Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 1): 46-56, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29372907

RESUMO

Thioredoxins are small ubiquitous proteins that participate in a diverse variety of redox reactions via the reversible oxidation of two cysteine thiol groups in a structurally conserved active site. Here, the NMR solution structures of a reduced and oxidized thioredoxin from Ehrlichia chaffeensis (Ec-Trx, ECH_0218), the etiological agent responsible for human monocytic ehrlichiosis, are described. The overall topology of the calculated structures is similar in both redox states and is similar to those of other thioredoxins: a five-stranded, mixed ß-sheet (ß1-ß3-ß2-ß4-ß5) surrounded by four α-helices. Unlike other thioredoxins studied by NMR in both redox states, the 1H-15N HSQC spectrum of reduced Ec-Trx was missing eight additional amide cross peaks relative to the spectrum of oxidized Ec-Trx. These missing amides correspond to residues Cys35-Glu39 in the active-site-containing helix (α2) and Ser72-Ile75 in a loop near the active site, and suggest a change in backbone dynamics on the millisecond-to-microsecond timescale associated with the breakage of an intramolecular Cys32-Cys35 disulfide bond in a thioredoxin. A consequence of the missing amide resonances is the absence of observable or unambiguous NOEs to provide the distance restraints necessary to define the N-terminal end of the α-helix containing the CPGC active site in the reduced state. This region adopts a well defined α-helical structure in other reported reduced thioredoxin structures, is mostly helical in oxidized Ec-Trx and CD studies of Ec-Trx in both redox states suggests there is no significant difference in the secondary structure of the protein. The NMR solution structure of reduced Ec-Trx illustrates that the absence of canonical structure in a region of a protein may be owing to unfavorable dynamics prohibiting NOE observations or unambiguous NOE assignments.


Assuntos
Proteínas de Bactérias/química , Ehrlichia chaffeensis/química , Tiorredoxinas/química , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Dicroísmo Circular , Sequência Conservada , Ehrlichia chaffeensis/genética , Humanos , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Oxirredução , Prolina/química , Estabilidade Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homologia de Sequência de Aminoácidos , Soluções , Termodinâmica , Tiorredoxinas/genética
2.
Bioorg Med Chem Lett ; 25(24): 5699-704, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26584881

RESUMO

The fragment FOL7185 (compound 17) was found to be a hit against IspD and IspE enzymes isolated from bacteria, and a series of analogs containing the pyrazolopyrimidine core were synthesized. The majority of these compounds inhibited the growth of Burkholderia thailandensis (Bt) and Pseudomonas aeruginosa (Pa) in the Kirby­Bauer disk diffusion susceptibility test. Compound 29 shows inhibitory activity at 0.1 mM (32.2 lg/mL), which is comparable to the control compound kanamycin (48.5 lg/mL). Compound 29 also shows inhibitory activity at 0.5 mM against kanamycin resistant P. aeruginosa. Saturation transfer difference NMR (STD-NMR) screening of these compounds against BtIspD and BtIspE indicated that most of these compounds significantly interact with BtIspE, suggesting that the compounds may inhibit the growth of Bt by disrupting isoprenoid biosynthesis. Ligand epitope mapping of compound 29 with BtIspE indicated that hydrogens on 2,4-dichlorophenyl group have higher proximity to the surface of the enzyme than hydrogens on the pyrazolopyrimidine ring.


