Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Biotechnol ; 42(3): 458-469, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37127662

ABSTRACT

Inefficient knock-in of transgene cargos limits the potential of cell-based medicines. In this study, we used a CRISPR nuclease that targets a site within an exon of an essential gene and designed a cargo template so that correct knock-in would retain essential gene function while also integrating the transgene(s) of interest. Cells with non-productive insertions and deletions would undergo negative selection. This technology, called SLEEK (SeLection by Essential-gene Exon Knock-in), achieved knock-in efficiencies of more than 90% in clinically relevant cell types without impacting long-term viability or expansion. SLEEK knock-in rates in T cells are more efficient than state-of-the-art TRAC knock-in with AAV6 and surpass more than 90% efficiency even with non-viral DNA cargos. As a clinical application, natural killer cells generated from induced pluripotent stem cells containing SLEEK knock-in of CD16 and mbIL-15 show substantially improved tumor killing and persistence in vivo.


Subject(s)
CRISPR-Cas Systems , Gene Editing , CRISPR-Cas Systems/genetics , Gene Knock-In Techniques , Transgenes/genetics
2.
Nat Commun ; 14(1): 4928, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37582836

ABSTRACT

Mutations in Atp2b2, an outer hair cell gene, cause dominant hearing loss in humans. Using a mouse model Atp2b2Obl/+, with a dominant hearing loss mutation (Oblivion), we show that liposome-mediated in vivo delivery of CRISPR-Cas9 ribonucleoprotein complexes leads to specific editing of the Obl allele. Large deletions encompassing the Obl locus and indels were identified as the result of editing. In vivo genome editing promotes outer hair cell survival and restores their function, leading to hearing recovery. We further show that in a double-dominant mutant mouse model, in which the Tmc1 Beethoven mutation and the Atp2b2 Oblivion mutation cause digenic genetic hearing loss, Cas9/sgRNA delivery targeting both mutations leads to partial hearing recovery. These findings suggest that liposome-RNP delivery can be used as a strategy to recover hearing with dominant mutations in OHC genes and with digenic mutations in the auditory hair cells, potentially expanding therapeutics of gene editing to treat hearing loss.


Subject(s)
Deafness , Hearing Loss , Humans , CRISPR-Cas Systems/genetics , Ribonucleoproteins/genetics , Liposomes , RNA, Guide, CRISPR-Cas Systems , Hearing Loss/genetics , Hearing Loss/therapy , Deafness/genetics
4.
Nat Commun ; 12(1): 3908, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162850

ABSTRACT

Though AsCas12a fills a crucial gap in the current genome editing toolbox, it exhibits relatively poor editing efficiency, restricting its overall utility. Here we isolate an engineered variant, "AsCas12a Ultra", that increased editing efficiency to nearly 100% at all sites examined in HSPCs, iPSCs, T cells, and NK cells. We show that AsCas12a Ultra maintains high on-target specificity thereby mitigating the risk for off-target editing and making it ideal for complex therapeutic genome editing applications. We achieved simultaneous targeting of three clinically relevant genes in T cells at >90% efficiency and demonstrated transgene knock-in efficiencies of up to 60%. We demonstrate site-specific knock-in of a CAR in NK cells, which afforded enhanced anti-tumor NK cell recognition, potentially enabling the next generation of allogeneic cell-based therapies in oncology. AsCas12a Ultra is an advanced CRISPR nuclease with significant advantages in basic research and in the production of gene edited cell medicines.


Subject(s)
Acidaminococcus/enzymology , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Endonucleases/metabolism , Gene Editing/methods , Acidaminococcus/genetics , Bacterial Proteins/genetics , CRISPR-Associated Proteins/genetics , Cells, Cultured , Endonucleases/genetics , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Jurkat Cells , Killer Cells, Natural/metabolism , Reproducibility of Results , T-Lymphocytes/metabolism
5.
CRISPR J ; 3(3): 177-187, 2020 06.
Article in English | MEDLINE | ID: mdl-32584143

