Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 1002, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36864019

ABSTRACT

In two-component systems, the information gathered by histidine kinases (HKs) are relayed to cognate response regulators (RRs). Thereby, the phosphoryl group of the auto-phosphorylated HK is transferred to the receiver (Rec) domain of the RR to allosterically activate its effector domain. In contrast, multi-step phosphorelays comprise at least one additional Rec (Recinter) domain that is typically part of the HK and acts as an intermediary for phosphoryl-shuttling. While RR Rec domains have been studied extensively, little is known about discriminating features of Recinter domains. Here we study the Recinter domain of the hybrid HK CckA by X-ray crystallography and NMR spectroscopy. Strikingly, all active site residues of the canonical Rec-fold are pre-arranged for phosphoryl-binding and BeF3- binding does not alter secondary or quaternary structure, indicating the absence of allosteric changes, the hallmark of RRs. Based on sequence-covariation and modeling, we analyze the intra-molecular DHp/Rec association in hybrid HKs.


Subject(s)
Histidine Kinase , Crystallography, X-Ray , Histidine Kinase/chemistry
2.
Sci Rep ; 13(1): 2727, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36810577

ABSTRACT

Bacterial second messengers c-di-GMP and (p)ppGpp have broad functional repertoires ranging from growth and cell cycle control to the regulation of biofilm formation and virulence. The recent identification of SmbA, an effector protein from Caulobacter crescentus that is jointly targeted by both signaling molecules, has opened up studies on how these global bacterial networks interact. C-di-GMP and (p)ppGpp compete for the same SmbA binding site, with a dimer of c-di-GMP inducing a conformational change that involves loop 7 of the protein that leads to downstream signaling. Here, we report a crystal structure of a partial loop 7 deletion mutant, SmbA∆loop in complex with c-di-GMP determined at 1.4 Å resolution. SmbA∆loop binds monomeric c-di-GMP indicating that loop 7 is required for c-di-GMP dimerization. Thus the complex probably represents the first step of consecutive c-di-GMP binding to form an intercalated dimer as has been observed in wild-type SmbA. Considering the prevalence of intercalated c-di-GMP molecules observed bound to proteins, the proposed mechanism may be generally applicable to protein-mediated c-di-GMP dimerization. Notably, in the crystal, SmbA∆loop forms a 2-fold symmetric dimer via isologous interactions with the two symmetric halves of c-di-GMP. Structural comparisons of SmbA∆loop with wild-type SmbA in complex with dimeric c-di-GMP or ppGpp support the idea that loop 7 is critical for SmbA function by interacting with downstream partners. Our results also underscore the flexibility of c-di-GMP, to allow binding to the symmetric SmbA∆loop dimer interface. It is envisaged that such isologous interactions of c-di-GMP could be observed in hitherto unrecognized targets.


Subject(s)
Cyclic GMP , Guanosine Pentaphosphate , Dimerization , Ligands , Guanosine Pentaphosphate/metabolism , Cyclic GMP/metabolism , Bacterial Proteins/metabolism
3.
Anal Biochem ; 639: 114523, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34906539

ABSTRACT

We describe a quenching-free, 'online' ion exchange chromatography (oIEC) method for the quantitative analysis of enzymatic reactions in real-time. We show that separate quenching of the ongoing reaction performed conventionally is not required, since enzymatic reactions are interrupted upon immobilization of the reaction compounds by binding to the stationary phase of the ion exchange column. The reaction mix samples are directly injected into the column, thereby improving data consistency and allowing automation of the process. The method allows reliable and efficient acquisition of enzymatic progress curves by automatic loading of aliquots of an ongoing reaction at predefined timepoints. We demonstrate the applicability of this method for a variety of enzymatic reactions. SUBJECT: Enzymatic assays and analysis.


