Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters

Publication year range
1.
J Bacteriol ; 206(2): e0034023, 2024 02 22.
Article in English | MEDLINE | ID: mdl-38214528

ABSTRACT

Glycerol utilization as a carbohydrate source by Borreliella burgdorferi, the Lyme disease spirochete, is critical for its successful colonization and persistence in the tick vector. The expression of the glpFKD (glp) operon, which encodes proteins for glycerol uptake/utilization, must be tightly regulated during the enzootic cycle of B. burgdorferi. Previous studies have established that the second messenger cyclic di-GMP (c-di-GMP) is required for the activation of glp expression, while an alternative sigma factor RpoS acts as a negative regulator for glp expression. In the present study, we report identification of a cis element within the 5´ untranslated region of glp that exerts negative regulation of glp expression. Further genetic screen of known and predicted DNA-binding proteins encoded in the genome of B. burgdorferi uncovered that overexpressing Borrelia host adaptation regulator (BadR), a known global regulator, dramatically reduced glp expression. Similarly, the badR mutant significantly increased glp expression. Subsequent electrophoretic mobility shift assay analyses demonstrated that BadR directly binds to this cis element, thereby repressing glp independent of RpoS-mediated repression. The efficiency of BadR binding was further assessed in the presence of c-di-GMP and various carbohydrates. This finding highlights multi-layered positive and negative regulatory mechanisms employed by B. burgdorferi to synchronize glp expression throughout its enzootic cycle.IMPORTANCEBorreliella burgdorferi, the Lyme disease pathogen, must modulate its gene expression differentially to adapt successfully to its two disparate hosts. Previous studies have demonstrated that the glycerol uptake and utilization operon, glpFKD, plays a crucial role in spirochetal survival within ticks. However, the glpFKD expression must be repressed when B. burgdorferi transitions to the mammalian host. In this study, we identified a specific cis element responsible for the repression of glpFKD. We further pinpointed Borrelia host adaptation regulator as the direct binding protein to this cis element, thereby repressing glpFKD expression. This discovery paves the way for a deeper exploration of how zoonotic pathogens sense distinct hosts and switch their carbon source utilization during transmission.


Subject(s)
Borrelia burgdorferi , Borrelia , Lyme Disease , Ticks , Animals , Borrelia/genetics , Borrelia/metabolism , Glycerol/metabolism , Host Adaptation , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Borrelia burgdorferi/genetics , Borrelia burgdorferi/metabolism , Operon , Gene Expression Regulation, Bacterial , Mammals/genetics , Mammals/metabolism
2.
PLoS Pathog ; 18(3): e1010338, 2022 03.
Article in English | MEDLINE | ID: mdl-35303742

ABSTRACT

Immune evasion facilitates survival of Borrelia, leading to infections like relapsing fever and Lyme disease. Important mechanism for complement evasion is acquisition of the main host complement inhibitor, factor H (FH). By determining the 2.2 Å crystal structure of Factor H binding protein A (FhbA) from Borrelia hermsii in complex with FH domains 19-20, combined with extensive mutagenesis, we identified the structural mechanism by which B. hermsii utilizes FhbA in immune evasion. Moreover, structure-guided sequence database analysis identified a new family of FhbA-related immune evasion molecules from Lyme disease and relapsing fever Borrelia. Conserved FH-binding mechanism within the FhbA-family was verified by analysis of a novel FH-binding protein from B. duttonii. By sequence analysis, we were able to group FH-binding proteins of Borrelia into four distinct phyletic types and identified novel putative FH-binding proteins. The conserved FH-binding mechanism of the FhbA-related proteins could aid in developing new approaches to inhibit virulence and complement resistance in Borrelia.


Subject(s)
Borrelia , Lyme Disease , Relapsing Fever , Borrelia/metabolism , Carrier Proteins/metabolism , Humans , Immune Evasion , Relapsing Fever/metabolism
3.
PLoS Pathog ; 18(3): e1010370, 2022 03.
Article in English | MEDLINE | ID: mdl-35286343

ABSTRACT

Borrelia species are amino acid auxotrophs that utilize di- and tri- peptides obtained through their oligopeptide transport system to supply amino acids for replicative growth during their enzootic cycles. However, Borrelia species from both the Lyme disease (LD) and relapsing fever (RF) groups harbor an amino acid transport and catabolism system, the Arginine Deiminase System (ADI), that could potentially augment intracellular L-arginine required for growth. RF spirochetes contain a "complete", four gene ADI (arcA, B, D, and C) while LD spirochetes harbor arcA, B, and sometimes D but lack arcC (encoding carbamate kinase). In this study, we evaluated the role of the ADI system in bacterial survival and virulence and discovered important differences in RF and LD ADIs. Both in vitro and in a murine model of infection, B. hermsii cells significantly reduced extracellular L-arginine levels and that reduction was dependent on arginine deiminase expression. Conversely, B. burgdorferi did not reduce the concentration of L-arginine during in vitro growth experiments nor during infection of the mammalian host, suggesting a fundamental difference in the ability to directly utilize L-arginine compared to B. hermsii. Further experiments using a panel of mutants generated in both B. burgdorferi and B. hermsii, identified important differences in growth characteristics and ADI transcription and protein expression. We also found that the ADI system plays a key role in blood and spleen colonization in RF spirochetes. In this study we have identified divergent metabolic strategies in two closely related human pathogens, that ultimately impacts the host-pathogen interface during infection.


Subject(s)
Borrelia burgdorferi , Borrelia , Lyme Disease , Relapsing Fever , Animals , Arginine/metabolism , Borrelia/genetics , Borrelia/metabolism , Borrelia burgdorferi/genetics , Humans , Lyme Disease/microbiology , Mammals , Mice , Relapsing Fever/microbiology
4.
Proteins ; 89(5): 588-594, 2021 05.
Article in English | MEDLINE | ID: mdl-32949018

ABSTRACT

Lyme disease is the most widespread vector-transmitted disease in North America and Europe, caused by infection with Borrelia burgdorferi sensu lato complex spirochetes. We report the solution NMR structure of the B. burgdorferi outer surface lipoprotein BBP28, a member of the multicopy lipoprotein (mlp) family. The structure comprises a tether peptide, five α-helices and an extended C-terminal loop. The fold is similar to that of Borrelia turicatae outer surface protein BTA121, which is known to bind lipids. These results contribute to the understanding of Lyme disease pathogenesis by revealing the molecular structure of a protein from the widely found mlp family.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Borrelia burgdorferi/metabolism , Lipoproteins/chemistry , Amino Acid Sequence , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Borrelia/chemistry , Borrelia/metabolism , Borrelia burgdorferi/chemistry , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Lipoproteins/genetics , Lipoproteins/metabolism , Lyme Disease/microbiology , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
5.
Proteins ; 88(11): 1423-1433, 2020 11.
Article in English | MEDLINE | ID: mdl-32519353

ABSTRACT

Structural characterization of alternatively folded and partially disordered protein conformations remains challenging. Outer surface protein A (OspA) is a pivotal protein in Borrelia infection, which is the etiological agent of Lyme disease. OspA exists in equilibrium with intermediate conformations, in which the central and the C-terminal regions of the protein have lower stabilities than the N-terminal. Here, we characterize pressure- and temperature-stabilized intermediates of OspA by nuclear magnetic resonance spectroscopy combined with paramagnetic relaxation enhancement (PRE). We found that although the C-terminal region of the intermediate was partially disordered, it retains weak specific contact with the N-terminal region, owing to a twist of the central ß-sheet and increased flexibility in the polypeptide chain. The disordered C-terminal region of the pressure-stabilized intermediate was more compact than that of the temperature-stabilized form. Further, molecular dynamics simulation demonstrated that temperature-induced disordering of the ß-sheet was initiated at the C-terminal region and continued through to the central region. An ensemble of simulation snapshots qualitatively described the PRE data from the intermediate and indicated that the intermediate structures of OspA may expose tick receptor-binding sites more readily than does the basic folded conformation.


Subject(s)
Antigens, Surface/chemistry , Arthropod Proteins/chemistry , Bacterial Outer Membrane Proteins/chemistry , Bacterial Vaccines/chemistry , Borrelia/chemistry , Intrinsically Disordered Proteins/chemistry , Lipoproteins/chemistry , Receptors, Cell Surface/chemistry , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Arthropod Proteins/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Vaccines/genetics , Bacterial Vaccines/metabolism , Binding Sites , Borrelia/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Folding , Protein Interaction Domains and Motifs , Receptors, Cell Surface/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics , Ticks/microbiology
6.
J Cutan Pathol ; 47(1): 76-97, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31237707

ABSTRACT

Cutaneous pseudolymphomas (PSLs) belong to a group of lymphocytic infiltrates that histopathologically and/or clinically simulate lymphomas. Different causative agents (e.g., Borrelia sp., injected substances, tattoo, arthropod bite) have been described, but in many cases no cause can be identified, hence the term idiopathic PSL. Clinicopathological correlation is important to make the diagnosis. Four main groups of cutaneous PSL can be distinguished based on histopathologic and/or clinical presentation: (a) nodular PSL; (b) pseudo-mycosis fungoides (pseudo-MF) and simulators of other CTCLs; (c) other PSL (representing distinct clinical entities); and (d) intravascular PSL. This article gives an overview of the histopathologic and clinical characteristics of cutaneous PSLs and proposes a new classification.


Subject(s)
Pseudolymphoma , Skin Neoplasms , Borrelia/metabolism , Borrelia Infections/classification , Borrelia Infections/metabolism , Borrelia Infections/pathology , Humans , Pseudolymphoma/classification , Pseudolymphoma/metabolism , Pseudolymphoma/pathology , Skin Neoplasms/classification , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tattooing/adverse effects
7.
J Biol Chem ; 293(22): 8600-8613, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29669808

ABSTRACT

The plasminogen system is essential for dissolution of fibrin clots, and in addition, it is involved in a wide variety of other physiological processes, including proteolytic activation of growth factors, cell migration, and removal of protein aggregates. On the other hand, uncontrolled plasminogen activation contributes to many pathological processes (e.g. tumor cells' invasion in cancer progression). Moreover, some virulent bacterial species (e.g. Streptococci or Borrelia) bind human plasminogen and hijack the host's plasminogen system to penetrate tissue barriers. Thus, the conversion of plasminogen to the active serine protease plasmin must be tightly regulated. Here, we show that human lactoferrin, an iron-binding milk glycoprotein, blocks plasminogen activation on the cell surface by direct binding to human plasminogen. We mapped the mutual binding sites to the N-terminal region of lactoferrin, encompassed also in the bioactive peptide lactoferricin, and kringle 5 of plasminogen. Finally, lactoferrin blocked tumor cell invasion in vitro and also plasminogen activation driven by Borrelia Our results explain many diverse biological properties of lactoferrin and also suggest that lactoferrin may be useful as a potential tool for therapeutic interventions to prevent both invasive malignant cells and virulent bacteria from penetrating host tissues.


Subject(s)
Borrelia/metabolism , Fibrinolysin/metabolism , Fibrinolysis , Lactoferrin/metabolism , Plasminogen/antagonists & inhibitors , Streptococcus/metabolism , Cell Movement , Cells, Cultured , Crystallography, X-Ray , Humans , Lactoferrin/chemistry , Lactoferrin/genetics , Plasminogen/metabolism , Protein Conformation
8.
BMC Evol Biol ; 19(1): 54, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30760200

ABSTRACT

BACKGROUND: The evolutionary history of a species is frequently derived from molecular sequences, and the resulting phylogenetic trees do not include explicit functional information. Here, we aimed to assess the functional relationships among bacteria in the Spirochaetes phylum, based on the biological processes of 42,489 proteins in reference proteomes of 34 Spirochaetes species. We tested the hypothesis that the species in the genus Borrelia might be sufficiently different to warrant splitting them into two separate genera. RESULTS: A detrended canonical analysis demonstrated that the presence/absence of biological processes among selected bacteria contained a strong phylogenetic signal, which did not separate species of Borrelia. We examined the ten biological processes in which most proteins were involved consistently. This analysis demonstrated that species in Borrelia were more similar to each other than to free-life species (Sediminispirochaeta, Spirochaeta, Sphaerochaeta) or to pathogenic species without vectors (Leptospira, Treponema, Brachyspira), which are highly divergent. A dendrogram based on the presence/absence of proteins in the reference proteomes demonstrated that distances between species of the same genus among free-life or pathogenic non-vector species were higher than the distances between the 19 species (27 strains) of Borrelia. A phyloproteomic network supported the close functional association between species of Borrelia. In the proteome of 27 strains of Borrelia, only a few proteins had evolved separately, in the relapsing fever and Lyme borreliosis groups. The most prominent Borrelia proteins and processes were a subset of those also found in free-living and non-vectored pathogenic species. In addition, the functional innovation (i.e., unique biological processes or proteins) of Borrelia was very low, compared to other genera of Spirochaetes. CONCLUSIONS: We found only marginal functional differences among Borrelia species. Phyloproteomic networks that included all pairwise combinations between species, proteins, and processes were more effective than other methods for evaluating the evolutionary relationships among taxa. With the limitations of data availability, our results did not support a split of the arthropod-transmitted spirochaetes into the proposed genera, Borrelia and Borreliella.


Subject(s)
Bacterial Proteins/metabolism , Borrelia/metabolism , Phylogeny , Proteomics , Animals , Biodiversity , Multivariate Analysis , Proteome/metabolism , Species Specificity
9.
Appl Environ Microbiol ; 83(5)2017 03 01.
Article in English | MEDLINE | ID: mdl-27986725

ABSTRACT

Relapsing fever (RF) spirochetes colonize and are transmitted to mammals primarily by Ornithodoros ticks, and little is known regarding the pathogen's life cycle in the vector. To further understand vector colonization and transmission of RF spirochetes, Borrelia turicatae expressing a green fluorescent protein (GFP) marker (B. turicatae-gfp) was generated. The transformants were evaluated during the tick-mammal infectious cycle, from the third nymphal instar to adult stage. B. turicatae-gfp remained viable for at least 18 months in starved fourth-stage nymphal ticks, and the studies indicated that spirochete populations persistently colonized the tick midgut and salivary glands. Our generation of B. turicatae-gfp also revealed that within the salivary glands, spirochetes are localized in the ducts and lumen of acini, and after tick feeding, the tissues remained populated with spirochetes. The B. turicatae-gfp generated in this study is an important tool to further understand and define the mechanisms of vector colonization and transmission.IMPORTANCE In order to interrupt the infectious cycle of tick-borne relapsing fever spirochetes, it is important to enhance our understanding of vector colonization and transmission. Toward this, we generated a strain of Borrelia turicatae that constitutively produced the green fluorescent protein, and we evaluated fluorescing spirochetes during the entire infectious cycle. We determined that the midgut and salivary glands of Ornithodoros turicata ticks maintain the pathogens throughout the vector's life cycle and remain colonized with the spirochetes for at least 18 months. We also determined that the tick's salivary glands were not depleted after a transmission blood feeding. These findings set the framework to further understand the mechanisms of midgut and salivary gland colonization.


Subject(s)
Borrelia/metabolism , Digestive System/microbiology , Green Fluorescent Proteins/biosynthesis , Nymph/microbiology , Ornithodoros/microbiology , Relapsing Fever/transmission , Salivary Glands/microbiology , Animals , Arthropod Vectors/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biomarkers , Borrelia/genetics , Borrelia/growth & development , DNA, Bacterial , Disease Models, Animal , Gene Expression Regulation, Bacterial , Genes, Bacterial , Green Fluorescent Proteins/genetics , Mice , Relapsing Fever/blood , Relapsing Fever/microbiology , Salivary Glands/pathology
10.
Crit Rev Microbiol ; 42(4): 573-87, 2016 Aug.
Article in English | MEDLINE | ID: mdl-25914944

ABSTRACT

The pathogenic spirochetes Borrelia burgdorferi, B. hermsii, B. recurrentis, Treponema denticola and Leptospira spp. are the etiologic agents of Lyme disease, relapsing fever, periodontitis and leptospirosis, respectively. Lyme borreliosis is a multi-systemic disorder and the most prevalent tick-borne disease in the northern hemisphere. Tick-borne relapsing fever is persistent in endemic areas worldwide, representing a significant burden in some African regions. Periodontal disease, a chronic inflammatory disorder that often leads to tooth loss, is caused by several potential pathogens found in the oral cavity including T. denticola. Leptospirosis is considered the most widespread zoonosis, and the predominant human disease in tropical, undeveloped regions. What these diseases have in common is that they are a significant burden to healthcare costs in the absence of prophylactic measures. This review addresses the interaction of these spirochetes with the fibrinolytic system, plasminogen (Plg) binding to the surface of bacteria and the generation of plasmin (Pla) on their surface. The consequences on host-pathogen interactions when the spirochetes are endowed with this proteolytic activity are discussed on the basis of the results reported in the literature. Spirochetes equipped with Pla activity have been shown to degrade extracellular matrix (ECM) components, in addition to digesting fibrin, facilitating bacterial invasion and dissemination. Pla generation triggers the induction of matrix metalloproteases (MMPs) in a cascade of events that enhances the proteolytic capacity of the spirochetes. These activities in concert with the interference exerted by the Plg/Pla on the complement system - helping the bacteria to evade the immune system - should illuminate our understanding of the mechanisms involved in host infection.


Subject(s)
Borrelia/pathogenicity , Fibrinolysis , Host-Pathogen Interactions , Leptospira/pathogenicity , Treponema denticola/pathogenicity , Borrelia/metabolism , Fibrinolysin/metabolism , Humans , Immune Evasion , Leptospira/metabolism , Matrix Metalloproteinases/metabolism , Plasminogen/metabolism , Protein Binding , Proteolysis , Treponema denticola/metabolism
11.
J Transl Med ; 13: 346, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26537892

ABSTRACT

OBJECTIVES: Prompt antibiotic treatment of early stage Lyme borreliosis (LB) prevents progression to severe multisystem disease. There is a clinical need to improve the diagnostic specificity of early stage Lyme assays in the period prior to the mounting of a robust serology response. Using a novel analyte harvesting nanotechnology, Nanotrap particles, we evaluated urinary Borrelia Outer surface protein A (OspA) C-terminus peptide in early stage LB before and after treatment, and in patients suspected of late stage disseminated LB. METHOD: We employed Nanotrap particles to concentrate urinary OspA and used a highly specific anti-OspA monoclonal antibody (mAb) as a detector of the C-terminus peptides. We mapped the mAb epitope to a narrow specific OspA C-terminal domain OspA236-239 conserved across infectious Borrelia species but with no homology to human proteins and no cross-reactivity with relevant viral and non-Borrelia bacterial proteins. 268 urine samples from patients being evaluated for all categories of LB were collected in a LB endemic area. The urinary OspA assay, blinded to outcome, utilized Nanotrap particle pre-processing, western blotting to evaluate the OspA molecular size, and OspA peptide competition for confirmation. RESULTS: OspA test characteristics: sensitivity 1.7 pg/mL (lowest limit of detection), % coefficient of variation (CV) = 8 %, dynamic range 1.7-30 pg/mL. Pre-treatment, 24/24 newly diagnosed patients with an erythema migrans (EM) rash were positive for urinary OspA while false positives for asymptomatic patients were 0/117 (Chi squared p < 10(-6)). For 10 patients who exhibited persistence of the EM rash during the course of antibiotic therapy, 10/10 were positive for urinary OspA. Urinary OspA of 8/8 patients switched from detectable to undetectable following symptom resolution post-treatment. Specificity of the urinary OspA test for the clinical symptoms was 40/40. Specificity of the urinary OspA antigen test for later serology outcome was 87.5 % (21 urinary OspA positive/24 serology positive, Chi squared p = 4.072e(-15)). 41 of 100 patients under surveillance for persistent LB in an endemic area were positive for urinary OspA protein. CONCLUSIONS: OspA urinary shedding was strongly linked to concurrent active symptoms (e.g. EM rash and arthritis), while resolution of these symptoms after therapy correlated with urinary conversion to OspA negative.


Subject(s)
Antigens, Surface/urine , Bacterial Outer Membrane Proteins/urine , Bacterial Vaccines/urine , Lipoproteins/urine , Lyme Disease/diagnosis , Lyme Disease/urine , Nanotechnology/methods , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Antibodies, Monoclonal/chemistry , Borrelia/metabolism , Case-Control Studies , Epitope Mapping , Epitopes/chemistry , Female , Humans , Immunoglobulin G/chemistry , Male , Mass Spectrometry , Molecular Sequence Data , Peptides/chemistry , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Reproducibility of Results , Sensitivity and Specificity , Sequence Homology, Amino Acid
12.
PLoS Pathog ; 9(4): e1003308, 2013.
Article in English | MEDLINE | ID: mdl-23637600

ABSTRACT

To cause infections microbes need to evade host defense systems, one of these being the evolutionarily old and important arm of innate immunity, the alternative pathway of complement. It can attack all kinds of targets and is tightly controlled in plasma and on host cells by plasma complement regulator factor H (FH). FH binds simultaneously to host cell surface structures such as heparin or glycosaminoglycans via domain 20 and to the main complement opsonin C3b via domain 19. Many pathogenic microbes protect themselves from complement by recruiting host FH. We analyzed how and why different microbes bind FH via domains 19-20 (FH19-20). We used a selection of FH19-20 point mutants to reveal the binding sites of several microbial proteins and whole microbes (Haemophilus influenzae, Bordetella pertussis, Pseudomonas aeruginosa, Streptococcus pneumonia, Candida albicans, Borrelia burgdorferi, and Borrelia hermsii). We show that all studied microbes use the same binding region located on one side of domain 20. Binding of FH to the microbial proteins was inhibited with heparin showing that the common microbial binding site overlaps with the heparin site needed for efficient binding of FH to host cells. Surprisingly, the microbial proteins enhanced binding of FH19-20 to C3b and down-regulation of complement activation. We show that this is caused by formation of a tripartite complex between the microbial protein, FH, and C3b. In this study we reveal that seven microbes representing different phyla utilize a common binding site on the domain 20 of FH for complement evasion. Binding via this site not only mimics the glycosaminoglycans of the host cells, but also enhances function of FH on the microbial surfaces via the novel mechanism of tripartite complex formation. This is a unique example of convergent evolution resulting in enhanced immune evasion of important pathogens via utilization of a "superevasion site."


Subject(s)
Bacteria/metabolism , Candida albicans/metabolism , Complement Factor H/metabolism , Bacteria/genetics , Bacteria/immunology , Bacteria/pathogenicity , Binding Sites , Bordetella pertussis/genetics , Bordetella pertussis/immunology , Bordetella pertussis/metabolism , Bordetella pertussis/pathogenicity , Borrelia/genetics , Borrelia/immunology , Borrelia/metabolism , Borrelia/pathogenicity , Candida albicans/genetics , Candida albicans/immunology , Candida albicans/pathogenicity , Cell Membrane/metabolism , Complement Activation , Complement Factor H/chemistry , Haemophilus influenzae/genetics , Haemophilus influenzae/immunology , Haemophilus influenzae/metabolism , Haemophilus influenzae/pathogenicity , Humans , Protein Binding , Protein Structure, Tertiary , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/metabolism , Streptococcus pneumoniae/pathogenicity
13.
Infect Immun ; 82(1): 453-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24218480

ABSTRACT

T cell-independent antibody responses develop rapidly, within 3 to 4 days, and are critical for preventing blood-borne pathogens from evolving into life-threatening infections. The interaction of BAFF, also known as BLyS, with its receptors BAFFR and TACI on B cells is critical for B cell homeostasis and function. Using a synthetic polysaccharide antigen, it has previously been shown that TACI is critical for T cell-independent antibody responses. To examine the role of BAFFR and TACI in T cell-independent antibody responses to an active infection, we utilized the Borrelia hermsii infection system. In this infection system, T cell-independent responses mediated by the B1b cell subset are critical for controlling bacteremia. We found that B1b cells express BAFFR and TACI and that the surface expression of both receptors is upregulated on B1b cells following exposure to whole B. hermsii cells. Surprisingly, we found that TACI(-/-) mice are not impaired either in specific antibody responses to B. hermsii or in controlling B. hermsii bacteremia. In contrast, TACI-deficient mice immunized with heat-killed type 3 serotype pneumococcus cells are impaired in generating pneumococcal polysaccharide-specific responses and succumb to challenge with live type 3 serotype pneumococcus, indicating that TACI is required for T cell-independent antibody responses to bacterial-associated polysaccharides. Although we have found that TACI is dispensable for controlling B. hermsii infection, mice deficient in BAFFR or BAFF exhibit impairment in B. hermsii-specific IgM responses and clearance of bacteremia. Collectively, these data indicate a disparity in the roles for TACI and BAFFR in primary T cell-independent antibody responses to bacterial pathogens.


Subject(s)
B-Cell Activating Factor/physiology , B-Cell Activation Factor Receptor/physiology , B-Lymphocytes/immunology , Borrelia/immunology , Lyme Disease/immunology , Transmembrane Activator and CAML Interactor Protein/physiology , Analysis of Variance , Animals , B-Cell Activating Factor/deficiency , B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/deficiency , B-Cell Activation Factor Receptor/metabolism , B-Lymphocytes/metabolism , Borrelia/metabolism , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Mice , Mice, Inbred C57BL , Transmembrane Activator and CAML Interactor Protein/deficiency , Transmembrane Activator and CAML Interactor Protein/metabolism , Up-Regulation
14.
Proc Natl Acad Sci U S A ; 108(51): 20707-12, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22143787

ABSTRACT

Rodents are natural reservoirs for a variety of species of Borrelia that cause relapsing fever (RF) in humans. The murine model of this disease recapitulates many of the clinical manifestations of the human disease and has revealed that T cell-independent antibody responses are required to resolve the bacteremic episodes. However, it is not clear whether such protective humoral responses are mounted in humans. We examined Borrelia hermsii infection in human hematopoietic stem cell-engrafted nonobese diabetic/SCID/IL-2Rγ(null) mice: "human immune system mice" (HISmice). Infection of these mice, which are severely deficient in lymphoid and myeloid compartments, with B. hermsii resulted in persistent bacteremia. In contrast, this infection in HISmice resulted in recurrent episodes of bacteremia, the hallmark of RF. The resolution of the primary episode of bacteremia was concurrent with the generation of B. hermsii-specific human IgM. Remarkably, HISmice generated antibody responses to the B. hermsii outer-membrane protein Factor H binding protein A. Sera from humans infected by B. hermsii have a similar reactivity, and studies in mice have shown that this response is generated by the B1b cell subset. HISmice contain several B-cell subsets, including those with the phenotype CD20(+)CD27(+)CD43(+)CD70(-), a proposed human equivalent of mouse B1 cells. Reduction of B cells by administration of anti-human CD20 antibody resulted in diminished anti-B. hermsii responses and persistent bacteremia in HISmice. These data indicate that analysis of B. hermsii infection in HISmice will serve as a model in which to study the cellular and molecular mechanisms involved in controlling human RF.


Subject(s)
Borrelia Infections/metabolism , Borrelia/metabolism , Hematopoietic Stem Cells/cytology , Relapsing Fever/microbiology , Animals , Antigens/metabolism , Antigens, CD34/biosynthesis , Borrelia Infections/microbiology , Flow Cytometry/methods , Hematopoietic Stem Cell Transplantation , Humans , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Mice, SCID , Relapsing Fever/pathology , Spirochaetales/metabolism , Spleen/metabolism , Splenomegaly
15.
Front Immunol ; 15: 1390468, 2024.
Article in English | MEDLINE | ID: mdl-38726006

ABSTRACT

Introduction: Relapsing fever (RF) remains a neglected human disease that is caused by a number of diverse pathogenic Borrelia (B.) species. Characterized by high cell densities in human blood, relapsing fever spirochetes have developed plentiful strategies to avoid recognition by the host defense mechanisms. In this scenario, spirochetal lipoproteins exhibiting multifunctional binding properties in the interaction with host-derived molecules are known to play a key role in adhesion, fibrinolysis and complement activation. Methods: Binding of CihC/FbpC orthologs to different human proteins and conversion of protein-bound plasminogen to proteolytic active plasmin were examined by ELISA. To analyze the inhibitory capacity of CihC/FbpC orthologs on complement activation, a microtiter-based approach was performed. Finally, AlphaFold predictions were utilized to identified the complement-interacting residues. Results and discussion: Here, we elucidate the binding properties of CihC/FbpC-orthologs from distinct RF spirochetes including B. parkeri, B. hermsii, B. turicatae, and B. recurrentis to human fibronectin, plasminogen, and complement component C1r. All CihC/FbpC-orthologs displayed similar binding properties to fibronectin, plasminogen, and C1r, respectively. Functional studies revealed a dose dependent binding of plasminogen to all borrelial proteins and conversion to active plasmin. The proteolytic activity of plasmin was almost completely abrogated by tranexamic acid, indicating that lysine residues are involved in the interaction with this serine protease. In addition, a strong inactivation capacity toward the classical pathway could be demonstrated for the wild-type CihC/FbpC-orthologs as well as for the C-terminal CihC fragment of B. recurrentis. Pre-incubation of human serum with borrelial molecules except CihC/FbpC variants lacking the C-terminal region protected serum-susceptible Borrelia cells from complement-mediated lysis. Utilizing AlphaFold2 predictions and existing crystal structures, we mapped the putative key residues involved in C1r binding on the CihC/FbpC orthologs attempting to explain the relatively small differences in C1r binding affinity despite the substitutions of key residues. Collectively, our data advance the understanding of the multiple binding properties of structural and functional highly similar molecules of relapsing fever spirochetes proposed to be involved in pathogenesis and virulence.


Subject(s)
Bacterial Proteins , Borrelia , Fibrinolysis , Host-Pathogen Interactions , Plasminogen , Humans , Bacterial Adhesion , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Borrelia/immunology , Borrelia/metabolism , Complement Activation , Complement System Proteins/immunology , Complement System Proteins/metabolism , Fibrinolysin/metabolism , Fibronectins/metabolism , Host-Pathogen Interactions/immunology , Immune Evasion , Plasminogen/metabolism , Protein Binding , Relapsing Fever/immunology , Relapsing Fever/microbiology
16.
Proc Natl Acad Sci U S A ; 107(4): 1535-40, 2010 Jan 26.
Article in English | MEDLINE | ID: mdl-20080535

ABSTRACT

Invariant natural killer T cells (iNKT cells) respond to CD1d-presented glycolipids from Borrelia burgdorferi, the causative agent of Lyme disease. Although mouse and human iNKT cells respond to different antigens based on subtle differences in their fatty acids, the mechanism by which fatty acid structure determines antigenic potency is not well understood. Here we show that the mouse and human CD1d present glycolipids having different fatty acids, based in part upon a difference at a single amino acid position that is involved in positioning the sugar epitope. CD1d also can bind nonantigenic lipids, however, but unexpectedly, mouse CD1d orients the two aliphatic chains of a nonantigenic lipid rotated 180 degrees, causing a dramatic repositioning of the exposed sugar. Therefore, our data reveal the biochemical basis for the high degree of antigenic specificity of iNKT cells for certain fatty acids, and they suggest how microbes could alter fatty acid biosynthesis as an immune evasion mechanism.


Subject(s)
Antigens, Bacterial/immunology , Antigens, CD1d/immunology , Borrelia/immunology , Glycolipids/immunology , Immune Evasion , Natural Killer T-Cells/immunology , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Antigens, CD1d/chemistry , Antigens, CD1d/metabolism , Borrelia/chemistry , Borrelia/metabolism , Fatty Acids/biosynthesis , Fatty Acids/immunology , Glycolipids/chemistry , Glycolipids/metabolism , Humans , Mice , Models, Molecular , Natural Killer T-Cells/chemistry , Natural Killer T-Cells/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/immunology
17.
J Clin Invest ; 133(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36649080

ABSTRACT

The RNA polymerase alternative σ factor RpoS in Borrelia burgdorferi (Bb), the Lyme disease pathogen, is responsible for programmatic-positive and -negative gene regulation essential for the spirochete's dual-host enzootic cycle. RpoS is expressed during tick-to-mammal transmission and throughout mammalian infection. Although the mammalian-phase RpoS regulon is well described, its counterpart during the transmission blood meal is unknown. Here, we used Bb-specific transcript enrichment by tick-borne disease capture sequencing (TBDCapSeq) to compare the transcriptomes of WT and ΔrpoS Bb in engorged nymphs and following mammalian host-adaptation within dialysis membrane chambers. TBDCapSeq revealed dramatic changes in the contours of the RpoS regulon within ticks and mammals and further confirmed that RpoS-mediated repression is specific to the mammalian-phase of Bb's enzootic cycle. We also provide evidence that RpoS-dependent gene regulation, including repression of tick-phase genes, is required for persistence in mice. Comparative transcriptomics of engineered Bb strains revealed that the Borrelia oxidative stress response regulator (BosR), a noncanonical Fur family member, and the cyclic diguanosine monophosphate (c-di-GMP) effector PlzA reciprocally regulate the function of RNA polymerase complexed with RpoS. BosR is required for RpoS-mediated transcription activation and repression in addition to its well-defined role promoting transcription of rpoS by the RNA polymerase alternative σ factor RpoN. During transmission, ligand-bound PlzA antagonizes RpoS-mediated repression, presumably acting through BosR.


Subject(s)
Borrelia burgdorferi , Borrelia , Lyme Disease , Ticks , Mice , Animals , Borrelia burgdorferi/genetics , Borrelia/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Ticks/genetics , Sigma Factor/genetics , Sigma Factor/metabolism , Lyme Disease/genetics , Mammals/metabolism , Gene Expression Regulation, Bacterial
18.
J Bacteriol ; 194(14): 3723-4, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22740657

ABSTRACT

We announce the draft genome sequence of Borrelia crocidurae (strain Achema). The 1,557,560-bp genome (27% GC content) comprises one 919,477-bp linear chromosome and 638,083-bp plasmids that together carry 1,472 open reading frames, 32 tRNAs, and three complete rRNAs, with almost complete colinearity between B. crocidurae and Borrelia duttonii chromosomes.


Subject(s)
Borrelia/genetics , Africa/epidemiology , Animals , Arachnid Vectors/microbiology , Borrelia/metabolism , Gene Expression Regulation, Bacterial/physiology , Genome, Bacterial , Humans , Molecular Sequence Data , Ornithodoros/microbiology , Relapsing Fever/epidemiology , Relapsing Fever/microbiology , Species Specificity , Tick-Borne Diseases , Zoonoses
19.
Infect Immun ; 80(5): 1881-90, 2012 May.
Article in English | MEDLINE | ID: mdl-22354035

ABSTRACT

Borrelia hermsii and other relapsing fever (RF) species are noted for their highly polymorphic surface antigens, the variable major proteins (VMP). Less is known about other surface proteins of these pathogens in either their vertebrate reservoirs or arthropod vectors. To further characterize these proteins, we elicited antibodies against VMP-less cells, noted antibody reactions against whole cells and cell components, and then subjected selected antigens to mass spectroscopy for amino acid sequencing for comparison against a B. hermsii genome database. One of the derived monoclonal antibodies, H0120, agglutinated spirochetes, and in Western blot analyses, it bound to a 14-kDa protein of whole cells and their membrane fractions but not after protease treatment. A search of open reading frames of the B. hermsii genome with extracted peptides identified the 14-kDa protein with bha128, a 453-nucleotide gene of the 175-kb linear plasmid. The bha128 gene was synthesized and expressed in Escherichia coli. The protein product was bound by antibody H0120. Genes homologous to bha128 occur in the RF species Borrelia turicatae, B. duttonii, and B. recurrentis but not in Lyme disease Borrelia species or other organisms. The following findings indicated an association of BHA128, renamed Alp, with the tick environment: (i) Alp was produced at higher levels at 23°C than at 34 °C; (ii) almost all spirochetes in tick salivary glands were bound by the H0120 antibody, but only ~1% of spirochetes in the blood of infected mice were bound; and (iii) infected mice produced antibodies to several B. hermsii antigens but not detectably to native or recombinant Alp.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Borrelia Infections/microbiology , Borrelia/metabolism , Amino Acid Sequence , Animals , Antibodies, Bacterial , Antibody Specificity , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/genetics , Base Sequence , Borrelia Infections/immunology , Female , Genome, Bacterial , Mice , Mice, SCID , Molecular Sequence Data , Mutation , Phylogeny , Salivary Glands/microbiology , Temperature , Ticks
20.
Cell Microbiol ; 13(5): 731-41, 2011 May.
Article in English | MEDLINE | ID: mdl-21276171

ABSTRACT

Some Borrelia species are the causative agents of tick-borne Lyme disease responsible for different disabilities depending on species and hosts. Borrelia are highly motile bacterial cells, and light microscopy shows that these spirochetes can associate with each other during movement. Using cryo-electron tomography, we observed closely associated Borrelia cells. Some of these showed a single outer membrane surrounding two longitudinally arranged cytoplasmic cylinders. We also observed fusion of two cytoplasmic cylinders and differences in the surface layer density of fused spirochetes. These processes could play a role in the interaction of Borrelia species with the host's immune system.


Subject(s)
Borrelia/ultrastructure , Cell Membrane/ultrastructure , Antigenic Variation/genetics , Antigenic Variation/immunology , Borrelia/genetics , Borrelia/metabolism , Cell Fusion , Cell Membrane/metabolism , Electron Microscope Tomography , Gene Transfer, Horizontal
SELECTION OF CITATIONS
SEARCH DETAIL