Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(4): e0231716, 2020.
Article in English | MEDLINE | ID: mdl-32298334

ABSTRACT

RNA-guided endonucleases such as Cas9 provide efficient on-target genome editing in cells but may also cleave at off-target loci throughout the genome. Engineered variants of Streptococcus pyogenes Cas9 (SpCas9) have been developed to globally reduce off-target activity, but individual off-targets may remain, or on-target activity may be compromised. In order to evolve against activity at specific off-targets while maintaining strong on-target editing, we developed a novel M13 bacteriophage-mediated selection method. Using this method, sequential rounds of positive and negative selection are used to identify mutations to Cas9 that enhance or diminish editing activity at particular genomic sequences. We also introduce scanning mutagenesis of oligo-directed targets (SMOOT), a comprehensive mutagenesis method to create highly diverse libraries of Cas9 variants that can be challenged with phage-based selection. Our platform identifies novel SpCas9 mutants which mitigate cleavage against off-targets both in biochemical assays and in T-cells while maintaining higher on-target activity than previously described variants. We describe an evolved variant, S. pyogenes Adapted to Reduce Target Ambiguity Cas9 (SpartaCas), composed of the most enriched mutations, each of unknown function. This evolved Cas9 mutant reduces off-target cleavage while preserving efficient editing at multiple therapeutically relevant targets. Directed evolution of Cas9 using our system demonstrates an improved structure-independent methodology to effectively engineer nuclease activity.


Subject(s)
Bacteriophage M13/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Directed Molecular Evolution/methods , Gene Editing/methods , Streptococcus pyogenes/genetics , CRISPR-Associated Protein 9/chemistry , High-Throughput Screening Assays , Mutagenesis , Mutation , Streptococcus pyogenes/enzymology , Substrate Specificity , T-Lymphocytes/metabolism
2.
CRISPR J ; 2: 172-185, 2019 06.
Article in English | MEDLINE | ID: mdl-31225747

ABSTRACT

Considerable effort has been devoted to developing a comprehensive understanding of CRISPR nuclease specificity. In silico predictions and multiple genome-wide cellular and biochemical approaches have revealed a basic understanding of the Cas9 specificity profile. However, none of these approaches has delivered a model that allows accurate prediction of a CRISPR nuclease's ability to cleave a site based entirely on the sequence of the guide RNA (gRNA) and the target. We describe a library-based biochemical assay that directly reports the cleavage efficiency of a particular Cas9-guide complex by measuring both uncleaved and cleaved target molecules over a wide range of mismatched library members. We applied our assay using libraries of targets to evaluate the specificity of Staphylococcus aureus Cas9 under a variety of experimental conditions. Surprisingly, our data show an unexpectedly high variation in the random gRNA:target DNA mismatch tolerance when cleaving with different gRNAs, indicating guide-intrinsic mismatch permissiveness and challenging the assumption of universal specificity models. We use data generated by our assay to create the first off-target, guide-specific cleavage models. The barcoded libraries of targets approach is rapid, highly modular, and capable of generating protein- and guide-specific models, as well as illuminating the biophysics of Cas9 binding versus cutting. These models may be useful in identifying potential off-targets, and the gRNA-intrinsic nature of mismatch tolerance argues for coupling these specificity models with orthogonal methods for a more complete assessment of gRNA specificity.


Subject(s)
CRISPR-Associated Protein 9/metabolism , Staphylococcus aureus/enzymology , CRISPR-Cas Systems , DNA/metabolism , Substrate Specificity
3.
Nat Med ; 25(2): 229-233, 2019 02.
Article in English | MEDLINE | ID: mdl-30664785

ABSTRACT

Leber congenital amaurosis type 10 is a severe retinal dystrophy caused by mutations in the CEP290 gene1,2. We developed EDIT-101, a candidate genome-editing therapeutic, to remove the aberrant splice donor created by the IVS26 mutation in the CEP290 gene and restore normal CEP290 expression. Key to this therapeutic, we identified a pair of Staphylococcus aureus Cas9 guide RNAs that were highly active and specific to the human CEP290 target sequence. In vitro experiments in human cells and retinal explants demonstrated the molecular mechanism of action and nuclease specificity. Subretinal delivery of EDIT-101 in humanized CEP290 mice showed rapid and sustained CEP290 gene editing. A comparable surrogate non-human primate (NHP) vector also achieved productive editing of the NHP CEP290 gene at levels that met the target therapeutic threshold, and demonstrated the ability of CRISPR/Cas9 to edit somatic primate cells in vivo. These results support further development of EDIT-101 for LCA10 and additional CRISPR-based medicines for other inherited retinal disorders.


Subject(s)
Gene Editing , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/physiopathology , Animals , Cell Line , Gene Knock-In Techniques , Humans , Mice , Primates , Reproducibility of Results , Vision, Ocular
4.
J Med Chem ; 61(20): 9301-9315, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30289257

ABSTRACT

The biological functions of the dual bromodomains of human transcription-initiation-factor TFIID subunit 1 (TAF1(1,2)) remain unknown, although TAF1 has been identified as a potential target for oncology research. Here, we describe the discovery of a potent and selective in vitro tool compound for TAF1(2), starting from a previously reported lead. A cocrystal structure of lead compound 2 bound to TAF1(2) enabled structure-based design and structure-activity-relationship studies that ultimately led to our in vitro tool compound, 27 (GNE-371). Compound 27 binds TAF1(2) with an IC50 of 10 nM while maintaining excellent selectivity over other bromodomain-family members. Compound 27 is also active in a cellular-TAF1(2) target-engagement assay (IC50 = 38 nM) and exhibits antiproliferative synergy with the BET inhibitor JQ1, suggesting engagement of endogenous TAF1 by 27 and further supporting the use of 27 in mechanistic and target-validation studies.


Subject(s)
Benzimidazoles/metabolism , Drug Design , Molecular Probes/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/metabolism , Humans , Models, Molecular , Protein Conformation , Protein Domains
6.
BMC Genomics ; 19(1): 212, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29562890

ABSTRACT

BACKGROUND: Understanding the diversity of repair outcomes after introducing a genomic cut is essential for realizing the therapeutic potential of genomic editing technologies. Targeted PCR amplification combined with Next Generation Sequencing (NGS) or enzymatic digestion, while broadly used in the genome editing field, has critical limitations for detecting and quantifying structural variants such as large deletions (greater than approximately 100 base pairs), inversions, and translocations. RESULTS: To overcome these limitations, we have developed a Uni-Directional Targeted Sequencing methodology, UDiTaS, that is quantitative, removes biases associated with variable-length PCR amplification, and can measure structural changes in addition to small insertion and deletion events (indels), all in a single reaction. We have applied UDiTaS to a variety of samples, including those treated with a clinically relevant pair of S. aureus Cas9 single guide RNAs (sgRNAs) targeting CEP290, and a pair of S. pyogenes Cas9 sgRNAs at T-cell relevant loci. In both cases, we have simultaneously measured small and large edits, including inversions and translocations, exemplifying UDiTaS as a valuable tool for the analysis of genome editing outcomes. CONCLUSIONS: UDiTaS is a robust and streamlined sequencing method useful for measuring small indels as well as structural rearrangements, like translocations, in a single reaction. UDiTaS is especially useful for pre-clinical and clinical application of gene editing to measure on- and off-target editing, large and small.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Gene Rearrangement , Genome, Human , INDEL Mutation , Osteosarcoma/diagnosis , Antigens, Neoplasm/genetics , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Cell Cycle Proteins , Cells, Cultured , Cytoskeletal Proteins , Genomics/methods , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Osteosarcoma/genetics , Sequence Deletion , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
7.
J Med Chem ; 60(24): 10151-10171, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29155580

ABSTRACT

The epigenetic regulator CBP/P300 presents a novel therapeutic target for oncology. Previously, we disclosed the development of potent and selective CBP bromodomain inhibitors by first identifying pharmacophores that bind the KAc region and then building into the LPF shelf. Herein, we report the "hybridization" of a variety of KAc-binding fragments with a tetrahydroquinoline scaffold that makes optimal interactions with the LPF shelf, imparting enhanced potency and selectivity to the hybridized ligand. To demonstrate the utility of our hybridization approach, two analogues containing unique Asn binders and the optimized tetrahydroquinoline moiety were rapidly optimized to yield single-digit nanomolar inhibitors of CBP with exquisite selectivity over BRD4(1) and the broader bromodomain family.


Subject(s)
High-Throughput Screening Assays/methods , p300-CBP Transcription Factors/antagonists & inhibitors , Animals , Asparagine/chemistry , Asparagine/metabolism , Binding Sites , Cell Cycle Proteins , Crystallography, X-Ray , Female , Fluorescence Resonance Energy Transfer/methods , Mice, Inbred Strains , Molecular Docking Simulation , Nuclear Proteins/antagonists & inhibitors , Protein Domains , Pyrazoles/chemistry , Pyridines/chemistry , Quinolines/chemistry , Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/metabolism
8.
ACS Med Chem Lett ; 8(7): 737-741, 2017 Jul 13.
Article in English | MEDLINE | ID: mdl-28740608

ABSTRACT

The biological function of bromodomains, epigenetic readers of acetylated lysine residues, remains largely unknown. Herein we report our efforts to discover a potent and selective inhibitor of the bromodomain of cat eye syndrome chromosome region candidate 2 (CECR2). Screening of our internal medicinal chemistry collection led to the identification of a pyrrolopyridone chemical lead, and subsequent structure-based drug design led to a potent and selective CECR2 bromodomain inhibitor (GNE-886) suitable for use as an in vitro tool compound.

9.
Nat Commun ; 8: 13905, 2017 01 09.
Article in English | MEDLINE | ID: mdl-28067217

ABSTRACT

The CRISPR-Cas9 system provides a versatile toolkit for genome engineering that can introduce various DNA lesions at specific genomic locations. However, a better understanding of the nature of these lesions and the repair pathways engaged is critical to realizing the full potential of this technology. Here we characterize the different lesions arising from each Cas9 variant and the resulting repair pathway engagement. We demonstrate that the presence and polarity of the overhang structure is a critical determinant of double-strand break repair pathway choice. Similarly, single nicks deriving from different Cas9 variants differentially activate repair: D10A but not N863A-induced nicks are repaired by homologous recombination. Finally, we demonstrate that homologous recombination is required for repairing lesions using double-stranded, but not single-stranded DNA as a template. This detailed characterization of repair pathway choice in response to CRISPR-Cas9 enables a more deterministic approach for designing research and therapeutic genome engineering strategies.


Subject(s)
BRCA2 Protein/genetics , CRISPR-Cas Systems , DNA/genetics , Gene Editing/methods , Genome, Human , Rad51 Recombinase/genetics , Recombinational DNA Repair , BRCA2 Protein/antagonists & inhibitors , BRCA2 Protein/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Protein 9 , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/metabolism , DNA Breaks, Double-Stranded , Endonucleases/genetics , Endonucleases/metabolism , HEK293 Cells , Humans , K562 Cells , Osteoblasts/cytology , Osteoblasts/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/metabolism
10.
J Med Chem ; 59(23): 10549-10563, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27682507

ABSTRACT

The single bromodomain of the closely related transcriptional regulators CBP/EP300 is a target of much recent interest in cancer and immune system regulation. A co-crystal structure of a ligand-efficient screening hit and the CBP bromodomain guided initial design targeting the LPF shelf, ZA loop, and acetylated lysine binding regions. Structure-activity relationship studies allowed us to identify a more potent analogue. Optimization of permeability and microsomal stability and subsequent improvement of mouse hepatocyte stability afforded 59 (GNE-272, TR-FRET IC50 = 0.02 µM, BRET IC50 = 0.41 µM, BRD4(1) IC50 = 13 µM) that retained the best balance of cell potency, selectivity, and in vivo PK. Compound 59 showed a marked antiproliferative effect in hematologic cancer cell lines and modulates MYC expression in vivo that corresponds with antitumor activity in an AML tumor model.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Pyrazoles/pharmacology , Pyridones/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dogs , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Madin Darby Canine Kidney Cells , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyridones/chemical synthesis , Pyridones/chemistry , Structure-Activity Relationship
11.
Proc Natl Acad Sci U S A ; 113(22): 6182-7, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27185940

ABSTRACT

Lysine to methionine (K-to-M) mutations in genes encoding histone H3 are thought to drive a subset of pediatric brain and bone cancers. These high-frequency K-to-M mutations occur at sites of methylation on histone H3, and tumors containing the mutant histones exhibit a global loss of specific histone methylation marks. Previous studies showed that K-to-M mutant histones, also known as oncohistones, are potent orthosteric inhibitors of specific Su(var)3-9, Enhancer-of-zeste, Trithorax (SET) domain methyltransferases. However, the biochemical and biophysical details of the interaction between K-to-M mutant histones and the respective SET domain methyltransferases are currently unknown. Here, we use the histone H3K9-directed methyltransferase G9a as a model to explore the mechanism of inhibition by K-to-M oncohistones. X-ray cocrystal structures revealed that the K9M residue of histone H3 occupies the active site cavity of G9a, and kinetic analysis indicates competitive inhibition of G9a by histone H3K9M. Additionally, we find that the cofactor S-adenosyl methionine (SAM) is necessary for stable interaction between G9a and H3K9M histone. Consistent with the formation of a ternary complex, we find that the inhibitory peptide is uncompetitive with regard to SAM. These data and others indicate that K-to-M oncohistones promote global loss of specific lysine methylation through sequestration and inhibition of SAM-bound SET domain methyltransferases.


Subject(s)
Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histones/genetics , Lysine/genetics , Methionine/genetics , Mutation/genetics , S-Adenosylmethionine/pharmacology , Crystallography, X-Ray , Histone-Lysine N-Methyltransferase/genetics , Histones/chemistry , Humans , Lysine/chemistry , Methionine/chemistry , Peptide Fragments/chemistry , Substrate Specificity
12.
ACS Med Chem Lett ; 7(5): 531-6, 2016 May 12.
Article in English | MEDLINE | ID: mdl-27190605

ABSTRACT

CBP and EP300 are highly homologous, bromodomain-containing transcription coactivators involved in numerous cellular pathways relevant to oncology. As part of our effort to explore the potential therapeutic implications of selectively targeting bromodomains, we set out to identify a CBP/EP300 bromodomain inhibitor that was potent both in vitro and in cellular target engagement assays and was selective over the other members of the bromodomain family. Reported here is a series of cell-potent and selective probes of the CBP/EP300 bromodomains, derived from the fragment screening hit 4-methyl-1,3,4,5-tetrahydro-2H-benzo[b][1,4]diazepin-2-one.

13.
J Med Chem ; 59(11): 5391-402, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27219867

ABSTRACT

The biological role played by non-BET bromodomains remains poorly understood, and it is therefore imperative to identify potent and highly selective inhibitors to effectively explore the biology of individual bromodomain proteins. A ligand-efficient nonselective bromodomain inhibitor was identified from a 6-methyl pyrrolopyridone fragment. Small hydrophobic substituents replacing the N-methyl group were designed directing toward the conserved bromodomain water pocket, and two distinct binding conformations were then observed. The substituents either directly displaced and rearranged the conserved solvent network, as in BRD4(1) and TAF1(2), or induced a narrow hydrophobic channel adjacent to the lipophilic shelf, as in BRD9 and CECR2. The preference of distinct substituents for individual bromodomains provided selectivity handles useful for future lead optimization efforts for selective BRD9, CECR2, and TAF1(2) inhibitors.


Subject(s)
Histone Acetyltransferases/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Pyridones/pharmacology , Pyrroles/pharmacology , TATA-Binding Protein Associated Factors/antagonists & inhibitors , Transcription Factor TFIID/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Water/chemistry , Binding Sites/drug effects , Cell Cycle Proteins , Dose-Response Relationship, Drug , Fluorescence Resonance Energy Transfer , Fluorometry , Histone Acetyltransferases/metabolism , Humans , Ligands , Models, Molecular , Molecular Conformation , Nuclear Proteins/metabolism , Pyridones/chemical synthesis , Pyridones/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Transcription Factors/metabolism
14.
J Med Chem ; 59(4): 1330-9, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26815195

ABSTRACT

In recent years, inhibition of the interaction between the bromodomain and extra-terminal domain (BET) family of chromatin adaptors and acetyl-lysine residues on chromatin has emerged as a promising approach to regulate the expression of important disease-relevant genes, including MYC, BCL-2, and NF-κB. Here we describe the identification and characterization of a potent and selective benzoisoxazoloazepine BET bromodomain inhibitor that attenuates BET-dependent gene expression in vivo, demonstrates antitumor efficacy in an MV-4-11 mouse xenograft model, and is currently undergoing human clinical trials for hematological malignancies (CPI-0610).


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Azepines/chemistry , Azepines/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Azepines/pharmacokinetics , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Clinical Trials as Topic , Dogs , Genes, myc/drug effects , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Rats , Transcription Factors/chemistry , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
15.
Bioorg Med Chem Lett ; 25(9): 1842-8, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25851940

ABSTRACT

In this report we detail the evolution of our previously reported thiophene isoxazole BET inhibitor chemotype exemplified by CPI-3 to a novel bromodomain selective chemotype (the methyl isoxazoleazepine chemotype) exemplified by carboxamide 23. The methyl isoxazoleazepine chemotype provides potent inhibition of the bromodomains of the BET family, excellent in vivo PK across species, low unbound clearance, and target engagement in a MYC PK-PD model.


Subject(s)
Azepines/pharmacology , Drug Design , Nuclear Proteins/antagonists & inhibitors , Oxazoles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Azepines/chemical synthesis , Azepines/chemistry , Cell Cycle Proteins , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Oxazoles/chemical synthesis , Oxazoles/chemistry , Structure-Activity Relationship
16.
ACS Med Chem Lett ; 4(9): 835-40, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24900758

ABSTRACT

The identification of a novel series of small molecule BET inhibitors is described. Using crystallographic binding modes of an amino-isoxazole fragment and known BET inhibitors, a structure-based drug design effort lead to a novel isoxazole azepine scaffold. This scaffold showed good potency in biochemical and cellular assays and oral activity in an in vivo model of BET inhibition.

17.
Biochemistry ; 50(5): 788-94, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21174448

ABSTRACT

X-ray crystal structures have been previously determined for three CLC-type transporter homologues, but the absolute unitary transport rate is known for only one of these. The Escherichia coli Cl(-)/H(+) antiporter (EC) moves ∼2000 Cl(-) ions/s, an exceptionally high rate among membrane-transport proteins. It is not known whether such rapid turnover is characteristic of ClCs in general or if the E. coli homologue represents a functional outlier. Here, we characterize a CLC Cl(-)/H(+) antiporter from the cyanobacterium Synechocystis sp. PCC6803 (SY) and determine its crystal structure at 3.2 Šresolution. The structure of SY is nearly identical to that of EC, with all residues involved in Cl(-) binding and proton coupling structurally similar to their equivalents in EC. SY actively pumps protons into liposomes against a gradient and moves Cl(-) at ∼20 s(-1), 1% of the EC rate. Electrostatic calculations, used to identify residues contributing to ion binding energetics in SY and EC, highlight two residues flanking the external binding site that are destabilizing for Cl(-) binding in SY and stabilizing in EC. Mutation of these two residues in SY to their counterparts in EC accelerates transport to ∼150 s(-1), allowing measurement of Cl(-)/H(+) stoichiometry of 2/1. SY thus shares a similar structure and a common transport mechanism to EC, but it is by comparison slow, a result that refutes the idea that the transport mechanism of CLCs leads to intrinsically high rates.


Subject(s)
Antiporters/chemistry , Bacterial Proteins/chemistry , Synechocystis/enzymology , Amino Acid Sequence , Antiporters/genetics , Antiporters/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chloride Channels/chemistry , Chloride Channels/genetics , Chloride Channels/metabolism , Chlorides/metabolism , Crystallography, X-Ray , Escherichia coli/chemistry , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Hydrogen/metabolism , Kinetics , Molecular Sequence Data , Mutation , Protein Structure, Secondary , Sequence Alignment , Synechocystis/chemistry , Synechocystis/genetics
18.
Nature ; 460(7258): 1040-3, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19578361

ABSTRACT

To reach the mammalian gut, enteric bacteria must pass through the stomach. Many such organisms survive exposure to the harsh gastric environment (pH 1.5-4) by mounting extreme acid-resistance responses, one of which, the arginine-dependent system of Escherichia coli, has been studied at levels of cellular physiology, molecular genetics and protein biochemistry. This multiprotein system keeps the cytoplasm above pH 5 during acid challenge by continually pumping protons out of the cell using the free energy of arginine decarboxylation. At the heart of the process is a 'virtual proton pump' in the inner membrane, called AdiC, that imports L-arginine from the gastric juice and exports its decarboxylation product agmatine. AdiC belongs to the APC superfamily of membrane proteins, which transports amino acids, polyamines and organic cations in a multitude of biological roles, including delivery of arginine for nitric oxide synthesis, facilitation of insulin release from pancreatic beta-cells, and, when inappropriately overexpressed, provisioning of certain fast-growing neoplastic cells with amino acids. High-resolution structures and detailed transport mechanisms of APC transporters are currently unknown. Here we describe a crystal structure of AdiC at 3.2 A resolution. The protein is captured in an outward-open, substrate-free conformation with transmembrane architecture remarkably similar to that seen in four other families of apparently unrelated transport proteins.


Subject(s)
Amino Acid Transport Systems/chemistry , Antiporters/chemistry , Bacterial Proteins/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Salmonella typhi/chemistry , Amino Acid Sequence , Amino Acid Transport Systems/metabolism , Antiporters/metabolism , Crystallography, X-Ray , Escherichia coli Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Multigene Family , Protein Conformation , Structural Homology, Protein
19.
Proc Natl Acad Sci U S A ; 105(32): 11194-9, 2008 Aug 12.
Article in English | MEDLINE | ID: mdl-18678918

ABSTRACT

The CLC family of Cl(-)-transporting proteins includes both Cl(-) channels and Cl(-)/H(+) exchange transporters. CLC-ec1, a structurally known bacterial homolog of the transporter subclass, exchanges two Cl(-) ions per proton with strict, obligatory stoichiometry. Point mutations at two residues, Glu(148) and Tyr(445), are known to impair H(+) movement while preserving Cl(-) transport. In the x-ray crystal structure of CLC-ec1, these residues form putative "gates" flanking an ion-binding region. In mutants with both of the gate-forming side chains reduced in size, H(+) transport is abolished, and unitary Cl(-) transport rates are greatly increased, well above values expected for transporter mechanisms. Cl(-) transport rates increase as side-chain volume at these positions is decreased. The crystal structure of a doubly ungated mutant shows a narrow conduit traversing the entire protein transmembrane width. These characteristics suggest that Cl(-) flux through uncoupled, ungated CLC-ec1 occurs via a channel-like electrodiffusion mechanism rather than an alternating-exposure conformational cycle that has been rendered proton-independent by the gate mutations.


Subject(s)
Chloride Channels/chemistry , Chlorides/chemistry , Protons , Amino Acid Substitution , Animals , Chloride Channels/genetics , Chloride Channels/metabolism , Chlorides/metabolism , Crystallography, X-Ray , Humans , Ion Transport/physiology , Point Mutation , Protein Structure, Tertiary/genetics
20.
J Virol ; 81(22): 12272-84, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17804496

ABSTRACT

Rotavirus, the major pathogen of infantile gastroenteritis, carries a nonstructural protein, NSP2, essential for viroplasm formation and genome replication/packaging. In addition to RNA-binding and helix-destabilizing properties, NSP2 exhibits nucleoside triphosphatase activity. A conserved histidine (H225) functions as the catalytic residue for this enzymatic activity, and mutation of this residue abrogates genomic double-stranded RNA synthesis without affecting viroplasm formation. To understand the structural basis of the phosphatase activity of NSP2, we performed crystallographic analyses of native NSP2 and a functionally defective H225A mutant in the presence of nucleotides. These studies showed that nucleotides bind inside a cleft between the two domains of NSP2 in a region that exhibits structural similarity to ubiquitous cellular HIT (histidine triad) proteins. Only minor conformational alterations were observed in the cleft upon nucleotide binding and hydrolysis. This hydrolysis involved the formation of a stable phosphohistidine intermediate. These observations, reminiscent of cellular nucleoside diphosphate (NDP) kinases, prompted us to investigate whether NSP2 exhibits phosphoryl-transfer activity. Bioluminometric assay showed that NSP2 exhibits an NDP kinase-like activity that transfers the bound phosphate to NDPs. However, NSP2 is distinct from the highly conserved cellular NDP kinases in both its structure and catalytic mechanism, thus making NSP2 a potential target for antiviral drug design. With structural similarities to HIT proteins, which are not known to exhibit NDP kinase activity, NSP2 represents a unique example among structure-activity relationships. The newly observed phosphoryl-transfer activity of NSP2 may be utilized for homeostasis of nucleotide pools in viroplasms during genome replication.


Subject(s)
Nucleoside-Diphosphate Kinase/chemistry , Nucleotides/chemistry , RNA-Binding Proteins/chemistry , Viral Nonstructural Proteins/chemistry , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Animals , Binding Sites , Catalysis , Catalytic Domain , Humans , Kinetics , Molecular Sequence Data , Nucleoside-Diphosphate Kinase/genetics , Nucleoside-Diphosphate Kinase/physiology , Phosphorylation , Protein Conformation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/physiology , Rotavirus/enzymology , Rotavirus/physiology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/physiology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...