CRYSTAL STRUCTURE OF A MarR FAMILY POLYPEPTIDE

The crystal structure of the product, crystals of the MarR protein, a regulator of multiple antibiotic resistance in Escherichia coli, and methods of crystallization of the MarR protein are described.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
RELATED APPLICATIONS

This application is a continuation application of U.S. application Ser. No. 12/488,275, filed Jun. 19, 2009; which is a continuation application of U.S. application Ser. No. 11/707,404, filed Feb. 16, 2007; which is a continuation application of U.S. application Ser. No. 10/196,655, filed Jul. 15, 2002, now U.S. Pat. No. 7,202,339, issued Apr. 10, 2007; which claims priority to U.S. Provisional Patent Application Ser. No. 60/388,622, filed on Jun. 13, 2002; and U.S. Provisional Patent Application Ser. No. 60/305,404, filed on Jul. 13, 2001. The entire contents of all of the above applications and patents are hereby incorporated herein by reference.

GOVERNMENT FUNDING

This invention was made with government support under GM51661 awarded by The National Institutes of Health. The government may, therefore, have certain rights in the invention.

BACKGROUND OF THE INVENTION

The Mar phenotype in E. coli is attributed largely to the action of MarA, the expression of which is regulated by MarR (Alekshun, M. N. supra (1997)). MarA is a transcription factor that autoactivates expression of the marRAB operon and regulates the expression of a global network of more than 60 chromosomal genes (Martin, R. G. et al. J. Bact. 178, 2216-2223 (1996); Barbosa, T. M. & Levy, S. B. J. Bact. 182, 3467-3474 (2000)). Mar mutants in isolates of clinical origin have now been identified (Maneewannakul, K. & Levy, S. B. Antimicrob. Agents Chemother. 40, 1695-1698 (1996); Oethinger, M. et al. Antimicrob. Agents Chemother. 42, 2089-2094 (1998); Linde, H. J. et al. Antimicrob. Agents Chemother. 44, 1865-1868 (2000); Ziha-Zarifi, I., et al. Antimicrob. Agents Chemother. 43, 287-291 (1999); Koutsolioutsou, A et al. Antimicrob. Agents Chemother. 45, 38-43 (2001)). Constitutive overexpression of MarA or a MarA homolog in many of these strains is a key contributor to the maintenance of the resistance phenotype, particularly with respect to the fluoroquinolones, and recent studies have documented the selection of Mar mutants, bearing mutations in MarR, MexR, or other homologous loci, in E. coli, Pseudomonas aeruginosa, and other organisms during antimicrobial chemotherapy (Oethinger, M supra; Linde, H. J. et al.; supra; Ziha-Zarifi, I. et al. supra; Kern, W. V., et al. Antimicrob. Agents Chemother. 44, 814-820 (2000)).

MarR is a regulator of multiple antibiotic resistance in Escherichia coli. It is the prototypic member of a family of regulatory proteins found in the Bacteria and the Archaea that play important roles in the development of antibiotic resistance, a global health problem. In the absence of an appropriate stimulus, MarR negatively regulates expression of the marRAB operon (Cohen, S. P., et al. 1993. J. Bacteriol. 175: 1484-1492; Martin, R. G. and Rosner, J. L. 1995. Proc. Natl. Acad. Sci. 92: 5456-5460; Seoane, A. S. and Levy, S. B. 1995. J. Bacteriol. 177: 414-3419, 1995). DNA footprinting experiments suggest that MarR dimerizes at two locations, sites I and II, health problem. In the absence of an appropriate stimulus, MarR negatively regulates expression of the marRAB operon (Cohen, S. P., et al. 1993. J. Bacteria 175: 1484-1492.; Martin, R. G. and Rosner, J. L. 1995. Proc. Natl. Acad. Sci. 92: 5456-5460; Seoane, A. S. and Levy, S. B. 1995. J. Bacteria 177: 3414-3419., 1995). DNA footprinting experiments suggest that MarR dimerizes at two locations, sites I and II, within the mar operator (marO) (Martin and Rosner, 1995, supra). Site I is positioned among the −35 and −10 hexamers and site II spans the putative MarR ribosome binding site (reviewed in Alekshun, M. N. and Levy, S. B. 1997. Antimicrob. Agents Chemother. 10: 2067-2075).

MarR is a member of a newly recognized family of regulatory proteins (Alekshun, M. N. and Levy, S. B. 1997. Antimicrob. Agents Chemother. 10:. 2067-2075. Sulavik, M. C., et al. 1995. Mol. Med. 1: 436-446) and many functional homologues have been identified in a variety of important human pathogens and have been found to regulate a variety of different processes. For example, some MarR homologues have been found to control expression of multiple antibiotic resistance operons, some regulate tissue-specific adhesive properties, some control expression of a cryptic hemolysin, some regulate protease production, and some regulate sporulation. Proteins of the MarR family control an assortment of biological functions including resistance to multiple antibiotics, organic solvents, household disinfectants, and oxidative stress agents, collectively termed the multiple antibiotic resistance (Mar) phenotype (Alekshun, M. N. & Levy, S. B. Trends Microbiol. 7, 410-413 (1999)). These proteins also regulate the synthesis of pathogenic factors in microbes that infect humans and plants (Miller, P. F. & Sulavik, M. C. Mol. Microbiol. 21, 441-448 (1996)). Insight into the three dimensional structure of MarR family proteins would be of great value in designing drugs that interact with this family of proteins and modulate MarR function, for example, antibiotic resistance and virulence.

SUMMARY OF THE INVENTION

The instant invention advances the prior art by providing the crystal structure of a MarR family polypeptide, MarR. The crystal structure was solved for both the MarR polypeptide and the MarR polypeptide bound to salicylate.

The invention pertains at least in part to a crystallized MarR family polypeptide. The MarR family polypeptide is crystallized under appropriate conditions such that its three dimensional structure can be determined. In another embodiments, the crystallized MarR family polypeptide is given by the atomic coordinates given in FIG. 7 or 8.

In another embodiment, the invention pertains, at least in part, to a method for determining the three dimensional structure of MarR family polypeptide. The method includes crystallizing the MarR family polypeptide under appropriate conditions such that crystals are formed, and analyzing it, such that its three dimensional structure is determined.

In yet another embodiment, the invention pertains to a method of making a crystal of a MarR family polypeptide. The method includes contacting MarR with a modulator to form a complex and allowing crystals of the complex to grow, e.g., by using hanging droplet vapor diffusion.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 shows the sequence alignment of MarR with representative members of the MarR family (SEQ ID NOS 2-8, respectively, in order of appearance).

FIG. 2 is a ribbon representation of the cocrystal structure of the MarR dimer with salicylate viewed with the subunit two-fold axis near vertical.

FIG. 3 is an electrostatic surface representation of the MarR dimer.

FIG. 4 is a Cα trace of a MarR subunit in stereo representation.

FIG. 5 is a representation of the N-/C-terminal domain represented by a surface around the van der Waals radii of the side chain atoms only of the hydrophobic core residues. Helices leading to and from the domain are shown in ribbon representation.

FIG. 6 is a diagram which shows interactions between the DNA-binding domains of the dimer in the region of the Arg 73-Asp 67′ salt bridges. The stereo view is coincident with the 2-fold rotation axis of the dimer. Electron density shown is a 2FO-FC map contoured at 1σ.

FIG. 7 shows the atomic coordinates of the MarR—salicylate co-crystal (residues 7-144 of SEQ ID NO:2).

FIG. 8 shows the atomic coordinates of the MarR crystal without salicylate (residues 12-144 and 9-144 of SEQ ID NO:2).

FIG. 9 is a ribbon representation of the MarR dimer with the two-fold axis near vertical.

DETAILED DESCRIPTION OF THE INVENTION

The invention pertains, at least in part, to crystallized MarR family polypeptides. The crystallized MarR family polypeptides are crystallized under appropriate conditions such that the three dimensional structure can be determined using methods described herein and/or art recognized techniques.

The term “MarR family polypeptide” includes molecules related to MarR, e.g., having certain shared structural and functional features. MarR family polypeptides also include those which are structural homologs of MarR. The structural homologs include those having a crystallized form which are structurally similar to that of crystallized MarR. MarR family members, in addition to having similarity to MarR, may bind to DNA and regulate transcription. While some MarR family members negatively control transcription (e.g., MarR), others have positive/activator functions (e.g., SlyA, BadR, NhhD, and MexR). MarR family polypeptides comprise DNA and protein binding domains. In addition, MarR family polypeptides can interact with a variety of structurally unrelated compounds that regulate their activity.

Exemplary MarR family members are taught in the art and can be found, e.g., in Sulavik et al. (1995. Molecular Medicine. 1:436), Miller and Sulavik (1996. Molecular Microbiology. 21:441) in which alignments of MarR and related proteins are shown, or through the use of BLAST searches and other techniques known in the art. Exemplary MarR family polypeptides are also illustrated in the following chart:

MarR Family Polypeptides Gram-negative Escherichia coli MarR SlyA EmrR (MprA) PapX PrsX HpcR Ec17 kD Salmonella typhimurium MarR SlyA EmrR Pseudomonas aeruginosa MexR Erwinia chrysanthemi PecS Rhodopseudomonas palustris BadR Burkholderia pseudomallei OrfE Gram-positive Bacillus subtilus YdcH YhbI YkmA YkoM Orf7 YfiV YetL YdgJ YwoH YwaE YwhA Hpr YybA YxaD YsmB YusO YpoP YkvE Bacillus firmus Orf7 Staphylococcus sciuri Orf145 Orf141 Butyrivibrio fibrisolvens CinR Sphingomonas aromaticivorans Orf158 Rhodococcus rhodochrous NhhD Streptomyces peucetius Orf1 Acid-fast Mycobacterium tuberculosis 14.7 kD Rv1404 Rv0737 Rv0042c Yz08 (15.6 kD) Mycobacterium leprae Yz08 (15.6 kD) Archaea Methanobacterium thermoautotrophicum MTH313 Sulfolobus solfataricus Lrs14 Archaeoglobus fulgidus CinR Purple non-sulfur Rhodobacter capsulatus PetP Sinorhizobium meliloti SlyA (E293909)

Preferably, the MarR family polypeptide is MarR. Other preferred MarR family polypeptides include: EmrR, Ec17kD, and MexR.

In a further embodiment, the MarR family polypeptide has a winged-helix structure, such as the three dimensional structure of MarR.

FIG. 1 shows a sequence alignment of MarR with representative MarR family polypeptides. The MarR secondary structure elements were identified in its crystal structure and are illustrated in FIG. 1 (e.g., as tubes for α-helices (α) and arrows for β-sheets (β) and the single wing region (W1)). The numbering in FIG. 1 is according to the MarR primary sequence. The MarR family polypeptides used for the alignment were from the following organisms: MarR, E. coli; MprA (EmrR), E. coli; MexR, Pseudomonas aeruginosa; YS87, Mycobacterium tuberculosis; SlyA, Salmonella typhimurium; PecS, Erwinia chrysanthemi; CinR, Butyrivibrio fibrisolvens.

In a further embodiment, MarR comprises, consists essentially of, or consists of the polypeptide sequence shown in Sequence Listing SEQ ID NO:1. Other MarR family polypeptides of interest include EmrR, YS87, PecS, CinR, SlyA, Ec17kD, MexR, etc.

In another embodiment, the MarR family polypeptide is found, for example, in one of the following organisms Escherichia coli, Salmonella typhimurium, Salmonella enterica, Enterobacter cloacae, Enterobacter aerogenes, Erwinia chrysanthemi, Yrsinia pestis, Yersinia enterocolitica, Kluyvera cryocrescens, Edwardsiella tarda, Pseudomonas aeruginosa, Vibrio cholera, Xanthomonas axonopodis, Xanthomonas campestris, Ralstonia solanacearum, Burkholderia pseudomallei, Burkholderia cepacia, Vogesella indigofera, Mesorhizobium loti, Agrobacterium tumefaciens, Sinorhizobium meliloti, Brucella melitensis, Caulobacter crescentus, Bacillus anthracis, Bacillus subtilis, Bacillus halodurans, Listeria monocytogenes, Listeria innocua, Listeria welshimeri, Staphylococcus sciuri, Streptococcus criceti, Streptococcus pneumoniae, Clostridium perfringens, Clostridium difficile, Streptomyces coelicolor, Streptomyces avermitilis, Mycobacterium tuberculosis, Mycobacterium leprae, Corynebacterium glutamicum, Thermotoga maritima, Methanosarcina acetivorans, Methanosarcina mazei, and Sulfolobus solfataricus.

In another embodiment, the MarR family polypeptide is from an organism belonging to one of the following biological classifications: Enterobacteriaceae, Enterobacter, Yersinia, Kluyvera, Edwardsiella, Xanthomonas group, Xanthomonadales, Pseudomonaceae/Moraxellaceae group, Pseudomonadaceae, Vibrionaceae group, Burkholderia/Oxalobacter/Ralstonia group, Ralstonia group, Burkholderia group, Neisseriaceae, Vogesella, Rhizobiaceae group, Phyllobacteriaceae, Mesorhizobium, Rhizobiaceae, Sinorhizobium, Brucellaceae, Brucella, Caulobacter group, Firmicutes, Bacillus/Clostridium group, Bacilli, Bacillales, Bacillus, Bacillaceae, Bacillus cereus group, Listeria, Listeriaceae, Staphylococcaceae, Staphylococcus, Streptococcus, Lactobacillales, Streptococcaceae, Clostridium, Clostridiaceae, Clostridiales, Clostridia, Actinomycetales, Actinobacteria, Actinobacteridae, Streptomyces, Streptomycineae, Streptomycetaceae, Corynebacterineae, Mycobacterium, Mycobacteriaceae, Corynebacteriaceae, Corynebacterium, Nostocales, Nostocaceae, Nostoc, Thermotogae, Thermotogales, Thermotogaceae; Thermotoga, Methanosarcina, Euryarchaeota, Methanococci; Methanosarcinales, Methanosarcinaceae, Crenarchaeota, Thermoprotei; Sulfolobales, Sulfolobaceae, Sulfolobus, Proteobacteria, Pectobacterium, Cyanobacteria, or Archaea.

In one embodiment, the MarR family polypeptides of the invention are naturally occurring. In another embodiment, the subject crystal structures can be generated using non-naturally occurring forms of MarR family polypeptides, e.g. mutants or synthetic forms of MarR family polypeptides not found in nature.

In one embodiment, the MarR family polypeptide comprises one or more conservative mutations as compared to the wild type protein for the particular MarR family polypeptide. The term “MarR family polypeptide” also includes fragments of MarR family polypeptides which minimally retain at least a portion of the tertiary structure of the MarR family protein.

MarR family member polypeptide sequences are “structurally related” to one or more known MarR family members, preferably to MarR. This structural relatedness is shown by sequence similarity between two MarR family polypeptide sequences or between two MarR family nucleotide sequences. Sequence similarity can be shown, e.g., by optimally aligning MarR family member sequences using an alignment program for purposes of comparison and comparing corresponding positions. To determine the degree of similarity between sequences, they will be aligned for optimal comparison purposes (e.g., gaps may be introduced in the sequence of one protein for nucleic acid molecule for optimal alignment with the other protein or nucleic acid molecules). The amino acid residues or bases and corresponding amino acid positions or bases are then compared. When a position in one sequence is occupied by the same amino acid residue or by the same base as the corresponding position in the other sequence, then the molecules are identical at that position. If amino acid residues are not identical, they may be similar. As used herein, an amino acid residue is “similar” to another amino acid residue if the two amino acid residues are members of the same family of residues having similar side chains. Families of amino acid residues having similar side chains have been defined in the art (see, for example, Altschul et al. 1990. J. Mol. Biol. 215:403) including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan). The degree (percentage) of identity or similarity between sequences, therefore, can be calculated as a function of the number of identical or similar positions shared by two sequences (i.e., % homology=# of identical or similar positions/total # of positions×100). Alignment strategies are well known in the art; see, for example, Altschul et al. supra for optimal sequence alignment.

MarR family polypeptides share some amino acid sequence similarity with MarR. The nucleic acid and amino acid sequences of MarR as well as other MarR family polypeptides are available in the art. For example, the nucleic acid and amino acid sequence of MarR can be found, e.g., on GeneBank (accession number M96235 or in Cohen et al. 1993. J. Bacteriol. 175:1484, or in SEQ ID NO:1).

The nucleic acid and protein sequences of MarR can be used as “query sequences” to perform a search against databases (e.g., either public or private) to, for example, identify other MarR family members having related sequences. Such searches can be performed, e.g., using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. Mol. Biol. 215:403-10. BLAST nucleotide searches can be performed with the NBLAST program, score=100, wordlength=12 to obtain nucleotide sequences homologous to MarR family nucleic acid molecules. BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to MarR protein molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res. 25(17):3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., BLAST and NBLAST) can be used. See http://www.ncbi.nlm.nih.gov.

MarR family members can also be identified as being similar based on their ability to specifically hybridize to the complement of nucleic acid sequences specifying MarR. Such stringent conditions are known to those skilled in the art and can be found e.g., in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. A preferred, non-limiting example of stringent hybridization conditions are hybridization in 6× sodium chloride/sodium citrate (SSC) at about 45° C., followed by one or more washes in 0.2X SSC, 0.1% SDS at 50-65° C. Conditions for hybridizations are largely dependent on the melting temperature Tm that is observed for half of the molecules of a substantially pure population of a double-stranded nucleic acid. Tm is the temperature in ° C. at which half the molecules of a given sequence are melted or single-stranded. For nucleic acids of sequence 11 to 23 bases, the Tm can be estimated in degrees C. as 2(number of A+T residues)+4(number of C+G residues).

Hybridization or annealing of nucleic acid molecules should be conducted at a temperature lower than the Tm, e.g., 15° C., 20° C., 25° C. or 30° C. lower than the Tm. The effect of salt concentration (in M of NaCl) can also be calculated, see for example, Brown, A., “Hybridization” pp. 503-506, in The Encyclopedia of Molec. Biol., J. Kendrew, Ed., Blackwell, Oxford (1994).

Preferably, the nucleic acid sequence of a MarR family member identified in this way is at least about 10%, 20%, more preferably at least about 30%, more preferably at least about 40% identical and most preferably at least about 50%, or 60% identical or more with a MarR nucleotide sequence. Preferably, MarR family members have an amino acid sequence at least about 20%, more preferably at least about 30%, more preferably at least about 40% identical and most preferably at least about 50%, or 60% or more identical with a MarR amino acid sequence. However, it will be understood that the level of sequence similarity among microbial regulators of gene transcription, even though members of the same family, is not necessarily high. This is particularly true in the case of divergent genomes where the level of sequence identity may be low, e.g., less than 20% (e.g., B. burgdorferi as compared e.g., to B. subtilis). For example, the level of amino acid sequence homology between MarR and PecS is about 31% and the level of amino acid sequence homology between MarR and PapX is about 28% when determined as described above. Accordingly, structural similarity among MarR family members can also be determined based on “three-dimensional correspondence” of amino acid residues. As used herein, the language “three-dimensional correspondence” is meant to includes residues which spatially correspond, e.g., are in the same functional position of a MarR family protein member as determined, e.g., by x-ray crystallography, but which may not correspond when aligned using a linear alignment program. The language “three-dimensional correspondence” also includes residues which perform the same function, e.g., bind to DNA or bind the same cofactor, as deteiinined, e.g., by mutational analysis.

Preferred MarR family polypeptides include: MarR, EmrR, Ec17kD, MexR, PapX, SlyA, Hpr, PecS, Hpr, MprA, (EmrR), as well as the other peptides listed in the chart above or known in the art. In a more preferred embodiment, a MarR family polypeptide is selected from the group consisting of MarR, EmrR, Ec171kD, and MexR. In a particularly preferred embodiment, a MarR family polypeptide is MarR.

In addition to sharing structural similarity, MarR family members have a MarR family polypeptide activity, i.e., they bind to DNA and regulate transcription. Some MarR family members positively regulate transcription (e.g., SlyA, BadR, NhhD, or MexR), while others negatively regulate transcription (e.g., MarR). While all MarR family members bind to DNA and regulate transcription, the different loci controlled by each family member regulate different processes in microbes. For example, MarR family polypeptides can control the expression of microbial loci involved in: regulation of antibiotic resistance [e.g., MarR (Cohen et al. 1993. J. Bacteriol. 175:1484), EmrR (Lomovskaya and Lewis. 1992. Proc. Natl. Acad. Sci. 89:8938), and Ec17kD (Sulavik et al. 1995. Mol. Med. 1:436), and MexR (Poole et al. 1996. Antimicrob. Agents. Chemother. 40:2021)], regulation of tissue-specific adhesive properties [e.g., PapX (Marklund et al., 1992. Mol. Microbiol. 6:2225)], regulation of expression of a cryptic hemolysin [e.g., SlyA (Ludwig et al. 1995 249:4740)], regulation of protease production [e.g., Hpr from B. subtilis (Perago and Hoch. 1988. J. Bacteriol. 170:2560) and PecS from Erwinia chrysanthemi (Reverchon et al., 1994. Mol. Microbiol. 11:1127)] and regulation of sporulation [e.g., Hpr (Perego and Hoch. 1988. J. Bacteriol. 170:2560)], regulation of the breakdown of plant materials [e.g., CinR (Dalymple and Swadling 1997 Microbiology)] sensing of phenolic compounds [(e.g., Sulvik et al. 1995. Mol. Med. 1:436], and repress marRAB expression when introduced into E. coli [e.g., Ec17kd (Markhmd et al. 1992. Mol. Microbiol. 6:2225) and MprA (EmrR) (del Castillo et al., 1991. J. Bacteriol. 173:3924)]. The activity of MarR family polypeptides is antagonized by salicylate (Lomovskaya et al., 1995. J. Bacteriol. 177:2328; Sulavik et al. 1995. Mol. Med. 1:436).

Preferred MarR family polypeptide activities include regulation of multiple drug resistance and/or regulation of virulence.

In addition to full length MarR family polypeptide fragments MarR family polypeptide which are useful in making crystals are also within the scope of the invention. Accordingly, MarR family polypeptides for use in the instant invention can be full length MarR family member proteins or fragments thereof. Thus, a MarR family polypeptide can comprise, consist essentially of, or consist of an amino acid sequence derived from the full length amino acid sequence of a MarR family member. For example, in one embodiment, a polypeptide comprising a MarR family polypeptide DNA interacting domain or a polypeptide comprising a MarR family member protein interacting domain can be used.

In addition, naturally or non-naturally occurring variants of these polypeptides and nucleic acid molecules which retain the same functional activity, e.g., the ability to bind to DNA and regulate transcription. Such variants can be made, e.g., by mutation using techniques which are known in the art. Alternatively, variants can be chemically synthesized.

For example, it will be understood that the MarR family polypeptides described herein, are also meant to include equivalents thereof. For instance, mutant forms of MarR family polypeptides which are functionally equivalent, (e.g., have the ability to bind to DNA and to regulate transcription from an operon) can be made using techniques which are well known in the art. Mutations can include, e.g., at least one of a discrete point mutation which can give rise to a substitution, or by at least one deletion or insertion. For example, random mutagenesis can be used. Mutations can be made by random mutagenesis or using cassette mutagenesis. For the former, the entire coding region of a molecule is mutagenized by one of several methods (chemical, PCR, doped oligonucleotide synthesis) and that collection of randomly mutated molecules is subjected to selection or screening procedures. In the latter, discrete regions of a protein, corresponding either to defined structural or functional determinants (e.g., the first or second helix of a helix-turn-helix domain) are subjected to saturating or semi-random mutagenesis and these mutagenized cassettes are re-introduced into the context of the otherwise wild type allele. In one embodiment, PCR mutagenesis can be used. For example, Megaprimer PCR can be used (O. H. Landt, Gene 96:125-128).

In addition, other portions of the above described polypeptides suitable for use in the claimed assays, such as those which retain their function (e.g., the ability to bind to DNA, to regulate transcription from an operon) or those which are critical for binding to regulatory molecules (such as compounds) can be easily determined by one of ordinary skill in the art, e.g., using standard truncation or mutagenesis techniques and used in the instant assays. Exemplary techniques are described by Gallegos et al. (1996. J. Bacteriol. 178:6427).

It shall be understood that the instant invention also pertains to isolated MarR family member polypeptides, portions thereof, and the nucleic acid molecules encoding them, including naturally occurring and mutant forms.

Preparation of MarR Family Polypeptides

Preferred MarR family polypeptides for use in the instant invention are synthesized, isolated or recombinant polypeptides. In one embodiment, MarR family polypeptides can be made from nucleic acid molecules. Nucleic acid molecules encoding MarR family polypeptides can be used to produce MarR family polypeptides for use in the instant assays. For example, nucleic acid molecules encoding a MarR family polypeptide can be isolated (e.g., isolated from the sequences which naturally flank it in the genome and from cellular components) and can be used to produce a MarR family polypeptide. In one embodiment, a nucleic acid molecule which has been (1) amplified in vitro by, for example, polymerase chain reaction (PCR); (2) recombinantly produced by cloning, or (3) purified, as by cleavage and gel separation; or (4) synthesized by, for example, chemical synthesis can be used to produce MarR family polypeptides. As used herein, the term “nucleic acid” refers to polynucleotides such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA).

Nucleic acid molecules specifying MarR family polypeptides can be placed in a vector. The term “vector” refers to a nucleic acid molecule capable of transporting another nucleic acid molecule to which it has been linked. The term “expression vector” includes any vector, (e.g., a plasmid, cosmid or phage chromosome) containing a gene construct in a form suitable for expression by a cell (e.g., linked to a promoter). In the present specification, “plasmid” and “vector” are used interchangeably, as a plasmid is a commonly used form of vector. Moreover, the invention is intended to include other vectors which serve equivalent functions.

Exemplary expression vectors for expression of a gene encoding a MarR family polypeptide and capable of replication in a bacterium, such a bacterium from a genus selected from the group consisting of Escherichia, Bacillus, Streptomyces, Streptococcus, or in a cell of a simple eukaryotic fungus such as a Saccharomyces or, Pichia, or in a cell of a eukaryotic organism such as an insect, a bird, a mammal, or a plant, are known in the art. Such vectors may carry functional replication-specifying sequences (replicons) both for a host for expression, for example a Streptomyces, and for a host, for example, E. coli, for genetic manipulations and vector construction. See e.g. U.S. Pat. No. 4,745,056. Suitable vectors for a variety of organisms are described in Ausubel, F. et al., Short Protocols in Molecular Biology, Wiley, New York (1995), and for example, for Pichia, can be obtained from Invitrogen (Carlsbad, Calif.).

Useful expression control sequences, include, for example, the early and late promoters of SV40, adenovirus or cytomegalovirus immediate early promoter, the lac system, the trp system, the TAC or TRC system, T7 promoter whose expression is directed by T7 RNA polymerase, the major operator and promoter regions of phage lambda, the control regions for fd coat protein, the promoter for 3-phosphoglycerate kinase or other glycolytic enzymes, the promoters of acid phosphatase, e.g., Pho5, the promoters of the yeast α-mating factors, the polyhedron promoter of the baculovirus system and other sequences known to control the expression of genes of prokaryotic or eukaryotic cells or their viruses, and various combinations thereof. A useful translational enhancer sequence is described in U.S. Pat. No. 4,820,639.

It should be understood that the design of the expression vector may depend on such factors as the choice of the host cell to be transformed and/or the type of protein desired to be expressed.

“Transcriptional regulatory sequence” is a generic term to refer to DNA sequences, such as initiation signals, enhancers, operators, and promoters, which induce or control transcription of nucleic acid sequences with which they are operably linked. It will also be understood that a recombinant gene encoding a MarR family polypeptide can be under the control of transcriptional regulatory sequences which are the same or which are different from those sequences which control transcription of the naturally-occurring MarR family gene. Exemplary regulatory sequences are described in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990). For instance, any of a wide variety of expression control sequences, that control the expression of a DNA sequence when operatively linked to it, may be used in these vectors to express DNA sequences encoding the MarR family proteins of this invention.

Appropriate vectors are widely available commercially and it is within the knowledge and discretion of one of ordinary skill in the art to choose a vector which is appropriate for use with a given microbial cell. The sequences encoding MarR family polypeptides can be introduced into a cell on a self-replicating vector or may be introduced into the chromosome of a microbe using homologous recombination or by an insertion element such as a transposon.

Such vectors can be introduced into cells using standard techniques, e.g., transformation or transfection. The terms “transformation” and “transfection” mean the introduction of a nucleic acid, e.g., an expression vector, into a recipient or “host” cell. The term “transduction” means transfer of a nucleic acid sequence, preferably DNA, from a donor to a recipient cell, by means of infection with a virus previously grown in the donor, preferably a bacteriophage. Nucleic acids can also be introduced into microbial cells by transformation using calcium chloride or electroporation.

“Cells,” “host cells,” “recipient cells, are terms used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. In preferred embodiments, cells used to express MarR family polypeptides for purification, e.g., host cells, comprise a mutation which renders any endogenous MarR family polypeptide nonfunctional or causes the endogenous polypeptide to not be expressed. In other embodiments, mutations may also be made in other related genes of the host cell, such that there will be no interference from the endogenous host loci.

Purification of a MarR family polypeptides, e.g., recombinantly expressed polypeptides, can be accomplished using techniques known in the art. For example, if the MarR family polypeptide is expressed in a form that is secreted from cells, the medium can be collected. Alternatively, if the MarR family polypeptide is expressed in a form that is retained by cells, the host cells can be lysed to release the MarR family polypeptide. Such spent medium or cell lysate can be used to concentrate and purify the MarR family polypeptide. For example, the medium or lysate can be passed over a column, e.g., a column to which antibodies specific for the MarR family member polypeptide have been bound. Alternatively, such antibodies can be specific for a non-MarR family member polypeptide which has been fused to the MarR family polypeptide (e.g., as a tag) to facilitate purification of the MarR family member polypeptide. Other means of purifying MarR family member polypeptides are known in the art.

Architecture of the MarR-Salicylate Co-Crystal Structure

The term “three dimensional structure” includes both pictorial representations of MarR family polypeptides (e.g., such as those shown for MarR with salicylate and MarR without salicylate in the Figures) as well as atomic coordinates (e.g., such as those given in FIG. 7 for MarR-salicylate cocrystal, or in FIG. 8 for MarR without salicylate) and other renditions of the shape, size, or symmetry of a MarR family polypeptide of interest. In a further embodiment, the three dimensional structure of the crystallized MarR family polypeptide is determined to a resolution of 5 Å or better, 3 Å or better, 2.5 Å or better, or, advantageously, 2.3 Å or better. The three dimensional structure of MarR, a MarR family polypeptide, is described in greater detail below.

MarR consists of a dimer with approximate overall dimensions of 50×55×45 Å (corresponding to the width, height and depth of the molecule in the orientation shown in FIG. 2). There is one monomer in the asymmetric unit of the crystal with the dimer composed of subunits related by a crystallographic two-fold rotation. The dimeric structure is consistent with the results of earlier in vitro experiments suggesting that MarR binds the mar operator (marO) as a dimer (Martin, R. G. et al. supra (1996); Martin, R. G. & Rosner, J. L. Proc. Natl. Acad. Sci. U.S.A. 92, 5456-5460 (1995)). Another family member, MprA (EmrR) (FIG. 1) is also believed to function as a dimer (Brooun, A., et al. J. Bact. 181, 5131-5133 (1999)).

Each MarR subunit is an α/β protein with approximate dimensions of 35×25×60 Å and can be divided into two domains as shown in FIG. 2. FIG. 2 is a ribbon representation of the co-crystal structure of the MarR dimer viewed with the subunit 2-fold axis near vertical. The secondary structure elements of one subunit are colored according to the scheme used in FIG. 1. The N- and C-terminal regions are closely juxtaposed and intertwine with the equivalent regions of the second subunit to form a domain that holds the subunits together (FIG. 5). This N-/C-terminal domain is linked to the remainder of the protein by two long antiparallel helices in each subunit. These helices lead to a globular domain that is likely to be responsible for DNA binding (see below). Although the globular DNA-binding domains of the dimer are adjacent to one another, they make minimal contact with each other and are situated to function independently. The overall organization of the N-/C-terminal domain and the two DNA-binding domains results in the formation of an approximately 6 Å wide channel through the center of the dimer (FIGS. 3 and 4). The electrostatic surface potential (FIG. 3) is consistent with the putative DNA-binding regions being strongly electropositive, as observed in other such winged-helix DNA-binding proteins (Gajiwala, K. S. & Burley, S. K. Curr. Opin. Str. Biol. 10, 110-116 (2000)).

Genetic and biochemical data have previously identified the N-terminus of MarR to be important for mediating protein-protein contacts between repressor subunits and have demonstrated that the C-terminus is important for protein function (Alekshun, M. N., et al. Mol. Microbiol. 35, 1394-404 (2000); Linde, H. J. et al. supra). The present structure shows that a-helices in the N- and C-terminal regions of each monomer fold around and interdigitate with those of the other subunit to form a well-packed hydrophobic core (FIG. 5) burying a surface area of 3,570 Å2 (the total buried surface area for the whole dimer is 3,700 Å2). The dimer is further stabilized in this region by several intermolecular hydrogen bonds, notably that between the c-amino group of Lys 24 and the main chain carbonyl oxygen of Pro 144′ in the C-terminus of the second subunit and that between the main chain carbonyl oxygen of Glu 10 and the side chain amino group of Lys 140′.

While the DNA-binding lobe of each subunit also forms a well-packed hydrophobic core, the only interactions between these lobes of the two subunits are salt bridges formed between Asp 67 and Arg 73′ and the reciprocal pair (FIG. 6). These salt bridges stabilize the relationship between the two lobes of the dimer in the crystal form of the protein but if disrupted by other interactions, such as might occur during the binding of MarR to marO, the two lobes would be able to act independently. Relative movement of the lobes would require distortion of the helices that link them to the N-/C-terminal domain. The long linker helix region encompassing residues 103-126 (α5/α5′) (FIG. 2) appears poorly ordered in the region of Gly 116, as is the loop (residues 128-131) that connects this helix to the C-terminal helix (α6/α6′) (FIGS. 1 and 2). It is possible that flexibility at these sites in MarR helps to accommodate relative shifts of the two lobes of the dimer that might occur on binding to DNA.

Architecture of the MarR Crystal Structure

The MarR without salicylate structure is a dimer (FIG. 9) and both subunits of the dimer are in the asymmetric unit. These individual subunits are joined by extensive protein-protein interactions mediated by amino acids within both the N- and C-termini of the monomers (FIG. 9). Like the MarR-salicylate structure, MarR without salicylate is an α/β protein. The MarR without salicylate structure is, however, conformationally different from the salicylate bound protein in that the caliper created by the dimer is more closed in the form of the protein without salicylate. Thus, the channel through the center of the dimer has been lost.

The overall architecture of the MarR without salicylate structure is comparable to that of the salicylate bound protein. The presumed DNA binding lobes or domains are linked to the remainder of the protein by two long α-helices. The positioning of the two DNA binding lobes in the MarR without salicylate structure is fixed by hydrogen bonds between the two lobes. This arrangement is believed to be mediated by interactions between Asp 67 and Arg 77′. In addition, Asp 26 is involved in hydrogen bonds with the side chains of Lys 44 and Lys 25. Together, the presumed recognition helices within the DNA binds lobes overlap by approximately one helical turn.

The DNA Binding Domain

Previous studies have shown the region spanning amino acids 61-121 in MarR to be required for its DNA binding activity (Alekshun, M. N et al., supra, (2000)). In the crystal structure, amino acids 55-100 [β1-α3-α4-β2-W1 (wing)-β3] adopt the winged-helix fold (Clark, K. L. et al. Nature 364, 412-420 (1993)). The overall topology [H1 (α2)-S1 (β1)-H2 (α3)-H3 (α4, recognition helix)-S2 (β2)-W1-S3 (β3)] of this region is similar to other winged-helix DNA binding proteins (the terminology applied for these and subsequent structural elements is according to Gajiwala and Burley, supra (2000)) except that a third strand of sheet present in most members of the group appears to be represented in this MarR structure by an interaction with Ile 55 (β1) (FIG. 1). The presence of this single residue as the third component in the sheet interaction is very similar to that observed in OmpR (Martinez-Hackert, E. & Stock, A. M. Structure 5, 109-124 (1997)), a winged helix protein, where Leu 180 interacts with the two strands of the antiparallel sheet that forms part of the “wing” in this transcription factor.

Within the winged-helix family of DNA-binding proteins, there are multiple modes of DNA binding. Members such as HNF-3γ use the recognition helix (H3) of the motif as the primary determinant for DNA-protein interactions in the major groove, and a wing region(s) (W1) to form minor groove or phosphodiester backbone nucleoprotein contacts (Clark, K. L. et al. supra (1993)). Others, such as hRFX1, use W1 to interact with the major groove and the H3 helix makes only a single minor groove contact (Gajiwala, K. S. et al. Nature 403, 916-921 (2000)). The juxtaposition of the DNA-binding lobes in the present structure does not allow for modeling of the whole dimer onto a B-DNA representation of the operator. However, since mutations in both α4 (H3) and W1 affect the DNA binding activity of MarR it is expected that amino acids from each of these regions would contribute to the DNA binding activity of the protein. For example, mutations in α4, including an R73C change, abolish MarR DNA binding activity in whole cells and in vitro (Alekshun, M. N et al., supra, (2000)). In the present crystal structure, it is the side chain of Arg 73 that is hydrogen bonded to Asp 67′ of the other subunit, an interaction that stabilizes the relative orientation of the two DNA-binding lobes (FIG. 6). Also, an R94C mutation at the tip of W1 is inactive in a whole cell assay while a G95S “superrepressor” mutation increases the DNA binding activity of MarR 30-fold in vitro (Alekshun, M. N et al., supra, (2000); Alekshun, M. N. & Levy, S. B. J. Bact. 181, 3303-3306 (1999)). In the absence of protein-DNA co-crystal structures, the precise mechanism by which these mutations affect the DNA binding activity of the protein is uncertain.

Footprinting experiments have suggested that MarR binds as a dimer at two separate but very similar sites in marO, the protein protects ˜21-bp of DNA on both strands at a single site, and does not bend its target (Martin, R. G. et al., supra (1996); Martin, R. G. et al. Proc. Natl. Acad. Sci. U.S.A. 92, 5456-5460 (1995)). Each MarR binding site is composed of two half-sites whose organization is such that they are on different faces of the DNA double helix (Alekshun, M. N. et al. Mol. Microbiol. 35, 1394-404 (2000)), an arrangement that is very similar to the hRFX1 binding site (Gajiwala, K. S. et al. Nature 403, 916-921 (2000)). For MarR to bind as a dimer, with each winged-helix DNA binding domain contacting one half-site on B-DNA, geometric constraints suggest only a few possible modes of binding. One scenario, involving the binding of a single dimer to one MarR binding site, would require reorientation of the DNA binding lobes so that each could reach one half-site. This would be analogous to the binding of an E2F-DP heterodimer (a eukaryotic transcription factor in which each subunit also has a winged-helix DNA binding domain) to its cognate binding site (Zheng, N. et al. Genes Dev. 13, 666-74. (1999)). A second scenario would involve the binding of two dimers, on opposite faces of the double helix, to a single MarR binding site. This model would be analogous to the binding of DtxR (a bacterial protein with a winged-helix DNA binding domain) to its target, although in DtxR the half-sites are on the same face of the DNA helix (Pohl, E. et al. J. Biol. Chem. 273, 22420-22427 (1998); White, A. et al. Nature 394, 502-506 (1998)).

The term “appropriate conditions” include those conditions which result in the formation of a crystal which can by analyzed to a resolution of 5.0 Å or less. The crystals may be formed using suitable art recognized techniques, such as hanging droplet vapor diffusion. In one embodiment, the temperature of crystallization of the MarR family polypeptide is from about 1° C. to about 30° C., from about 10° C. to about 25° C., from about 15° C. to about 20° C., or abut 17° C. In a further embodiment, the conditions are selected such that crystals of said MarR family polypeptide grow within an acceptable time and reach dimensions which are suitable for structural determination, e.g., by using X-ray diffraction. In one embodiment, the acceptable time is 8 weeks or less, 6 weeks or less, 4 weeks or less, or 3 weeks or less. In an embodiment, the dimensions of the crystal are approximately 0.1 mm or greater per side, 0.2 mm or greater per said, or approximately. 0.3 mm per side or greater.

In a further embodiment, the appropriate conditions include a cocrystallization agent which interacts with the protein such that the three dimensional structure of the protein can be determined.

The term “cocrystallization agent” includes substances which can be crystallized with the MarR family polypeptide such that the three dimensional structure can be determined. In an embodiment, the coocrystallization agent is a MarR family polypeptide modulator. The term “MarR family polypeptide modulator” includes compounds which interact with MarR, either to inhibit or enhance the activity of MarR, such that they alter its activity in its non-crystallized form. In one embodiment, the MarR family polypeptide modulator is a MarR inhibitor (e.g., salicylate, plombagin, or DNP). In an embodiment, the concentration of the salicylate is about 100 mM or less, 150 mM or less, 200 mM or less, or 250 mM or less.

The crystal structure or MarR has been solved using crystals grown in the presence and in the absence of high concentrations (250 mM) of sodium salicylate. This agent, at millimolar concentrations, is known to inhibit MarR activity both in vitro and in whole cells (Alekshun, M. N. supra (1999)). It is routinely used as a model inhibitor of MarR to induce MarA expression in E. coli and S. typhimurium (Cohen, S. P. et al. J. Bact. 175, 7856-7862 (1993); Sulavik, M. C. et al. J. Bact. 179, 1857-1866 (1997)) and thus, to confer a Mar phenotype (Alekshun, M. N. supra (1999)). In one example, salicylate was included in the current crystal growth conditions to provide stable crystals. In another example, the crystal structure of MarR was determined using MarR without salicylate.

Electron density that is consistent with bound salicylate is apparent at two sites on each subunit in the present structure (FIG. 6). These sites are on the surface of the molecule on either side of the proposed DNA-binding helix α4 (H3). In one site (SAL-A), the salicylate hydroxyl is hydrogen bonded to the hydroxyl side chain of Thr 72 in the α4 (H3) helix and the salicylate carboxylate hydrogen bonds to the guanidinium group of Arg 86. In the other site (SAL-B), the salicylate hydroxyl hydrogen bonds to the backbone carbonyl of Ala 70 and its carboxyl hydrogen bonds to Arg 77. In each of these sites, the salicylate ring sits over a hydrophobic side chain in the pocket; Pro 57 in SAL-A and Met 74 in SAL-B and other surface hydrophobes are also located laterally within 3.5 Å of the unsubstituted side of the ring. Although SAL-B is solvent exposed, SAL-A packs in the crystal with Val 96 of a symmetry mate situated 3.6 Å above the salicylate ring and adjacent to the SAL-A site of this symmetry mate. Since both SAL-A and SAL-B are close to the DNA binding helix, they may be positioned to influence DNA binding.

The crystal structure of MarR was solved by multiwavelength anomalous dispersion methods using protein containing selenomethionine. Diffraction data were collected to 2.3 Å from crystals of both seleno and native protein.

The invention is further illustrated by the following examples, which should not be construed as further limiting. The contents of all references, pending patent applications and published patents, cited throughout this application are hereby expressly incorporated by reference.

EXEMPLIFICATION. OF THE. INVENTION EXAMPLE 1 Crystallization of MarR with Salicylate

Protein Production and Purification

Native and selenomethionine (Se-Met) containing MarR was prepared from E. coli BL21(DE3) (Novagen) bearing pMarR-WT, a wild type MarR expression vector that has been previously described (Alekshun, M. N. & Levy, S. B. J. Bact. 181, 4669-4672 (1999)). Native MarR was produced in whole cells according to previous methods (Alekshun, M. N. & Levy, S. B. J. Bact. 181, 4669-4672 (1999)). Se-Met MarR was produced by diluting an overnight culture of E. coli BL21(DE3)+pMarR-WT 1:1000 in M9 medium supplemented with 2 mM MgSO4, 0.2% glucose, 0.1 mM CaCl2, 0.00005% thiamine, 0.04 mg ml−1 each of the following amino acids phenylalanine, leucine, isoleucine, valine, serine, threonine, tyrosine, histidine, lysine, aspartic acid, glutamic acid, tryptophan, and tryptophan, and kanamycin (Miller, J. H. In Experiments in Molecular Genetics. (Cold Spring Harbor Laboratories, Cold Spring Harbor, N.Y.; 1972). This culture was grown at 37° C. to an OD600≈0.6 and 100 mg each of amino acids threonine, lysine-hydrochloride, phenylalanine, 50 mg each of amino acids leucine, isoleucine, and valine (single letter abbreviations), and 60 mg L-(+)-selenomethionine (Sigma) were then added. The culture was grown for 15 min at 37° C.; IPTG was subsequently added to a final concentration of 1 mM and protein production was allowed to proceed for 14.5 hr at 37° C. Cell pellets were collected and processed as previously described (Alekshun, M. N. & Levy, S. B. J. Bact. 181, 4669-4672 (1999)).

Frozen cell pellets containing native or Se-Met MarR were resuspended in 100 mM sodium phosphate buffer (pH 7.4) containing a bacterial protease inhibitor cocktail (Sigma) and sonicated on ice. All buffers contained 2 mM DTT when Se-Met MarR was prepared. Insoluble matter was removed by centrifugation at 4° C. at 30,000×g for 40 min. The supernatant was passed over prepacked 5 ml SP-sepharose HiTrap columns (Amersham Pharmacia Biotech) previously equilibrated with 10 mM sodium phosphate buffer (pH 7.4). The column was washed with 50 ml of 10 mM sodium phosphate buffer (pH 7.4) and the pure proteins were eluted with a linear gradient (0-0.5 M) of NaCl in 10 mM sodium phosphate buffer (pH 7.4). Protein containing fractions were dialyzed vs. 10 mM HEPES (pH 7.4), 200 mM NaCl, and 1 mM DTT, or 2 mM DTT in the case of Se-Met MarR, and the protein in these samples was judged to be greater than 99% pure via SDS-PAGE and electrospray ionization mass spectrophotometry. The latter also demonstrated that more than 95% of the three methionine residues in Se-Met MarR were substituted with selenomethionine.

Crystallization:

MarR crystals were originally grown in 18% PEG MME 5000, 200 mM ammonium sulfate, 100 mM citrate buffer (pH 5.6) but showed anisotropic disorder in the diffraction data that made them unsuitable for structure determination. To stabilize the protein, the citrate was substituted by the known inhibitor salicylate. Crystals of the MarR-salicylate complex were grown at 17° C. by hanging droplet vapor diffusion. 6 μl of a 11.4 mg ml−1 protein solution in 200 mM NaCl, 20 mM HEPES (pH 7.4), and 10 mM DTT were added to 2 μl of reservoir buffer (18% PEG MME 5000, 50 mM ammonium sulfate, 250 mM sodium salicylate, 10 mM DTT, and 15% glycerol, pH 5.5), and 0.8 μl 15% heptanetriol. The droplets were equilibrated with 1 ml of reservoir buffer. Crystals grew within 1 week reaching dimensions of approximately 0.3 mm per side.

X-Ray Data Collection, Structure Determination, and Refinement:

Diffraction data were collected at the Brookhaven National Synchrotron Light Source, beamline X8C. Crystals were flash frozen in mother liquor at the beam line before data collection. All data were processed and reduced using DENZO and SCALEPACK (Otwinowski, Z. In CCP4 Proceedings. 56-62 (Daresbury Laboratory, Warrington, UK, 1993). The space group of the MarR-salicylate co-crystals was determined to be I4122 with one molecule in the asymmetric unit and with unit cell dimensions of a=b=62.0 Å, c=132.9 Å, α=β=γ=90° for both the native and the selenoprotein. Data were collected on the selenoprotein crystals at three wavelengths to enable MAD phasing. Phases were determined from the MAD data using the program SOLVE (Terwilliger, T. C. & Berendzen, J. Acta. Crystallogr. D. 55, 849-861 (1999)). This showed two selenium sites per asymmetric unit, with the third selenomethionine, at the N-terminus, apparently disordered. Maps were solvent-flattened using the program DM and the model was built into density using the program O (Collaborative Computational Project, Number 4. Acta Crystallogr. D. 50, 760-763 (1994); Jones, T. A. et al. Acta Crystallogr. A 47, 110-119 (1991)). Model and refinement parameters for salicylate were obtained from the Hetero Compound Information Center (Kleywegt, G. J. & Jones, T. A. Acta Crystallogr. D. 54, 1119-1131 (1998)). Model refinement was performed using CNS and cycles of rebuilding and refinement continued to give the final model (Brunger, A. T. et al. Acta. Crystallogr. D. 54, 905-921 (1998)). Model quality was assessed by sa-omit, Fo-Fc, maps generated over the whole molecule omitting no more than 7% of the structure at a time. The model extends from residue 6 to the C-terminus at residue 144. In common with several other transcription factors (e.g. TetR, (1A6I), ArgR (1B4B) and TreR (1BYK)), MarR shows relatively high thermal mobility throughout the structure, as reflected by the B-factors. Certain regions appear to be particularly mobile, including the extended structure at the N-terminus, the tip of the “wing” (residues 91-94), parts of the α5 helix, especially around Gly 116 and the connecting loop (128-131) between the α5 and the C-terminal α6 helix. Consistent with the high B-factors, the molecule shows few well-ordered solvent molecules. PROCHECK reports overall g-factors of 0.25 (dihedrals) and 0.55 (main chain covalent forces) and shows that 91% of the residues fall within the most favored region of the Ramachandran plot, with only residue Ala 53 in a disallowed region. This residue is located at the start of the loop connecting the α2 and α3 helices.

The coordinates of the MarR-salicylate cocrystal are shown in FIG. 7. Table 1 shows data collection, phasing, and refinement statistics for the MarR-salicylate co-crystal.

TABLE 1 Data set Native Se-met edge Se-met peak Se-met remote Wavelength (Å) 1.072 0.9795 0.9793 0.9500 Resolution range (Å) 50-2.3 50-2.3 50-2.3 50-2.3 Measured reflections 56,495 84,173 96,582 87,365 Unique reflections 6,069 5,534 5,564 5,472 Completeness (%) 99.5 (100) 91.3 (99.8) 91.7 (99.8) 90.4 (99.7) overall (final <I/σI> (final shell) 21.1 (12.0) 12.2 (7.2) 12.0 (7.0) 12.9 (7.9) Rmerge(%) (final shell) 6.0 (20.0) 6.4 (29.7) 5.7 (30.3) 4.9 (25.5) Rano (%) 4.9 5.0 3.5 Overall FOM 0.59/0.71 (centric/acentric) Resolution 50-2.3 Rfree 28.7% Rcryst 24.7% Atoms/AU Protein 1078 Salicylate 20 Water 18 Average B (Å2) main chain 49.7 side chain 59.2 salicylate 42.7 water 50.0 R.m.s. deviation Bonds (Å) 0.009 Angles (°) 1.3 indicates data missing or illegible when filed

EXAMPLE 2 Crystallization of MarR

MarR was produced and purified as described in Example 1.

Crystallization:

Crystals of MarR were grown by hanging droplet vapor diffusion. 3 μl of a 10 mg ml−1 2:1 (mol:mol) DNA-protein solution in 200 mM NaCl, 20 mM HEPES, pH 7.4, 20 mM TRIS-HCl, pH 8.0, and 2 mM MgCl2 was added to 1 μl of reservoir buffer (23% PEG MME 5000, 100 mM sodium citrate, 200 mM ammonium sulfate, 10 mM DTT, 10% glycerol, 5% Isopropanol, pH 5.6), and 0.4 μl 15% heptanetriol. The droplets were equilibrated with 0.5 ml of reservoir buffer.

X-Ray Data Collection, Structure Determination, and Refinement:

Diffraction data were collected at the Brookhaven National Synchrotron Light Source, beamline X8C. Crystals were flash frozen in mother liquor at the beam line before data collection. All data were processed and reduced using DENZO and

SCALEPACK (Otwinowski, Z. In CCP4 Proceedings. 56-62 (Daresbury Laboratory, Warrington, UK, 1993).

The coordinates of the MarR crystal without salicylate are shown in FIG. 8. Table 2 shows data collection, phasing, and refinement statistics for the MarR crystal.

TABLE 2 Space group C222 Unit cell (Å) a = 65.8, b = 137.7, c = 96.4 Resolution 50-2.7 Rfree 26.7% Rcryst 23.2% Atoms/AU Protein 2093 Water 14 Average B (Å2) main chain 40.0 side chain 48.0 Water 32.6 R.m.s. deviation Bonds (Å) 0.009 Angles (°) 1.3

EQUIVALENTS

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments and methods described herein. Such equivalents are intended to be encompassed by the scope of the following claims.

All patents, patent applications, and literature references cited herein are hereby expressly incorporated by reference. The entire contents of Alekshun et al. “The Crystal Structure of MarR a Regulator of Multiple Antibiotic Resistance at 2.3 Å resolution,” Nature Structural Biology 8(8) is hereby incorporated herein by reference.

Claims

1. A crystallized MarR family polypeptide, wherein said crystallized MarR family polypeptide is crystallized under appropriate conditions such that the three dimensional structure of said MarR family polypeptide can be determined.

2. The crystallized MarR family polypeptide of claim 1, wherein the crystallized MarR family polypeptide is a structural homolog of crystallized MarR.

3. The crystallized MarR family polypeptide of claim 1, wherein said MarR family polypeptide is selected from the group consisting of MarR, SlyA, EmrR (MprA), PapX, PrsX, HpcR, Ec17kD of Escherichia coli; MarR, SlyA, EmrR of Salmonella typhimurium; MexR of Pseudomonas aeruginosa; PecS of Erwinia chrysanthemi; BadR of Rhodopseudomonas palustris; OrfE of Burkholderia pseudomallei; YdcH, YhbI, YkmA, YkoM, Orf7, YfiV, YetL, YdgJ, YwoH, YwaE, YwhA, Hpr, YybA, YxaD, YsmB, YusO, YpoP, YkvE of Bacillus subtilus;: Orf7 of Bacillus firmus;: Orf145, Orf141 of Staphylococcus sciuri;: CinR of Butyrivibrio fibrisolvens;: Orf158 of Sphingomonas aromaticivorans;: NhhD of Rhodococcus rhodochrous; Orf1 of Streptomyces peucetius;: 14.7 kD, Rv1404, Rv0737, Rv0042c, Yz08 (15.6 kD) of Mycobacterium tuberculosis; Yz08 (15.6 kD) of Mycobacterium leprae;: MTH313 of Methanobacterium thermoautotrophicum; Lrs14 of Sulfolobus solfataricus; CinR of Archaeoglobus fulgidus; PetP of Rhodobacter capsulatus; and SlyA (E293909) of Sinorhizobium meliloti.

4. The crystallized MarR family polypeptide of claim 1, wherein said MarR family polypeptide is MarR.

5. The crystallized MarR family polypeptide of claim 4, wherein the peptide sequence of MarR comprises the amino acid sequence shown in SEQ ID NO:2.

6. The crystallized MarR family polypeptide of claim 4, wherein said three dimensional structure has the space group of I4122.

7. The crystallized MarR family polypeptide of claim 1, wherein said appropriate conditions include the presence of a MarR family polypeptide modulator.

8. The crystallized MarR family polypeptide of claim 7, wherein said MarR family polypeptide inhibitor is salicylate.

9. The crystallized MarR family polypeptide of claim 1, wherein said three dimensional structure is determined to a resolution of 5 Å or better.

10.-12. (canceled)

13. The crystallized MarR family polypeptide of claim 4, wherein said crystallized MarR family polypeptide has a structure defined by the atomic coordinates given in FIG. 7 or FIG. 8.

14. (canceled)

15. The crystallized MarR family polypeptide of claim 1, wherein said MarR family polypeptide has a winged-helix structure.

16. A crystallized MarR family polypeptide defined by the atomic coordinates given in FIG. 7 or FIG. 8.

17. (canceled)

18. A method for determining the three dimensional structure of MarR family polypeptide, comprising

crystallizing the MarR family polypeptide under appropriate conditions such that crystals are formed; and
analyzing the crystallized MarR family polypeptide, such that the three dimensional structure of the MarR family polypeptide is determined.

19. The method of claim 18, wherein said crystallized MarR family polypeptide is analyzed by x-ray diffraction techniques.

20. The method of claim 18, wherein said MarR family polypeptide is MarR.

21. The method of claim 20, wherein MarR comprises the amino acid sequence set forth in SEQ ID NO:2.

22. The method of claim 18, wherein said appropriate conditions comprise the presence of a MarR family polypeptide modulator.

23. The method of claim 22, wherein said MarR family polypeptide modulator is salicylate.

24. The method of claim 18, wherein said three dimensional structure is determined to a resolution of 5 Å or better.

25.-27. (canceled)

28. The method of claim 18, wherein said MarR family polypeptide is selected from the group consisting of: MarR, SlyA, EmrR (MprA), PapX, PrsX, HpcR, Ec17kD of Escherichia coli; MarR, SlyA, EmrR of Salmonella typhimurium; MexR of Pseudomonas aeruginosa; PecS of Erwinia chrysanthemi; BadR of Rhodopseudomonas palustris; OrfE of Burkholderia pseudomallei; YdcH, YhbI, YkmA, YkoM, Orf7, YfiV, YetL, YdgJ, YwoH, YwaE, YwhA, Hpr, YybA, YxaD, YsmB, YusO, YpoP, YkvE of Bacillus subtilus;: Orf7 of Bacillus firmus;: Orf145, Orf141 of Staphylococcus sciuri;: CinR of Butyrivibrio fibrisolvens;: Orf158 of Sphingomonas aromaticivorans;: NhhD of Rhodococcus rhodochrous; Orf1 of Streptomyces peucetius;: 14.7 kD, Ry1404, Rv0737, Rv0042c, Yz08 (15.6 kD) of Mycobacterium tuberculosis; Yz08 (15.6 kD) of Mycobacterium leprae;: MTH313 of Methanobacterium thermoautotrophicum; Lrs14 of Sulfolobus solfataricus; CinR of Archaeoglobus fulgidus; PetP of Rhodobacter capsulatus; and SlyA (E293909) of Sinorhizobium meliloti.

29. (canceled)

Patent History
Publication number: 20130144038
Type: Application
Filed: May 15, 2012
Publication Date: Jun 6, 2013
Applicants: Trustees of Boston University (Boston, MA), Trustees of Tufts College (Medford, MA)
Inventors: Michael N. Alekshun (Marlboro, NJ), Stuart B. Levy (Boston, MA), James F. Head (Newton, MA), Barbara A. Seaton (Newton, MA)
Application Number: 13/472,114
Classifications
Current U.S. Class: Proteins, I.e., More Than 100 Amino Acid Residues (530/350); Diffraction, Reflection, Or Scattering Analysis (378/70)
International Classification: C07K 14/245 (20060101); G01N 23/20 (20060101);