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










Publication year range
1.
Genes Environ ; 46(1): 11, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38659010

ABSTRACT

BACKGROUND: An in vitro micronucleus assay is a standard genotoxicity test. Although the technique and interpretation of the results are simple, manual counting of the total and micronucleus-containing cells in a microscopic field is tedious. To address this issue, several systems have been developed for quick and efficient micronucleus counting, including flow cytometry and automated detection based on specialized software and detection systems that analyze images. RESULTS: Here, we present a simple and effective method for automated micronucleus counting using image recognition technology. Our process involves separating the RGB channels in a color micrograph of cells stained with acridine orange. The cell nuclei and micronuclei were detected by scaling the G image, whereas the cytoplasm was recognized from a composite image of the R and G images. Finally, we identified cells with overlapping cytoplasm and micronuclei as micronucleated cells, and the application displayed the number of micronucleated cells and the total number of cells. Our method yielded results that were comparable to manually measured values. CONCLUSIONS: Our micronucleus detection (MN/cell detection software) system can accurately detect the total number of cells and micronucleus-forming cells in microscopic images with the same level of precision as achieved through manual counting. The accuracy of micronucleus numbers depends on the cell staining conditions; however, the software has options by which users can easily manually optimize parameters such as threshold, denoise, and binary to achieve the best results. The optimization process is easy to handle and requires less effort, making it an efficient way to obtain accurate results.

2.
Front Mol Biosci ; 10: 1091677, 2023.
Article in English | MEDLINE | ID: mdl-36845542

ABSTRACT

Protein folding is often hampered by intermolecular protein aggregation, which can be prevented by a variety of chaperones in the cell. Bacterial chaperonin GroEL is a ring-shaped chaperone that forms complexes with its cochaperonin GroES, creating central cavities to accommodate client proteins (also referred as substrate proteins) for folding. GroEL and GroES (GroE) are the only indispensable chaperones for bacterial viability, except for some species of Mollicutes such as Ureaplasma. To understand the role of chaperonins in the cell, one important goal of GroEL research is to identify a group of obligate GroEL/GroES clients. Recent advances revealed hundreds of in vivo GroE interactors and obligate chaperonin-dependent clients. This review summarizes the progress on the in vivo GroE client repertoire and its features, mainly for Escherichia coli GroE. Finally, we discuss the implications of the GroE clients for the chaperone-mediated buffering of protein folding and their influences on protein evolution.

3.
Microscopy (Oxf) ; 70(3): 289-296, 2021 Jun 06.
Article in English | MEDLINE | ID: mdl-33173948

ABSTRACT

Escherichia coli chaperonin GroEL, which is a large cylindrical protein complex comprising two heptameric rings with cavities of 4.5 nm each in the center, assists in intracellular protein folding with the aid of GroES and adenosine triphosphate (ATP). Here, we investigated the possibility that GroEL can also encapsulate metal nanoparticles (NPs) up to ∼5 nm in diameter into the cavities with the aid of GroES and ATP. The slow ATP-hydrolyzing GroELD52A/D398A mutant, which forms extremely stable complexes with GroES (half-time of ∼6 days), made it possible to analyze GroEL/GroES complexes containing metal NPs. Scanning transmission electron microscopy-energy-dispersive X-ray spectroscopy analysis proved distinctly that FePt NPs and Au NPs were encapsulated in the GroEL/GroES complexes. Dynamic light scattering measurements showed that the NPs in the GroEL/GroES complex were able to maintain their dispersibility in solution. We previously described that the incubation of GroEL and GroES in the presence of ATP·BeFx and adenosine diphosphate·BeFx resulted in the formation of symmetric football-shaped and asymmetric bullet-shaped complexes, respectively. Based on this knowledge, we successfully constructed the football-shaped complex in which two compartments were occupied by Pt or Au NPs (first compartment) and FePt NPs (second compartment). This study showed that metal NPs were sequentially encapsulated according to the GroEL reaction in a step-by-step manner. In light of these results, chaperonin can be used as a tool for handling nanomaterials.


Subject(s)
Chaperonin 10/chemistry , Chaperonin 10/metabolism , Chaperonin 60/chemistry , Chaperonin 60/metabolism , Escherichia coli/chemistry , Escherichia coli/metabolism , Metal Nanoparticles/chemistry , Adenosine Triphosphate/metabolism , Chaperonin 60/genetics , Chaperonins/chemistry , Chaperonins/genetics , Chaperonins/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gold/chemistry , Gold/metabolism , Iron/chemistry , Iron/metabolism , Microscopy, Electron, Scanning Transmission , Microscopy, Electron, Transmission , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Platinum/chemistry , Platinum/metabolism , Protein Binding , Protein Folding
4.
J Infect Chemother ; 21(1): 70-3, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25305808

ABSTRACT

Linezolid pharmacokinetic profile in pediatric patients has not been fully characterized, and the dose needed to achieve a pharmacokinetic-pharmacodynamic (PK-PD) target has yet to be established because its efficacy is associated with the area under the plasma drug concentration-time curve (AUC24)/minimum inhibitory concentration (MIC) ratio. The present study aimed to define the pharmacokinetic parameters of intravenous linezolid in pediatric patients and assess the rationale for the approved dosage recommendations. Linezolid was safe, tolerated well, and clinically effective for treating Gram-positive bacteria in five pediatric patients (3-11 years). The mean values for the volume of distribution and total clearance (CL) in a one-compartment model were estimated to be 0.646 ± 0.239 l/kg and 0.171 ± 0.068 l/h/kg, respectively (mean ± S.D.). Based on this analysis, the AUC24 and trough drug concentration in plasma (C(min)) for linezolid doses were predicted to be 175.4 µg h/ml and 3.4 µg/ml for 30 mg/kg/day, 204.7 µg h/ml and 4.3 µg/ml for 35 mg/kg/day, and 263.2 µg h/ml and 6.2 µg/ml for 45 mg/kg/day, respectively. Taking into account that AUC24 should be ≥ 200 µg h/ml for MIC of 2.0 µg/ml (to achieve an AUC24/MIC ratio of ≥ 100) and C(min) should be approximately 7 µg/ml (to avoid thrombocytopenia), we consider the approved dosage of 30 mg/kg/day to be fundamentally rational, but can be underdosed against bacteria with MIC of 2.0 µg/ml; therefore, a dose of 35-45 mg/kg/day is more appropriate to ensure the efficacy and safety of linezolid in pediatric patients.


Subject(s)
Acetamides/administration & dosage , Acetamides/pharmacokinetics , Oxazolidinones/administration & dosage , Oxazolidinones/pharmacokinetics , Acetamides/therapeutic use , Child , Child, Preschool , Female , Glomerular Filtration Rate , Humans , Linezolid , Male , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Oxazolidinones/therapeutic use , Staphylococcal Infections/drug therapy
5.
J Mol Biol ; 426(21): 3634-41, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25174333

ABSTRACT

The chaperonin GroEL is an essential chaperone that assists in protein folding with the aid of GroES and ATP. GroEL forms a double-ring structure, and both rings can bind GroES in the presence of ATP. Recent progress on the GroEL mechanism has revealed the importance of a symmetric 1:2 GroEL:GroES2 complex (the "football"-shaped complex) as a critical intermediate during the functional GroEL cycle. We determined the crystal structure of the football GroEL:GroES2-ATP14 complex from Escherichia coli at 3.8Å, using a GroEL mutant that is extremely defective in ATP hydrolysis. The overall structure of the football complex resembled the GroES-bound GroEL ring of the asymmetric 1:1 GroEL:GroES complex (the "bullet" complex). However, the two GroES-bound GroEL rings form a modified interface by an ~7° rotation about the 7-fold axis. As a result, the inter-ring contacts between the two GroEL rings in the football complex differed from those in the bullet complex. The differences provide a structural basis for the apparently impaired inter-ring negative cooperativity observed in several biochemical analyses.


Subject(s)
Adenosine Triphosphate/chemistry , Chaperonin 10/chemistry , Chaperonin 60/chemistry , Crystallography, X-Ray/methods , Escherichia coli/chemistry , Crystallization , Hydrolysis , Models, Molecular , Mutation , Nanotechnology , Protein Binding , Protein Folding , Protein Structure, Tertiary
6.
J Biol Chem ; 289(43): 30005-11, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25202010

ABSTRACT

The Escherichia coli chaperonin GroEL is a double-ring chaperone that assists protein folding with the aid of GroES and ATP. Asp-398 in GroEL is known as one of the critical residues on ATP hydrolysis because GroEL(D398A) mutant is deficient in ATP hydrolysis (<2% of the wild type) but not in ATP binding. In the archaeal Group II chaperonin, another aspartate residue, Asp-52 in the corresponding E. coli GroEL, in addition to Asp-398 is also important for ATP hydrolysis. We investigated the role of Asp-52 in GroEL and found that ATPase activity of GroEL(D52A) and GroEL(D52A/D398A) mutants was ∼ 20% and <0.01% of wild-type GroEL, respectively, indicating that Asp-52 in E. coli GroEL is also involved in the ATP hydrolysis. GroEL(D52A/D398A) formed a symmetric football-shaped GroEL-GroES complex in the presence of ATP, again confirming the importance of the symmetric complex during the GroEL ATPase cycle. Notably, the symmetric complex of GroEL(D52A/D398A) was extremely stable, with a half-time of ∼ 150 h (∼ 6 days), providing a good model to characterize the football-shaped complex.


Subject(s)
Adenosine Triphosphate/metabolism , Aspartic Acid/metabolism , Chaperonin 60/metabolism , Escherichia coli/metabolism , Binding Sites , Chaperonin 10/metabolism , Chaperonin 10/ultrastructure , Chaperonin 60/chemistry , Chaperonin 60/ultrastructure , Hydrolysis , Malate Dehydrogenase/metabolism , Mutant Proteins/metabolism , Negative Staining , Protein Folding , Protein Stability , Protein Subunits/chemistry , Protein Subunits/metabolism , Structure-Activity Relationship , Thiosulfate Sulfurtransferase/metabolism
7.
Int J Antimicrob Agents ; 44(3): 242-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108880

ABSTRACT

The pharmacokinetic/pharmacodynamic (PK/PD) index for the efficacy of linezolid is a 24-h area under the plasma drug concentration-time curve (AUC24)/minimum inhibitory concentration (MIC) ratio of ≥100. The main adverse event associated with administration of linezolid is thrombocytopenia. Therefore, the aims of the present study were to define PD thresholds that would minimise linezolid-induced thrombocytopenia and to perform a population PK analysis to identify factors influencing the pharmacokinetics of linezolid. Population PK analysis revealed that creatinine clearance (CLCr) significantly affected linezolid pharmacokinetics: the mean parameter estimate of drug clearance (CL; in L/h)=0.0258 × CLCr + 2.03. A strong correlation (r=0.970) was found between AUC24 and trough plasma concentrations (Cmin) [AUC24=18.2 × Cmin + 134.4]. The Cmin value for AUC24=200 (in the case of MIC=2 µg/mL) was estimated to be 3.6 µg/mL. Regarding safety, Cmin was a significant predictor of thrombocytopenia during treatment, and its threshold to minimise linezolid-induced thrombocytopenia was 8.2 µg/mL. A Kaplan-Meier plot revealed that the median time from initiation of therapy to the development of thrombocytopenia was 15 days. Therefore, the target Cmin range was 3.6-8.2 µg/mL. The following formula to achieve a target Cmin in patients with different degrees of renal function was proposed based on these results: initial daily dose (mg/day)=CL × AUC24=(0.0258 × CLCr + 2.03)×(18.2 × Cmin + 134.4). This recommended initial dosage and subsequent dosage adjustment for the target concentration range should avoid adverse events, thereby enabling effective linezolid-based therapies to be continued.


Subject(s)
Acetamides/adverse effects , Acetamides/pharmacology , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Oxazolidinones/adverse effects , Oxazolidinones/pharmacology , Thrombocytopenia/chemically induced , Acetamides/administration & dosage , Acetamides/pharmacokinetics , Adult , Aged , Aged, 80 and over , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Area Under Curve , Female , Humans , Linezolid , Male , Metabolic Clearance Rate , Microbial Sensitivity Tests , Middle Aged , Models, Statistical , Oxazolidinones/administration & dosage , Oxazolidinones/pharmacokinetics , Plasma/chemistry
8.
Genes Cells ; 16(5): 545-56, 2011 May.
Article in English | MEDLINE | ID: mdl-21453424

ABSTRACT

We report a new type of interaction between two yeast prion proteins, Sup35 and New1. New1 consists of an N-terminal prion region (New1N) and a C-terminal region homologous to a translation elongation factor with two ATP-binding motifs. Amyloid formation of the Sup35 prion region (Sup35NM) was accelerated by a small amount of sonicated New1N amyloid (New1N-seeds) to produce Sup35NM[New1] amyloid. New1N amyloid formation was accelerated by Sup35NM[New1]-seeds but not by spontaneously generated Sup35NM-seeds, indicating that the structural features of the New1N amyloid were transmitted via the Sup35NM amyloid. Surprisingly, full-length New1 broke the Sup35NM amyloid fibrils in an ATP-dependent manner. This activity of New1 was independent from Hsp104. It was lost by a mutation in the second ATP-binding motif, by the truncation of the N-terminal prion region of New1 and by the pre-incubation of New1 with New1N-seeds. When New1 was overproduced in yeast [PSI(+)] cells carrying GFP-fused Sup35NM, diverse morphological changes in fluorescent foci occurred. Thus, New1 potentially has a regulatory role in prion state in yeast, working independently of the Hsp104 system.


Subject(s)
Adenosine Triphosphate/metabolism , Amyloid/metabolism , Peptide Termination Factors/metabolism , Prions/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Sequence , Amyloid/genetics , Asparagine/genetics , Asparagine/metabolism , Binding Sites/genetics , Blotting, Western , Glutamine/genetics , Glutamine/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Mutation , Peptide Termination Factors/genetics , Prions/genetics , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
9.
Int J Antimicrob Agents ; 36(2): 179-81, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20392606

ABSTRACT

The major adverse event associated with linezolid treatment is reversible myelosuppression, mostly thrombocytopenia. Recent studies have reported that the incidence of linezolid-induced thrombocytopenia was higher in patients with renal failure than in patients with normal renal function, although the underlying mechanisms of this toxicity are still unknown. The present study thus aimed to investigate the relationship between renal function and linezolid exposure as well as the effects of drug exposure on thrombocytopenia. A statistically significant (P<0.01) strong correlation (r=0.933) was observed between linezolid clearance and creatinine clearance. A negative correlation (r=-0.567) was also shown between linezolid clearance and blood urea nitrogen, although the correlation was not statistically significant. In thrombocytopenic patients, the trough concentration was 14.4-35.6 mg/L and the area under the plasma linezolid concentration-time curve for 24h (AUC(24h)) was 513.1-994.6 mg h/L; in non-thrombocytopenic patients, drug exposure was relatively low (6.9 mg/L and 7.2mg/L for trough concentration and 294.3 mg h/L and 323.6 mg h/L for AUC(24h)). These results provide a pharmacokinetic explanation for the mechanism of the adverse event that renal dysfunction increased linezolid trough concentration and AUC and that higher drug exposure induced thrombocytopenia.


Subject(s)
Acetamides/adverse effects , Acetamides/pharmacokinetics , Anti-Infective Agents/adverse effects , Anti-Infective Agents/pharmacokinetics , Oxazolidinones/adverse effects , Oxazolidinones/pharmacokinetics , Renal Insufficiency/physiopathology , Thrombocytopenia/chemically induced , Acetamides/administration & dosage , Administration, Oral , Aged , Anti-Infective Agents/administration & dosage , Blood Urea Nitrogen , Creatinine/blood , Female , Humans , Injections, Intravenous , Japan , Linezolid , Male , Oxazolidinones/administration & dosage , Risk Factors
10.
Structure ; 17(2): 287-93, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19217399

ABSTRACT

The chaperonin GroEL interacts with various proteins, leading them to adopt their correct conformations with the aid of GroES and ATP. The actual mechanism is still being debated. In this study, by use of cryo-electron microscopy, we determined the solution structure of the Thermus thermophilus GroEL-GroES complex encapsulating its substrate proteins. We observed the averaged density of substrate proteins in the center of the GroEL-GroES cavity. The position of the averaged substrate density in the cavity suggested a repulsive interaction between a majority of the substrate proteins and the interior wall of the cavity, which is suitable for substrate release. In addition, we observed a distortion of the cis-GroEL ring, especially at the position near the substrate, which indicated that the interaction between the encapsulated proteins and the GroEL ring results in an adjustment in the cavity's shape to accommodate the substrate.


Subject(s)
Chaperonin 10/chemistry , Chaperonin 10/metabolism , Chaperonin 60/chemistry , Chaperonin 60/metabolism , Thermus thermophilus/metabolism , Adenosine Triphosphate/metabolism , Binding Sites/physiology , Cryoelectron Microscopy , Crystallography, X-Ray , Ligands , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism
12.
J Biol Chem ; 283(35): 23774-81, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18567584

ABSTRACT

The Escherichia coli chaperonin GroEL is a double-ring chaperone that assists in protein folding with the aid of GroES and ATP. It is believed that GroEL alternates the folding-active rings and that the substrate protein (and GroES) can bind to the open trans-ring only after ATP in the cis-ring is hydrolyzed. However, we found that a substrate protein prebound to the trans-ring remained bound during the first ATP cycle, and this substrate was assisted by GroEL-GroES when the second cycle began. Moreover, a slow ATP-hydrolyzing GroEL mutant (D398A) in the ATP-bound form bound a substrate protein and GroES to the trans-ring. The apparent discrepancy with the results from an earlier study (Rye, H. S., Roseman, A. M., Chen, S., Furtak, K., Fenton, W. A., Saibil, H. R., and Horwich, A. L. (1999) Cell 97, 325-338) can be explained by the previously unnoticed fact that the ATP-bound form of the D398A mutant exists as a symmetric 1:2 GroEL-GroES complex (the "football"-shaped complex) and that the substrate protein (and GroES) in the medium is incorporated into the complex only after the slow turnover. In light of these results, the current model of the GroEL-GroES reaction cycle via the asymmetric 1:1 GroEL-GroES complex deserves reexamination.


Subject(s)
Chaperonin 10/chemistry , Chaperonin 60/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Multiprotein Complexes/chemistry , Mutation, Missense , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Amino Acid Substitution , Chaperonin 10/genetics , Chaperonin 10/metabolism , Chaperonin 60/genetics , Chaperonin 60/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fluorescence Resonance Energy Transfer , Hydrolysis , Models, Chemical , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Binding/physiology , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
J Biol Chem ; 281(2): 962-7, 2006 Jan 13.
Article in English | MEDLINE | ID: mdl-16239229

ABSTRACT

In the crystal structure of the native GroEL.GroES.substrate protein complex from Thermus thermophilus, one GroEL subunit makes contact with two GroES subunits. One contact is through the H-I helices, and the other is through a novel GXXLE region. The side chain of Leu, in the GXXLE region, forms a hydrophobic cluster with residues of the H helix (Shimamura, T., Koike-Takeshita, A., Yokoyama, K., Masui, R., Murai, N., Yoshida, M., Taguchi, H., and Iwata, S. (2004) Structure (Camb.) 12, 1471-1480). Here, we investigated the functional role of Leu in the GXXLE region, using Escherichia coli GroEL. The results are as follows: (i) cross-linking between introduced cysteines confirmed that the GXXLE region in the E. coli GroEL.GroES complex is also in contact with GroES; (ii) when Leu was replaced by Lys (GroEL(L309K)) or other charged residues, chaperone activity was largely lost; (iii) the GroEL(L309K).substrate complex failed to bind GroES to produce a stable GroEL(L309K).GroES.substrate complex, whereas free GroEL(L309K) bound GroES normally; (iv) the GroEL(L309K).GroES.substrate complex was stabilized with BeF(x), but the substrate protein in the complex was readily digested by protease, indicating that it was not properly encapsulated into the internal cavity of the complex. Thus, conformational communication between the two GroES contact sites, the H helix and the GXXLE region (through Leu(309)), appears to play a critical role in encapsulation of the substrate.


Subject(s)
Chaperonin 60/chemistry , Leucine/chemistry , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Bacterial Proteins/chemistry , Binding Sites , Chaperonin 10/chemistry , Chromatography, Gel , Cross-Linking Reagents/pharmacology , Cysteine/chemistry , Escherichia coli/metabolism , Models, Molecular , Molecular Chaperones/chemistry , Molecular Conformation , Mutation , Peptide Hydrolases/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation , Protein Folding , Protein Structure, Tertiary , Substrate Specificity , Thermus thermophilus/metabolism , Time Factors
14.
J Biol Chem ; 279(44): 45737-43, 2004 Oct 29.
Article in English | MEDLINE | ID: mdl-15347650

ABSTRACT

Coupling with ATP hydrolysis and cooperating with GroES, the double ring chaperonin GroEL assists the folding of other proteins. Here we report novel GroEL-GroES complexes formed in fluoroberyllate (BeF(x)) that can mimic the phosphate part of the enzyme-bound nucleotides. In ATP, BeF(x) stops the functional turnover of GroEL by preventing GroES release and produces a symmetric 1:2 GroEL-GroES complex in which both GroEL rings contain ADP.BeF(x) and an encapsulated substrate protein. In ADP, the substrate protein-loaded GroEL cannot bind GroES. In ADP plus BeF(x), however, it can bind GroES to form a stable 1:1 GroEL-GroES complex in which one of GroEL rings contains ADP.BeF(x) and an encapsulated substrate protein. This 1:1 GroEL-GroES complex is converted into the symmetric 1:2 GroEL-GroES complex when GroES is supplied in ATP plus BeF(x). Thus, BeF(x) stabilizes two GroEL-GroES complexes; one with a single folding chamber and the other with double folding chambers. These results shed light on the intermediate ADP.P(i) nucleotide states in the functional cycle of GroEL.


Subject(s)
Beryllium/pharmacology , Chaperonin 10/metabolism , Chaperonin 60/metabolism , Fluorides/pharmacology , Protein Folding , Adenosine Triphosphate/metabolism , Chaperonin 10/chemistry , Chaperonin 60/chemistry , Hydrolysis
15.
Structure ; 12(8): 1471-80, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15296740

ABSTRACT

The chaperonins GroEL and GroES are essential mediators of protein folding. GroEL binds nonnative protein, ATP, and GroES, generating a ternary complex in which protein folding occurs within the cavity capped by GroES (cis-cavity). We determined the crystal structure of the native GroEL-GroES-ADP homolog from Thermus thermophilus, with substrate proteins in the cis-cavity, at 2.8 A resolution. Twenty-four in vivo substrate proteins within the cis-cavity were identified from the crystals. The structure around the cis-cavity, which encapsulates substrate proteins, shows significant differences from that observed for the substrate-free Escherichia coli GroEL-GroES complex. The apical domain around the cis-cavity of the Thermus GroEL-GroES complex exhibits a large deviation from the 7-fold symmetry. As a result, the GroEL-GroES interface differs considerably from the previously reported E. coli GroEL-GroES complex, including a previously unknown contact between GroEL and GroES.


Subject(s)
Chaperonin 10/chemistry , Chaperonin 60/chemistry , Macromolecular Substances/chemistry , Models, Molecular , Amino Acid Sequence , Crystallography, X-Ray , Escherichia coli/chemistry , Molecular Sequence Data , Sequence Homology, Amino Acid , Thermus thermophilus/chemistry
16.
Mol Cell Biol ; 24(3): 1313-23, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14729975

ABSTRACT

Most cis-acting regulatory elements have generally been assumed to activate a single nearby gene. However, many genes are clustered together, raising the possibility that they are regulated through a common element. We show here that a single peroxisome proliferator response element (PPRE), located between the mouse PEX11 alpha and perilipin genes, confers on both genes activation by peroxisome proliferator-activated receptor alpha (PPAR alpha) and PPAR gamma. A functional PPRE 8.4 kb downstream of the promoter of PEX11 alpha, a PPAR alpha target gene, was identified by a gene transfection study. This PPRE was positioned 1.9 kb upstream of the perilipin gene and also functioned with the perilipin promoter. In addition, this PPRE, when combined with the natural promoters of the PEX11 alpha and perilipin genes, conferred subtype-selective activation by PPAR alpha and PPAR gamma 2. The PPRE sequence specifically bound to the heterodimer of RXR alpha and PPAR alpha or PPAR gamma 2, as assessed by electrophoretic gel mobility shift assays. Furthermore, tissue-selective binding of PPAR alpha and PPAR gamma to the PPRE was demonstrated in hepatocytes and adipocytes, respectively, by chromatin immunoprecipitation assay. Hence, the expression of these genes is induced through the same PPRE in the liver and adipose tissue, where the two PPAR subtypes are specifically expressed.


Subject(s)
Genes, Regulator , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Animals , Base Sequence , Carrier Proteins , Conserved Sequence , Dimerization , Genes, Reporter , Humans , Mice , Molecular Sequence Data , Organ Specificity , Perilipin-1 , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors , Transcription Factors/metabolism , Transfection
17.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 9): 1632-4, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12925795

ABSTRACT

The chaperonin GroEL-GroES (GroEL/ES) complex from a thermophilic eubacteria, Thermus thermophilus HB8, has been purified and crystallized. The GroEL/ES complex is known to be composed of 14 identical GroEL subunits (58 kDa) and seven identical GroES subunits (11 kDa). The GroEL/ES complex crystals belong to the triclinic space group P1, with unit-cell parameters a = 140.4, b = 156.4, c = 273.1 A, alpha = 82.9, beta = 85.4, gamma = 68.5 degrees. The crystal asymmetric unit contains two molecules (MW = 885 kDa). One data set to 3.0 A resolution, with 383 652 independent observations (89.3% complete) and an R(merge) of 0.08, has been collected from a single crystal. A molecular-replacement solution was obtained using the structure of the GroEL/ES complex from Escherichia coli as a search model.


Subject(s)
Chaperonin 10/chemistry , Chaperonin 60/chemistry , Crystallization , Thermus thermophilus/chemistry , Protein Binding , Protein Conformation , X-Ray Diffraction
18.
Biosci Biotechnol Biochem ; 66(7): 1538-45, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12224638

ABSTRACT

A gene (btrC2) encoding the 20-kDa subunit of 2-deoxy-scyllo-inosose (DOI) synthase, a key enzyme in the biosynthesis of 2-deoxystreptamine, was identified from the butirosin-producer Bacillus circulans by reverse genetics. The deduced amino acid sequence of BtrC2 closely resembled that of YaaE of B. subtilis, but the function of the latter has not been known to date. Instead, BtrC2 appeared to show sequence similarity to a certain extent with HisH of B. subtilis, an amidotransferase subunit of imidazole glycerol phosphate synthase. Disruption of btrC2 reduced the growth rate compared with the wild type, and simultaneously antibiotic producing activity was lost. Addition of NH4Cl to the medium complemented only the growth rate of the disruptant, and both the growth rate and antibiotic production were restored by addition of yeast extract. In addition, a heterologous co-expression system of btrC2 with btrC was constructed in Escherichia coli. The simultaneously over-expressed BtrC2 and BtrC constituted a heterodimer, the biochemical features of which resembled those of DOI synthase from B. circulans more than those of the recombinant homodimeric BtrC. Despite the similarity of BtrC2 to HisH the heterodimer showed neither aminotransfer nor amidotransfer activity for 2-deoxy-scyllo-inosose as a substrate. All the observations suggest that BtrC2 is involved not only in the secondary metabolism, but also in the primary metabolism in B. circulans. The function of BtrC2 in the butirosin biosynthesis appears to be indirect, and may be involved in stabilization of DOI synthase and in regulation of its enzyme activity.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Bacillus/genetics , Bacillus/metabolism , Butirosin Sulfate/biosynthesis , Lyases/genetics , Lyases/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Butirosin Sulfate/pharmacology , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Genes, Bacterial/genetics , Kinetics , Lyases/isolation & purification , Microbial Sensitivity Tests , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...