Assuntos
Antibacterianos/síntese química , Pirazóis/química , Piridinas/química , Antibacterianos/química , Antibacterianos/farmacologia , Burkholderia/efeitos dos fármacos , Espectroscopia de Ressonância Magnética , Testes de Sensibilidade Microbiana , Pseudomonas aeruginosa/efeitos dos fármacos , Pirazóis/síntese química , Pirazóis/farmacologia , Piridinas/síntese química , Piridinas/farmacologia , Relação Estrutura-Atividade
3.
Biomol NMR Assign ; 9(2): 381-5, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25957069

RESUMO

Using a deuterated sample, all the observable backbone (1)H(N), (15)N, (13)C(a), and (13)C' chemical shifts for the dimeric, periplasmic sensor domain of the Burkholderia pseudomallei histidine kinase RisS were assigned. Approximately one-fifth of the amide resonances are "missing" in the (1)H-(15)N HSQC spectrum and map primarily onto α-helices at the dimer interface observed in a crystal structure suggesting this region either undergoes intermediate timescale motion (µs-ms) and/or is heterogeneous.


Assuntos
Burkholderia pseudomallei/metabolismo , Ressonância Magnética Nuclear Biomolecular , Proteínas Quinases/química , Multimerização Proteica , Sequência de Aminoácidos , Histidina Quinase , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Espectroscopia de Prótons por Ressonância Magnética
4.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 5): 522-30, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25945704

RESUMO

Cryptosporidiosis is an infectious disease caused by protozoan parasites of the Cryptosporidium genus. Infection is associated with mild to severe diarrhea that usually resolves spontaneously in healthy human adults, but may lead to severe complications in young children and in immunocompromised patients. The genome of C. parvum contains a gene, CUTA_CRYPI, that may play a role in regulating the intracellular concentration of copper, which is a toxic element in excess. Here, the crystal structure of this CutA1 protein, Cp-CutA1, is reported at 2.0 Šresolution. As observed for other CutA1 structures, the 117-residue protein is a trimer with a core ferrodoxin-like fold. Circular dichroism spectroscopy shows little, in any, unfolding of Cp-CutA1 up to 353 K. This robustness is corroborated by (1)H-(15)N HSQC spectra at 333 K, which are characteristic of a folded protein, suggesting that NMR spectroscopy may be a useful tool to further probe the function of the CutA1 proteins. While robust, Cp-CutA1 is not as stable as the homologous protein from a hyperthermophile, perhaps owing to a wide ß-bulge in ß2 that protrudes Pro48 and Ser49 outside the ß-sheet.


Assuntos
Cátions Bivalentes/química , Criptosporidiose/genética , Cryptosporidium parvum/química , Cryptosporidium parvum/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Sequência de Aminoácidos , Animais , Cristalização , Humanos , Dados de Sequência Molecular , Parasitos/química , Parasitos/genética , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
5.
Tuberculosis (Edinb) ; 95(2): 142-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25613812

RESUMO

High-resolution three-dimensional structures of essential Mycobacterium tuberculosis (Mtb) proteins provide templates for TB drug design, but are available for only a small fraction of the Mtb proteome. Here we evaluate an intra-genus "homolog-rescue" strategy to increase the structural information available for TB drug discovery by using mycobacterial homologs with conserved active sites. Of 179 potential TB drug targets selected for x-ray structure determination, only 16 yielded a crystal structure. By adding 1675 homologs from nine other mycobacterial species to the pipeline, structures representing an additional 52 otherwise intractable targets were solved. To determine whether these homolog structures would be useful surrogates in TB drug design, we compared the active sites of 106 pairs of Mtb and non-TB mycobacterial (NTM) enzyme homologs with experimentally determined structures, using three metrics of active site similarity, including superposition of continuous pharmacophoric property distributions. Pair-wise structural comparisons revealed that 19/22 pairs with >55% overall sequence identity had active site Cα RMSD <1 Å, >85% side chain identity, and ≥80% PSAPF (similarity based on pharmacophoric properties) indicating highly conserved active site shape and chemistry. Applying these results to the 52 NTM structures described above, 41 shared >55% sequence identity with the Mtb target, thus increasing the effective structural coverage of the 179 Mtb targets over three-fold (from 9% to 32%). The utility of these structures in TB drug design can be tested by designing inhibitors using the homolog structure and assaying the cognate Mtb enzyme; a promising test case, Mtb cytidylate kinase, is described. The homolog-rescue strategy evaluated here for TB is also generalizable to drug targets for other diseases.


