Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters










Publication year range
1.
Mol Microbiol ; 111(4): 918-937, 2019 04.
Article in English | MEDLINE | ID: mdl-30556355

ABSTRACT

In bacteria, disulfide bonds confer stability on many proteins exported to the cell envelope or beyond, including bacterial virulence factors. Thus, proteins involved in disulfide bond formation represent good targets for the development of inhibitors that can act as antibiotics or anti-virulence agents, resulting in the simultaneous inactivation of several types of virulence factors. Here, we present evidence that the disulfide bond forming enzymes, DsbB and VKOR, are required for Pseudomonas aeruginosa pathogenicity and Mycobacterium tuberculosis survival respectively. We also report the results of a HTS of 216,767 compounds tested against P. aeruginosa DsbB1 and M. tuberculosis VKOR using Escherichia coli cells. Since both P. aeruginosa DsbB1 and M. tuberculosis VKOR complement an E. coli dsbB knockout, we screened simultaneously for inhibitors of each complemented E. coli strain expressing a disulfide-bond sensitive ß-galactosidase reported previously. The properties of several inhibitors obtained from these screens suggest they are a starting point for chemical modifications with potential for future antibacterial development.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Disulfides/metabolism , Membrane Proteins/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Pseudomonas aeruginosa/enzymology , Animals , Bacterial Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Female , High-Throughput Screening Assays , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/pathogenicity , Pseudomonas aeruginosa/pathogenicity , Small Molecule Libraries/pharmacology , Virulence , Virulence Factors/metabolism
2.
J Bacteriol ; 200(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29784887

ABSTRACT

Disulfide bonds influence the stability and activity of many proteins. In Escherichia coli, the DsbA and DsbB enzymes promote disulfide bond formation. Other bacteria, including the Actinobacteria, use instead of DsbB the enzyme vitamin K epoxide reductase (VKOR), whose gene is found either fused to or in the same operon as a dsbA-like gene. Mycobacterium tuberculosis and other Gram-positive actinobacteria secrete many proteins with even numbers of cysteines to the cell envelope. These organisms have predicted oxidoreductases and VKOR orthologs. These findings indicate that such bacteria likely form disulfide bonds in the cell envelope. The M. tuberculosisvkor gene complements an E. colidsbB deletion strain, restoring the oxidation of E. coli DsbA. While we have suggested that the dsbA gene linked to the vkor gene may express VKOR's partner in mycobacteria, others have suggested that two other extracytoplasmic oxidoreductases (DsbE or DsbF) may be catalysts of protein disulfide bond formation. However, there is no direct evidence for interactions of VKOR with either DsbA, DsbE, or DsbF. To identify the actual substrate of VKOR, we identified two additional predicted extracytoplasmic DsbA-like proteins using bioinformatics analysis of the M. tuberculosis genome. Using the five potential DsbAs, we attempted to reconstitute disulfide bond pathways in E. coli and in Mycobacterium smegmatis, a close relative of M. tuberculosis Our results show that only M. tuberculosis DsbA is oxidized by VKOR. Comparison of the properties of dsbA- and vkor-null mutants in M. smegmatis shows parallels to the properties of dsb mutations in E. coliIMPORTANCE Disulfide bond formation has a great impact on bacterial pathogenicity. Thus, disulfide-bond-forming proteins represent new targets for the development of antibacterials, since the inhibition of disulfide bond formation would result in the simultaneous loss of the activity of several classes of virulence factors. Here, we identified five candidate proteins encoded by the M. tuberculosis genome as possible substrates of the M. tuberculosis VKOR protein involved in disulfide bond formation. We then reconstituted the mycobacterial disulfide bond formation pathway in E. coli and showed that of the five candidates, only M. tuberculosis DsbA is efficiently oxidized by VKOR in E. coli We also present evidence for the involvement of VKOR in DsbA oxidation in M. smegmatis.


Subject(s)
Bacterial Proteins/genetics , Disulfides/metabolism , Mycobacterium tuberculosis/genetics , Thioredoxins/metabolism , Vitamin K Epoxide Reductases/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Models, Molecular , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/pathogenicity , Oxidation-Reduction , Oxidoreductases , Protein Disulfide-Isomerases/metabolism , Vitamin K Epoxide Reductases/metabolism
3.
Nat Microbiol ; 3(3): 270-280, 2018 03.
Article in English | MEDLINE | ID: mdl-29463925

ABSTRACT

Interest in protein disulfide bond formation has recently increased because of the prominent role of disulfide bonds in bacterial virulence and survival. The first discovered pathway that introduces disulfide bonds into cell envelope proteins consists of Escherichia coli enzymes DsbA and DsbB. Since its discovery, variations on the DsbAB pathway have been found in bacteria and archaea, probably reflecting specific requirements for survival in their ecological niches. One variation found amongst Actinobacteria and Cyanobacteria is the replacement of DsbB by a homologue of human vitamin K epoxide reductase. Many Gram-positive bacteria express enzymes involved in disulfide bond formation that are similar, but non-homologous, to DsbAB. While bacterial pathways promote disulfide bond formation in the bacterial cell envelope, some archaeal extremophiles express proteins with disulfide bonds both in the cytoplasm and in the extra-cytoplasmic space, possibly to stabilize proteins in the face of extreme conditions, such as growth at high temperatures. Here, we summarize the diversity of disulfide-bond-catalysing systems across prokaryotic lineages, discuss examples for understanding the biological basis of such systems, and present perspectives on how such systems are enabling advances in biomedical engineering and drug development.


Subject(s)
Disulfides/metabolism , Escherichia coli/enzymology , Gram-Negative Bacteria/enzymology , Membrane Proteins/metabolism , Actinobacteria/metabolism , Bacterial Proteins/metabolism , Catalysis , Cell Membrane , Cyanobacteria/metabolism , Escherichia coli/growth & development , Escherichia coli Proteins/metabolism , Gram-Negative Bacteria/genetics , Humans , Protein Disulfide-Isomerases/metabolism , Protein Folding , Vitamin K Epoxide Reductases/metabolism
4.
Microbiology (Reading) ; 163(12): 1864-1879, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29139344

ABSTRACT

Disulfide bonds confer stability and activity to proteins. Bioinformatic approaches allow predictions of which organisms make protein disulfide bonds and in which subcellular compartments disulfide bond formation takes place. Such an analysis, along with biochemical and protein structural data, suggests that many of the extremophile Crenarachaea make protein disulfide bonds in both the cytoplasm and the cell envelope. We have sought to determine the oxidative folding pathways in the sequenced genomes of the Crenarchaea, by seeking homologues of the enzymes known to be involved in disulfide bond formation in bacteria. Some Crenarchaea have two homologues of the cytoplasmic membrane protein VKOR, a protein required in many bacteria for the oxidation of bacterial DsbAs. We show that the two VKORs of Aeropyrum pernix assume opposite orientations in the cytoplasmic membrane, when expressed in E. coli. One has its active cysteines oriented toward the E. coli periplasm (ApVKORo) and the other toward the cytoplasm (ApVKORi). Furthermore, the ApVKORo promotes disulfide bond formation in the E. coli cell envelope, while the ApVKORi promotes disulfide bond formation in the E. coli cytoplasm via a co-expressed archaeal protein ApPDO. Amongst the VKORs from different archaeal species, the pairs of VKORs in each species are much more closely related to each other than to the VKORs of the other species. The results suggest two independent occurrences of the evolution of the two topologically inverted VKORs in archaea. Our results suggest a mechanistic basis for the formation of disulfide bonds in the cytoplasm of Crenarchaea.


Subject(s)
Aeropyrum/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Disulfides/chemistry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Aeropyrum/chemistry , Aeropyrum/genetics , Archaeal Proteins/genetics , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Cysteine/chemistry , Cysteine/metabolism , Cytoplasm/chemistry , Cytoplasm/genetics , Cytoplasm/metabolism , Disulfides/metabolism , Membrane Proteins/genetics , Periplasm/genetics , Periplasm/metabolism , Protein Folding
5.
J Biol Chem ; 292(16): 6529-6541, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28232484

ABSTRACT

Disulfide bonds contribute to protein stability, activity, and folding in a variety of proteins, including many involved in bacterial virulence such as toxins, adhesins, flagella, and pili, among others. Therefore, inhibitors of disulfide bond formation enzymes could have profound effects on pathogen virulence. In the Escherichia coli disulfide bond formation pathway, the periplasmic protein DsbA introduces disulfide bonds into substrates, and then the cytoplasmic membrane protein DsbB reoxidizes DsbA's cysteines regenerating its activity. Thus, DsbB generates a protein disulfide bond de novo by transferring electrons to the quinone pool. We previously identified an effective pyridazinone-related inhibitor of DsbB enzymes from several Gram-negative bacteria. To map the protein residues that are important for the interaction with this inhibitor, we randomly mutagenized by error-prone PCR the E. coli dsbB gene and selected dsbB mutants that confer resistance to this drug using two approaches. We characterized in vivo and in vitro some of these mutants that map to two areas in the structure of DsbB, one located between the two first transmembrane segments where the quinone ring binds and the other located in the second periplasmic loop of DsbB, which interacts with DsbA. In addition, we show that a mutant version of a protein involved in lipopolysaccharide assembly, lptD4213, is synthetically lethal with the deletion of dsbB as well as with DsbB inhibitors. This finding suggests that drugs decreasing LptD assembly may be synthetically lethal with inhibitors of the Dsb pathway, potentiating the antibiotic effects.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Proteins/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Membrane Proteins/chemistry , Mutation , Anti-Bacterial Agents/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Disulfides/chemistry , Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , Gene Library , Kinetics , Lipopolysaccharides/chemistry , Membrane Proteins/genetics , Mutagenesis , Polymerase Chain Reaction , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/genetics , Pyridazines/chemistry , Quinones/chemistry , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , Temperature , Virulence
6.
Mol Microbiol ; 103(3): 413-422, 2017 02.
Article in English | MEDLINE | ID: mdl-27785850

ABSTRACT

Disulfide bonds are found in many proteins associated with the cell wall of Escherichia coli, and for some of these proteins the disulfide bond is critical to their stability and function. One protein found to contain a disulfide bond is the essential cell division protein FtsN, but the importance of this bond to the protein's structural integrity is unclear. While it evidently plays a role in the proper folding of the SPOR domain of FtsN, this domain is non-essential, suggesting that the disulfide bond might also be dispensable. However, we find that FtsN mutants lacking cysteines give rise to filamentous growth. Furthermore, FtsN protein levels in strains expressing these mutants were significantly lower than in a strain expressing the wild-type allele, as were FtsN levels in strains incapable of making disulfide bonds (dsb- ) exposed to anaerobic conditions. These results strongly suggest that FtsN lacking a disulfide bond is unstable, thereby making this disulfide critical for function. We have previously found that dsb- strains fail to grow anaerobically, and the results presented here suggest that this growth defect may be due in part to misfolded FtsN. Thus, proper cell division in E. coli is dependent upon disulfide bond formation.


Subject(s)
Escherichia coli Proteins/metabolism , Membrane Proteins/metabolism , Cell Division/physiology , Cell Wall/metabolism , Disulfides , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/physiology , Membrane Proteins/genetics , Membrane Proteins/physiology , Protein Binding , Protein Domains , Structure-Activity Relationship
7.
Nat Chem Biol ; 11(4): 292-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25686372

ABSTRACT

In bacteria, disulfide bonds confer stability on many proteins exported to the cell envelope or beyond. These proteins include numerous bacterial virulence factors, and thus bacterial enzymes that promote disulfide bond formation represent targets for compounds inhibiting bacterial virulence. Here, we describe a new target- and cell-based screening methodology for identifying compounds that inhibit the disulfide bond-forming enzymes Escherichia coli DsbB (EcDsbB) or Mycobacterium tuberculosis VKOR (MtbVKOR), which can replace EcDsbB, although the two are not homologs. Initial screening of 51,487 compounds yielded six specifically inhibiting EcDsbB. These compounds share a structural motif and do not inhibit MtbVKOR. A medicinal chemistry approach led us to select related compounds, some of which are much more effective DsbB inhibitors than those found in the screen. These compounds inhibit purified DsbB and prevent anaerobic growth of E. coli. Furthermore, these compounds inhibit all but one of the DsbBs of nine other Gram-negative pathogenic bacteria tested.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Escherichia coli/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Mycobacterium tuberculosis/metabolism , Agar/chemistry , Anti-Bacterial Agents/chemistry , Catalytic Domain , Chemistry, Pharmaceutical/methods , Combinatorial Chemistry Techniques , Disulfides , Dose-Response Relationship, Drug , Drug Design , Electron Transport , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Mass Spectrometry , Microbial Sensitivity Tests , Mycobacterium smegmatis/metabolism , Protein Conformation , Protein Disulfide-Isomerases/antagonists & inhibitors , Protein Disulfide-Isomerases/chemistry , Pseudomonas aeruginosa/metabolism
8.
J Bacteriol ; 196(18): 3343-50, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25002543

ABSTRACT

Targeted, translational LacZ fusions provided the initial support for the signal sequence hypothesis in prokaryotes and allowed for selection of the mutations that identified the Sec translocon. Many of these selections relied on the fact that expression of targeted, translational lacZ fusions like malE-lacZ and lamB-lacZ42-1 causes lethal toxicity as folded LacZ jams the translocation pore. However, there is another class of targeted LacZ fusions that do not jam the translocon. These targeted, nonjamming fusions also show toxic phenotypes that may be useful for selecting mutations in genes involved in posttranslocational protein folding and targeting; however, they have not been investigated to the same extent as their jamming counterparts. In fact, it is still unclear whether LacZ can be fully translocated in these fusions. It may be that they simply partition into the inner membrane where they can no longer participate in folding or assembly. In the present study, we systematically characterize the nonjamming fusions and determine their ultimate localization. We report that LacZ can be fully translocated into the periplasm, where it is toxic. We show that this toxicity is likely due to LacZ misfolding and that, in the absence of the periplasmic disulfide bond catalyst DsbA, LacZ folds in the periplasm. Using the novel phenotype of periplasmic ß-galactosidase activity, we show that the periplasmic chaperone FkpA contributes to LacZ folding in this nonnative compartment. We propose that targeted, nonjamming LacZ fusions may be used to further study folding and targeting in the periplasm of Escherichia coli.


Subject(s)
Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/physiology , Lac Operon/physiology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Genotype , Membrane Proteins/genetics , Membrane Proteins/metabolism , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Protein Folding , Recombinant Proteins , Signal Transduction , Translocation, Genetic
10.
Biochim Biophys Acta ; 1844(8): 1402-14, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24576574

ABSTRACT

The formation of structural disulfide bonds is essential for the function and stability of a great number of proteins, particularly those that are secreted. There exists a variety of dedicated cellular catalysts and pathways from archaea to humans that ensure the formation of native disulfide bonds. In this review we describe the initial discoveries of these pathways and report progress in recent years in our understanding of the diversity of these pathways in prokaryotes, including those newly discovered in some archaea. We will also discuss the various successful efforts to achieve laboratory-based evolution and design of synthetic disulfide bond formation machineries in the bacterium Escherichia coli. These latter studies have also led to new more general insights into the redox environment of the cytoplasm and bacterial cell envelope. This article is part of a Special Issue entitled: Thiol-Based Redox Processes.


Subject(s)
Disulfides/chemistry , Prokaryotic Cells/metabolism , Animals , Humans , Oxidation-Reduction
11.
Curr Biol ; 23(10): R422-5, 2013 May 20.
Article in English | MEDLINE | ID: mdl-23841128
12.
Res Microbiol ; 164(6): 497-504, 2013.
Article in English | MEDLINE | ID: mdl-23538404

ABSTRACT

The Sec pathway for export of proteins across the cytoplasmic membrane to the bacterial periplasm and outer membrane was the first secretion pathway to be discovered in bacteria. A combination of bacterial genetics, development of an in vitro membrane vesicle system and the concurrent elaboration of the signal hypothesis from studies on eukaryotes led to the identification and characterization of two pathways leading to protein export through the SecYEG cytoplasmic membrane translocon. The Sec pathway is also required for assembly of proteins into the cytoplasmic membrane. Since the membrane translocon for Sec pathways is conserved across the three domains of life, the history of research progress in eukaryotes and bacteria was facilitated by the close interaction between those studying both classes of organisms.


Subject(s)
Bacteria/metabolism , Bacterial Proteins/metabolism , Bacterial Secretion Systems , Bacteria/genetics , Bacterial Proteins/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Protein Transport
13.
mBio ; 3(2)2012.
Article in English | MEDLINE | ID: mdl-22493033

ABSTRACT

UNLABELLED: The Escherichia coli membrane protein DsbD functions as an electron hub that dispatches electrons received from the cytoplasmic thioredoxin system to periplasmic oxidoreductases involved in protein disulfide isomerization, cytochrome c biogenesis, and sulfenic acid reduction. Here, we describe a new class of DsbD proteins, named ScsB, whose members are found in proteobacteria and Chlamydia. ScsB has a domain organization similar to that of DsbD, but its amino-terminal domain differs significantly. In DsbD, this domain directly interacts with substrates to reduce them, which suggests that ScsB acts on a different array of substrates. Using Caulobacter crescentus as a model organism, we searched for the substrates of ScsB. We discovered that ScsB provides electrons to the first peroxide reduction pathway identified in the bacterial cell envelope. The reduction pathway comprises a thioredoxin-like protein, TlpA, and a peroxiredoxin, PprX. We show that PprX is a thiol-dependent peroxidase that efficiently reduces both hydrogen peroxide and organic peroxides. Moreover, we identified two additional proteins that depend on ScsB for reduction, a peroxiredoxin-like protein, PrxL, and a novel protein disulfide isomerase, ScsC. Altogether, our results reveal that the array of proteins involved in reductive pathways in the oxidative cell envelope is significantly broader than was previously thought. Moreover, the identification of a new periplasmic peroxiredoxin indicates that in some bacteria, it is important to directly scavenge peroxides in the cell envelope even before they reach the cytoplasm. IMPORTANCE: Peroxides are reactive oxygen species (ROS) that damage cellular components such as lipids, proteins, and nucleic acids. The presence of protection mechanisms against ROS is essential for cell survival. Bacteria express cytoplasmic catalases and thiol-dependent peroxidases to directly scavenge harmful peroxides. We report the identification of a peroxide reduction pathway active in the periplasm of Caulobacter crescentus, which reveals that, in some bacteria, it is important to directly scavenge peroxides in the cell envelope even before they reach the cytoplasm. The electrons required for peroxide reduction are delivered to this pathway by ScsB, a new type of membrane electron transporter. We also identified two additional likely ScsB substrates, including a novel protein disulfide isomerase. Our results reveal that the array of proteins involved in reductive pathways in the oxidative environment of the cell envelope is significantly broader than was previously thought.


Subject(s)
Cell Membrane/enzymology , Chlamydia/metabolism , Electron Transport Chain Complex Proteins/metabolism , Periplasm/enzymology , Peroxiredoxins/metabolism , Thioredoxins/metabolism , Amino Acid Sequence , Caulobacter crescentus/enzymology , Caulobacter crescentus/genetics , Caulobacter crescentus/metabolism , Cell Membrane/metabolism , Chlamydia/genetics , Cluster Analysis , Electron Transport Chain Complex Proteins/genetics , Models, Biological , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction , Periplasm/metabolism , Phylogeny , Sequence Homology, Amino Acid , Substrate Specificity
14.
J Bacteriol ; 194(6): 1515-22, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22247510

ABSTRACT

Production of deoxyribonucleotides for DNA synthesis is an essential and tightly regulated process. The class Ia ribonucleotide reductase (RNR), the product of the nrdAB genes, is required for aerobic growth of Escherichia coli. In catalyzing the reduction of ribonucleotides, two of the cysteines of RNR become oxidized, forming a disulfide bond. To regenerate active RNR, the cell uses thioredoxins and glutaredoxins to reduce the disulfide bond. Strains that lack thioredoxins 1 and 2 and glutaredoxin 1 do not grow because RNR remains in its oxidized, inactive form. However, suppressor mutations that lead to RNR overproduction allow glutaredoxin 3 to reduce sufficient RNR for growth of these mutant strains. We previously described suppressor mutations in the dnaA and dnaN genes that had such effects. Here we report the isolation of new mutations that lead to increased levels of RNR. These include mutations that were not known to influence production of RNR previously, such as a mutation in the hda gene and insertions in the nrdAB promoter region of insertion elements IS1 and IS5. Bioinformatic analysis raises the possibility that IS element insertion in this region represents an adaptive mechanism in nrdAB regulation in E. coli and closely related species. We also characterize mutations altering different amino acids in DnaA and DnaN from those isolated before.


Subject(s)
Escherichia coli/enzymology , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Ribonucleotide Reductases/biosynthesis , Ribonucleotide Reductases/genetics , Suppression, Genetic , Bacterial Proteins/genetics , DNA Polymerase III/genetics , DNA-Binding Proteins/genetics , Escherichia coli Proteins/genetics , Genes, Bacterial , Mutagenesis, Insertional , Promoter Regions, Genetic , Ribonucleoside Diphosphate Reductase/genetics
15.
J Bacteriol ; 193(18): 4988-92, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21784946

ABSTRACT

FtsL and FtsB are two inner-membrane proteins that are essential constituents of the cell division apparatus of Escherichia coli. In this study, we demonstrate that the leucine zipper-like (LZ) motifs, located in the periplasmic domain of FtsL and FtsB, are required for an optimal interaction between these two essential proteins.


Subject(s)
Cell Cycle Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Leucine Zippers , Membrane Proteins/metabolism , Protein Interaction Mapping , Cell Cycle Proteins/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Membrane Proteins/genetics , Protein Binding
16.
J Mol Biol ; 409(1): 7-13, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21334345

ABSTRACT

The papers from the Jacob and Monod groups that presented the operon model and repressor control of gene expression provided a paradigm that opened up the field of gene regulation. Following the set of papers published between 1958 and 1961, there ensued almost immediately a period of "normal science" as Thomas Kuhn has defined it. Most researchers applied the repressor model to their own systems. I suggest a number of factors that were responsible for the rapid acceptance of the Jacob-Monod model and inhibited suggestions for alternative mechanisms of regulation. Nevertheless, I argue that this adherence to the paradigm, including specifically control by repressors, may well have been necessary for the field to progress. Ultimately, the evolution of this field of study was to reveal the unexpected complexity to genetic regulation.


Subject(s)
Gene Expression Regulation , Molecular Biology/history , Molecular Biology/trends , Operon , History, 20th Century , History, 21st Century , Repressor Proteins/metabolism
17.
Antioxid Redox Signal ; 14(8): 1413-20, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-20969481

ABSTRACT

We have presented evidence that a homologue of vertebrate membrane protein vitamin K epoxide reductase (VKOR) is an important component of the protein disulfide bond-forming pathway in many bacteria. Bacterial VKOR appears to take the place of the nonhomologous DsbB found in Escherichia coli. We also determined the structure of a VKOR from a Cyanobacterium and showed that two or four conserved cysteines are required, according to different reductants for activity in an in vitro assay. Here we present evidence for the topologic arrangement in the cytoplasmic membrane of the VKOR from Mycobacterium tuberculosis (Mtb). The results show that Mtb VKOR is a membrane protein that spans the membrane 5 times with its N-terminus in the cytoplasm, C-terminus in the periplasm, and all four cysteines facing the periplasm. The essentiality of the four conserved cysteine residues has also been demonstrated in promoting disulfide bond formation in vivo and a mixed disulfide between a cysteine of DsbA of E. coli, and one of the cysteines (Cys(57)) of the VKOR homologue has been identified to be a likely intermediate in the disulfide bond-forming pathway. These studies may inform future resolution of issues surrounding the functioning of human VKOR.


Subject(s)
Cell Membrane/enzymology , Cytoplasm/enzymology , Disulfides/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Biocatalysis , Cell Line , Cell Membrane/metabolism , Cysteine/genetics , Cytoplasm/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Mutation , Mycobacterium tuberculosis/metabolism , Vitamin K Epoxide Reductases
18.
Nat Chem Biol ; 7(2): 101-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21186348

ABSTRACT

The first and highly conserved step in glutathione (GSH) biosynthesis is formation of γ-glutamyl cysteine by the enzyme glutamate-cysteine ligase (GshA). However, bioinformatic analysis revealed that many prokaryotic species that encode GSH-dependent proteins lack the gene for this enzyme. To understand how bacteria cope without gshA, we isolated Escherichia coli ΔgshA multigenic suppressors that accumulated physiological levels of GSH. Mutations in both proB and proA, the first two genes in L-proline biosynthesis, provided a new pathway for γ-glutamyl cysteine formation via the selective interception of ProB-bound γ-glutamyl phosphate by amino acid thiols, likely through an S-to-N acyl shift mechanism. Bioinformatic analysis suggested that the L-proline biosynthetic pathway may have a second role in γ-glutamyl cysteine formation in prokaryotes. Also, we showed that this mechanism could be exploited to generate cytoplasmic redox buffers bioorthogonal to GSH.


Subject(s)
Evolution, Molecular , Glutathione/biosynthesis , Glutamate-Cysteine Ligase/chemistry , Glutamate-Cysteine Ligase/genetics , Mutation
19.
Trends Genet ; 26(11): 459-61, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20828856

ABSTRACT

Marketers of genetic tests often openly or implicitly misrepresent the utility of genetic information. Scientists who are well aware of the current limitations to the utility of such tests are best placed to publicly counter misrepresentations of the science.


Subject(s)
Consumer Health Information , Genetic Testing , Genome, Human , Biomedical Research , Consumer Health Information/trends , Humans
20.
Biochemistry ; 49(41): 8922-8, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20873718

ABSTRACT

Thioredoxin-1 from Escherichia coli has frequently been used as a model substrate in protein folding studies. However, for reasons of convenience, these studies have focused largely on oxidized thioredoxin and not on reduced thioredoxin, the more physiologically relevant species. Here we describe the first extensive characterization of the refolding kinetics and conformational thermodynamics of reduced thioredoxin. We have previously described a genetic screen that yielded mutant thioredoxin proteins that fold more slowly in both the oxidized and reduced forms. In this study, we apply our more detailed analysis of reduced thioredoxin folding to a larger number of folding mutants that includes those obtained from continuation of the genetic screen. We have identified mutant proteins that display folding defects specifically in the reduced state but not the oxidized state. Some of these substitutions represent unusual folding mutants in that they result in semiconservative substitutions at solvent-exposed positions in the folded conformation and do not appear to affect the conformational stability of the protein. Further, the genetic selection yields mutants at only a limited number of sites, pointing to perhaps the most critical amino acids in the folding pathway and underscoring, in particular, the role of the carboxy-terminal amino acids in the folding of thioredoxin. Our results demonstrate the importance of studying the physiologically relevant folding species.


Subject(s)
Escherichia coli K12/enzymology , Escherichia coli Proteins/chemistry , Protein Folding , Thioredoxins/chemistry , Amino Acid Substitution , Escherichia coli K12/genetics , Escherichia coli Proteins/genetics , Kinetics , Mutation, Missense , Thermodynamics , Thioredoxins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...