ABSTRACT

Multiplexed genome editing with DNA endonucleases has broad application, including for cellular therapies, but chromosomal translocations, natural byproducts of inducing simultaneous genomic breaks, have not been explored in detail. Here we apply various CRISPR-Cas nucleases to edit the T cell receptor alpha and beta 2 microglobulin genes in human primary T cells and comprehensively evaluate the frequency and stability of the resulting translocations. A thorough translocation frequency analysis using three orthogonal methods (droplet digital PCR, unidirectional sequencing, and metaphase fluorescence in situ hybridization) yielded comparable results and an overall translocation rate of ∼7% between two simultaneous CRISPR-Cas9 induced edits. In addition, we show that chromosomal translocations can be reduced when using different nuclease combinations, or by the presence of a homologous single stranded oligo donor for multiplexed genome editing. Importantly, the two different approaches for translocation reduction are compatible with cell therapy applications.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , T-Lymphocytes , Translocation, Genetic , CD4-Positive T-Lymphocytes , CRISPR-Associated Protein 9/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/genetics , Endonucleases/genetics , Genome, Human , Humans , In Situ Hybridization, Fluorescence , Multifactorial Inheritance , RNA, Guide, Kinetoplastida , Streptococcus pyogenes
6.
Nature ; 533(7603): 420-4, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27096365

ABSTRACT

Current genome-editing technologies introduce double-stranded (ds) DNA breaks at a target locus as the first step to gene correction. Although most genetic diseases arise from point mutations, current approaches to point mutation correction are inefficient and typically induce an abundance of random insertions and deletions (indels) at the target locus resulting from the cellular response to dsDNA breaks. Here we report the development of 'base editing', a new approach to genome editing that enables the direct, irreversible conversion of one target DNA base into another in a programmable manner, without requiring dsDNA backbone cleavage or a donor template. We engineered fusions of CRISPR/Cas9 and a cytidine deaminase enzyme that retain the ability to be programmed with a guide RNA, do not induce dsDNA breaks, and mediate the direct conversion of cytidine to uridine, thereby effecting a C→T (or G→A) substitution. The resulting 'base editors' convert cytidines within a window of approximately five nucleotides, and can efficiently correct a variety of point mutations relevant to human disease. In four transformed human and murine cell lines, second- and third-generation base editors that fuse uracil glycosylase inhibitor, and that use a Cas9 nickase targeting the non-edited strand, manipulate the cellular DNA repair response to favour desired base-editing outcomes, resulting in permanent correction of ~15-75% of total cellular DNA with minimal (typically ≤1%) indel formation. Base editing expands the scope and efficiency of genome editing of point mutations.


Subject(s)
CRISPR-Cas Systems , Cytidine Deaminase/metabolism , Cytidine/genetics , Genetic Engineering/methods , Genome/genetics , Point Mutation/genetics , Uridine/genetics , APOBEC-1 Deaminase , Animals , Apolipoprotein E4/genetics , Base Sequence , CRISPR-Associated Proteins/metabolism , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA/genetics , DNA/metabolism , DNA Cleavage , DNA Repair , Deoxyribonuclease I/metabolism , Genes, p53/genetics , Humans , INDEL Mutation/genetics , Mice , RNA, Guide, Kinetoplastida/genetics , Templates, Genetic , Uracil-DNA Glycosidase/antagonists & inhibitors
7.
Proc Natl Acad Sci U S A ; 113(11): 2868-73, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929348

ABSTRACT

A central challenge to the development of protein-based therapeutics is the inefficiency of delivery of protein cargo across the mammalian cell membrane, including escape from endosomes. Here we report that combining bioreducible lipid nanoparticles with negatively supercharged Cre recombinase or anionic Cas9:single-guide (sg)RNA complexes drives the electrostatic assembly of nanoparticles that mediate potent protein delivery and genome editing. These bioreducible lipids efficiently deliver protein cargo into cells, facilitate the escape of protein from endosomes in response to the reductive intracellular environment, and direct protein to its intracellular target sites. The delivery of supercharged Cre protein and Cas9:sgRNA complexed with bioreducible lipids into cultured human cells enables gene recombination and genome editing with efficiencies greater than 70%. In addition, we demonstrate that these lipids are effective for functional protein delivery into mouse brain for gene recombination in vivo. Therefore, the integration of this bioreducible lipid platform with protein engineering has the potential to advance the therapeutic relevance of protein-based genome editing.


Subject(s)
Gene Knockout Techniques , Genes, Synthetic , Genetic Engineering/methods , Lipids/chemistry , Nanoparticles , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Ceramides/chemistry , Cholesterol/chemistry , Drug Carriers , Endocytosis , Endonucleases/administration & dosage , Endonucleases/genetics , Endosomes/metabolism , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Hypothalamus/metabolism , Integrases/administration & dosage , Integrases/genetics , Lipids/administration & dosage , Lipids/chemical synthesis , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Molecular Structure , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/metabolism , Nanoparticles/toxicity , Phosphatidylethanolamines/chemistry , RNA/genetics , Recombinant Proteins/biosynthesis , Recombination, Genetic , Static Electricity , Structure-Activity Relationship , Thalamus/metabolism
8.
Nat Methods ; 12(10): 939-42, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26258293

ABSTRACT

Nucleases containing programmable DNA-binding domains can alter the genomes of model organisms and have the potential to become human therapeutics. Here we present DNA-binding phage-assisted continuous evolution (DB-PACE) as a general approach for the laboratory evolution of DNA-binding activity and specificity. We used this system to generate transcription activator-like effectors nucleases (TALENs) with broadly improved DNA cleavage specificity, establishing DB-PACE as a versatile approach for improving the accuracy of genome-editing agents.


Subject(s)
DNA-Binding Proteins/metabolism , Deoxyribonucleases/metabolism , Directed Molecular Evolution/methods , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Targeting/methods , High-Throughput Screening Assays/methods , Humans , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Protein Engineering/methods
9.
Nat Chem Biol ; 11(5): 316-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25848930

ABSTRACT

Directly modulating the activity of genome-editing proteins has the potential to increase their specificity by reducing activity following target locus modification. We developed Cas9 nucleases that are activated by the presence of a cell-permeable small molecule by inserting an evolved 4-hydroxytamoxifen-responsive intein at specific positions in Cas9. In human cells, conditionally active Cas9s modify target genomic sites with up to 25-fold higher specificity than wild-type Cas9.


Subject(s)
Endonucleases/genetics , Genome/drug effects , Bacterial Proteins/drug effects , Bacterial Proteins/genetics , Cells, Cultured , Endonucleases/drug effects , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Ligands , Models, Molecular , Protein Conformation , Small Molecule Libraries , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
10.
Nat Biotechnol ; 33(1): 73-80, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25357182

ABSTRACT

Efficient intracellular delivery of proteins is needed to fully realize the potential of protein therapeutics. Current methods of protein delivery commonly suffer from low tolerance for serum, poor endosomal escape and limited in vivo efficacy. Here we report that common cationic lipid nucleic acid transfection reagents can potently deliver proteins that are fused to negatively supercharged proteins, that contain natural anionic domains or that natively bind to anionic nucleic acids. This approach mediates the potent delivery of nM concentrations of Cre recombinase, TALE- and Cas9-based transcription activators, and Cas9:sgRNA nuclease complexes into cultured human cells in media containing 10% serum. Delivery of unmodified Cas9:sgRNA complexes resulted in up to 80% genome modification with substantially higher specificity compared to DNA transfection. This approach also mediated efficient delivery of Cre recombinase and Cas9:sgRNA complexes into the mouse inner ear in vivo, achieving 90% Cre-mediated recombination and 20% Cas9-mediated genome modification in hair cells.


Subject(s)
Lipids/administration & dosage , Proteins/administration & dosage , Cations , In Vitro Techniques , Trans-Activators/administration & dosage , Transfection
11.
Proc Natl Acad Sci U S A ; 111(14): 5177-82, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706857

ABSTRACT

Life requires orchestrated control of cell proliferation, cell maintenance, and cell death. Involved in these decisions are protein complexes that assimilate a variety of inputs that report on the status of the cell and lead to an output response. Among the proteins involved in this response are nutrient-deprivation autophagy factor-1 (NAF-1)- and Bcl-2. NAF-1 is a homodimeric member of the novel Fe-S protein NEET family, which binds two 2Fe-2S clusters. NAF-1 is an important partner for Bcl-2 at the endoplasmic reticulum to functionally antagonize Beclin 1-dependent autophagy [Chang NC, Nguyen M, Germain M, Shore GC (2010) EMBO J 29(3):606-618]. We used an integrated approach involving peptide array, deuterium exchange mass spectrometry (DXMS), and functional studies aided by the power of sufficient constraints from direct coupling analysis (DCA) to determine the dominant docked conformation of the NAF-1-Bcl-2 complex. NAF-1 binds to both the pro- and antiapoptotic regions (BH3 and BH4) of Bcl-2, as demonstrated by a nested protein fragment analysis in a peptide array and DXMS analysis. A combination of the solution studies together with a new application of DCA to the eukaryotic proteins NAF-1 and Bcl-2 provided sufficient constraints at amino acid resolution to predict the interaction surfaces and orientation of the protein-protein interactions involved in the docked structure. The specific integrated approach described in this paper provides the first structural information, to our knowledge, for future targeting of the NAF-1-Bcl-2 complex in the regulation of apoptosis/autophagy in cancer biology.


Subject(s)
Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Ribonucleoproteins/metabolism , Amino Acid Sequence , Humans , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Oligopeptides/chemistry , Protein Binding
12.
PLoS One ; 8(5): e61202, 2013.
Article in English | MEDLINE | ID: mdl-23717386

ABSTRACT

Nutrient-deprivation autophagy factor-1 (NAF-1) (synonyms: Cisd2, Eris, Miner1, and Noxp70) is a [2Fe-2S] cluster protein immune-detected both in endoplasmic reticulum (ER) and mitochondrial outer membrane. It was implicated in human pathology (Wolfram Syndrome 2) and in BCL-2 mediated antagonization of Beclin 1-dependent autophagy and depression of ER calcium stores. To gain insights about NAF-1 functions, we investigated the biochemical properties of its 2Fe-2S cluster and sensitivity of those properties to small molecules. The structure of the soluble domain of NAF-1 shows that it forms a homodimer with each protomer containing a [2Fe-2S] cluster bound by 3 Cys and one His. NAF-1 has shown the unusual abilities to transfer its 2Fe-2S cluster to an apo-acceptor protein (followed in vitro by spectrophotometry and by native PAGE electrophoresis) and to transfer iron to intact mitochondria in cell models (monitored by fluorescence imaging with iron fluorescent sensors targeted to mitochondria). Importantly, the drug pioglitazone abrogates NAF-1's ability to transfer the cluster to acceptor proteins and iron to mitochondria. Similar effects were found for the anti-diabetes and longevity-promoting antioxidant resveratrol. These results reveal NAF-1 as a previously unidentified cell target of anti-diabetes thiazolidinedione drugs like pioglitazone and of the natural product resveratrol, both of which interact with the protein and stabilize its labile [2Fe-2S] cluster.


Subject(s)
Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Ribonucleoproteins/metabolism , Cells, Cultured , Drug Delivery Systems/methods , Humans , Iron/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Protein Subunits/metabolism , Small Molecule Libraries/metabolism , Small Molecule Libraries/therapeutic use , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacology
13.
Proc Natl Acad Sci U S A ; 110(3): 948-53, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23271805

ABSTRACT

Metalloproteins (MPs) comprise one-third of all known protein structures. This diverse set of proteins contain a plethora of unique inorganic moieties capable of performing chemistry that would otherwise be impossible using only the amino acids found in nature. Most of the well-studied MPs are generally viewed as being very rigid in structure, and it is widely thought that the properties of the metal centers are primarily determined by the small fraction of amino acids that make up the local environment. Here we examine both theoretically and experimentally whether distal regions can influence the metal center in the diabetes drug target mitoNEET. We demonstrate that a loop (L2) 20 Å away from the metal center exerts allosteric control over the cluster binding domain and regulates multiple properties of the metal center. Mutagenesis of L2 results in significant shifts in the redox potential of the [2Fe-2S] cluster and orders of magnitude effects on the rate of [2Fe-2S] cluster transfer to an apo-acceptor protein. These surprising effects occur in the absence of any structural changes. An examination of the native basin dynamics of the protein using all-atom simulations shows that twisting in L2 controls scissoring in the cluster binding domain and results in perturbations to one of the cluster-coordinating histidines. These allosteric effects are in agreement with previous folding simulations that predicted L2 could communicate with residues surrounding the metal center. Our findings suggest that long-range dynamical changes in the protein backbone can have a significant effect on the functional properties of MPs.


Subject(s)
Metalloproteins/chemistry , Metalloproteins/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Allosteric Regulation , Binding Sites , Biophysical Phenomena , Crystallography, X-Ray , Histidine/chemistry , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Metalloproteins/genetics , Mitochondrial Proteins/genetics , Models, Molecular , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Conformation , Protein Stability
14.
Plant Cell ; 24(5): 2139-54, 2012 May.
Article in English | MEDLINE | ID: mdl-22562611

ABSTRACT

The NEET family is a newly discovered group of proteins involved in a diverse array of biological processes, including autophagy, apoptosis, aging, diabetes, and reactive oxygen homeostasis. They form a novel structure, the NEET fold, in which two protomers intertwine to form a two-domain motif, a cap, and a unique redox-active labile 2Fe-2S cluster binding domain. To accelerate the functional study of NEET proteins, as well as to examine whether they have an evolutionarily conserved role, we identified and characterized a plant NEET protein. Here, we show that the Arabidopsis thaliana At5g51720 protein (At-NEET) displays biochemical, structural, and biophysical characteristics of a NEET protein. Phenotypic characterization of At-NEET revealed a key role for this protein in plant development, senescence, reactive oxygen homeostasis, and Fe metabolism. A role in Fe metabolism was further supported by biochemical and cell biology studies of At-NEET in plant and mammalian cells, as well as mutational analysis of its cluster binding domain. Our findings support the hypothesis that NEET proteins have an ancient role in cells associated with Fe metabolism.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Iron/metabolism , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Molecular Sequence Data , Protein Structure, Secondary , Sequence Homology, Amino Acid
15.
J Biol Chem ; 287(15): 11649-55, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22351774

ABSTRACT

MitoNEET (mNT) is the founding member of the recently discovered CDGSH family of [2Fe-2S] proteins capable of [2Fe-2S] cluster transfer to apo-acceptor proteins. It is a target of the thiazolidinedione (TZD) class of anti-diabetes drugs whose binding modulate both electron transfer and cluster transfer properties. The [2Fe-2S] cluster in mNT is destabilized upon binding of NADPH, which leads to loss of the [2Fe-2S] cluster to the solution environment. Because mNT is capable of transferring [2Fe-2S] clusters to apo-acceptor proteins, we sought to determine whether NADPH binding also affects cluster transfer. We show that NADPH inhibits transfer of the [2Fe-2S] cluster to an apo-acceptor protein with an inhibition constant (K(i)) of 200 µm, which reflects that of NADPH concentrations expected under physiological conditions. In addition, we determined that the strictly conserved cluster interacting residue Asp-84 in the CDGSH domain is necessary for the NADPH-dependent inhibition of [2Fe-2S] cluster transfer. The most critical cellular function of NADPH is in the maintenance of a pool of reducing equivalents, which is essential to counteract oxidative damage. Taken together, our findings suggest that NADPH can regulate both mNT [2Fe-2S] cluster levels in the cell as well as the ability of the protein to transfer [2Fe-2S] clusters to cytosolic or mitochondrial acceptors.


Subject(s)
Apoproteins/chemistry , Ferredoxins/chemistry , Iron-Sulfur Proteins/chemistry , Mitochondrial Proteins/chemistry , NADP/chemistry , Amino Acid Motifs , Binding Sites , Binding, Competitive , Hydrogen-Ion Concentration , Hypoglycemic Agents/chemistry , Kinetics , Mitochondrial Proteins/genetics , Models, Molecular , Mutation, Missense , Oxidation-Reduction , Protein Binding , Protein Structure, Tertiary , Thiazolidinediones/chemistry
16.
Proc Natl Acad Sci U S A ; 108(32): 13047-52, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21788481

ABSTRACT

MitoNEET (mNT) is an outer mitochondrial membrane target of the thiazolidinedione diabetes drugs with a unique fold and a labile [2Fe-2S] cluster. The rare 1-His and 3-Cys coordination of mNT's [2Fe-2S] leads to cluster lability that is strongly dependent on the presence of the single histidine ligand (His87). These properties of mNT are similar to known [2Fe-2S] shuttle proteins. Here we investigated whether mNT is capable of cluster transfer to acceptor protein(s). Facile [2Fe-2S] cluster transfer is observed between oxidized mNT and apo-ferredoxin (a-Fd) using UV-VIS spectroscopy and native-PAGE, as well as with a mitochondrial iron detection assay in cells. The transfer is unidirectional, proceeds to completion, and occurs with a second-order-reaction rate that is comparable to known iron-sulfur transfer proteins. Mutagenesis of His87 with Cys (H87C) inhibits transfer of the [2Fe-2S] clusters to a-Fd. This inhibition is beyond that expected from increased cluster kinetic stability, as the equivalently stable Lys55 to Glu (K55E) mutation did not inhibit transfer. The H87C mutant also failed to transfer its iron to mitochondria in HEK293 cells. The diabetes drug pioglitazone inhibits iron transfer from WT mNT to mitochondria, indicating that pioglitazone affects a specific property, [2Fe-2S] cluster transfer, in the cellular environment. This finding is interesting in light of the role of iron overload in diabetes. Our findings suggest a likely role for mNT in [2Fe-2S] and/or iron transfer to acceptor proteins and support the idea that pioglitazone's antidiabetic mode of action may, in part, be to inhibit transfer of mNT's [2Fe-2S] cluster.


Subject(s)
Ferredoxins/metabolism , Hypoglycemic Agents/pharmacology , Iron-Sulfur Proteins/metabolism , Mitochondrial Proteins/metabolism , Ferredoxins/chemistry , HEK293 Cells , Histidine/metabolism , Humans , Iron/metabolism , Iron-Sulfur Proteins/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Oxidation-Reduction/drug effects , Permeability/drug effects , Pioglitazone , Structure-Activity Relationship , Thiazolidinediones/pharmacology
17.
Acta Crystallogr D Biol Crystallogr ; 67(Pt 6): 516-23, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21636891

ABSTRACT

MitoNEET is the only identified Fe-S protein localized to the outer mitochondrial membrane and a 1.5 Šresolution X-ray analysis has revealed a unique structure [Paddock et al. (2007), Proc. Natl Acad. Sci. USA, 104, 14342-14347]. The 2Fe-2S cluster is bound with a 3Cys-1His coordination which defines a new class of 2Fe-2S proteins. The hallmark feature of this class is the single noncysteine ligand His87, which when replaced by Cys decreases the redox potential (E(m)) by ∼300 mV and increases the stability of the cluster by around sixfold. Unexpectedly, the pH dependence of the lifetime of the 2Fe-2S cluster remains the same as in the wild-type protein. Here, the crystal structure of H87C mitoNEET was determined to 1.7 Šresolution (R factor = 18%) to investigate the structural basis of the changes in the properties of the 2Fe-2S cluster. In comparison to the wild type, structural changes are localized to the immediate vicinity of the cluster-binding region. Despite the increased stability, Cys87 displays two distinct conformations, with distances of 2.3 and 3.2 Šbetween the S(γ) and the outer Fe of the 2Fe-2S cluster. In addition, Lys55 exhibits multiple conformations in the H87C mutant protein. The structure and distinct characteristics of the H87C mutant provide a framework for further studies investigating the effects of mutation on the properties of the 2Fe-2S cluster in this new class of proteins.


Subject(s)
Iron-Sulfur Proteins/chemistry , Mitochondrial Proteins/chemistry , Mutation , Histidine/genetics , Histidine/metabolism , Humans , Ligands , Models, Molecular , Protein Folding , Protein Structure, Quaternary , Protein Structure, Tertiary
18.
Proc Natl Acad Sci U S A ; 108(6): 2240-5, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21266547

ABSTRACT

Regulation of protein function via cracking, or local unfolding and refolding of substructures, is becoming a widely recognized mechanism of functional control. Oftentimes, cracking events are localized to secondary and tertiary structure interactions between domains that control the optimal position for catalysis and/or the formation of protein complexes. Small changes in free energy associated with ligand binding, phosphorylation, etc., can tip the balance and provide a regulatory functional switch. However, understanding the factors controlling function in single-domain proteins is still a significant challenge to structural biologists. We investigated the functional landscape of a single-domain plant-type ferredoxin protein and the effect of a distal loop on the electron-transfer center. We find the global stability and structure are minimally perturbed with mutation, whereas the functional properties are altered. Specifically, truncating the L1,2 loop does not lead to large-scale changes in the structure, determined via X-ray crystallography. Further, the overall thermal stability of the protein is only marginally perturbed by the mutation. However, even though the mutation is distal to the iron-sulfur cluster (∼20 Å), it leads to a significant change in the redox potential of the iron-sulfur cluster (57 mV). Structure-based all-atom simulations indicate correlated dynamical changes between the surface-exposed loop and the iron-sulfur cluster-binding region. Our results suggest intrinsic communication channels within the ferredoxin fold, composed of many short-range interactions, lead to the propagation of long-range signals. Accordingly, protein interface interactions that involve L1,2 could potentially signal functional changes in distal regions, similar to what is observed in other allosteric systems.


Subject(s)
Ferredoxins/chemistry , Models, Molecular , Protein Folding , Allosteric Regulation/physiology , Amino Acid Motifs , Ferredoxins/genetics , Ferredoxins/metabolism , Humans , Iron/chemistry , Iron/metabolism , Mutation , Protein Stability , Protein Structure, Tertiary , Sulfur/chemistry , Sulfur/metabolism
19.
J Am Chem Soc ; 132(38): 13120-2, 2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20812736

ABSTRACT

MitoNEET is a newly discovered mitochondrial protein and a target of the TZD class of antidiabetes drugs. MitoNEET is homodimeric with each protomer binding a [2Fe-2S] center through a rare 3-Cys and 1-His coordination geometry. Both the fold and the coordination of the [2Fe-2S] centers suggest that it could have novel properties compared to other known [2Fe-2S] proteins. We tested the robustness of mitoNEET to mutation and the range over which the redox potential (E(M)) could be tuned. We found that the protein could tolerate an array of mutations that modified the E(M) of the [2Fe-2S] center over a range of ∼700 mV, which is the largest E(M) range engineered in an FeS protein and, importantly, spans the cellular redox range (+200 to -300 mV). These properties make mitoNEET potentially useful for both physiological studies and industrial applications as a stable, water-soluble, redox agent.


Subject(s)
Iron-Sulfur Proteins/chemistry , Models, Molecular , Oxidation-Reduction
20.
Biochemistry ; 48(43): 10193-5, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19791753

ABSTRACT

MitoNEET is a small mitochondrial protein that has been identified recently as a target for the thiazolidinedione (TZD) class of diabetes drugs. MitoNEET also binds a unique three-Cys- and one-His-ligated [corrected] [2Fe-2S] cluster. Here we use protein film voltammetry (PFV) as a means to probe the redox properties of mitoNEET and demonstrate the direct impact of TZD drug binding upon the redox chemistry of the FeS cluster. When TZDs bind, the midpoint potential at pH 7 is lowered by more than 100 mV, shifting from approximately 0 to -100 mV. In contrast, a His87Cys mutant negates the ability of TZDs to affect the midpoint potential, suggesting a model of drug binding in which His87 is critical to communication with the FeS center of mitoNEET.


Subject(s)
Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Thiazolidinediones/metabolism , Animals , Humans , Hydrogen-Ion Concentration , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Oxidation-Reduction , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...