Assuntos
Antituberculosos/farmacologia , Desenho de Fármacos , Terapia de Alvo Molecular/métodos , Mycobacterium tuberculosis/efeitos dos fármacos , Antituberculosos/química , Proteínas de Bactérias/química , Biologia Computacional/métodos , Cristalografia por Raios X/métodos , Bases de Dados de Proteínas , Ativação Enzimática , Genômica/métodos , Humanos , Modelos Moleculares , Mycobacterium/classificação , Mycobacterium/enzimologia , Mycobacterium/genética , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/genética , Relação Quantitativa Estrutura-Atividade , Especificidade da Espécie
7.
J Struct Funct Genomics ; 14(2): 47-57, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23709284

RESUMO

Macrophage migration inhibitory factor (MIF) is a eukaryotic cytokine that affects a broad spectrum of immune responses and its activation/inactivation is associated with numerous diseases. During protozoan infections MIF is not only expressed by the host, but, has also been observed to be expressed by some parasites and released into the host. To better understand the biological role of parasitic MIF proteins, the crystal structure of the MIF protein from Giardia lamblia (Gl-MIF), the etiological agent responsible for giardiasis, has been determined at 2.30 Å resolution. The 114-residue protein adopts an α/ß fold consisting of a four-stranded ß-sheet with two anti-parallel α-helices packed against a face of the ß-sheet. An additional short ß-strand aligns anti-parallel to ß4 of the ß-sheet in the adjacent protein unit to help stabilize a trimer, the biologically relevant unit observed in all solved MIF crystal structures to date, and form a discontinuous ß-barrel. The structure of Gl-MIF is compared to the MIF structures from humans (Hs-MIF) and three Plasmodium species (falciparum, berghei, and yoelii). The structure of all five MIF proteins are generally similar with the exception of a channel that runs through the center of each trimer complex. Relative to Hs-MIF, there are differences in solvent accessibility and electrostatic potential distribution in the channel of Gl-MIF and the Plasmodium-MIFs due primarily to two "gate-keeper" residues in the parasitic MIFs. For the Plasmodium MIFs the gate-keeper residues are at positions 44 (Y --> R) and 100 (V --> D) and for Gl-MIF it is at position 100 (V --> R). If these gate-keeper residues have a biological function and contribute to the progression of parasitemia they may also form the basis for structure-based drug design targeting parasitic MIF proteins.


Assuntos
Giardia lamblia/metabolismo , Fatores Inibidores da Migração de Macrófagos/química , Proteínas de Protozoários/química , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Oxirredutases Intramoleculares/química , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Plasmodium , Conformação Proteica , Proteínas de Protozoários/metabolismo
8.
J Clin Invest ; 122(6): 2301-5, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22565309

RESUMO

Effective control and eradication of malaria will require new tools to prevent transmission. Current antimalarial therapies targeting the asexual stage of Plasmodium do not prevent transmission of circulating gametocytes from infected humans to mosquitoes. Here, we describe a new class of transmission-blocking compounds, bumped kinase inhibitors (BKIs), which inhibit microgametocyte exflagellation. Oocyst formation and sporozoite production, necessary for transmission to mammals, were inhibited in mosquitoes fed on either BKI-1-treated human blood or mice treated with BKI-1. BKIs are hypothesized to act via inhibition of Plasmodium calcium-dependent protein kinase 4 and predicted to have little activity against mammalian kinases. Our data show that BKIs do not inhibit proliferation of mammalian cell lines and are well tolerated in mice. Used in combination with drugs active against asexual stages of Plasmodium, BKIs could prove an important tool for malaria control and eradication.


Assuntos
Anopheles/parasitologia , Quinase 2 de Adesão Focal/antagonistas & inibidores , Malária Falciparum , Plasmodium berghei/enzimologia , Plasmodium falciparum/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas de Protozoários/antagonistas & inibidores , Animais , Controle de Doenças Transmissíveis/métodos , Quinase 2 de Adesão Focal/metabolismo , Humanos , Malária Falciparum/enzimologia , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Camundongos
9.
J Med Chem ; 55(6): 2803-10, 2012 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-22369268

RESUMO

Diseases caused by the apicomplexan protozoans Toxoplasma gondii and Cryptosporidium parvum are a major health concern. The life cycle of these parasites is regulated by a family of calcium-dependent protein kinases (CDPKs) that have no direct homologues in the human host. Fortuitously, CDPK1 from both parasites contains a rare glycine gatekeeper residue adjacent to the ATP-binding pocket. This has allowed creation of a series of C3-substituted pyrazolopyrimidine compounds that are potent inhibitors selective for CDPK1 over a panel of human kinases. Here we demonstrate that selectivity is further enhanced by modification of the scaffold at the C1 position. The explanation for this unexpected result is provided by crystal structures of the inhibitors bound to CDPK1 and the human kinase c-SRC. Furthermore, the insight gained from these studies was applied to transform an alternative ATP-competitive scaffold lacking potency and selectivity for CDPK1 into a low nanomolar inhibitor of this enzyme with no activity against SRC.


Assuntos
Antiprotozoários/síntese química , Benzimidazóis/síntese química , Cálcio/fisiologia , Cryptosporidium parvum/enzimologia , Modelos Moleculares , Inibidores de Proteínas Quinases/síntese química , Proteínas Quinases/química , Proteínas de Protozoários/antagonistas & inibidores , Toxoplasma/enzimologia , Antiprotozoários/química , Benzimidazóis/química , Humanos , Estrutura Molecular , Inibidores de Proteínas Quinases/química , Proteínas de Protozoários/química , Relação Estrutura-Atividade
10.
PLoS One ; 7(12): e52482, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23285060

RESUMO

Insoluble recombinant proteins are a major issue for both structural genomics and enzymology research. Greater than 30% of recombinant proteins expressed in Escherichia coli (E. coli) appear to be insoluble. The prevailing view is that insolubly expressed proteins cannot be easily solubilized, and are usually sequestered into inclusion bodies. However, we hypothesize that small molecules added during the cell lysis stage can yield soluble protein from insoluble protein previously screened without additives or ligands. We present a novel screening method that utilized 144 additive conditions to increase the solubility of recombinant proteins expressed in E. coli. These selected additives are natural ligands, detergents, salts, buffers, and chemicals that have been shown to increase the stability of proteins in vivo. We present the methods used for this additive solubility screen and detailed results for 41 potential drug target recombinant proteins from infectious organisms. Increased solubility was observed for 80% of the recombinant proteins during the primary and secondary screening of lysis with the additives; that is 33 of 41 target proteins had increased solubility compared with no additive controls. Eleven additives (trehalose, glycine betaine, mannitol, L-Arginine, potassium citrate, CuCl(2), proline, xylitol, NDSB 201, CTAB and K(2)PO(4)) solubilized more than one of the 41 proteins; these additives can be easily screened to increase protein solubility. Large-scale purifications were attempted for 15 of the proteins using the additives identified and eight (40%) were prepared for crystallization trials during the first purification attempt. Thus, this protocol allowed us to recover about a third of seemingly insoluble proteins for crystallography and structure determination. If recombinant proteins are required in smaller quantities or less purity, the final success rate may be even higher.


Assuntos
Excipientes/farmacologia , Proteínas Recombinantes/metabolismo , Soluções Tampão , Fracionamento Celular , Eletroforese em Gel de Poliacrilamida , Excipientes/química , Genômica , Modelos Biológicos , Proteínas de Protozoários/isolamento & purificação , Solubilidade/efeitos dos fármacos
11.
Artigo em Inglês | MEDLINE | ID: mdl-21904041

RESUMO

Despite recent advances, the expression of heterologous proteins in Escherichia coli for crystallization remains a nontrivial challenge. The present study investigates the efficacy of maltose-binding protein (MBP) fusion as a general strategy for rescuing the expression of target proteins. From a group of sequence-verified clones with undetectable levels of protein expression in an E. coli T7 expression system, 95 clones representing 16 phylogenetically diverse organisms were selected for recloning into a chimeric expression vector with an N-terminal histidine-tagged MBP. PCR-amplified inserts were annealed into an identical ligation-independent cloning region in an MBP-fusion vector and were analyzed for expression and solubility by high-throughput nickel-affinity binding. This approach yielded detectable expression of 72% of the clones; soluble expression was visible in 62%. However, the solubility of most proteins was marginal to poor upon cleavage of the MBP tag. This study offers large-scale evidence that MBP can improve the soluble expression of previously non-expressing proteins from a variety of eukaryotic and prokaryotic organisms. While the behavior of the cleaved proteins was disappointing, further refinements in MBP tagging may permit the more widespread use of MBP-fusion proteins in crystallographic studies.


Assuntos
Escherichia coli/metabolismo , Proteínas Ligantes de Maltose/isolamento & purificação , Expressão Gênica , Proteínas Ligantes de Maltose/genética , Proteínas Ligantes de Maltose/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
12.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 9): 1044-50, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21904048

RESUMO

Burkholderia pseudomallei is a soil-dwelling bacterium endemic to Southeast Asia and Northern Australia. Burkholderia is responsible for melioidosis, a serious infection of the skin. The enzyme 2,3-bisphosphoglycerate-dependent phosphoglycerate mutase (PGAM) catalyzes the interconversion of 3-phosphoglycerate and 2-phosphoglycerate, a key step in the glycolytic pathway. As such it is an extensively studied enzyme and X-ray crystal structures of PGAM enzymes from multiple species have been elucidated. Vanadate is a phosphate mimic that is a powerful tool for studying enzymatic mechanisms in phosphoryl-transfer enzymes such as phosphoglycerate mutase. However, to date no X-ray crystal structures of phosphoglycerate mutase have been solved with vanadate acting as a substrate mimic. Here, two vanadate complexes together with an ensemble of substrate and fragment-bound structures that provide a comprehensive picture of the function of the Burkholderia enzyme are reported.


Assuntos
Burkholderia pseudomallei/enzimologia , Fosfoglicerato Mutase/química , Cristalografia por Raios X , Modelos Moleculares , Fosfoglicerato Mutase/metabolismo , Estrutura Terciária de Proteína , Especificidade por Substrato
13.
Artigo em Inglês | MEDLINE | ID: mdl-21904051

RESUMO

Glutaric acidemia type 1 is an inherited metabolic disorder which can cause macrocephaly, muscular rigidity, spastic paralysis and other progressive movement disorders in humans. The defects in glutaryl-CoA dehydrogenase (GCDH) associated with this disease are thought to increase holoenzyme instability and reduce cofactor binding. Here, the first structural analysis of a GCDH enzyme in the absence of the cofactor flavin adenine dinucleotide (FAD) is reported. The apo structure of GCDH from Burkholderia pseudomallei reveals a loss of secondary structure and increased disorder in the FAD-binding pocket relative to the ternary complex of the highly homologous human GCDH. After conducting a fragment-based screen, four small molecules were identified which bind to GCDH from B. pseudomallei. Complex structures were determined for these fragments, which cause backbone and side-chain perturbations to key active-site residues. Structural insights from this investigation highlight differences from apo GCDH and the utility of small-molecular fragments as chemical probes for capturing alternative conformational states of preformed protein crystals.


Assuntos
Burkholderia pseudomallei/enzimologia , Glutaril-CoA Desidrogenase/química , Apoenzimas/química , Domínio Catalítico , Cristalografia por Raios X , Humanos , Modelos Moleculares , Filogenia , Estrutura Quaternária de Proteína , Homologia Estrutural de Proteína
14.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 9): 1078-83, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21904053

RESUMO

Cat scratch fever (also known as cat scratch disease and bartonellosis) is an infectious disease caused by the proteobacterium Bartonella henselae following a cat scratch. Although the infection usually resolves spontaneously without treatment in healthy adults, bartonellosis may lead to severe complications in young children and immunocompromised patients, and there is new evidence suggesting that B. henselae may be associated with a broader range of clinical symptoms then previously believed. The genome of B. henselae contains genes for two putative Nudix hydrolases, BH02020 and BH01640 (KEGG). Nudix proteins play an important role in regulating the intracellular concentration of nucleotide cofactors and signaling molecules. The amino-acid sequence of BH02020 is similar to that of the prototypical member of the Nudix superfamily, Escherichia coli MutT, a protein that is best known for its ability to neutralize the promutagenic compound 7,8-dihydro-8-oxoguanosine triphosphate. Here, the crystal structure of BH02020 (Bh-MutT) in the Mg(2+)-bound state was determined at 2.1 Å resolution (PDB entry 3hhj). As observed in all Nudix hydrolase structures, the α-helix of the highly conserved `Nudix box' in Bh-MutT is one of two helices that sandwich a four-stranded mixed ß-sheet with the central two ß-strands parallel to each other. The catalytically essential divalent cation observed in the Bh-MutT structure, Mg(2+), is coordinated to the side chains of Glu57 and Glu61. The structure is not especially robust; a temperature melt obtained using circular dichroism spectroscopy shows that Bh-MutT irreversibly unfolds and precipitates out of solution upon heating, with a T(m) of 333 K.


Assuntos
Bartonella henselae/enzimologia , Magnésio/química , Pirofosfatases/química , Magnésio/metabolismo , Modelos Moleculares , Ligação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Pirofosfatases/metabolismo , Homologia Estrutural de Proteína , Nudix Hidrolases
15.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 9): 1129-36, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21904062

RESUMO

Brucella melitensis is the etiological agent responsible for brucellosis. Present in the B. melitensis genome is a 116-residue protein related to arsenate reductases (Bm-YffB; BR0369). Arsenate reductases (ArsC) convert arsenate ion (H(2)AsO(4)(-)), a compound that is toxic to bacteria, to arsenite ion (AsO(2)(-)), a product that may be efficiently exported out of the cell. Consequently, Bm-YffB is a potential drug target because if arsenate reduction is the protein's major biological function then disabling the cell's ability to reduce arsenate would make these cells more sensitive to the deleterious effects of arsenate. Size-exclusion chromatography and NMR spectroscopy indicate that Bm-YffB is a monomer in solution. The solution structure of Bm-YffB (PDB entry 2kok) shows that the protein consists of two domains: a four-stranded mixed ß-sheet flanked by two α-helices on one side and an α-helical bundle. The α/ß domain is characteristic of the fold of thioredoxin-like proteins and the overall structure is generally similar to those of known arsenate reductases despite the marginal sequence similarity. Chemical shift perturbation studies with (15)N-labeled Bm-YffB show that the protein binds reduced glutathione at a site adjacent to a region similar to the HX(3)CX(3)R catalytic sequence motif that is important for arsenic detoxification activity in the classical arsenate-reductase family of proteins. The latter observation supports the hypothesis that the ArsC-YffB family of proteins may function as glutathione-dependent thiol reductases. However, comparison of the structure of Bm-YffB with the structures of proteins from the classical ArsC family suggest that the mechanism and possibly the function of Bm-YffB and other related proteins (ArsC-YffB) may differ from those of the ArsC family of proteins.


Assuntos
Proteínas de Bactérias/química , Brucella melitensis/química , Cristalografia por Raios X , Modelos Moleculares , Estrutura Terciária de Proteína , Homologia Estrutural de Proteína
16.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 9): 1148-53, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21904065

RESUMO

Owing to the evolution of multi-drug-resistant and extremely drug-resistant Mycobacterium tuberculosis strains, there is an urgent need to develop new antituberculosis strategies to prevent TB epidemics in the industrial world. Among the potential new drug targets are two small nonheme iron-binding proteins, rubredoxin A (Rv3251c) and rubredoxin B (Rv3250c), which are believed to play a role in electron-transfer processes. Here, the solution structure and biophysical properties of one of these two proteins, rubredoxin B (Mt-RubB), determined in the zinc-substituted form are reported. The zinc-substituted protein was prepared by expressing Mt-RubB in minimal medium containing excess zinc acetate. Size-exclusion chromatography and NMR spectroscopy indicated that Mt-RubB was a monomer in solution. The structure (PDB entry 2kn9) was generally similar to those of other rubredoxins, containing a three-stranded antiparallel ß-sheet (ß2-ß1-ß3) and a metal tetrahedrally coordinated to the S atoms of four cysteine residues (Cys9, Cys12, Cys42 and Cys45). The first pair of cysteine residues is at the C-terminal end of the first ß-strand and the second pair of cysteine residues is towards the C-terminal end of the loop between ß2 and ß3. The structure shows the metal buried deeply within the protein, an observation that is supported by the inability to remove the metal with excess EDTA at room temperature. Circular dichroism spectroscopy shows that this stability extends to high temperature, with essentially no change being observed in the CD spectrum of Mt-RubB upon heating to 353 K.


Assuntos
Mycobacterium tuberculosis/química , Rubredoxinas/química , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Estabilidade Proteica , Estrutura Terciária de Proteína
17.
PLoS One ; 5(9)2010 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-20862217

RESUMO

BACKGROUND: Pathogenic bacteria adhere to the host cell surface using a family of outer membrane proteins called Trimeric Autotransporter Adhesins (TAAs). Although TAAs are highly divergent in sequence and domain structure, they are all conceptually comprised of a C-terminal membrane anchoring domain and an N-terminal passenger domain. Passenger domains consist of a secretion sequence, a head region that facilitates binding to the host cell surface, and a stalk region. METHODOLOGY/PRINCIPAL FINDINGS: Pathogenic species of Burkholderia contain an overabundance of TAAs, some of which have been shown to elicit an immune response in the host. To understand the structural basis for host cell adhesion, we solved a 1.35 A resolution crystal structure of a BpaA TAA head domain from Burkholderia pseudomallei, the pathogen that causes melioidosis. The structure reveals a novel fold of an intricately intertwined trimer. The BpaA head is composed of structural elements that have been observed in other TAA head structures as well as several elements of previously unknown structure predicted from low sequence homology between TAAs. These elements are typically up to 40 amino acids long and are not domains, but rather modular structural elements that may be duplicated or omitted through evolution, creating molecular diversity among TAAs. CONCLUSIONS/SIGNIFICANCE: The modular nature of BpaA, as demonstrated by its head domain crystal structure, and of TAAs in general provides insights into evolution of pathogen-host adhesion and may provide an avenue for diagnostics.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Burkholderia pseudomallei/química , Adesinas Bacterianas , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/fisiologia , Cristalografia por Raios X , Humanos , Melioidose/microbiologia , Conformação Molecular , Dados de Sequência Molecular , Multimerização Proteica , Estrutura Terciária de Proteína
18.
Biomol NMR Assign ; 3(2): 163-6, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19888681

RESUMO

Burkholderia pseudomallei is a NIAID Category B microorganism responsible for melioidosis. Here we report backbone and side chain NMR assignments for the 139-residue, homodimeric, organic hydroperoxide resistance protein (Ohr) from this organism.


Assuntos
Proteínas de Bactérias/química , Burkholderia , Ressonância Magnética Nuclear Biomolecular , Estrutura Secundária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA