globin compensation adds mechanistic insight to the poorly understood phenomenon of stress-induced globin compensation and could inform strategies to treat hemoglobinopathies.


Subject(s)
Activating Transcription Factor 4/metabolism , Gene Expression Regulation , Proto-Oncogene Proteins c-myb/genetics , beta-Globins/metabolism , gamma-Globins/genetics , Activating Transcription Factor 4/genetics , Base Sequence , Cell Differentiation/genetics , Cell Line , DNA, Intergenic/genetics , Down-Regulation/genetics , Enhancer Elements, Genetic/genetics , Fetal Hemoglobin/genetics , Hematopoietic Stem Cells/metabolism , Humans , Mutation/genetics , Protein Binding , Proto-Oncogene Proteins c-myb/metabolism , Repressor Proteins/metabolism , Time Factors , Transcription, Genetic , Transcriptome/genetics , Up-Regulation/genetics , gamma-Globins/metabolism
5.
PLoS One ; 14(1): e0208237, 2019.
Article in English | MEDLINE | ID: mdl-30645582

ABSTRACT

Sickle Cell Disease and ß-thalassemia, which are caused by defective or deficient adult ß-globin (HBB) respectively, are the most common serious genetic blood diseases in the world. Persistent expression of the fetal ß-like globin, also known as 𝛾-globin, can ameliorate both disorders by serving in place of the adult ß-globin as a part of the fetal hemoglobin tetramer (HbF). Here we use CRISPR-Cas9 gene editing to explore a potential 𝛾-globin silencer region upstream of the δ-globin gene identified by comparison of naturally-occurring deletion mutations associated with up-regulated 𝛾-globin. We find that deletion of a 1.7 kb consensus element or select 350 bp sub-regions from bulk populations of cells increases levels of HbF. Screening of individual sgRNAs in one sub-region revealed three single guides that caused increases in 𝛾-globin expression. Deletion of the 1.7 kb region in HUDEP-2 clonal sublines, and in colonies derived from CD34+ hematopoietic stem/progenitor cells (HSPCs), does not cause significant up-regulation of 𝛾-globin. These data suggest that the 1.7 kb region is not an autonomous 𝛾-globin silencer, and thus by itself is not a suitable therapeutic target for gene editing treatment of ß-hemoglobinopathies.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Erythroid Cells/metabolism , Fetal Hemoglobin/metabolism , Repressor Proteins/metabolism , Cell Line , DNA, Intergenic/genetics , Gene Editing , Gene Silencing , Genotype , Hematopoietic Stem Cells/metabolism , Humans , Phenotype , Sequence Deletion/genetics , Up-Regulation/genetics , gamma-Globins/genetics
6.
Stem Cells ; 37(2): 284-294, 2019 02.
Article in English | MEDLINE | ID: mdl-30372555

ABSTRACT

Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated system (Cas9)-mediated gene editing of human hematopoietic stem cells (hHSCs) is a promising strategy for the treatment of genetic blood diseases through site-specific correction of identified causal mutations. However, clinical translation is hindered by low ratio of precise gene modification using the corrective donor template (homology-directed repair, HDR) to gene disruption (nonhomologous end joining, NHEJ) in hHSCs. By using a modified version of Cas9 with reduced nuclease activity in G1 phase of cell cycle when HDR cannot occur, and transiently increasing the proportion of cells in HDR-preferred phases (S/G2), we achieved a four-fold improvement in HDR/NHEJ ratio over the control condition in vitro, and a significant improvement after xenotransplantation of edited hHSCs into immunodeficient mice. This strategy for improving gene editing outcomes in hHSCs has important implications for the field of gene therapy, and can be applied to diseases where increased HDR/NHEJ ratio is critical for therapeutic success. Stem Cells 2019;37:284-294.


Subject(s)
DNA Repair/genetics , Gene Editing/methods , Hematopoietic Stem Cell Transplantation/methods , Stem Cells/metabolism , Transplantation Conditioning/methods , Animals , Humans , Mice
7.
ACS Nano ; 11(7): 6773-6781, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28618223

ABSTRACT

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.


Subject(s)
Cadmium Compounds/chemistry , Kinesins/analysis , Optical Imaging/methods , Quantum Dots/chemistry , Selenium Compounds/chemistry , Sulfides/chemistry , HeLa Cells , Humans , Ligands , Nanotechnology , Water/chemistry
8.
Elife ; 62017 05 02.
Article in English | MEDLINE | ID: mdl-28462777

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems , DNA/chemistry , DNA/genetics , Gene Editing/methods , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Protein 9 , Endonucleases/metabolism
9.
Methods ; 121-122: 9-15, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28410976

ABSTRACT

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.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Cas Systems , Endonucleases/genetics , Gene Editing/methods , Gene Transfer Techniques , RNA, Guide, Kinetoplastida/genetics , Ribonucleoproteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Protein 9 , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/genetics , DNA/metabolism , Endonucleases/metabolism , Gene Targeting/methods , Genome, Human , HEK293 Cells , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Jurkat Cells , K562 Cells , Primary Cell Culture , RNA, Guide, Kinetoplastida/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonucleoproteins/metabolism
10.
Nat Biomed Eng ; 1: 889-901, 2017.
Article in English | MEDLINE | ID: mdl-29805845

ABSTRACT

CRISPR/Cas9-based therapeutics, especially those that can correct gene mutations via homology directed repair (HDR), have the potential to revolutionize the treatment of genetic diseases. However, HDR-based therapeutics are challenging to develop because they require 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.

11.
Sci Transl Med ; 8(360): 360ra134, 2016 10 12.
Article in English | MEDLINE | ID: mdl-27733558

ABSTRACT

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 ß-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.


Subject(s)
Adult Stem Cells/metabolism , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Gene Editing/methods , Hematopoietic Stem Cells/metabolism , Hemoglobin, Sickle/genetics , Adult , Animals , CRISPR-Cas Systems , Cell Line , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mutation , Polymorphism, Single Nucleotide , Translational Research, Biomedical
12.
Nat Biotechnol ; 34(3): 339-44, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26789497

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems/genetics , DNA Breaks, Double-Stranded , RNA Editing/genetics , Recombinational DNA Repair/genetics , Cell Line , DNA End-Joining Repair/genetics , DNA, Single-Stranded/genetics , Genetic Engineering , Genome , Humans
13.
Nat Struct Mol Biol ; 22(1): 73-80, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25486306

ABSTRACT

Cytoplasmic dynein is an AAA+ motor responsible for intracellular cargo transport and force generation along microtubules (MTs). Unlike kinesin and myosin, dynein contains multiple ATPase subunits, with AAA1 serving as the primary catalytic site. ATPase activity at AAA3 is also essential for robust motility, but its role in dynein's mechanochemical cycle remains unclear. Here, we introduced transient pauses in Saccharomyces cerevisiae dynein motility by using a slowly hydrolyzing ATP analog. Analysis of pausing behavior revealed that AAA3 hydrolyzes nucleotide an order of magnitude more slowly than AAA1, and the two sites do not coordinate. ATPase mutations to AAA3 abolish the ability of dynein to modulate MT release. Nucleotide hydrolysis at AAA3 lifts this 'MT gate' to allow fast motility. These results suggest that AAA3 acts as a switch that repurposes cytoplasmic dynein for fast cargo transport and MT-anchoring tasks in cells.


Subject(s)
Cytoplasmic Dyneins/metabolism , Macromolecular Substances/metabolism , Microtubules/metabolism , Saccharomyces cerevisiae/enzymology , Adenosine Triphosphate/metabolism , Catalytic Domain , Hydrolysis , Saccharomyces cerevisiae/metabolism
14.
Nat Commun ; 5: 4587, 2014 Aug 11.
Article in English | MEDLINE | ID: mdl-25109325

ABSTRACT

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. In addition, 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. On the basis of these results, we propose a quantitative model for the stepping characteristics of dynein and its response to chemical and mechanical perturbation.


Subject(s)
Adenosine Triphosphate/chemistry , Dyneins/chemistry , Microtubules/chemistry , Adenosine Triphosphatases/chemistry , Animals , Cytoplasm/metabolism , Glutathione Transferase/metabolism , Green Fluorescent Proteins/chemistry , Monte Carlo Method , Motion , Mutation , Nucleotides/chemistry , Nucleotides/genetics , Optics and Photonics , Protein Conformation , Protein Engineering/methods , Protein Multimerization , Protein Structure, Tertiary , Saccharomyces cerevisiae/metabolism , Sea Urchins , Stress, Mechanical , Thermus/metabolism
15.
Elife ; 3: e03205, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-25069614

ABSTRACT

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.


Subject(s)
Dyneins/chemistry , Microtubules/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Biological Transport , Cytoplasm/chemistry , Cytoplasm/metabolism , Dyneins/genetics , Dyneins/metabolism , Fluorescent Dyes , Gene Expression , Kinesins/chemistry , Kinesins/genetics , Kinesins/metabolism , Microtubules/genetics , Microtubules/metabolism , Models, Biological , Motion , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Tubulin/chemistry , Tubulin/genetics , Tubulin/metabolism
16.
Science ; 335(6065): 221-5, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22157083

ABSTRACT

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.


Subject(s)
Cytoplasm/metabolism , Dyneins/chemistry , Dyneins/metabolism , Microtubules/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphate/metabolism , Models, Biological , Models, Molecular , Protein Multimerization , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
17.
Biochemistry ; 49(45): 9722-31, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-20886836

ABSTRACT

Studying the interplay between protein structure and function remains a daunting task. Especially lacking are methods for measuring structural changes in real time. Here we report our most recent improvements to a method that can be used to address such challenges. This method, which we now call tryptophan-induced quenching (TrIQ), provides a straightforward, sensitive, and inexpensive way to address questions of conformational dynamics and short-range protein interactions. Importantly, TrIQ only occurs over relatively short distances (∼5-15 Å), making it complementary to traditional fluorescence resonance energy transfer (FRET) methods that occur over distances too large for precise studies of protein structure. As implied in the name, TrIQ measures the efficient quenching induced in some fluorophores by tryptophan (Trp). We present here our analysis of the TrIQ effect for five different fluorophores that span a range of sizes and spectral properties. Each probe was attached to four different cysteine residues on T4 lysozyme, and the extent of TrIQ caused by a nearby Trp was measured. Our results show that, at least for smaller probes, the extent of TrIQ is distance dependent. Moreover, we also demonstrate how TrIQ data can be analyzed to determine the fraction of fluorophores involved in a static, nonfluorescent complex with Trp. Based on this analysis, our study shows that each fluorophore has a different TrIQ profile, or "sphere of quenching", which correlates with its size, rotational flexibility, and the length of attachment linker. This TrIQ-based "sphere of quenching" is unique to every Trp-probe pair and reflects the distance within which one can expect to see the TrIQ effect. Thus,TrIQ provides a straightforward, readily accessible approach for mapping distances within proteins and monitoring conformational changes using fluorescence spectroscopy.


Subject(s)
Proteins/chemistry , Tryptophan/chemistry , Amino Acids/analysis , Anisotropy , Bacteriophage T4/enzymology , Bacteriophage T4/genetics , Fluorescence Resonance Energy Transfer , Models, Molecular , Muramidase/chemistry , Muramidase/genetics , Muramidase/isolation & purification , Protein Structure, Secondary , Spectrometry, Fluorescence/methods , Structure-Activity Relationship
18.
J Mol Biol ; 365(5): 1257-65, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17118401

ABSTRACT

The manganese transport regulator (MntR) from Bacillus subtilis binds cognate DNA sequences in response to elevated manganese concentrations. MntR functions as a homodimer that binds two manganese ions per subunit. Metal binding takes place at the interface of the two domains that comprise each MntR subunit: an N-terminal DNA-binding domain and a C-terminal dimerization domain. In order to elucidate the link between metal binding and activation, a crystallographic study of MntR in its metal-free state has been undertaken. Here we describe the structures of the native protein and a selenomethionine-containing variant, solved to 2.8 A. The two structures contain five crystallographically unique subunits of MntR, providing diverse views of the metal-free protein. In apo-MntR, as in the manganese complex, the dimer is formed by dyad-related C-terminal domains that provide a conserved structural core. Similarly, each DNA-binding domain largely retains the folded conformation found in metal bound forms of MntR. However, compared to metal-activated MntR, the DNA-binding domains move substantially with respect to the dimer interface in apo-MntR. Overlays of multiple apo-MntR structures indicate that there is a greater range of positioning allowed between N and C-terminal domains in the metal-free state and that the DNA-binding domains of the dimer are farther apart than in the activated complex. To further investigate the conformation of the DNA-binding domain of apo-MntR, a site-directed spin labeling experiment was performed on a mutant of MntR containing cysteine at residue 6. Consistent with the crystallographic results, EPR spectra of the spin-labeled mutant indicate that tertiary structure is conserved in the presence or absence of bound metals, though slightly greater flexibility is present in inactive forms of MntR.


Subject(s)
Bacillus subtilis/chemistry , Bacterial Proteins/chemistry , Manganese/metabolism , Apoproteins/chemistry , Apoproteins/metabolism , Bacterial Proteins/metabolism , Biological Transport , Crystallography, X-Ray , DNA, Bacterial/metabolism , Molecular Probes , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/metabolism , Spin Labels
SELECTION OF CITATIONS
SEARCH DETAIL
...