Subject(s)
Chromatography, Ion Exchange/methods , Enzyme Assays/methods , Chromatography, Ion Exchange/instrumentation , Enzyme Assays/instrumentation , Equipment Design , Fungal Proteins/metabolism , Hexokinase/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism
4.
Microorganisms ; 9(8)2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34442725

ABSTRACT

Proteins containing a FIC domain catalyze AMPylation and other post-translational modifications (PTMs). In bacteria, they are typically part of FicTA toxin-antitoxin modules that control conserved biochemical processes such as topoisomerase activity, but they have also repeatedly diversified into host-targeted virulence factors. Among these, Bartonella effector proteins (Beps) comprise a particularly diverse ensemble of FIC domains that subvert various host cellular functions. However, no comprehensive comparative analysis has been performed to infer molecular mechanisms underlying the biochemical and functional diversification of FIC domains in the vast Bep family. Here, we used X-ray crystallography, structural modelling, and phylogenetic analyses to unravel the expansion and diversification of Bep repertoires that evolved in parallel in three Bartonella lineages from a single ancestral FicTA toxin-antitoxin module. Our analysis is based on 99 non-redundant Bep sequences and nine crystal structures. Inferred from the conservation of the FIC signature motif that comprises the catalytic histidine and residues involved in substrate binding, about half of them represent AMP transferases. A quarter of Beps show a glutamate in a strategic position in the putative substrate binding pocket that would interfere with triphosphate-nucleotide binding but may allow binding of an AMPylated target for deAMPylation or another substrate to catalyze a distinct PTM. The ß-hairpin flap that registers the modifiable target segment to the active site exhibits remarkable structural variability. The corresponding sequences form few well-defined groups that may recognize distinct target proteins. The binding of Beps to promiscuous FicA antitoxins is well conserved, indicating a role of the antitoxin to inhibit enzymatic activity or to serve as a chaperone for the FIC domain before translocation of the Bep into host cells. Taken together, our analysis indicates a remarkable functional plasticity of Beps that is mostly brought about by structural changes in the substrate pocket and the target dock. These findings may guide future structure-function analyses of the highly versatile FIC domains.

5.
mBio ; 12(3): e0017321, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34154415

ABSTRACT

Bacteria that colonize animals must overcome, or coexist, with the reactive oxygen species products of inflammation, a front-line defense of innate immunity. Among these is the neutrophilic oxidant bleach, hypochlorous acid (HOCl), a potent antimicrobial that plays a primary role in killing bacteria through nonspecific oxidation of proteins, lipids, and DNA. Here, we report that in response to increasing HOCl levels, Escherichia coli regulates biofilm production via activation of the diguanylate cyclase DgcZ. We identify the mechanism of DgcZ sensing of HOCl to be direct oxidation of its regulatory chemoreceptor zinc-binding (CZB) domain. Dissection of CZB signal transduction reveals that oxidation of the conserved zinc-binding cysteine controls CZB Zn2+ occupancy, which in turn regulates the catalysis of c-di-GMP by the associated GGDEF domain. We find DgcZ-dependent biofilm formation and HOCl sensing to be regulated in vivo by the conserved zinc-coordinating cysteine. Additionally, point mutants that mimic oxidized CZB states increase total biofilm. A survey of bacterial genomes reveals that many pathogenic bacteria that manipulate host inflammation as part of their colonization strategy possess CZB-regulated diguanylate cyclases and chemoreceptors. Our findings suggest that CZB domains are zinc-sensitive regulators that allow host-associated bacteria to perceive host inflammation through reactivity with HOCl. IMPORTANCE Immune cells are well equipped to eliminate invading bacteria, and one of their primary tools is the synthesis of bleach, hypochlorous acid (HOCl), the same chemical used as a household disinfectant. In this work, we present findings showing that many host-associated bacteria possess a bleach-sensing protein that allows them to adapt to the presence of this chemical in their environment. We find that the bacterium Escherichia coli responds to bleach by hunkering down and producing a sticky matrix known as biofilm, which helps it aggregate and adhere to surfaces. This behavior may play an important role in pathogenicity for E. coli and other bacteria, as it allows the bacteria to detect and adapt to the weapons of the host immune system.


Subject(s)
Bacterial Adhesion/genetics , Biofilms/growth & development , Cyclic GMP/analogs & derivatives , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Inflammation/genetics , Signal Transduction , Bacteria/classification , Bacteria/genetics , Bacteria/immunology , Bacteria/metabolism , Bacterial Adhesion/immunology , Biofilms/drug effects , Cyclic GMP/genetics , Cyclic GMP/metabolism , Escherichia coli/drug effects , Escherichia coli/immunology , Escherichia coli/metabolism , Escherichia coli Proteins/immunology , Escherichia coli Proteins/metabolism , Genome, Bacterial , Hypochlorous Acid/pharmacology , Inflammation/immunology
6.
Nat Commun ; 12(1): 2162, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846343

ABSTRACT

Diguanylate cyclases synthesising the bacterial second messenger c-di-GMP are found to be regulated by a variety of sensory input domains that control the activity of their catalytical GGDEF domain, but how activation proceeds mechanistically is, apart from a few examples, still largely unknown. As part of two-component systems, they are activated by cognate histidine kinases that phosphorylate their Rec input domains. DgcR from Leptospira biflexa is a constitutively dimeric prototype of this class of diguanylate cyclases. Full-length crystal structures reveal that BeF3- pseudo-phosphorylation induces a relative rotation of two rigid halves in the Rec domain. This is coupled to a reorganisation of the dimeric structure with concomitant switching of the coiled-coil linker to an alternative heptad register. Finally, the activated register allows the two substrate-loaded GGDEF domains, which are linked to the end of the coiled-coil via a localised hinge, to move into a catalytically competent dimeric arrangement. Bioinformatic analyses suggest that the binary register switch mechanism is utilised by many diguanylate cyclases with N-terminal coiled-coil linkers.


Subject(s)
Escherichia coli Proteins/metabolism , Leptospira/enzymology , Phosphorus-Oxygen Lyases/metabolism , Allosteric Regulation , Amino Acid Sequence , Aspartic Acid/metabolism , Beryllium/chemistry , Enzyme Activation , Escherichia coli Proteins/chemistry , Feedback, Physiological , Fluorides/chemistry , Kinetics , Models, Molecular , Phosphorus-Oxygen Lyases/chemistry , Phosphorylation , Protein Domains , Protein Multimerization , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/metabolism , Rotation
7.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Article in English | MEDLINE | ID: mdl-33723071

ABSTRACT

Small GTPases of the Ras-homology (Rho) family are conserved molecular switches that control fundamental cellular activities in eukaryotic cells. As such, they are targeted by numerous bacterial toxins and effector proteins, which have been intensively investigated regarding their biochemical activities and discrete target spectra; however, the molecular mechanism of target selectivity has remained largely elusive. Here we report a bacterial effector protein that selectively targets members of the Rac subfamily in the Rho family of small GTPases but none in the closely related Cdc42 or RhoA subfamilies. This exquisite target selectivity of the FIC domain AMP-transferase Bep1 from Bartonella rochalimae is based on electrostatic interactions with a subfamily-specific pair of residues in the nucleotide-binding G4 motif and the Rho insert helix. Residue substitutions at the identified positions in Cdc42 enable modification by Bep1, while corresponding Cdc42-like substitutions in Rac1 greatly diminish modification. Our study establishes a structural understanding of target selectivity toward Rac-subfamily GTPases and provides a highly selective tool for their functional analysis.


Subject(s)
Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Protein Processing, Post-Translational , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/metabolism , Amino Acid Sequence , Bartonella , Binding Sites , Models, Molecular , Multigene Family , Protein Binding , Protein Conformation , Structure-Activity Relationship , rac GTP-Binding Proteins/genetics
8.
Nat Microbiol ; 6(1): 59-72, 2021 01.
Article in English | MEDLINE | ID: mdl-33168988

ABSTRACT

Bacteria use small signalling molecules such as (p)ppGpp or c-di-GMP to tune their physiology in response to environmental changes. It remains unclear whether these regulatory networks operate independently or whether they interact to optimize bacterial growth and survival. We report that (p)ppGpp and c-di-GMP reciprocally regulate the growth of Caulobacter crescentus by converging on a single small-molecule-binding protein, SmbA. While c-di-GMP binding inhibits SmbA, (p)ppGpp competes for the same binding site to sustain SmbA activity. We demonstrate that (p)ppGpp specifically promotes Caulobacter growth on glucose, whereas c-di-GMP inhibits glucose consumption. We find that SmbA contributes to this metabolic switch and promotes growth on glucose by quenching the associated redox stress. The identification of an effector protein that acts as a central regulatory hub for two global second messengers opens up future studies on specific crosstalk between small-molecule-based regulatory networks.


Subject(s)
Caulobacter crescentus/growth & development , Cyclic GMP/analogs & derivatives , Guanosine Pentaphosphate/metabolism , Second Messenger Systems/genetics , Transferases/metabolism , Binding Sites/physiology , Binding, Competitive/physiology , Caulobacter crescentus/genetics , Caulobacter crescentus/metabolism , Cyclic GMP/metabolism , Gene Expression Regulation, Bacterial/genetics , Glucose/metabolism , Oxidation-Reduction , Signal Transduction/genetics
9.
Proc Natl Acad Sci U S A ; 117(29): 17211-17220, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32611811

ABSTRACT

The bacterial second messenger cyclic diguanylate (c-di-GMP) regulates a wide range of cellular functions from biofilm formation to growth and survival. Targeting a second-messenger network is challenging because the system involves a multitude of components with often overlapping functions. Here, we present a strategy to intercept c-di-GMP signaling pathways by directly targeting the second messenger. For this, we developed a c-di-GMP-sequestering peptide (CSP) that was derived from a CheY-like c-di-GMP effector protein. CSP binds c-di-GMP with submicromolar affinity. The elucidation of the CSP⋅c-di-GMP complex structure by NMR identified a linear c-di-GMP-binding motif, in which a self-intercalated c-di-GMP dimer is tightly bound by a network of H bonds and π-stacking interactions involving arginine and aromatic residues. Structure-based mutagenesis yielded a variant with considerably higher, low-nanomolar affinity, which subsequently was shortened to 19 residues with almost uncompromised affinity. We demonstrate that endogenously expressed CSP intercepts c-di-GMP signaling and effectively inhibits biofilm formation in Pseudomonas aeruginosa, the most widely used model for serious biofilm-associated medical implications.


Subject(s)
Bacterial Proteins/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Peptides/metabolism , Second Messenger Systems , Signal Transduction , Biofilms/growth & development , Escherichia coli Proteins , Models, Molecular , Mutagenesis , Peptides/chemistry , Peptides/genetics , Point Mutation , Protein Conformation , Protein Domains , Protein Interaction Domains and Motifs , Pseudomonas aeruginosa/metabolism
10.
Nat Commun ; 11(1): 816, 2020 02 10.
Article in English | MEDLINE | ID: mdl-32041947

ABSTRACT

Bacteria adapt their growth rate to their metabolic status and environmental conditions by modulating the length of their G1 period. Here we demonstrate that a gradual increase in the concentration of the second messenger c-di-GMP determines precise gene expression during G1/S transition in Caulobacter crescentus. We show that c-di-GMP stimulates the kinase ShkA by binding to its central pseudo-receiver domain, activates the TacA transcription factor, and initiates a G1/S-specific transcription program leading to cell morphogenesis and S-phase entry. Activation of the ShkA-dependent genetic program causes c-di-GMP to reach peak levels, which triggers S-phase entry and promotes proteolysis of ShkA and TacA. Thus, a gradual increase of c-di-GMP results in precise control of ShkA-TacA activity, enabling G1/S-specific gene expression that coordinates cell cycle and morphogenesis.


Subject(s)
Caulobacter crescentus/cytology , Caulobacter crescentus/genetics , Cell Cycle/genetics , Cyclic GMP/analogs & derivatives , Histidine Kinase/metabolism , Morphogenesis/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Caulobacter crescentus/growth & development , Caulobacter crescentus/metabolism , Cyclic GMP/metabolism , Gene Expression Regulation, Bacterial , Histidine Kinase/chemistry , Histidine Kinase/genetics , Phosphorylation , Protein Binding , Protein Domains , Proteolysis , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism
11.
Proc Natl Acad Sci U S A ; 117(2): 1000-1008, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31882446

ABSTRACT

Cytosolic hybrid histidine kinases (HHKs) constitute major signaling nodes that control various biological processes, but their input signals and how these are processed are largely unknown. In Caulobacter crescentus, the HHK ShkA is essential for accurate timing of the G1-S cell cycle transition and is regulated by the corresponding increase in the level of the second messenger c-di-GMP. Here, we use a combination of X-ray crystallography, NMR spectroscopy, functional analyses, and kinetic modeling to reveal the regulatory mechanism of ShkA. In the absence of c-di-GMP, ShkA predominantly adopts a compact domain arrangement that is catalytically inactive. C-di-GMP binds to the dedicated pseudoreceiver domain Rec1, thereby liberating the canonical Rec2 domain from its central position where it obstructs the large-scale motions required for catalysis. Thus, c-di-GMP cannot only stabilize domain interactions, but also engage in domain dissociation to allosterically invoke a downstream effect. Enzyme kinetics data are consistent with conformational selection of the ensemble of active domain constellations by the ligand and show that autophosphorylation is a reversible process.


Subject(s)
Caulobacter crescentus/metabolism , Cyclic GMP/analogs & derivatives , Histidine Kinase/chemistry , Histidine Kinase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Caulobacter crescentus/genetics , Cell Cycle/physiology , Crystallography, X-Ray , Cyclic GMP/chemistry , Cyclic GMP/metabolism , Histidine Kinase/genetics , Models, Molecular , Molecular Dynamics Simulation , Phosphorylation , Protein Binding , Protein Conformation , Protein Domains , Second Messenger Systems
12.
J Am Chem Soc ; 141(13): 5275-5285, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30883103

ABSTRACT

Sulfoxide synthases are nonheme iron enzymes that catalyze oxidative carbon-sulfur bond formation between cysteine derivatives and N-α-trimethylhistidine as a key step in the biosynthesis of thiohistidines. The complex catalytic mechanism of this enzyme reaction has emerged as the controversial subject of several biochemical and computational studies. These studies all used the structure of the γ-glutamyl cysteine utilizing sulfoxide synthase, MthEgtB from Mycobacterium thermophilum (EC 1.14.99.50), as a structural basis. To provide an alternative model system, we have solved the crystal structure of CthEgtB from Chloracidobacterium thermophilum (EC 1.14.99.51) that utilizes cysteine as a sulfur donor. This structure reveals a completely different configuration of active site residues that are involved in oxygen binding and activation. Furthermore, comparison of the two EgtB structures enables a classification of all ergothioneine biosynthetic EgtBs into five subtypes, each characterized by unique active-site features. This active site diversity provides an excellent platform to examine the catalytic mechanism of sulfoxide synthases by comparative enzymology, but also raises the question as to why so many different solutions to the same biosynthetic problem have emerged.


Subject(s)
Acidobacteria/enzymology , Ergothioneine/biosynthesis , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Oxygen/metabolism , Binding Sites , Biocatalysis , Ergothioneine/chemistry , Molecular Structure , Oxidation-Reduction , Oxygen/chemistry
13.
Elife ; 62017 11 01.
Article in English | MEDLINE | ID: mdl-29091032

ABSTRACT

The flagellar motor is a sophisticated rotary machine facilitating locomotion and signal transduction. Owing to its important role in bacterial behavior, its assembly and activity are tightly regulated. For example, chemotaxis relies on a sensory pathway coupling chemical information to rotational bias of the motor through phosphorylation of the motor switch protein CheY. Using a chemical proteomics approach, we identified a novel family of CheY-like (Cle) proteins in Caulobacter crescentus, which tune flagellar activity in response to binding of the second messenger c-di-GMP to a C-terminal extension. In their c-di-GMP bound conformation Cle proteins interact with the flagellar switch to control motor activity. We show that individual Cle proteins have adopted discrete cellular functions by interfering with chemotaxis and by promoting rapid surface attachment of motile cells. This study broadens the regulatory versatility of bacterial motors and unfolds mechanisms that tie motor activity to mechanical cues and bacterial surface adaptation.


Subject(s)
Bacterial Adhesion , Bacterial Proteins/metabolism , Caulobacter crescentus/physiology , Chemotaxis , Cyclic GMP/analogs & derivatives , Flagella/physiology , Gene Expression Regulation, Bacterial , Caulobacter crescentus/chemistry , Cyclic GMP/metabolism , Flagella/chemistry , Protein Binding , Proteome/analysis
14.
Structure ; 25(1): 203-211, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27889208

ABSTRACT

The BID (Bep intracellular delivery) domain functions as secretion signal in a subfamily of protein substrates of bacterial type IV secretion (T4S) systems. It mediates transfer of (1) relaxases and the attached DNA during bacterial conjugation, and (2) numerous Bartonella effector proteins (Beps) during protein transfer into host cells infected by pathogenic Bartonella species. Furthermore, BID domains of Beps have often evolved secondary effector functions within host cells. Here, we provide crystal structures for three representative BID domains and describe a novel conserved fold characterized by a compact, antiparallel four-helix bundle topped with a hook. The conserved hydrophobic core provides a rigid scaffold to a surface that, despite a few conserved exposed residues and similarities in charge distribution, displays significant variability. We propose that the genuine function of BID domains as T4S signal may primarily depend on their rigid structure, while the plasticity of their surface may facilitate adaptation to secondary effector functions.


Subject(s)
Bartonella/metabolism , Type VI Secretion Systems/chemistry , Bartonella/chemistry , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Models, Molecular , Protein Domains , Protein Structure, Secondary
15.
PLoS One ; 11(9): e0163654, 2016.
Article in English | MEDLINE | ID: mdl-27657533

ABSTRACT

FIC domain proteins mediate post-translational modifications of target proteins, which typically results in their inactivation. Depending on the conservation of crucial active site residues, the FIC fold serves as structural scaffold for various enzymatic activities, mostly target adenylylation. The founding member of the vast Fic protein family, EcFicT, was identified in Escherichia coli some time ago. The G55R point mutant of EcFicT displays the "filamentation induced by cAMP" (Fic) phenotype at high 3',5'-cyclic adenosine monophosphate (cAMP) concentrations and elevated temperature, but the underlying molecular mechanism and any putative biochemical activity of EcFicT have remained unknown. EcFicT belongs to class I Fic toxin proteins that are encoded together with a small inhibitory protein (antitoxin), named EcFicA in E. coli. Here, we report the crystal structures of two mutant EcFicT/EcFicA complexes (EcFicTG55RA and EcFicTAE28G) both showing close resemblance with the structure of the AMP-transferase VbhT from Bartonella schoenbuchensis in complex with its cognate antitoxin VbhA. However, crucial differences in the active site of EcFicT compared to VbhT and other AMP-transferases rationalize the lack of evidence for adenylylation activity. Comprehensive bioinformatic analysis suggests that EcFicT has evolved from canonical AMP-transferases and has acquired a conserved binding site for a yet to be discovered novel substrate. The G55R mutation has no effect on structure or thermal stability of EcFicT, such that the molecular basis for its associated Fic phenotype remains elusive. We anticipate that this structure will inspire further bioinformatic and experimental analyses in order to characterize the enzymatic activity of EcFicT and help revealing its physiological role.

16.
Sci Adv ; 2(9): e1600823, 2016 09.
Article in English | MEDLINE | ID: mdl-27652341

ABSTRACT

Histidine kinases are key components of regulatory networks in bacteria. Although many of these enzymes are bifunctional, mediating both phosphorylation and dephosphorylation of downstream targets, the molecular details of this central regulatory switch are unclear. We showed recently that the universal second messenger cyclic di-guanosine monophosphate (c-di-GMP) drives Caulobacter crescentus cell cycle progression by forcing the cell cycle kinase CckA from its default kinase into phosphatase mode. We use a combination of structure determination, modeling, and functional analysis to demonstrate that c-di-GMP reciprocally regulates the two antagonistic CckA activities through noncovalent cross-linking of the catalytic domain with the dimerization histidine phosphotransfer (DHp) domain. We demonstrate that both c-di-GMP and ADP (adenosine diphosphate) promote phosphatase activity and propose that c-di-GMP stabilizes the ADP-bound quaternary structure, which allows the receiver domain to access the dimeric DHp stem for dephosphorylation. In silico analyses predict that c-di-GMP control is widespread among bacterial histidine kinases, arguing that it can replace or modulate canonical transmembrane signaling.


Subject(s)
Cyclic GMP/chemistry , Histidine Kinase/chemistry , Models, Molecular , Phosphoric Monoester Hydrolases/chemistry , Adenosine Diphosphate/chemistry , Catalytic Domain , Caulobacter crescentus/enzymology , Protein Structure, Tertiary , Signal Transduction
17.
J Mol Biol ; 428(19): 3683-701, 2016 09 25.
Article in English | MEDLINE | ID: mdl-27498163

ABSTRACT

Cellular levels of the second messenger cyclic di-guanosine monophosphate (c-di-GMP) are determined by the antagonistic activities of diguanylate cyclases and specific phosphodiesterases. In a given bacterial organism, there are often multiple variants of the two enzymes, which are tightly regulated by a variety of external and internal cues due to the presence of specialized sensory or regulatory domains. Dependent on the second messenger level, specific c-di-GMP receptors then control fundamental cellular processes, such as bacterial life style, biofilm formation, and cell cycle control. Here, I review the large body of data on structure-function relationships in diguanylate cyclases. Although the catalytic GGDEF domain is related to the respective domain of adenylate cyclases, the catalyzed intermolecular condensation reaction of two GTP molecules requires the formation of a competent GGDEF dimer with the two substrate molecules juxtaposed. This prerequisite appears to constitute the basis for GGDEF regulation with signal-induced changes within the homotypic dimer of the input domain (PAS, GAF, HAMP, etc.), which are structurally coupled with the arrangement of the GGDEF domains via a rigid coiled-coil linker. Alternatively, phosphorylation of a Rec input domain can drive GGDEF dimerization. Both mechanisms allow modular combination of input and output function that appears advantageous for evolution and rationalizes the striking similarities in domain architecture found in diguanylate cyclases and histidine kinases.


Subject(s)
Bacteria/enzymology , Cyclic GMP/analogs & derivatives , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Evolution, Molecular , Gene Expression Regulation , Metabolic Networks and Pathways/genetics , Phosphorus-Oxygen Lyases/chemistry , Phosphorus-Oxygen Lyases/metabolism , Cyclic GMP/biosynthesis , Escherichia coli Proteins/genetics , Guanosine Triphosphate/metabolism , Models, Molecular , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Phosphorus-Oxygen Lyases/genetics , Protein Conformation , Protein Multimerization
18.
Proc Natl Acad Sci U S A ; 113(5): E529-37, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787847

ABSTRACT

Filamentation induced by cyclic AMP (FIC)-domain enzymes catalyze adenylylation or other posttranslational modifications of target proteins to control their function. Recently, we have shown that Fic enzymes are autoinhibited by an α-helix (αinh) that partly obstructs the active site. For the single-domain class III Fic proteins, the αinh is located at the C terminus and its deletion relieves autoinhibition. However, it has remained unclear how activation occurs naturally. Here, we show by structural, biophysical, and enzymatic analyses combined with in vivo data that the class III Fic protein NmFic from Neisseria meningitidis gets autoadenylylated in cis, thereby autonomously relieving autoinhibition and thus allowing subsequent adenylylation of its target, the DNA gyrase subunit GyrB. Furthermore, we show that NmFic activation is antagonized by tetramerization. The combination of autoadenylylation and tetramerization results in nonmonotonic concentration dependence of NmFic activity and a pronounced lag phase in the progress of target adenylylation. Bioinformatic analyses indicate that this elaborate dual-control mechanism is conserved throughout class III Fic proteins.


Subject(s)
Bacterial Proteins/metabolism , Biopolymers/metabolism , Cyclic AMP/metabolism , Neisseria meningitidis/enzymology , Nucleotidyltransferases/metabolism , DNA Gyrase/metabolism , Models, Molecular
19.
J Bacteriol ; 198(3): 448-62, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26553851

ABSTRACT

UNLABELLED: Intracellular levels of the bacterial second messenger cyclic di-GMP (c-di-GMP) are controlled by antagonistic activities of diguanylate cyclases and phosphodiesterases. The phosphodiesterase PdeH was identified as a key regulator of motility in Escherichia coli, while deletions of any of the other 12 genes encoding potential phosphodiesterases did not interfere with motility. To analyze the roles of E. coli phosphodiesterases, we demonstrated that most of these proteins are expressed under laboratory conditions. We next isolated suppressor mutations in six phosphodiesterase genes, which reinstate motility in the absence of PdeH by reducing cellular levels of c-di-GMP. Expression of all mutant alleles also led to a reduction of biofilm formation. Thus, all of these proteins are bona fide phosphodiesterases that are capable of interfering with different c-di-GMP-responsive output systems by affecting the global c-di-GMP pool. This argues that E. coli possesses several phosphodiesterases that are inactive under laboratory conditions because they lack appropriate input signals. Finally, one of these phosphodiesterases, PdeL, was studied in more detail. We demonstrated that this protein acts as a transcription factor to control its own expression. Motile suppressor alleles led to a strong increase of PdeL activity and elevated pdeL transcription, suggesting that enzymatic activity and transcriptional control are coupled. In agreement with this, we showed that overall cellular levels of c-di-GMP control pdeL transcription and that this control depends on PdeL itself. We thus propose that PdeL acts both as an enzyme and as a c-di-GMP sensor to couple transcriptional activity to the c-di-GMP status of the cell. IMPORTANCE: Most bacteria possess multiple diguanylate cyclases and phosphodiesterases. Genetic studies have proposed that these enzymes show signaling specificity by contributing to distinct cellular processes without much cross talk. Thus, spatial separation of individual c-di-GMP signaling units was postulated. However, since most cyclases and phosphodiesterases harbor N-terminal signal input domains, it is equally possible that most of these enzymes lack their activating signals under laboratory conditions, thereby simulating signaling specificity on a genetic level. We demonstrate that a subset of E. coli phosphodiesterases can be activated genetically to affect the global c-di-GMP pool and thus influence different c-di-GMP-dependent processes. Although this does not exclude spatial confinement of individual phosphodiesterases, this study emphasizes the importance of environmental signals for activation of phosphodiesterases.


Subject(s)
Cyclic GMP/analogs & derivatives , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Phosphoric Diester Hydrolases/metabolism , Amino Acid Sequence , Catalytic Domain , Cyclic GMP/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Movement , Phosphoric Diester Hydrolases/genetics , Video Recording
20.
Cell Rep ; 12(9): 1497-507, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26299961

ABSTRACT

Toxin-antitoxin (TA) modules are ubiquitous molecular switches controlling bacterial growth via the release of toxins that inhibit cell proliferation. Most of these toxins interfere with protein translation, but a growing variety of other mechanisms hints at a diversity that is not yet fully appreciated. Here, we characterize a group of FIC domain proteins as toxins of the conserved and abundant FicTA family of TA modules, and we reveal that they act by suspending control of cellular DNA topology. We show that FicTs are enzymes that adenylylate DNA gyrase and topoisomerase IV, the essential bacterial type IIA topoisomerases, at their ATP-binding site. This modification inactivates both targets by blocking their ATPase activity, and, consequently, causes reversible growth arrest due to the knotting, catenation, and relaxation of cellular DNA. Our results give insight into the regulation of DNA topology and highlight the remarkable plasticity of FIC domain proteins.


Subject(s)
Bacterial Toxins/metabolism , DNA Gyrase/metabolism , DNA Topoisomerase IV/metabolism , DNA, Bacterial/metabolism , Escherichia coli Proteins/metabolism , DNA, Bacterial/chemistry , Escherichia coli/metabolism , Nucleic Acid Conformation , Pseudomonas aeruginosa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL