Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Uruguay Oncology Collection
Country/Region as subject
Publication year range
1.
Annu Rev Microbiol ; 73: 507-528, 2019 09 08.
Article in English | MEDLINE | ID: mdl-31226026

ABSTRACT

Perceiving environmental and internal information and reacting in adaptive ways are essential attributes of living organisms. Two-component systems are relevant protein machineries from prokaryotes and lower eukaryotes that enable cells to sense and process signals. Implicating sensory histidine kinases and response regulator proteins, both components take advantage of protein phosphorylation and flexibility to switch conformations in a signal-dependent way. Dozens of two-component systems act simultaneously in any given cell, challenging our understanding about the means that ensure proper connectivity. This review dives into the molecular level, attempting to summarize an emerging picture of how histidine kinases and cognate response regulators achieve required efficiency, specificity, and directionality of signaling pathways, properties that rely on protein:protein interactions. α helices that carry information through long distances, the fine combination of loose and specific kinase/regulator interactions, and malleable reaction centers built when the two components meet emerge as relevant universal principles.


Subject(s)
Allosteric Regulation/genetics , Histidine Kinase , Protein Interaction Domains and Motifs/genetics , Signal Transduction , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Genes, Bacterial , Histidine Kinase/genetics , Histidine Kinase/metabolism , Phosphorylation , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Biochem J ; 480(2): 127-140, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36688908

ABSTRACT

All living organisms include a set of signaling devices that confer the ability to dynamically perceive and adapt to the fluctuating environment. Two-component systems are part of this sensory machinery that regulates the execution of different genetic and/or biochemical programs in response to specific physical or chemical signals. In the last two decades, there has been tremendous progress in our molecular understanding on how signals are detected, the allosteric mechanisms that control intramolecular information transmission and the specificity determinants that guarantee correct wiring. All this information is starting to be exploited in the development of new synthetic networks. Connecting multiple molecular players, analogous to programming lines of code, can provide the resources to build new sophisticated biocomputing systems. The Synthetic Biology field is starting to revolutionize several scientific fields, such as biomedicine and agriculture, propelling the development of new solutions. Expanding the spectrum of available nanodevices in the toolbox is key to unleash its full potential. This review aims to discuss, from a structural perspective, how to take advantage of the vast array of sensor and effector protein modules involved in two-component systems for the construction of new synthetic circuits.


Subject(s)
Signal Transduction , Synthetic Biology , Proteins
3.
J Struct Biol ; 213(2): 107732, 2021 06.
Article in English | MEDLINE | ID: mdl-33819633

ABSTRACT

Protein Kinase A (PKA) is a widespread enzyme that plays a key role in many signaling pathways from lower eukaryotes to metazoans. In mammals, the regulatory (R) subunits sequester and target the catalytic (C) subunits to proper subcellular locations. This targeting is accomplished by the dimerization and docking (D/D) domain of the R subunits. The activation of the holoenzyme depends on the binding of the second messenger cAMP. The only available structures of the D/D domain proceed from mammalian sources. Unlike dimeric mammalian counterparts, the R subunit from Saccharomyces cerevisiae (Bcy1) forms tetramers in solution. Here we describe the first high-resolution structure of a non-mammalian D/D domain. The tetramer in the crystals of the Bcy1 D/D domain is a dimer of dimers that retain the classical D/D domain fold. By using phylogenetic and structural analyses combined with site-directed mutagenesis, we found that fungal R subunits present an insertion of a single amino acid at the D/D domain that shifts the position of a downstream, conserved arginine. This residue participates in intra-dimer interactions in mammalian D/D domains, while due to this insertion it is involved in inter-dimer contacts in Bcy1, which are crucial for the stability of the tetramer. This surprising finding challenges well-established concepts regarding the oligomeric state within the PKAR protein family and provides important insights into the yet unexplored structural diversity of the D/D domains and the molecular determinants of R subunit oligomerization.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Animals , Arginine/genetics , Circular Dichroism , Crystallography, X-Ray , Cyclic AMP-Dependent Protein Kinases/metabolism , Mammals , Models, Molecular , Mutagenesis, Site-Directed , Phylogeny , Protein Domains , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits/chemistry , Saccharomyces cerevisiae Proteins/genetics , Solutions
4.
Glycobiology ; 27(2): 140-153, 2017 01.
Article in English | MEDLINE | ID: mdl-27913570

ABSTRACT

Polypeptide GalNAc-transferases (GalNAc-Ts) constitute a family of 20 human glycosyltransferases (comprising 9 subfamilies), which initiate mucin-type O-glycosylation. The O-glycoproteome is thought to be differentially regulated via the different substrate specificities and expression patterns of each GalNAc-T isoforms. Here, we present a comprehensive in vitro analysis of the peptide substrate specificity of GalNAc-T13, showing that it essentially overlaps with the ubiquitous expressed GalNAc-T1 isoform found in the same subfamily as T13. We have also identified and partially characterized nine splice variants of GalNAc-T13, which add further complexity to the GalNAc-T family. Two variants with changes in their lectin domains were characterized by in vitro glycosylation assays, and one (Δ39Ex9) was inactive while the second one (Ex10b) had essentially unaltered activity. We used reverse transcription-polymerase chain reaction analysis of human neuroblastoma cell lines, normal brain and a small panel of neuroblastoma tumors to demonstrate that several splice variants (Ex10b, ΔEx9, ΔEx2-7 and ΔEx6/8-39bpEx9) were highly expressed in tumor cell lines compared with normal brain, although the functional implications remain to be unveiled. In summary, the GalNAc-T13 isoform is predicted to function similarly to GalNAc-T1 against peptide substrates in vivo, in contrast to a prior report, but is unique by being selectively expressed in the brain.


Subject(s)
Glycopeptides/genetics , N-Acetylgalactosaminyltransferases/genetics , Peptides/genetics , Protein Isoforms/genetics , Amino Acid Sequence , Brain/metabolism , Gene Expression Regulation , Glycopeptides/metabolism , Glycosylation , Humans , Lectins/genetics , Lectins/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Peptides/metabolism , Protein Isoforms/metabolism , Substrate Specificity , Polypeptide N-acetylgalactosaminyltransferase
5.
Mol Microbiol ; 101(3): 457-70, 2016 08.
Article in English | MEDLINE | ID: mdl-27113476

ABSTRACT

Leptospira are unique among bacteria based on their helical cell morphology with hook-shaped ends and the presence of periplasmic flagella (PF) with pronounced spontaneous supercoiling. The factors that provoke such supercoiling, as well as the role that PF coiling plays in generating the characteristic hook-end cell morphology and motility, have not been elucidated. We have now identified an abundant protein from the pathogen L. interrogans, exposed on the PF surface, and named it Flagellar-coiling protein A (FcpA). The gene encoding FcpA is highly conserved among Leptospira and was not found in other bacteria. fcpA(-) mutants, obtained from clinical isolates or by allelic exchange, had relatively straight, smaller-diameter PF, and were not able to produce translational motility. These mutants lost their ability to cause disease in the standard hamster model of leptospirosis. Complementation of fcpA restored the wild-type morphology, motility and virulence phenotypes. In summary, we identified a novel Leptospira 36-kDa protein, the main component of the spirochete's PF sheath, and a key determinant of the flagella's coiled structure. FcpA is essential for bacterial translational motility and to enable the spirochete to penetrate the host, traverse tissue barriers, disseminate to cause systemic infection and reach target organs.


Subject(s)
Flagella/physiology , Leptospira/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cricetinae , Dogs , Flagella/genetics , Flagella/metabolism , Flagellin/genetics , Flagellin/metabolism , Genetic Complementation Test , Leptospira/genetics , Leptospira/metabolism , Leptospira/pathogenicity , Leptospirosis/microbiology , Madin Darby Canine Kidney Cells , Male , Mesocricetus , Mutation , Periplasm/metabolism , Protein Structural Elements , Virulence
6.
Mol Microbiol ; 98(2): 258-71, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26172072

ABSTRACT

The thermosensor histidine kinase DesK from Bacillus subtilis senses changes in membrane fluidity initiating an adaptive response. Structural changes in DesK have been implicated in transmembrane signaling, but direct evidence is still lacking. On the basis of structure-guided mutagenesis, we now propose a mechanism of DesK-mediated signal sensing and transduction. The data indicate that stabilization/destabilization of a 2-helix coiled coil, which connects the transmembrane sensory domain of DesK to its cytosolic catalytic region, is crucial to control its signaling state. Computational modeling and simulations reveal couplings between protein, water and membrane mechanics. We propose that membrane thickening is the main driving force for signal sensing and that it acts by inducing helix stretching and rotation prompting an asymmetric kinase-competent state. Overall, the known structural changes of the sensor kinase, as well as further dynamic rearrangements that we now predict, consistently link structure determinants to activity modulation.


Subject(s)
Bacillus subtilis/physiology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Protein Kinases/chemistry , Protein Kinases/metabolism , Signal Transduction , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Cell Membrane/metabolism , Cold Temperature , Computer Simulation , Histidine Kinase , Membrane Fluidity , Membrane Proteins/metabolism , Mutagenesis , Protein Conformation , Protein Kinases/genetics , Signal Transduction/genetics
7.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 3): 388-97, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23519414

ABSTRACT

Despite being the most abundant class of immunoglobulins in humans and playing central roles in the adaptive immune response, high-resolution structural data are still lacking for the antigen-binding region of human isotype A antibodies (IgAs). The crystal structures of a human Fab fragment of IgA1 in three different crystal forms are now reported. The three-dimensional organization is similar to those of other Fab classes, but FabA1 seems to be more rigid, being constrained by a hydrophobic core in the interface between the variable and constant domains of the heavy chain (VH-CH1) as well as by a disulfide bridge that connects the light and heavy chains, influencing the relative heavy/light-chain orientation. The crystal structure of the same antibody but with a G-isotype CH1 which is reported to display different antigen affinity has also been solved. The differential structural features reveal plausible mechanisms for constant/variable-domain long-distance effects whereby antibody class switching could alter antigen affinity.


Subject(s)
Antigen-Antibody Reactions , Antigens/chemistry , Binding Sites, Antibody , Immunoglobulin A/chemistry , Immunoglobulin Constant Regions/chemistry , Immunoglobulin Fab Fragments/chemistry , Serine Endopeptidases/chemistry , Antigen-Antibody Reactions/physiology , Antigens/physiology , Clostridium/enzymology , Crystallography, X-Ray , Humans , Immunoglobulin A/physiology , Immunoglobulin Constant Regions/physiology , Immunoglobulin Fab Fragments/physiology , Neisseria gonorrhoeae/enzymology , Protein Structure, Tertiary , Serine Endopeptidases/physiology
8.
Sci Signal ; 16(769): eabo7588, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36693130

ABSTRACT

Phosphorylation carries chemical information in biological systems. In two-component systems (TCSs), the sensor histidine kinase and the response regulator are connected through phosphoryl transfer reactions that may be uni- or bidirectional. Directionality enables the construction of complex regulatory networks that optimize signal propagation and ensure the forward flow of information. We combined x-ray crystallography, hybrid quantum mechanics/molecular mechanics (QM/MM) simulations, and systems-integrative kinetic modeling approaches to study phosphoryl flow through the Bacillus subtilis thermosensing TCS DesK-DesR. The allosteric regulation of the histidine kinase DesK was critical to avoid back transfer of phosphoryl groups and futile phosphorylation-dephosphorylation cycles by isolating phosphatase, autokinase, and phosphotransferase activities. Interactions between the kinase's ATP-binding domain and the regulator's receiver domain placed the regulator in two distinct positions in the phosphotransferase and phosphatase complexes, thereby determining whether a key glutamine residue in DesK was properly situated to assist in the dephosphorylation reaction. Moreover, an energetically unfavorable phosphotransferase conformation when DesK was not phosphorylated minimized reverse phosphoryl transfer. DesR dimerization and a dissociative phosphoryl transfer reaction also enforced the direction of phosphoryl flow. Shorter or longer distances between the phosphoryl acceptor and donor residues shifted the phosphoryl transfer equilibrium by modulating the stabilizing effect of the Mg2+ cofactor. These mechanisms control the directionality of signal transmission and show how structure-encoded allostery stores and transmits information in signaling systems.


Subject(s)
Bacillus subtilis , Signal Transduction , Histidine Kinase/metabolism , Bacillus subtilis/genetics , Phosphorylation , Phosphoric Monoester Hydrolases , Bacterial Proteins/metabolism
9.
Proc Natl Acad Sci U S A ; 106(38): 16185-90, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19805278

ABSTRACT

Temperature sensing is essential for the survival of living cells. A major challenge is to understand how a biological thermometer processes thermal information to optimize cellular functions. Using structural and biochemical approaches, we show that the thermosensitive histidine kinase, DesK, from Bacillus subtilis is cold-activated through specific interhelical rearrangements in its central four-helix bundle domain. As revealed by the crystal structures of DesK in different functional states, the plasticity of this helical domain influences the catalytic activities of the protein, either by modifying the mobility of the ATP-binding domains for autokinase activity or by modulating binding of the cognate response regulator to sustain the phosphotransferase and phosphatase activities. The structural and biochemical data suggest a model in which the transmembrane sensor domain of DesK promotes these structural changes through conformational signals transmitted by the membrane-connecting two-helical coiled-coil, ultimately controlling the alternation between output autokinase and phosphatase activities. The structural comparison of the different DesK variants indicates that incoming signals can take the form of helix rotations and asymmetric helical bends similar to those reported for other sensing systems, suggesting that a similar switching mechanism could be operational in a wide range of sensor histidine kinases.


Subject(s)
Bacillus subtilis/enzymology , Bacterial Proteins/chemistry , Protein Kinases/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Substitution , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites/genetics , Catalysis , Chromatography, Gel , Crystallization , Crystallography, X-Ray , Histidine Kinase , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship , Temperature
10.
J Biol Chem ; 285(32): 24892-903, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20507988

ABSTRACT

DesK is a sensor histidine kinase (HK) that allows Bacillus subtilis to respond to cold shock, triggering the adaptation of membrane fluidity via transcriptional control of a fatty acid desaturase. It belongs to the HK family HPK7, which includes the nitrogen metabolism regulators NarX/Q and the antibiotic sensor LiaS among other important sensor kinases. Structural information on different HK families is still scarce and several questions remain, particularly concerning the molecular features that determine HK specificity during its catalytic autophosphorylation and subsequent response-regulator phosphotransfer reactions. To analyze the ATP-binding features of HPK7 HKs and dissect their mechanism of autophosphorylation at the molecular level, we have studied DesK in complex with ATP using high resolution structural approaches in combination with biochemical studies. We report the first crystal structure of an HK in complex with its natural nucleotidic substrate. The general fold of the ATP-binding domain of DesK is conserved, compared with well studied members of other families. Yet, DesK displays a far more compact structure at the ATP-binding pocket: the ATP lid loop is much shorter with no secondary structural organization and becomes ordered upon ATP loading. Sequence conservation mapping onto the molecular surface, semi-flexible protein-protein docking simulations, and structure-based point mutagenesis allow us to propose a specific domain-domain geometry during autophosphorylation catalysis. Supporting our hypotheses, we have been able to trap an autophosphorylating intermediate state, by protein engineering at the predicted domain-domain interaction surface.


Subject(s)
Adenosine Triphosphate/chemistry , Bacterial Proteins/chemistry , Protein Kinases/chemistry , Bacillus subtilis/enzymology , Catalysis , Disulfides/chemistry , Histidine Kinase , Kinetics , Molecular Conformation , Mutagenesis , Phosphorylation , Protein Binding , Protein Conformation , Protein Engineering/methods , Protein Structure, Tertiary , Signal Transduction
11.
Methods Mol Biol ; 2077: 1-18, 2020.
Article in English | MEDLINE | ID: mdl-31707648

ABSTRACT

The ability to perceive the environment, an essential attribute in living organisms, is linked to the evolution of signaling proteins that recognize specific signals and execute predetermined responses. Such proteins constitute concerted systems that can be as simple as a unique protein, able to recognize a ligand and exert a phenotypic change, or extremely complex pathways engaging dozens of different proteins which act in coordination with feedback loops and signal modulation. To understand how cells sense their surroundings and mount specific adaptive responses, we need to decipher the molecular workings of signal recognition, internalization, transfer, and conversion into chemical changes inside the cell. Protein allostery and dynamics play a central role. Here, we review recent progress on the study of two-component systems, important signaling machineries of prokaryotes and lower eukaryotes. Such systems implicate a sensory histidine kinase and a separate response regulator protein. Both components exploit protein flexibility to effect specific conformational rearrangements, modulating protein-protein interactions, and ultimately transmitting information accurately. Recent work has revealed how histidine kinases switch between discrete functional states according to the presence or absence of the signal, shifting key amino acid positions that define their catalytic activity. In concert with the cognate response regulator's allosteric changes, the phosphoryl-transfer flow during the signaling process is exquisitely fine-tuned for proper specificity, efficiency and directionality.


Subject(s)
Proteins/metabolism , Signal Transduction , Allosteric Regulation , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Eukaryotic Cells/metabolism , Histidine Kinase/chemistry , Histidine Kinase/metabolism , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Phosphorylation , Prokaryotic Cells/metabolism , Protein Binding , Protein Conformation , Proteins/chemistry , Structure-Activity Relationship
12.
Nat Commun ; 11(1): 3703, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32710080

ABSTRACT

Mycobacterium tuberculosis is a pathogen with a unique cell envelope including very long fatty acids, implicated in bacterial resistance and host immune modulation. FasR is a TetR-like transcriptional activator that plays a central role in sensing mycobacterial long-chain fatty acids and regulating lipid biosynthesis. Here we disclose crystal structures of M. tuberculosis FasR in complex with acyl effector ligands and with DNA, uncovering its molecular sensory and switching mechanisms. A long tunnel traverses the entire effector-binding domain, enabling long fatty acyl effectors to bind. Only when the tunnel is entirely occupied, the protein dimer adopts a rigid configuration with its DNA-binding domains in an open state, leading to DNA dissociation. The protein-folding hydrophobic core connects the two domains, and is completed into a continuous spine when the effector binds. Such a transmission spine is conserved in a large number of TetR-like regulators, offering insight into effector-triggered allosteric functional control.


Subject(s)
Acyl Coenzyme A/chemistry , Bacterial Proteins/chemistry , DNA-Binding Proteins/chemistry , Mycobacterium tuberculosis/metabolism , Transcription Factors/chemistry , Acyl Coenzyme A/metabolism , Allosteric Site , Bacterial Proteins/metabolism , Cell Wall/metabolism , Crystallography, X-Ray , DNA, Bacterial/chemistry , DNA-Binding Proteins/metabolism , Fatty Acids/metabolism , Ligands , Models, Molecular , Protein Conformation , Transcription Factors/metabolism
13.
Elife ; 92020 03 11.
Article in English | MEDLINE | ID: mdl-32157997

ABSTRACT

Spirochete bacteria, including important pathogens, exhibit a distinctive means of swimming via undulations of the entire cell. Motility is powered by the rotation of supercoiled 'endoflagella' that wrap around the cell body, confined within the periplasmic space. To investigate the structural basis of flagellar supercoiling, which is critical for motility, we determined the structure of native flagellar filaments from the spirochete Leptospira by integrating high-resolution cryo-electron tomography and X-ray crystallography. We show that these filaments are coated by a highly asymmetric, multi-component sheath layer, contrasting with flagellin-only homopolymers previously observed in exoflagellated bacteria. Distinct sheath proteins localize to the filament inner and outer curvatures to define the supercoiling geometry, explaining a key functional attribute of this spirochete flagellum.


Subject(s)
Bacterial Proteins/physiology , Flagella/physiology , Leptospira/physiology , Movement , Rotation
14.
Nat Plants ; 5(7): 755-765, 2019 07.
Article in English | MEDLINE | ID: mdl-31235877

ABSTRACT

In C4 grasses of agronomical interest, malate shuttled into the bundle sheath cells is decarboxylated mainly by nicotinamide adenine dinucleotide phosphate (NADP)-malic enzyme (C4-NADP-ME). The activity of C4-NADP-ME was optimized by natural selection to efficiently deliver CO2 to Rubisco. During its evolution from a plastidic non-photosynthetic NADP-ME, C4-NADP-ME acquired increased catalytic efficiency, tetrameric structure and pH-dependent inhibition by its substrate malate. Here, we identified specific amino acids important for these C4 adaptions based on strict differential conservation of amino acids, combined with solving the crystal structures of maize and sorghum C4-NADP-ME. Site-directed mutagenesis and structural analyses show that Q503, L544 and E339 are involved in catalytic efficiency; E339 confers pH-dependent regulation by malate, F140 is critical for the stabilization of the oligomeric structure and the N-terminal region is involved in tetramerization. Together, the identified molecular adaptations form the basis for the efficient catalysis and regulation of one of the central biochemical steps in C4 metabolism.


Subject(s)
Malate Dehydrogenase/chemistry , Malate Dehydrogenase/metabolism , Plant Proteins/chemistry , Plant Proteins/metabolism , Sorghum/enzymology , Zea mays/enzymology , Amino Acid Motifs , Biocatalysis , Catalytic Domain , Hydrogen-Ion Concentration , Malate Dehydrogenase/genetics , Malates/metabolism , Photosynthesis , Plant Proteins/genetics , Sorghum/chemistry , Sorghum/genetics , Zea mays/chemistry , Zea mays/genetics
15.
J Cell Biochem ; 105(2): 381-90, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18553460

ABSTRACT

Survivin is recognized as an attractive target in cancer therapy because of its selective overexpression in the majority of tumors. Upregulated expression of this protein correlates with increased tumor grade, recurrence risk and decreased cancer patients survival. In this study, we assessed the efficacy of two survivin-specific small interfering RNA (siRNA) constructs to inhibit T47D human breast cancer cell growth. After siRNA transfection, T47D cells showed a significant reduction in proliferation and survival exhibiting clear signs of apoptosis. pSil_1 that targeted exon 1 exhibited a stronger inhibitory effect on cell growth, and increased cell apoptosis compared to pSil_30 that targeted exon 4. Cell apoptosis was found to be mediated by translocation of the mitochondrial apoptosis inducing factor (AIF), while no changes were observed in caspase-3 activation and Bid cleavage. Thus, silencing survivin expression using siRNA strategies represents a suitable therapeutic approach to selectively modulate the survival and growth of human breast cancer cells.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Gene Silencing/drug effects , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Caspases , Female , Humans , Inhibitor of Apoptosis Proteins , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Survivin , Transfection
16.
Article in English | MEDLINE | ID: mdl-29868490

ABSTRACT

The spirochete endoflagellum is a unique motility apparatus among bacteria. Despite its critical importance for pathogenesis, the full composition of the flagellum remains to be determined. We have recently reported that FcpA is a novel flagellar protein and a major component of the sheath of the filament of the spirochete Leptospira. By screening a library of random transposon mutants in the spirochete Leptospira biflexa, we found a motility-deficient mutant harboring a disruption in a hypothetical gene of unknown function. Here, we show that this gene encodes a surface component of the endoflagellar filament and is required for typical hook- and spiral-shaped ends of the cell body, coiled structure of the endoflagella, and high velocity phenotype. We therefore named the gene fcpB for flagellar-coiling protein B. fcpB is conserved in all members of the Leptospira genus, but not present in other organisms including other spirochetes. Complementation of the fcpB- mutant restored the wild-type morphology and motility phenotypes. Immunoblotting with anti-FcpA and anti-FcpB antisera and cryo-electron microscopy of the filament indicated that FcpB assembled onto the surface of the sheath of the filament and mostly located on the outer (convex) side of the coiled filament. We provide evidence that FcpB, together with FcpA, are Leptospira-specific novel components of the sheath of the filament, key determinants of the coiled and asymmetric structure of the endoflagella and are essential for high velocity. Defining the components of the endoflagella and their functions in these atypical bacteria should greatly enhance our understanding of the mechanisms by which these bacteria produce motility.


Subject(s)
Cell Movement/physiology , Flagella/physiology , Flagellin/metabolism , Leptospira/physiology , Amino Acid Sequence , Cell Movement/genetics , Cryoelectron Microscopy , DNA Transposable Elements , Flagella/ultrastructure , Flagellin/genetics , Leptospira/genetics , Leptospira/ultrastructure , Microscopy, Video , Phenotype , Sequence Alignment , Sequence Deletion
17.
Bio Protoc ; 7(16): e2510, 2017 Aug 20.
Article in English | MEDLINE | ID: mdl-34541173

ABSTRACT

We have developed protocols to generate site-specific variants of the histidine-kinase DesK and its cognate response regulator DesR, conducive to trapping different signaling states of the proteins. Co-expression of both partners in E. coli, ensuring an excess of the regulator, was essential for soluble production of the DesK:DesR complexes and further purification. The 3D structures of the complex trapped in the phosphotransferase and in the phosphatase reaction steps, were solved by X-ray crystallography using molecular replacement. The solution was not trivial, and we found that in silico-generated models used as search probes, were instrumental to succeeding in placing a large portion of the complex in the asymmetric unit. Electron density maps were then clear enough to allow for manual model building attaining complete atomic models. These methods contribute to tackling a major challenge in the bacterial signaling field, namely obtaining stable kinase:regulator complexes, in distinct conformational states, amenable for high-resolution crystallographic studies.

18.
Acta Crystallogr F Struct Biol Commun ; 73(Pt 3): 123-129, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28291747

ABSTRACT

The protein FcpA is a unique component of the flagellar filament of spirochete bacteria belonging to the genus Leptospira. Although it plays an essential role in translational motility and pathogenicity, no structures of FcpA homologues are currently available in the PDB. Its three-dimensional structure will unveil the novel motility mechanisms that render pathogenic Leptospira particularly efficient at invading and disseminating within their hosts, causing leptospirosis in humans and animals. FcpA from L. interrogans was purified and crystallized, but despite laborious attempts no useful X ray diffraction data could be obtained. This challenge was solved by expressing a close orthologue from the related saprophytic species L. biflexa. Three different crystal forms were obtained: a primitive and a centred monoclinic form, as well as a hexagonal variant. All forms diffracted X-rays to suitable resolutions for crystallographic analyses, with the hexagonal type typically reaching the highest limits of 2.0 Šand better. A variation of the quick-soaking procedure resulted in an iodide derivative that was instrumental for single-wavelength anomalous diffraction methods.


Subject(s)
Bacterial Proteins/chemistry , Flagella/chemistry , Leptospira interrogans/chemistry , Leptospira/chemistry , Plasmids/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Flagella/metabolism , Gene Expression , Leptospira/metabolism , Leptospira interrogans/metabolism , Plasmids/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
19.
J Histochem Cytochem ; 54(3): 317-28, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16260590

ABSTRACT

Mucin O-glycosylation is characterized in cancer by aberrant expression of immature carbohydrate structures (Tn, T, and sialyl-Tn antigens). The UDP-N-acetyl-D-galactosamine: polypeptide N-acetylgalactosaminyltransferases (ppGalNAc-T) family enzymes regulate the initial steps of mucin O-glycosylation and could be responsible for the altered glycosylation observed in cancer. Considering that we recently found the ppGalNAc-T6 mRNA expressed in breast carcinomas, we produced a highly specific monoclonal antibody (MAb T6.3) to assess the expression profile of ppGalNAc-T6 protein product in breast tissues. The expression of ppGalNAc-T6 by breast carcinoma cells was confirmed on MCF-7 and T47D cell lines. In formalin-fixed tissues, ppGalNAc-T6 expression was observed in 60/74 (81%) breast cancers, 21/23 (91.3%) adjacent ductal carcinoma in situ (DCIS), 4/20 benign breast lesions (2/2 sclerosing adenosis and 2/13 fibroadenoma), and in 0/5 normal breast samples. We observed a statistically significant association of ppGalNAc-T6 expression with T1 tumor stage. This fact, as well as the observation that ppGalNAc-T6 was strongly expressed in sclerosing adenosis and in most DCIS, suggests that ppGalNAc-T6 expression could be an early event during human breast carcinogenesis. Considering that an abnormal O-glycosylation greatly contributes to the phenotype and biology of breast cancer cells, ppGalNAc-T6 expression could provide new insights about breast cancer glycobiology.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , N-Acetylgalactosaminyltransferases/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal , Breast Diseases/enzymology , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Lobular/enzymology , Carcinoma, Papillary/enzymology , Female , Humans , Immunohistochemistry , Mammary Glands, Human/enzymology , Metaplasia , Mice , Middle Aged , N-Acetylgalactosaminyltransferases/immunology
20.
Elife ; 52016 12 12.
Article in English | MEDLINE | ID: mdl-27938660

ABSTRACT

Two-component systems (TCS) are protein machineries that enable cells to respond to input signals. Histidine kinases (HK) are the sensory component, transferring information toward downstream response regulators (RR). HKs transfer phosphoryl groups to their specific RRs, but also dephosphorylate them, overall ensuring proper signaling. The mechanisms by which HKs discriminate between such disparate directions, are yet unknown. We now disclose crystal structures of the HK:RR complex DesK:DesR from Bacillus subtilis, comprising snapshots of the phosphotransfer and the dephosphorylation reactions. The HK dictates the reactional outcome through conformational rearrangements that include the reactive histidine. The phosphotransfer center is asymmetric, poised for dissociative nucleophilic substitution. The structural bases of HK phosphatase/phosphotransferase control are uncovered, and the unexpected discovery of a dissociative reactional center, sheds light on the evolution of TCS phosphotransfer reversibility. Our findings should be applicable to a broad range of signaling systems and instrumental in synthetic TCS rewiring.


Subject(s)
Bacillus subtilis/enzymology , Histidine Kinase/chemistry , Histidine Kinase/metabolism , Signal Transduction , Transcription Factors/chemistry , Transcription Factors/metabolism , Crystallography, X-Ray , Models, Molecular , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL