Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Publication year range
1.
Biochem Biophys Res Commun ; 717: 150057, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38718568

ABSTRACT

Leptospirosis is a widespread zoonotic infectious disease of human and veterinary concern caused by pathogenic spirochetes of the genus Leptospira. To date, little progress towards understanding leptospiral pathogenesis and identification of virulence factors has been made, which is the main bottleneck for developing effective measures against the disease. Some leptospiral proteins, including LipL32, Lig proteins, LipL45, and LipL21, are being considered as potential virulence factors or vaccine candidates. However, their function remains to be established. LipL45 is the most expressed membrane lipoprotein in leptospires, upregulated when the bacteria are transferred to temperatures resembling the host, expressed during infection, suppressed after culture attenuation, and known to suffer processing in vivo and in vitro, generating fragments. Based on body of evidence, we hypothesized that the LipL45 processing might occur by an auto-cleavage event, deriving two fragments. The results presented here, based on bioinformatics, structure modeling analysis, and experimental data, corroborate that LipL45 processing probably includes a self-catalyzed non-proteolytic event and suggest the participation of LipL45 in cell-surface signaling pathways, as the protein shares structural similarities with bacterial sigma regulators. Our data indicate that LipL45 might play an important role in response to environmental conditions, with possible function in the adaptation to the host.


Subject(s)
Leptospira , Lipoproteins , Lipoproteins/metabolism , Lipoproteins/chemistry , Lipoproteins/genetics , Leptospira/metabolism , Leptospira/chemistry , Sigma Factor/metabolism , Sigma Factor/chemistry , Sigma Factor/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Models, Molecular , Leptospirosis/metabolism , Leptospirosis/microbiology
2.
Proteins ; 88(7): 840-852, 2020 07.
Article in English | MEDLINE | ID: mdl-31998983

ABSTRACT

Leucine rich repeats (LRRs) are present in over 430 000 proteins from viruses to eukaryotes. The LRRs are 20 to 30 residues long and occur in tandem. Individual LRRs are separated into a highly conserved segment with the consensus of LxxLxLxxNxL or LxxLxLxxNxxL (HCS) and a variable segment (VS). In LRRs parallel stacking of short ß-strands (at positions 3-5 in HCS) form a super helix arrangement called a solenoid structure. Many classes have been recognized. All three classes of Plant specific, Leptospira-like, and SDS22-like LRRs which are 24, 23, and 22 residues long, respectively, form a 3(10)-helix in the VS part. To get a deeper understanding of sequence, structure correlations in LRR structures, we utilized secondary structure assignment and HELFIT analysis (calculating helix axis, pitch, radius, residues per turn, and handedness) based on the atomic coordinates in crystal structures of 43 LRR proteins. We also defined three structural parameters using the three unit vectors of the helix axes of 3(10)-helix, ß-turn, and LRR-domain calculated by HELFIT. The combination of the secondary structure assignment and HELFIT reveals that their LRRs adopt unique super secondary structures consisting of a 3(10)-helix and one or two Type I ß-turns. We propose one structural parameter as a geometrical invariant of LRR solenoid structures. The common LxxLxxL sequence (where "L" is Leu, Ile, Val, Phe or Cys) in the three classes is an essential determinant for the super secondary structures providing a medium range interaction.


Subject(s)
Leucine/chemistry , Protein Phosphatase 1/chemistry , Repetitive Sequences, Amino Acid , Animals , Conserved Sequence , Crystallography, X-Ray , Humans , Leptospira/chemistry , Models, Molecular , Plants/chemistry , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Structure-Activity Relationship , Viruses/chemistry
3.
Nature ; 515(7527): 448-452, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25186729

ABSTRACT

SWEETs and their prokaryotic homologues are monosaccharide and disaccharide transporters that are present from Archaea to plants and humans. SWEETs play crucial roles in cellular sugar efflux processes: that is, in phloem loading, pollen nutrition and nectar secretion. Their bacterial homologues, which are called SemiSWEETs, are among the smallest known transporters. Here we show that SemiSWEET molecules, which consist of a triple-helix bundle, form symmetrical, parallel dimers, thereby generating the translocation pathway. Two SemiSWEET isoforms were crystallized, one in an apparently open state and one in an occluded state, indicating that SemiSWEETs and SWEETs are transporters that undergo rocking-type movements during the transport cycle. The topology of the triple-helix bundle is similar yet distinct to that of the basic building block of animal and plant major facilitator superfamily (MFS) transporters (for example, GLUTs and SUTs). This finding indicates two possibilities: that SWEETs and MFS transporters evolved from an ancestral triple-helix bundle or that the triple-helix bundle represents convergent evolution. In SemiSWEETs and SWEETs, two triple-helix bundles are arranged in a parallel configuration to produce the 6- and 6 + 1-transmembrane-helix pores, respectively. In the 12-transmembrane-helix MFS transporters, four triple-helix bundles are arranged into an alternating antiparallel configuration, resulting in a much larger 2 × 2 triple-helix bundle forming the pore. Given the similarity of SemiSWEETs and SWEETs to PQ-loop amino acid transporters and to mitochondrial pyruvate carriers (MPCs), the structures characterized here may also be relevant to other transporters in the MtN3 clan. The insight gained from the structures of these transporters and from the analysis of mutations of conserved residues will improve the understanding of the transport mechanism, as well as allow comparative studies of the different superfamilies involved in sugar transport and the evolution of transporters in general.


Subject(s)
Bacterial Proteins/chemistry , Leptospira/chemistry , Monosaccharide Transport Proteins/chemistry , Vibrio/chemistry , Arabidopsis/chemistry , Bacterial Proteins/metabolism , Crystallography, X-Ray , Evolution, Molecular , Glucose/metabolism , Leptospira/genetics , Models, Molecular , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Movement , Protein Conformation , Protein Multimerization , Structure-Activity Relationship
4.
Proteins ; 86(7): 712-722, 2018 07.
Article in English | MEDLINE | ID: mdl-29633350

ABSTRACT

Proteomes of pathogenic Leptospira interrogans and L. borgpetersenii and the saprophytic L. biflexa were filtered through computational tools to identify Outer Membrane Proteins (OMPs) that satisfy the required biophysical parameters for their presence on the outer membrane. A total of 133, 130, and 144 OMPs were identified in L. interrogans, L. borgpetersenii, and L. biflexa, respectively, which forms approximately 4% of proteomes. A holistic analysis of transporting and pathogenic characteristics of OMPs together with Clusters of Orthologous Groups (COGs) among the OMPs and their distribution across 3 species was made and put forward a set of 21 candidate OMPs specific to pathogenic leptospires. It is also found that proteins homologous to the candidate OMPs were also present in other pathogenic species of leptospires. Six OMPs from L. interrogans and 2 from L. borgpetersenii observed to have similar COGs while those were not found in any intermediate or saprophytic forms. These OMPs appears to have role in infection and pathogenesis and useful for anti-leptospiral strategies.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Leptospira/chemistry , Proteome , Bacterial Outer Membrane Proteins/metabolism , Biological Transport , Databases, Protein , Leptospira/metabolism , Lipoproteins/metabolism
5.
Biochemistry ; 55(44): 6138-6149, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27749038

ABSTRACT

We explored the photoisomerization mechanisms in novel homologues of photoactive yellow protein (PYP) from Leptospira biflexa (Lbif) to identify conserved features and functional diversity in the primary photochemistry of this family of photoreceptors. In close agreement with the prototypical PYP from Halorhodospira halophila (Hhal), we observe excited-state absorbance near 375 nm and stimulated emission near 500 nm, with triphasic excited-state decay. While the excited-state decay for Lbif PYP is the slowest among those of known PYPs due to the redistribution of the amplitudes of the three decay components, the quantum yield for productive photocycle entry is very similar to that of Hhal PYP. Pro68 is highly conserved in PYPs and is important for the high photochemical quantum yield in Hhal PYP, but this residue is Ile in wild-type Lbif PYP. The level of photoproduct formation is slightly increased in I68P Lbif PYP, indicating that this residue regulates the photochemical quantum yield in the entire PYP family. Lbif PYP also exhibited a blue-shifted photoproduct previously undiscovered in ultrafast studies of PYP, which we have named pUV. We posit that pUV is a detour in the PYP photocycle with a twisted protonated pCAH configuration. Cryokinetic experiments with Hhal PYP confirmed the presence of pUV, but the population of this state in room-temperature ultrafast experiments is very small. These results resolve the long-standing inconsistency in the literature regarding the existence of a bifurcation in the room-temperature photocycle of PYP.


Subject(s)
Bacterial Proteins/chemistry , Halorhodospira halophila/chemistry , Leptospira/chemistry , Photoreceptors, Microbial/chemistry , Hydrogen Bonding , Spectrophotometry, Ultraviolet
6.
Curr Top Microbiol Immunol ; 387: 187-221, 2015.
Article in English | MEDLINE | ID: mdl-25388136

ABSTRACT

The outer membrane (OM) is the front line of leptospiral interactions with their environment and the mammalian host. Unlike most invasive spirochetes, pathogenic leptospires must be able to survive in both free-living and host-adapted states. As organisms move from one set of environmental conditions to another, the OM must cope with a series of conflicting challenges. For example, the OM must be porous enough to allow nutrient uptake, yet robust enough to defend the cell against noxious substances. In the host, the OM presents a surface decorated with adhesins and receptors for attaching to, and acquiring, desirable host molecules such as the complement regulator, Factor H.Factor H. On the other hand, the OM must enable leptospires to evade detection by the host's immune system on their way from sites of invasion through the bloodstream to the protected niche of the proximal tubule. The picture that is emerging of the leptospiral OM is that, while it shares many of the characteristics of the OMs of spirochetes and Gram-negative bacteria, it is also unique and different in ways that make it of general interest to microbiologists. For example, unlike most other pathogenic spirochetes, the leptospiral OM is rich in lipopolysaccharide (LPS). Leptospiral LPS is similar to that of Gram-negative bacteria but has a number of unique structural features that may explain why it is not recognized by the LPS-specific Toll-like receptor 4 of humans. As in other spirochetes, lipoproteins are major components of the leptospiral OM, though their roles are poorly understood. The functions of transmembrane outer membrane proteins (OMPs) in many cases are better understood, thanks to homologies with their Gram-negative counterparts and the emergence of improved genetic techniques. This chapter will review recent discoveries involving the leptospiral OM and its role in leptospiral physiology and pathogenesis.


Subject(s)
Cell Membrane/chemistry , Leptospira/chemistry , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/physiology , Immunity, Innate , Leptospira/ultrastructure , Lipopolysaccharides/chemistry , Lipopolysaccharides/physiology
7.
J Biol Chem ; 289(40): 27966-78, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25128527

ABSTRACT

The committed step of leucine biosynthesis, converting acetyl-CoA and α-ketoisovalerate into α-isopropylmalate, is catalyzed by α-isopropylmalate synthase (IPMS), an allosteric enzyme subjected to feedback inhibition by the end product L-leucine. We characterized the short form IPMS from Leptospira biflexa (LbIPMS2), which exhibits a catalytic activity comparable with that of the long form IPMS (LbIPMS1) and has a similar N-terminal domain followed by subdomain I and subdomain II but lacks the whole C-terminal regulatory domain. We found that partial deletion of the regulatory domain of LbIPMS1 resulted in a loss of about 50% of the catalytic activity; however, when the regulatory domain was deleted up to Arg-385, producing a protein that is almost equivalent to the intact LbIPMS2, about 90% of the activity was maintained. Moreover, in LbIPMS2 or LbIPMS1, further deletion of several residues from the C terminus of subdomain II significantly impaired or completely abolished the catalytic activity, respectively. These results define a complete and independently functional catalytic module of IPMS consisting of both the N-terminal domain and the two subdomains. Structural comparison of LbIPMS2 and the Mycobacterium tuberculosis IPMS revealed two different conformations of subdomain II that likely represent two substrate-binding states related to cooperative catalysis. The biochemical and structural analyses together with the previously published hydrogen-deuterium exchange data led us to propose a conformation transition mechanism for feedback inhibition mediated by subdomains I and II that might associated with alteration of the binding affinity toward acetyl-CoA.


Subject(s)
2-Isopropylmalate Synthase/chemistry , 2-Isopropylmalate Synthase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Feedback, Physiological , Leptospira/enzymology , 2-Isopropylmalate Synthase/genetics , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Binding Sites , Crystallography, X-Ray , Kinetics , Leptospira/chemistry , Leptospira/genetics , Leucine/chemistry , Leucine/metabolism , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Alignment , Substrate Specificity
8.
Infect Immun ; 83(3): 888-97, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534939

ABSTRACT

Upon infection, pathogenic Leptospira species bind several complement regulators in order to overcome host innate immunity. We previously characterized a 20-kDa leptospiral surface protein which interacts with C4b binding protein (C4BP): leptospiral complement regulator-acquiring protein A (LcpA). Here we show that LcpA also interacts with human factor H (FH), which remains functionally active once bound to the protein. Antibodies directed against short consensus repeat 20 (SCR20) inhibited binding of FH to LcpA by approximately 90%, thus confirming that this particular domain is involved in the interaction. We have also shown for the first time that leptospires bind human vitronectin and that the interaction is mediated by LcpA. Coincubation with heparin blocked LcpA-vitronectin interaction in a dose-dependent manner, strongly suggesting that binding may occur through the heparin binding domains of vitronectin. LcpA also bound to the terminal pathway component C9 and inhibited Zn(2+)-induced polymerization and membrane attack complex (MAC) formation. Competitive binding assays indicated that LcpA interacts with C4BP, FH, and vitronectin through distinct sites. Taken together, our findings indicate that LcpA may play a role in leptospiral immune evasion.


Subject(s)
Bacterial Proteins/chemistry , Leptospira interrogans/chemistry , Leptospira/chemistry , Peptide Fragments/chemistry , Vitronectin/chemistry , Antibodies, Monoclonal/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/immunology , Binding Sites , Binding, Competitive , Complement Activation , Complement C4b-Binding Protein/chemistry , Complement C4b-Binding Protein/immunology , Complement C9/chemistry , Complement C9/immunology , Complement Factor H/chemistry , Complement Factor H/immunology , Complement Membrane Attack Complex/chemistry , Heparin/chemistry , Humans , Immune Evasion , Leptospira/immunology , Leptospira/pathogenicity , Leptospira interrogans/immunology , Leptospira interrogans/pathogenicity , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/immunology , Protein Binding , Vitronectin/immunology , Zinc/chemistry
9.
BMC Microbiol ; 15: 244, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26518696

ABSTRACT

BACKGROUND: Lipopolysaccharides (LPS) are complex, amphipathic biomolecules that constitute the major surface component of Gram-negative bacteria. Leptospira, unlike other human-pathogenic spirochetes, produce LPS, which is fundamental to the taxonomy of the genus, involved in host-adaption and also the target of diagnostic antibodies. Despite its significance, little is known of Leptospira LPS composition and carbohydrate structure among different serovars. RESULTS: LPS from Leptospira interrogans serovar Copenhageni strain L1-130, a pathogenic species, and L. licerasiae serovar Varillal strain VAR 010, an intermediately pathogenic species, were studied. LPS prepared from aqueous and phenol phases were analyzed separately. L. interrogans serovar Copenhageni has additional sugars not found in L. licerasiae serovar Varillal, including fucose (2.7%), a high amount of GlcNAc (12.3%), and two different types of dideoxy HexNAc. SDS-PAGE indicated that L. interrogans serovar Copenhageni LPS had a far higher molecular weight and complexity than that of L. licerasiae serovar Varillal. Chemical composition showed that L. interrogans serovar Copenhageni LPS has an extended O-antigenic polysaccharide consisting of sugars, not present in L. licerasiae serovar Varillal. Arabinose, xylose, mannose, galactose and L-glycero-D-mannoheptose were detected in both the species. Fatty acid analysis by gas chromatography-mass spectrometry (GC-MS) showed the presence of hydroxypalmitate (3-OH-C16:0) only in L. interrogans serovar Copenhageni. Negative staining electron microscopic examination of LPS showed different filamentous morphologies in L. interrogans serovar Copenhageni vs. L. licerasiae serovar Varillal. CONCLUSIONS: This comparative biochemical analysis of pathogenic and intermediately pathogenic Leptospira LPS reveals important carbohydrate and lipid differences that underlie future work in understanding the mechanisms of host-adaptation, pathogenicity and vaccine development in leptospirosis.


Subject(s)
Leptospira/chemistry , Lipopolysaccharides/analysis , Carbohydrates/analysis , Electrophoresis, Polyacrylamide Gel , Fatty Acids/analysis , Gas Chromatography-Mass Spectrometry , Leptospira/pathogenicity , Leptospira/ultrastructure , Microscopy, Electron, Transmission , Molecular Weight
10.
J Infect Dis ; 209(6): 876-86, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24163418

ABSTRACT

Leptospirosis is an infectious disease of public health importance. To successfully colonize the host, pathogens have evolved multiple strategies to escape the complement system. Here we demonstrate that the culture supernatant of pathogenic but not saprophytic Leptospira inhibit the three complement pathways. We showed that the proteolytic activity in the supernatants of pathogenic strains targets the central complement molecule C3 and specific proteins from each pathway, such as factor B, C2, and C4b. The proteases cleaved α and ß chains of C3 and work in synergy with host regulators to inactivate C3b. Proteolytic activity was inhibited by 1,10-phenanthroline, suggesting the participation of metalloproteases. A recombinant leptospiral metalloprotease from the thermolysin family cleaved C3 in serum and could be one of the proteases responsible for the supernatant activity. We conclude that pathogenic leptospiral proteases can deactivate immune effector molecules and represent potential targets to the development of new therapies in leptospirosis.


Subject(s)
Bacterial Proteins/metabolism , Complement C3/metabolism , Leptospira/immunology , Leptospirosis/microbiology , Peptide Hydrolases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Complement Pathway, Classical , Humans , Immune Evasion , Leptospira/chemistry , Leptospira/enzymology , Leptospira/pathogenicity , Leptospirosis/immunology , Models, Biological , Peptide Hydrolases/immunology , Peptide Hydrolases/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thermolysin/chemistry , Thermolysin/metabolism
11.
Article in Russian | MEDLINE | ID: mdl-26950986

ABSTRACT

AIM: Creation of a classification model of Leptospira spp. serovar model using ClinProTools 3.0 software and evaluation of use of MALDI-TOF MS as a method of quality control of reference strains of leptospira. MATERIALS AND METHODS: 10 reference strains of Leptospira spp. were used in the study according to microscopic agglutination reaction from the collection of Pasteur RIEM. All the strains were cultivated for 10 days in Terskikh medium at 28 degrees C. Cell extracts were obtained by ethanol/formic acid method. α-cyano-4-hydroxycinnamic acid solution was used as a matrix. Mass-spectra were obtained in Microflex mass-spectrometer (Bruker Daltonics, Germany). External validation of the test-model was carried out using novel spectra of every reference strain during their repeated reseeding. RESULTS: Values of cross-validation and confirmatory ability of the optimal model, built on a genetic algorithm, was 99.14 and 100%, respectively. This model contained 11 biomarker peaks (m/z 2959, 3447, 3548, 3764, 3895, 5221, 5917, 6173, 6701, 7013, 8364) for serovar classification. Results of the external validation have shown a 100% correct classification in serovar classesin Sejroe, Ballum, Tarassovi; Copenhageni, Mozdoc, Grippotyphosa and Patoc, that indicates a high prognostic ability of the model in these classes. However, data from verification matrix have shown, that 50%.of the spectra from Canicola and Pomona serovars were classified as Patoc class, that could be associated with cross serological activity of Patoc serovar L. biflexa with pathogenic leptospirae. CONCLUSION: MALDI-TOF mass-spectrometry method combined with building and using the classification model could be a useful instrument for intra-laboratory control of leptospira reseeding.


Subject(s)
Coumaric Acids/chemistry , Leptospira/classification , Phylogeny , Serotyping/methods , Software , Culture Media/chemistry , Humans , Leptospira/chemistry , Leptospira/growth & development , Leptospira/metabolism , Principal Component Analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
Curr Microbiol ; 68(4): 524-30, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24343176

ABSTRACT

The C-terminal region of the Leptospiral immunoglobulin-like A protein (LigA) contains six carboxy-terminal Ig-like repeat domains (LigANI). Subunit vaccine preparations based on recombinant LigANI produced in Escherichia coli, are promising vaccine candidates, albeit with variable efficacy. In the present study, LigANI was expressed in the methylotrophic yeast Pichia pastoris using a 12 L bioreactor to produce mannosylated LigANI (mLigANI) for use in a vaccine preparation against leptospirosis. Hamsters immunized with a mLigANI vaccine preparation produced a significant IgG antibody response (P < 0.001) and were protected (83.3 %; P < 0.001) against lethal challenge with 36× LD50 of a virulent strain of L. interrogans serovar Copenhageni. A vaccine preparation based on demannosylated mLigANI (nmLigANI) elicited an immune response in hamsters, but did not afford protection. The production of mLigANI in bioreactor by P. pastoris yielded ~50 mg L(-1) of recombinant protein. P. pastoris is a potential platform for the production of leptospiral antigens on an industrial scale. The results demonstrate that LigANI secreted by P. pastoris on mannosylated form (mLigANI) protect hamsters as subunit vaccine of L. interrogans lethal infection.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Bioreactors , Leptospira/chemistry , Leptospirosis/prevention & control , Recombinant Proteins/immunology , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cricetinae , Female , Leptospira/genetics , Leptospirosis/immunology , Pichia/genetics , Pichia/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Survival Analysis
13.
J Biol Chem ; 287(7): 4826-34, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22147698

ABSTRACT

LipL32 is the most abundant outer membrane protein from pathogenic Leptospira and has been shown to bind extracellular matrix (ECM) proteins as well as Ca(2+). Recent crystal structures have been obtained for the protein in the apo- and Ca(2+)-bound forms. In this work, we produced three LipL32 mutants (D163-168A, Q67A, and S247A) and evaluated their ability to interact with Ca(2+) and with ECM glycoproteins and human plasminogen. The D163-168A mutant modifies aspartate residues involved in Ca(2+) binding, whereas the other two modify residues in a cavity on the other side of the protein structure. Loss of calcium binding in the D163-D168A mutant was confirmed using intrinsic tryptophan fluorescence, circular dichroism, and thermal denaturation whereas the Q67A and S247A mutants presented the same Ca(2+) affinity as the wild-type protein. We then evaluated if Ca(2+) binding to LipL32 would be crucial for its interaction with collagen type IV and plasma proteins fibronectin and plasminogen. Surprisingly, the wild-type protein and all three mutants, including the D163-168A variant, bound to these ECM proteins with very similar affinities, both in the presence and absence of Ca(2+) ions. In conclusion, calcium binding to LipL32 may be important to stabilize the protein, but is not necessary to mediate interaction with host extracellular matrix proteins.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Calcium/metabolism , Collagen Type IV/metabolism , Fibronectins/metabolism , Leptospira/metabolism , Lipoproteins/metabolism , Plasminogen/metabolism , Amino Acid Substitution , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Cations, Divalent , Collagen Type IV/chemistry , Collagen Type IV/genetics , Fibronectins/chemistry , Fibronectins/genetics , Humans , Leptospira/chemistry , Leptospira/genetics , Lipoproteins/chemistry , Lipoproteins/genetics , Mutation, Missense , Plasminogen/chemistry , Plasminogen/genetics , Protein Binding , Protein Stability
14.
J Biomol Struct Dyn ; 41(20): 10347-10367, 2023 12.
Article in English | MEDLINE | ID: mdl-36510668

ABSTRACT

Leptospirosis is one of the neglected zoonosis, affecting human and animal populations worldwide. Reliable effective therapeutics and concerns to look for more research into the molecular analysis of its genome is therefore needed. In the genomic pool of the Leptospira interrogans many hypothetical proteins are still uncharacterized. In the current research, we performed extensive in silico analysis to prioritize the potential hypothetical proteins of L. interrogans serovar Copenhageni via stepwise reducing the available hypothetical proteins (Total 3606) of the assembly to only 15, based on non-homologous to homosapien, essential, functional, virulent, cellular localization. Out of them, only two proteins WP_000898918.1 (Hypothetical Protein 1) & WP_001014594.1 (Hypothetical Protein 2) were found druggable and involved in protein-protein interaction network. The 3 D structures of these two target proteins were predicted via ab initio homology modeling followed by structures refinement and validation, as no structures were available till date. The analysis also revealed that the functional domains, families and protein-protein interacting partners identified in both proteins are crucial for the survival of the bacteria. The binding cavities were predicted for both the proteins through blind and specific protein-ligand docking with their respective ligands and inhibitors and were found to be in accordance with the druggable sites predicted by DoGSiteScorer. The docking interactions were found within the active functional domains for both the proteins while for Hypothetical Protein 2, the same residues were involved in interactions with Cytidine-5'-triphosphate in blind and specific docking. Furthermore, the simulations of molecular dynamics and free binding energy revealed the stable substrate binding and efficient binding energies, and were in accordance to our docking results. The work predicted two unique hypothetical proteins of L. interrogans as a potential druggable targets for designing of inhibitors for them.Communicated by Ramaswamy H. Sarma.


Subject(s)
Leptospira interrogans , Leptospira , Leptospirosis , Animals , Humans , Serogroup , Leptospira interrogans/genetics , Leptospira interrogans/metabolism , Leptospirosis/drug therapy , Leptospirosis/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Computational Biology , Leptospira/chemistry , Leptospira/metabolism
15.
FEBS J ; 290(18): 4513-4532, 2023 09.
Article in English | MEDLINE | ID: mdl-37243454

ABSTRACT

Leptospirosis is a commonly overlooked zoonotic disease that occurs in tropical and subtropical regions. Recent studies have divided the Leptospira spp. into three groups based on virulence, including pathogenic, intermediate, and saprophytic species. Pathogenic species express a protein family with leucine-rich repeat (LRR) domains, which are less expressed or absent in nonpathogenic species, highlighting the importance of this protein family in leptospirosis. However, the role of LRR domain proteins in the pathogenesis of leptospirosis is still unknown and requires further investigation. In this study, the 3D structure of LSS_01692 (rLRR38) was obtained using X-ray crystallography at a resolution of 3.2 Å. The results showed that rLRR38 forms a typical horseshoe structure with 11 α-helices and 11 ß-sheets and an antiparallel dimeric structure. The interactions of rLRR38 with extracellular matrix and cell surface receptors were evaluated using ELISA and single-molecule atomic force microscopy. The results showed that rLRR38 interacted with fibronectin, collagen IV, and Toll-like receptor 2 (TLR2). Incubating HK2 cells with rLRR38 induced two downstream inflammation responses (IL-6 and MCP-1) in the TLR2 signal transduction pathway. The TLR2-TLR1 complex showed the most significant upregulation effects under rLRR38 treatment. Inhibitors also significantly inhibited nuclear factor κB and mitogen-activated protein kinases signals transduction under rLRR38 stimulation. In conclusion, rLRR38 was determined to be a novel LRR domain protein in 3D structure and demonstrated as a TLR2-binding protein that induces inflammatory responses. These structural and functional studies provide a deeper understanding of the pathogenesis of leptospirosis.


Subject(s)
Leptospira , Leptospirosis , Humans , Leptospira/genetics , Leptospira/chemistry , Leptospira/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Signal Transduction , Leptospirosis/genetics , Leptospirosis/metabolism
16.
BMC Microbiol ; 12: 185, 2012 Aug 27.
Article in English | MEDLINE | ID: mdl-22925589

ABSTRACT

BACKGROUND: In this study mass spectrometry was used for evaluating extracted leptospiral protein samples and results were compared with molecular typing methods. For this, an extraction protocol for Leptospira spp. was independently established in two separate laboratories. Reference spectra were created with 28 leptospiral strains, including pathogenic, non-pathogenic and intermediate strains. This set of spectra was then evaluated on the basis of measurements with well-defined, cultured leptospiral strains and with 16 field isolates of veterinary or human origin. To verify discriminating peaks for the applied pathogenic strains, statistical analysis of the protein spectra was performed using the software tool ClinProTools. In addition, a dendrogram of the reference spectra was compared with phylogenetic trees of the 16S rRNA gene sequences and multi locus sequence typing (MLST) analysis. RESULTS: Defined and reproducible protein spectra using MALDI-TOF MS were obtained for all leptospiral strains. Evaluation of the newly-built reference spectra database allowed reproducible identification at the species level for the defined leptospiral strains and the field isolates. Statistical analysis of three pathogenic genomospecies revealed peak differences at the species level and for certain serovars analyzed in this study. Specific peak patterns were reproducibly detected for the serovars Tarassovi, Saxkoebing, Pomona, Copenhageni, Australis, Icterohaemorrhagiae and Grippotyphosa. Analysis of the dendrograms of the MLST data, the 16S rRNA sequencing, and the MALDI-TOF MS reference spectra showed comparable clustering. CONCLUSIONS: MALDI-TOF MS analysis is a fast and reliable method for species identification, although Leptospira organisms need to be produced in a time-consuming culture process. All leptospiral strains were identified, at least at the species level, using our described extraction protocol. Statistical analysis of the three genomospecies L. borgpetersenii, L. interrogans and L. kirschneri revealed distinctive, reproducible differentiating peaks for seven leptospiral strains which represent seven serovars. Results obtained by MALDI-TOF MS were confirmed by MLST and 16S rRNA gene sequencing.


Subject(s)
Bacteriological Techniques/methods , Leptospira/chemistry , Leptospira/classification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Genes, rRNA , Genotype , Humans , Leptospira/genetics , Molecular Sequence Data , RNA, Ribosomal, 16S/genetics , Reproducibility of Results , Sequence Analysis, DNA
17.
Front Cell Infect Microbiol ; 12: 966370, 2022.
Article in English | MEDLINE | ID: mdl-36081769

ABSTRACT

Extracellular proteolytic enzymes are produced by a variety of pathogenic microorganisms, and contribute to host colonization by modulating virulence. Here, we present a first characterization of leptolysin, a Leptospira metalloprotease of the pappalysin family identified in a previous exoproteomic study. Comparative molecular analysis of leptolysin with two other pappalysins from prokaryotes, ulilysin and mirolysin, reveals similarities regarding calcium, zinc, and arginine -binding sites conservation within the catalytic domain, but also discloses peculiarities. Variations observed in the primary and tertiary structures may reflect differences in primary specificities. Purified recombinant leptolysin of L. interrogans was obtained as a ~50 kDa protein. The protease exhibited maximal activity at pH 8.0 and 37°C, and hydrolytic activity was observed in the presence of different salts with maximum efficiency in NaCl. Substrate specificity was assessed using a small number of FRET peptides, and showed a marked preference for arginine residues at the P1 position. L. interrogans leptolysin proteolytic activity on proteinaceous substrates such as proteoglycans and plasma fibronectin was also evaluated. All proteins tested were efficiently degraded over time, confirming the protease´s broad-spectrum activity in vitro. In addition, leptolysin induced morphological alterations on HK-2 cells, which may be partially attributed to extracellular matrix (ECM) degradation. Hemorrhagic foci were observed in the dorsal skin of mice intradermally injected with leptolysin, as a plausible consequence of ECM disarray and vascular endothelium glycocalyx damage. Assuming that leptospiral proteases play an important role in all stages of the infectious process, characterizing their functional properties, substrates and mechanisms of action is of great importance for therapeutic purposes.


Subject(s)
Leptospira , Metalloproteases , Animals , Arginine/metabolism , Leptospira/chemistry , Leptospira/metabolism , Leptospirosis , Metalloproteases/metabolism , Metalloproteases/pharmacology , Mice , Peptide Hydrolases/metabolism
18.
J Bacteriol ; 193(22): 6266-75, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21926230

ABSTRACT

Leptospira spp. are thin, highly motile, slow-growing spirochetes that can be distinguished from other bacteria on the basis of their unique helical shape. Defining the mechanisms by which these bacteria generate and maintain this atypical morphology should greatly enhance our understanding of the fundamental physiology of these pathogens. In this study, we showed that peptidoglycan sacculi from Leptospira spp. retain the helical shape of intact cells. Interestingly, the distribution of muropeptides was different from that in the Escherichia coli model, indicating that specific enzymes might be active on the peptidoglycan macromolecule. We could alter the shape of Leptospira biflexa with the broad-spectrum ß-lactam antibiotic penicillin G and with amdinocillin and aztreonam, which are ß-lactams that preferentially target penicillin-binding protein 2 (PBP2) and PBP3, respectively, in some species. Although genetic manipulations of Leptospira spp. are scarce, we were able to obtain mutants with alterations in genes encoding PBPs, including PBP3. Loss of this protein resulted in cell elongation. We also generated an L. biflexa strain that conditionally expresses MreB. Loss of the MreB function was correlated with morphological abnormalities such as a localized increased diameter and heterogeneous length. A prolonged depletion of MreB resulted in cell lysis, suggesting that this protein is essential. These findings indicate that important aspects of leptospiral cell morphology are determined by the cytoskeleton and the murein layer, thus providing a starting point for a better understanding of the morphogenesis in these atypical bacteria.


Subject(s)
Leptospira/chemistry , Leptospira/cytology , Peptidoglycan/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Leptospira/genetics , Leptospira/metabolism , Molecular Structure , Mutation , Peptidoglycan/metabolism
19.
Infect Immun ; 79(3): 1134-42, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21173310

ABSTRACT

Leptospirosis can activate inflammatory responses through Toll-like receptors (TLRs) and may cause renal tubulointerstitial fibrosis characterized by the accumulation of extracellular matrix (ECM). We have previously demonstrated that Leptospira santorosai serovar Shermani detergent extract stimulates ECM accumulation in vitro. The aim of this study was to examine the mechanistic basis of these previous observations and, in particular, to examine the potential involvement of TLRs. The addition of serovar Shermani detergent extract led to an increase in fibronectin gene expression and production. Inhibition of TLR2 but not TLR4 expression abrogated serovar Shermani detergent extract-mediated increases in fibronectin production. This response was also blocked by the knockdown of the gene expression of the TLR2 downstream transducers myeloid differentiation factor 88 (MyD88) and tumor necrosis factor receptor-associated factor 6 (TRAF6). Serovar Shermani detergent extract also activated nuclear factor-κB, and its inhibition by curcumin-attenuated serovar Shermani detergent extract induced increases in fibronectin production. These effects were also mimicked by the specific TLR2 agonist, Pam(3)CsK(4), a response that was also abrogated by the knockdown of MyD88 and TRAF6. Similarly, the administration of live leptospires to cells also induced fibronectin production that was blocked by inhibition of TLR2 and MyD88 expression. In conclusion, serovar Shermani detergent extract can induce fibronectin production through the TLR2-associated cascade, providing evidence of an association between TLRs and leptospirosis-mediated ECM deposition.


Subject(s)
Complex Mixtures/pharmacology , Fibronectins/biosynthesis , Leptospira/metabolism , Leptospirosis/metabolism , Toll-Like Receptor 2/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Blotting, Western , Cell Line , Detergents , Electrophoretic Mobility Shift Assay , Gene Knockdown Techniques , Humans , Immunohistochemistry , Kidney/metabolism , Kidney/pathology , Leptospira/chemistry , Myeloid Differentiation Factor 88/drug effects , Myeloid Differentiation Factor 88/metabolism , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , TNF Receptor-Associated Factor 6/drug effects , TNF Receptor-Associated Factor 6/metabolism
20.
Curr Microbiol ; 62(5): 1478-82, 2011 May.
Article in English | MEDLINE | ID: mdl-21305291

ABSTRACT

Leptospira infection involves the adhesion of the bacteria followed by invasion of the host crossing the extracellular matrix barrier. In an effort to understand the molecular mechanism of this process, the possibility of occurrence of matrix degrading enzymes from Leptospira was investigated. Zymographic analysis showed that the outer membrane of Leptospires contains a gelatinase of average molecular size of 46 kDa. The gelatinase exhibited maximum activity at neutral pH and was inhibited by metal chelators such as EGTA, EDTA, and Orthophenanthroline and was activated by calcium, magnesium, zinc, and copper, suggesting that it is a membrane-associated neutral matrix metalloproteinase. Analysis of the production of the enzyme by various serovars showed that the pathogenic serovars expressed significant amount of this enzyme while nonpathogenic forms either did not express or showed only very low activity, suggesting that this enzyme may be associated with pathogenesis of leptospirosis.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Gelatinases/metabolism , Leptospira/enzymology , Bacterial Outer Membrane Proteins/chemistry , Enzyme Stability , Gelatinases/chemistry , Gelatinases/genetics , Humans , Leptospira/chemistry , Leptospira/genetics , Leptospirosis/microbiology , Molecular Weight
SELECTION OF CITATIONS
SEARCH DETAIL