Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Biol Chem ; 299(8): 104972, 2023 08.
Article in English | MEDLINE | ID: mdl-37380082

ABSTRACT

Borrelial pathogens are vector-borne etiological agents known to cause Lyme disease, relapsing fever, and Borrelia miyamotoi disease. These spirochetes each encode several surface-localized lipoproteins that bind components of the human complement system to evade host immunity. One borrelial lipoprotein, BBK32, protects the Lyme disease spirochete from complement-mediated attack via an alpha helical C-terminal domain that interacts directly with the initiating protease of the classical complement pathway, C1r. In addition, the B. miyamotoi BBK32 orthologs FbpA and FbpB also inhibit C1r, albeit via distinct recognition mechanisms. The C1r-inhibitory activities of a third ortholog termed FbpC, which is found exclusively in relapsing fever-causing spirochetes, remains unknown. Here, we report the crystal structure of the C-terminal domain of Borrelia hermsii FbpC to a limiting resolution of 1.5 Å. We used surface plasmon resonance and assays of complement function to demonstrate that FbpC retains potent BBK32-like anticomplement activities. Based on the structure of FbpC, we hypothesized that conformational dynamics of the complement inhibitory domains of borrelial C1r inhibitors may differ. To test this, we utilized the crystal structures of the C-terminal domains of BBK32, FbpA, FbpB, and FbpC to carry out molecular dynamics simulations, which revealed borrelial C1r inhibitors adopt energetically favored open and closed states defined by two functionally critical regions. Taken together, these results advance our understanding of how protein dynamics contribute to the function of bacterial immune evasion proteins and reveal a surprising plasticity in the structures of borrelial C1r inhibitors.


Subject(s)
Bacterial Proteins , Borrelia , Complement C1 Inactivator Proteins , Lyme Disease , Relapsing Fever , Humans , Bacterial Proteins/chemistry , Lyme Disease/immunology , Lyme Disease/microbiology , Relapsing Fever/immunology , Relapsing Fever/microbiology , Complement C1 Inactivator Proteins/chemistry , Protein Domains , Crystallography, X-Ray
2.
bioRxiv ; 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36909632

ABSTRACT

Borrelial pathogens are vector-borne etiological agents of Lyme disease, relapsing fever, and Borrelia miyamotoi disease. These spirochetes each encode several surface-localized lipoproteins that bind to components of the human complement system. BBK32 is an example of a borrelial lipoprotein that protects the Lyme disease spirochete from complement-mediated attack. The complement inhibitory activity of BBK32 arises from an alpha helical C-terminal domain that interacts directly with the initiating protease of the classical pathway, C1r. Borrelia miyamotoi spirochetes encode BBK32 orthologs termed FbpA and FbpB, and these proteins also inhibit C1r, albeit via distinct recognition mechanisms. The C1r-inhibitory activities of a third ortholog termed FbpC, which is found exclusively in relapsing fever spirochetes, remains unknown. Here we report the crystal structure of the C-terminal domain of B. hermsii FbpC to a limiting resolution of 1.5 Å. Surface plasmon resonance studies and assays of complement function demonstrate that FbpC retains potent BBK32-like anti-complement activities. Based on the structure of FbpC, we hypothesized that conformational dynamics of the complement inhibitory domains of borrelial C1r inhibitors may differ. To test this, we utilized the crystal structures of the C-terminal domains of BBK32, FbpA, FbpB, and FbpC to carry out 1 µs molecular dynamics simulations, which revealed borrelial C1r inhibitors adopt energetically favored open and closed states defined by two functionally critical regions. This study advances our understanding of how protein dynamics contribute to the function of bacterial immune evasion proteins and reveals a surprising plasticity in the structures of borrelial C1r inhibitors.

3.
J Thromb Haemost ; 20(11): 2656-2665, 2022 11.
Article in English | MEDLINE | ID: mdl-35996342

ABSTRACT

BACKGROUND: Anti-platelet factor 4 (PF4)/heparin immune complexes that cause heparin-induced thrombocytopenia (HIT) activate complement via the classical pathway. Previous studies have shown that the alternative pathway of complement substantially amplifies the classical pathway of complement activation through the C3b feedback cycle. OBJECTIVES: These studies sought to examine the contributions of the alternative pathway to complement activation by HIT antibodies. METHODS: Using IgG monoclonal (KKO) and/or patient-derived HIT antibodies, we compared the effects of classical pathway (BBK32 and C1-esterase inhibitor [C1-INH]), alternative pathway (anti-factor B [fB] or factor D [fD] inhibitor) or combined classical and alternative pathway inhibition (soluble complement receptor 1 [sCR1]) in whole blood or plasma. RESULTS: Classical pathway inhibitors BBK32 and C1-INH and the combined classical/alternative pathway inhibitor sCR1 prevented KKO/HIT immune complex-induced complement activation, including release of C3 and C5 activation products, binding of immune complexes to B cells, and neutrophil activation. The alternative pathway inhibitors fB and fD, however, did not affect complement activation by KKO/HIT immune complexes. Similarly, alternative pathway inhibition had no effect on complement activation by unrelated immune complexes consisting of anti-dinitrophenyl (DNP) antibody and the multivalent DNP--keyhole limpet hemocyanin antigen. CONCLUSIONS: Collectively, these findings suggest the alternative pathway contributes little in support of complement activation by HIT immune complexes. Additional in vitro and in vivo studies are required to examine if this property is shared by most IgG-containing immune complexes or if predominance of the classic pathway is limited to immune complexes composed of multivalent antigens.


Subject(s)
Antigen-Antibody Complex , Thrombocytopenia , Humans , Complement Factor D , Heparin/adverse effects , Complement Activation , Complement System Proteins , Immunoglobulin G , Receptors, Complement , Esterases/adverse effects
4.
Front Immunol ; 13: 886733, 2022.
Article in English | MEDLINE | ID: mdl-35693799

ABSTRACT

Pathogens that traffic in the blood of their hosts must employ mechanisms to evade the host innate immune system, including the complement cascade. The Lyme disease spirochete, Borreliella burgdorferi, has evolved numerous outer membrane lipoproteins that interact directly with host proteins. Compared to Lyme disease-associated spirochetes, relatively little is known about how an emerging tick-borne spirochetal pathogen, Borrelia miyamotoi, utilizes surface lipoproteins to interact with a human host. B. burgdorferi expresses the multifunctional lipoprotein, BBK32, that inhibits the classical pathway of complement through interaction with the initiating protease C1r, and also interacts with fibronectin using a separate intrinsically disordered domain. B. miyamotoi encodes two separate bbk32 orthologs denoted fbpA and fbpB; however, the activities of these proteins are unknown. Here, we show that B. miyamotoi FbpA binds human fibronectin in a manner similar to B. burgdorferi BBK32, whereas FbpB does not. FbpA and FbpB both bind human complement C1r and protect a serum-sensitive B. burgdorferi strain from complement-mediated killing, but surprisingly, differ in their ability to recognize activated C1r versus zymogen states of C1r. To better understand the observed differences in C1r recognition and inhibition properties, high-resolution X-ray crystallography structures were solved of the C1r-binding regions of B. miyamotoi FbpA and FbpB at 1.9Å and 2.1Å, respectively. Collectively, these data suggest that FbpA and FbpB have partially overlapping functions but are functionally and structurally distinct. The data presented herein enhances our overall understanding of how bloodborne pathogens interact with fibronectin and modulate the complement system.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi , Borrelia , Lyme Disease , Borrelia/physiology , Complement System Proteins/metabolism , Fibronectins , Humans , Lipoproteins
5.
J Immunol ; 207(11): 2856-2867, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34759015

ABSTRACT

Complement evasion is a hallmark of extracellular microbial pathogens such as Borrelia burgdorferi, the causative agent of Lyme disease. Lyme disease spirochetes express nearly a dozen outer surface lipoproteins that bind complement components and interfere with their native activities. Among these, BBK32 is unique in its selective inhibition of the classical pathway. BBK32 blocks activation of this pathway by selectively binding and inhibiting the C1r serine protease of the first component of complement, C1. To understand the structural basis for BBK32-mediated C1r inhibition, we performed crystallography and size-exclusion chromatography-coupled small angle X-ray scattering experiments, which revealed a molecular model of BBK32-C in complex with activated human C1r. Structure-guided site-directed mutagenesis was combined with surface plasmon resonance binding experiments and assays of complement function to validate the predicted molecular interface. Analysis of the structures shows that BBK32 inhibits activated forms of C1r by occluding substrate interaction subsites (i.e., S1 and S1') and reveals a surprising role for C1r B loop-interacting residues for full inhibitory activity of BBK32. The studies reported in this article provide for the first time (to our knowledge) a structural basis for classical pathway-specific inhibition by a human pathogen.


Subject(s)
Bacterial Proteins/immunology , Borrelia burgdorferi/chemistry , Complement C1r/immunology , Lyme Disease/immunology , Peptide Hydrolases/immunology , Bacterial Proteins/chemistry , Borrelia burgdorferi/immunology , Humans , Models, Molecular
6.
Curr Issues Mol Biol ; 42: 473-518, 2021.
Article in English | MEDLINE | ID: mdl-33353871

ABSTRACT

Lyme disease Borrelia are obligately parasitic, tick- transmitted, invasive, persistent bacterial pathogens that cause disease in humans and non-reservoir vertebrates primarily through the induction of inflammation. During transmission from the infected tick, the bacteria undergo significant changes in gene expression, resulting in adaptation to the mammalian environment. The organisms multiply and spread locally and induce inflammatory responses that, in humans, result in clinical signs and symptoms. Borrelia virulence involves a multiplicity of mechanisms for dissemination and colonization of multiple tissues and evasion of host immune responses. Most of the tissue damage, which is seen in non-reservoir hosts, appears to result from host inflammatory reactions, despite the low numbers of bacteria in affected sites. This host response to the Lyme disease Borrelia can cause neurologic, cardiovascular, arthritic, and dermatologic manifestations during the disseminated and persistent stages of infection. The mechanisms by which a paucity of organisms (in comparison to many other infectious diseases) can cause varied and in some cases profound inflammation and symptoms remains mysterious but are the subjects of diverse ongoing investigations. In this review, we provide an overview of virulence mechanisms and determinants for which roles have been demonstrated in vivo, primarily in mouse models of infection.


Subject(s)
Borrelia , Disease Susceptibility , Lyme Disease/microbiology , Animals , Arthropod Vectors/microbiology , Borrelia/genetics , Disease Models, Animal , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions , Humans , Lyme Disease/transmission , Ticks/microbiology , Virulence , Virulence Factors/genetics
7.
Curr Issues Mol Biol ; 42: 385-408, 2021.
Article in English | MEDLINE | ID: mdl-33310914

ABSTRACT

Being able to vizualize a pathogen at a site of interaction with a host is an aesthetically appealing idea and the resulting images can be both informative as well as enjoyable to view. Moreover, the approaches used to derive these images can be powerful in terms of offering data unobtainable by other methods. In this article, we review three primary modalities for live imaging Borrelia spirochetes: whole animal imaging, intravital microscopy and live cell imaging. Each method has strengths and weaknesses, which we review, as well as specific purposes for which they are optimally utilized. Live imaging borriliae is a relatively recent development and there was a need of a review to cover the area. Here, in addition to the methods themselves, we also review areas of spirochete biology that have been significantly impacted by live imaging and present a collection of images associated with the forward motion in the field driven by imaging studies.


Subject(s)
Borrelia/cytology , Microscopy , Animals , Bacterial Physiological Phenomena , Borrelia/physiology , Humans , Microscopy/methods , Optical Imaging/methods
8.
Trends Microbiol ; 28(11): 889-899, 2020 11.
Article in English | MEDLINE | ID: mdl-32482556

ABSTRACT

The complement system is an ancient arm of the innate immune system that plays important roles in pathogen recognition and elimination. Upon activation by microbes, complement opsonizes bacterial surfaces, recruits professional phagocytes, and causes bacteriolysis. Borreliella species are spirochetal bacteria that are transmitted to vertebrate hosts via infected Ixodes ticks and are the etiologic agents of Lyme disease. Pathogens that traffic in blood and other body fluids, like Borreliella, have evolved means to evade complement. Lyme disease spirochetes interfere with complement by producing a small arsenal of outer-surface lipoproteins that bind host complement components and manipulate their native activities. Here we review the current landscape of complement evasion by Lyme disease spirochetes and provide an update on recent discoveries.


Subject(s)
Borrelia burgdorferi/immunology , Complement System Proteins/immunology , Immune Evasion , Lyme Disease/immunology , Lyme Disease/microbiology , Animals , Borrelia burgdorferi/genetics , Borrelia burgdorferi/physiology , Humans , Immunity, Innate , Ixodes/immunology , Ixodes/microbiology , Ixodes/physiology , Lyme Disease/transmission
9.
PLoS Pathog ; 16(5): e1008423, 2020 05.
Article in English | MEDLINE | ID: mdl-32365143

ABSTRACT

Post-transcriptional regulation via small regulatory RNAs (sRNAs) has been implicated in diverse regulatory processes in bacteria, including virulence. One class of sRNAs, termed trans-acting sRNAs, can affect the stability and/or the translational efficiency of regulated transcripts. In this study, we utilized a collaborative approach that employed data from infection with the Borrelia burgdorferi Tn library, coupled with Tn-seq, together with borrelial sRNA and total RNA transcriptomes, to identify an intergenic trans-acting sRNA, which we designate here as ittA for infectivity-associated and tissue-tropic sRNA locus A. The genetic inactivation of ittA resulted in a significant attenuation in infectivity, with decreased spirochetal load in ear, heart, skin and joint tissues. In addition, the ittA mutant did not disseminate to peripheral skin sites or heart tissue, suggesting a role for ittA in regulating a tissue-tropic response. RNA-Seq analysis determined that 19 transcripts were differentially expressed in the ittA mutant relative to its genetic parent, including vraA, bba66, ospD and oms28 (bba74). Subsequent proteomic analyses also showed a significant decrease of OspD and Oms28 (BBA74) proteins. To our knowledge this is the first documented intergenic sRNA that alters the infectivity potential of B. burgdorferi.


Subject(s)
Borrelia burgdorferi/genetics , RNA, Small Untranslated/metabolism , Tropism/genetics , Borrelia burgdorferi/metabolism , Borrelia burgdorferi/pathogenicity , Gene Expression Regulation, Bacterial/genetics , Gene Library , Genome, Bacterial , Lyme Disease/microbiology , Proteomics , RNA, Bacterial/genetics , RNA, Small Untranslated/genetics , Transcriptome/genetics , Virulence
10.
PLoS Pathog ; 15(5): e1007644, 2019 05.
Article in English | MEDLINE | ID: mdl-31086414

ABSTRACT

Borrelia burgdorferi, the causative agent of Lyme disease in humans, is maintained in a complex biphasic life cycle, which alternates between tick and vertebrate hosts. To successfully survive and complete its enzootic cycle, B. burgdorferi adapts to diverse hosts by regulating genes required for survival in specific environments. Here we describe the first ever use of transposon insertion sequencing (Tn-seq) to identify genes required for B. burgdorferi survival in its tick host. We found that insertions into 46 genes resulted in a complete loss of recovery of mutants from larval Ixodes ticks. Insertions in an additional 56 genes resulted in a >90% decrease in fitness. The screen identified both previously known and new genes important for larval tick survival. Almost half of the genes required for survival in the tick encode proteins of unknown function, while a significant portion (over 20%) encode membrane-associated proteins or lipoproteins. We validated the results of the screen for five Tn mutants by performing individual competition assays using mutant and complemented strains. To better understand the role of one of these genes in tick survival, we conducted mechanistic studies of bb0017, a gene previously shown to be required for resistance against oxidative stress. In this study we show that BB0017 affects the regulation of key borrelial virulence determinants. The application of Tn-seq to in vivo screening of B. burgdorferi in its natural vector is a powerful tool that can be used to address many different aspects of the host pathogen interaction.


Subject(s)
Bacterial Proteins/genetics , Borrelia burgdorferi/growth & development , DNA Transposable Elements , Gene Expression Regulation, Bacterial , Lyme Disease/microbiology , Ticks/growth & development , Virulence Factors/genetics , Animals , Bacterial Proteins/metabolism , Borrelia burgdorferi/genetics , Borrelia burgdorferi/immunology , Disease Models, Animal , Disease Vectors , High-Throughput Nucleotide Sequencing , Host-Pathogen Interactions , Lyme Disease/immunology , Mice , Ticks/microbiology , Virulence Factors/metabolism
11.
PLoS Pathog ; 15(3): e1007659, 2019 03.
Article in English | MEDLINE | ID: mdl-30897158

ABSTRACT

The carboxy-terminal domain of the BBK32 protein from Borrelia burgdorferi sensu stricto, termed BBK32-C, binds and inhibits the initiating serine protease of the human classical complement pathway, C1r. In this study we investigated the function of BBK32 orthologues of the Lyme-associated Borrelia burgdorferi sensu lato complex, designated BAD16 from B. afzelii strain PGau and BGD19 from B. garinii strain IP90. Our data show that B. afzelii BAD16-C exhibits BBK32-C-like activities in all assays tested, including high-affinity binding to purified C1r protease and C1 complex, and potent inhibition of the classical complement pathway. Recombinant B. garinii BGD19-C also bound C1 and C1r with high-affinity yet exhibited significantly reduced in vitro complement inhibitory activities relative to BBK32-C or BAD16-C. Interestingly, natively produced BGD19 weakly recognized C1r relative to BBK32 and BAD16 and, unlike these proteins, BGD19 did not confer significant protection from serum killing. Site-directed mutagenesis was performed to convert BBK32-C to resemble BGD19-C at three residue positions that are identical between BBK32 and BAD16 but different in BGD19. The resulting chimeric protein was designated BXK32-C and this BBK32-C variant mimicked the properties observed for BGD19-C. To query the disparate complement inhibitory activities of BBK32 orthologues, the crystal structure of BBK32-C was solved to 1.7Å limiting resolution. BBK32-C adopts an anti-parallel four-helix bundle fold with a fifth alpha-helix protruding from the helical core. The structure revealed that the three residues targeted in the BXK32-C chimera are surface-exposed, further supporting their potential relevance in C1r binding and inhibition. Additional binding assays showed that BBK32-C only recognized C1r fragments containing the serine protease domain. The structure-function studies reported here improve our understanding of how BBK32 recognizes and inhibits C1r and provide new insight into complement evasion mechanisms of Lyme-associated spirochetes of the B. burgdorferi sensu lato complex.


Subject(s)
Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Complement Pathway, Classical/genetics , Bacterial Proteins/immunology , Bacterial Proteins/ultrastructure , Borrelia burgdorferi/immunology , Borrelia burgdorferi Group , Complement C1r/metabolism , Complement Pathway, Classical/physiology , Complement System Proteins/metabolism , Humans , Lyme Disease/physiopathology , Protein Domains/physiology , Recombinant Proteins , Sequence Analysis, Protein
12.
Front Immunol ; 9: 959, 2018.
Article in English | MEDLINE | ID: mdl-29867944

ABSTRACT

Activation of the classical complement pathway occurs to varying degrees within strains of the Borrelia burgdorferi sensu lato complex, which contain a group of pathogenic spirochetes that cause tick-borne Lyme borreliosis, including the agent of Lyme disease in the United States, B. burgdorferi. Despite this information, details related to the control of B. burgdorferi by the classical pathway are not clear. To address this question, we infected C1qα-/- mice, which cannot assemble the C1 complex and thus fail to activate the classical pathway, with B. burgdorferi sensu stricto strain B31. Using bioluminescent in vivo imaging, we found that C1qα-/- mice harbored more B. burgdorferi following 10 days of infection relative to their isogenic C57BL/6 parent. Quantitative PCR (qPCR) demonstrated that C1qα-/- mice harbored significantly more B. burgdorferi than parent mice did within lymph nodes, skin, heart, and joints. The increased B. burgdorferi load in C1qα-/- mice was observed at 21 and 28 days of infection, consistent with the classical pathway promoting complement-dependent, antibody-mediated killing following the development of a B. burgdorferi-specific humoral immune response. In addition, circulating borrelial-specific IgM was higher in C1qα-/- mice relative to their parent mouse strain and did not decrease at 21 and 28 days post-infection, indicating that IgG class switching was delayed in C1qα-/- mice. At day 28, both Borrelia-specific IgG1 and IgG3 levels were higher in infected C1qα-/- mice, but that these antibodies were not sufficient to control borrelial infection in the absence of the classical pathway. Furthermore, the lack of C1q also altered the balance of the Th1/Th2 response, as both circulating Th1 (MIP-1α, IL-2, IL-12, and TNFα), Th2 (IL-4, IL-10, and MCP-1), and Th17 (IL-17) cytokines were elevated in infected C1qα-/- mice. These data imply that C1q and the classical pathway play important roles in controlling borrelial infection via antibody and complement-dependent killing, as well as altering both antibody maturation processes and the T cell response following exposure to infectious B. burgdorferi.


Subject(s)
Antibodies, Bacterial/blood , Complement C1q/immunology , Complement Pathway, Classical/immunology , Lyme Disease/immunology , Animals , Borrelia burgdorferi , Complement C1q/genetics , Cytokines/immunology , Heart/microbiology , Immunoglobulin Class Switching , Immunoglobulin G/blood , Immunoglobulin M/blood , Luminescent Measurements , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Skin/microbiology
13.
Methods Mol Biol ; 1690: 241-257, 2018.
Article in English | MEDLINE | ID: mdl-29032549

ABSTRACT

Borrelia burgdorferi, etiologic agent of Lyme disease, is the leading tick-borne disease in the United States with approximately 300,000 cases diagnosed annually. Disease occurs in stages beginning localized infection at the site of a tick bite and progresses to disseminated infection when antibiotic treatment is not administered in a timely manner. A multi-systemic infection develops following dissemination to numerous immunoprotective tissues, such as the heart, bladder, and joints, resulting in late Lyme disease. B. burgdorferi undergoes dynamic genetic regulation throughout mammalian infection and defining the exact role of virulence genes at distinct stages of disease is challenging. The murine model allows for the characterization of the pathogenic function of genes in B. burgdorferi, but traditional end point studies limit the ability to gather data throughout an infection study and greatly increase the required number of mice. Molecular genetic techniques to evaluate and quantitate B. burgdorferi infection are laborious and costly. To partly circumvent these issues, a codon optimized firefly luciferase, under the control of a constitutive borrelial promoter, was introduced into B. burgdorferi enabling the characterization of mutant or modified strains under in vitro growth conditions and throughout murine infection. The detection of bioluminescent B. burgdorferi is highly sensitive and allows for the repeated real-time quantitative evaluation of borrelial load during murine infection. Furthermore, bioluminescence has also been utilized to evaluate alteration in tissue localization and tissue-specific gene expression of B. burgdorferi. In this chapter, we describe the generation of bioluminescent borrelial strains along with methods for in vitro, in vivo, and ex vivo B. burgdorferi studies.


Subject(s)
Borrelia burgdorferi/isolation & purification , Luminescent Measurements/methods , Lyme Disease/diagnostic imaging , Lyme Disease/microbiology , Optical Imaging/methods , Animals , Borrelia burgdorferi/genetics , Disease Models, Animal , Female , Fireflies/enzymology , Gene Expression Regulation, Bacterial , Luciferases, Firefly/analysis , Luciferases, Firefly/genetics , Luminescent Agents/analysis , Luminescent Agents/metabolism , Lyme Disease/pathology , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic , Whole Body Imaging/methods
14.
PLoS Pathog ; 13(2): e1006225, 2017 02.
Article in English | MEDLINE | ID: mdl-28212410

ABSTRACT

Borrelia burgdorferi, the causative agent of Lyme disease in humans, is exposed to reactive oxygen and nitrogen species (ROS and RNS) in both the tick vector and vertebrate reservoir hosts. B. burgdorferi contains a limited repertoire of canonical oxidative stress response genes, suggesting that novel gene functions may be important for protection of B. burgdorferi against ROS or RNS exposure. Here, we use transposon insertion sequencing (Tn-seq) to conduct an unbiased search for genes involved in resistance to nitric oxide, hydrogen peroxide, and tertiary-butyl hydroperoxide in vitro. The screens identified 66 genes whose disruption resulted in increased susceptibility to at least one of the stressors. These genes include previously characterized mediators of ROS and RNS resistance (including components of the nucleotide excision repair pathway and a subunit of a riboflavin transporter), as well as novel putative resistance candidates. DNA repair mutants were among the most sensitive to RNS in the Tn-seq screen, and survival assays with individual Tn mutants confirmed that the putative ribonuclease BB0839 is involved in resistance to nitric oxide. In contrast, mutants lacking predicted inner membrane proteins or transporters were among the most sensitive to ROS, and the contribution of three such membrane proteins (BB0017, BB0164, and BB0202) to ROS sensitivity was confirmed using individual Tn mutants and complemented strains. Further analysis showed that levels of intracellular manganese are significantly reduced in the Tn::bb0164 mutant, identifying a novel role for BB0164 in B. burgdorferi manganese homeostasis. Infection of C57BL/6 and gp91phox-/- mice with a mini-library of 39 Tn mutants showed that many of the genes identified in the in vitro screens are required for infectivity in mice. Collectively, our data provide insight into how B. burgdorferi responds to ROS and RNS and suggests that this response is relevant to the in vivo success of the organism.


Subject(s)
Borrelia burgdorferi/genetics , Borrelia burgdorferi/immunology , Genes, Bacterial/immunology , Lyme Disease/microbiology , Animals , Disease Models, Animal , High-Throughput Nucleotide Sequencing , Lyme Disease/immunology , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
15.
Cell Rep ; 16(10): 2593-2604, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27568563

ABSTRACT

Systemic dissemination of microbes is critical for progression of many infectious diseases and is associated with most mortality due to bacterial infection. The physical mechanisms mediating a key dissemination step, bacterial association with vascular endothelia in blood vessels, remain unknown. Here, we show that endothelial interactions of the Lyme disease spirochete Borrelia burgdorferi under physiological shear stress mechanistically resemble selectin-dependent leukocyte rolling. Specifically, these interactions are mediated by transfer of mechanical load along a series of adhesion complexes and are stabilized by tethers and catch bond properties of the bacterial adhesin BBK32. Furthermore, we found that the forces imposed on adhesive bonds under flow may be small enough to permit active migration driven by bacterial flagellar motors. These findings provide insight into the biomechanics of bacterial-vascular interactions and demonstrate that disseminating bacteria and circulating host immune cells share widely conserved mechanisms for interacting with endothelia under physiological shear stress.


Subject(s)
Blood Vessels/microbiology , Blood Vessels/pathology , Borrelia burgdorferi/physiology , Host-Pathogen Interactions , Bacterial Adhesion , Bacterial Proteins/metabolism , Biomechanical Phenomena , Endothelial Cells/microbiology , Endothelial Cells/pathology , Humans , Leukocyte Rolling , Models, Biological , Multiprotein Complexes/metabolism , Rotation , Stress, Mechanical , Torque , Venules/pathology , Venules/virology
16.
PLoS Pathog ; 12(1): e1005404, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26808924

ABSTRACT

Pathogens that traffic in blood, lymphatics, or interstitial fluids must adopt strategies to evade innate immune defenses, notably the complement system. Through recruitment of host regulators of complement to their surface, many pathogens are able to escape complement-mediated attack. The Lyme disease spirochete, Borrelia burgdorferi, produces a number of surface proteins that bind to factor H related molecules, which function as the dominant negative regulator of the alternative pathway of complement. Relatively less is known about how B. burgdorferi evades the classical pathway of complement despite the observation that some sensu lato strains are sensitive to classical pathway activation. Here we report that the borrelial lipoprotein BBK32 potently and specifically inhibits the classical pathway by binding with high affinity to the initiating C1 complex of complement. In addition, B. burgdorferi cells that produce BBK32 on their surface bind to both C1 and C1r and a serum sensitive derivative of B. burgdorferi is protected from killing via the classical pathway in a BBK32-dependent manner. Subsequent biochemical and biophysical approaches localized the anti-complement activity of BBK32 to its globular C-terminal domain. Mechanistic studies reveal that BBK32 acts by entrapping C1 in its zymogen form by binding and inhibiting the C1 subcomponent, C1r, which serves as the initiating serine protease of the classical pathway. To our knowledge this is the first report of a spirochetal protein acting as a direct inhibitor of the classical pathway and is the only example of a biomolecule capable of specifically and noncovalently inhibiting C1/C1r. By identifying a unique mode of complement evasion this study greatly enhances our understanding of how pathogens subvert and potentially manipulate host innate immune systems.


Subject(s)
Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Complement Activation/immunology , Complement Pathway, Classical/immunology , Host-Parasite Interactions/immunology , Immune Evasion/immunology , Complement C1/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Immunoprecipitation , Lyme Disease/immunology
17.
Infect Immun ; 83(9): 3693-703, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26150534

ABSTRACT

Borrelia burgdorferi, the etiologic agent of Lyme disease, produces a variety of proteins that promote survival and colonization in both the Ixodes species vector and various mammalian hosts. We initially identified BB0744 (also known as p83/100) by screening for B. burgdorferi strain B31 proteins that bind to α1ß1 integrin and hypothesized that, given the presence of a signal peptide, BB0744 may be a surface-exposed protein. In contrast to this expectation, localization studies suggested that BB0744 resides in the periplasm. Despite its subsurface location, we were interested in testing whether BB0744 is required for borrelial pathogenesis. To this end, a bb0744 deletion was isolated in a B. burgdorferi strain B31 infectious background, complemented, and queried for the role of BB0744 following experimental infection. A combination of bioluminescent imaging, cultivation of infected tissues, and quantitative PCR (qPCR) demonstrated that Δbb0744 mutant B. burgdorferi bacteria were attenuated in the ability to colonize heart tissue, as well as skin locations distal to the site of infection. Furthermore, qPCR indicated a significantly reduced spirochetal load in distal skin and joint tissue infected with Δbb0744 mutant B. burgdorferi. Complementation with bb0744 restored infectivity, indicating that the defect seen in Δbb0744 mutant B. burgdorferi was due to the loss of BB0744. Taken together, these results suggest that BB0744 is necessary for tissue tropism, particularly in heart tissue, alters the ability of B. burgdorferi to disseminate efficiently, or both. Additional studies are warranted to address the mechanism employed by BB0744 that alters the pathogenic potential of B. burgdorferi.


Subject(s)
Adhesins, Bacterial/metabolism , Borrelia burgdorferi/pathogenicity , Lyme Disease/microbiology , Animals , Borrelia burgdorferi/metabolism , Disease Models, Animal , Female , Gene Knockdown Techniques , Immunoblotting , Luminescent Measurements , Lyme Disease/metabolism , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
18.
Mol Microbiol ; 96(1): 68-83, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25560615

ABSTRACT

Borrelia burgdorferi, the etiologic agent of Lyme disease, adapts to the mammalian hosts by differentially expressing several genes in the BosR and Rrp2-RpoN-RpoS dependent pathways, resulting in a distinct protein profile relative to that seen for survival in the Ixodes spp. tick. Previous studies indicate that a putative lipoprotein, BBA33, is produced in an RpoS-dependent manner under conditions that mimic the mammalian component of the borrelial lifecycle. However, the significance and function for BBA33 is not known. Given its linkage to the BosR/Rrp2-RpoN-RpoS regulatory cascade, we hypothesized that BBA33 facilitates B. burgdorferi infection in the mammalian host. The deletion of bba33 eliminated B. burgdorferi infectivity in C3H mice, which was rescued by genetic complementation with intact bba33. With regard to function, a combinatorial peptide approach, coupled with subsequent in vitro binding assays, indicated that BBA33 binds to collagen type VI and, to a lesser extent, collagen type IV. Whole cell binding assays demonstrated BBA33-dependent binding to human collagen type VI. Taken together, these results suggest that BBA33 interacts with collagenous structures and may function as an adhesin in a process that is required to prevent bacterial clearance.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Borrelia burgdorferi/pathogenicity , Collagen/metabolism , Gene Expression Regulation, Bacterial , Lipoproteins/metabolism , Adhesins, Bacterial/metabolism , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Gene Deletion , Gene Expression Profiling , Genetic Complementation Test , Humans , Lipoproteins/genetics , Mice , Mice, Inbred C3H , Protein Binding
19.
Mol Microbiol ; 86(5): 1116-31, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23095033

ABSTRACT

Systemic dissemination of microbial pathogens permits microbes to spread from the initial site of infection to secondary target tissues and is responsible for most mortality due to bacterial infections. Dissemination is a critical stage of disease progression by the Lyme spirochaete, Borrelia burgdorferi. However, many mechanistic features of the process are not yet understood. A key step is adhesion of circulating microbes to vascular surfaces in the face of the shear forces present in flowing blood. Using real-time microscopic imaging of the Lyme spirochaete in living mice we previously identified the first bacterial protein (B. burgdorferi BBK32) shown to mediate vascular adhesion in vivo. Vascular adhesion is also dependent on host fibronectin (Fn) and glycosaminoglycans (GAGs). In the present study, we investigated the mechanisms of BBK32-dependent vascular adhesion in vivo. We determined that BBK32-Fn interactions (tethering) function as a molecular braking mechanism that permits the formation of more stable BBK32-GAG interactions (dragging) between circulating bacteria and vascular surfaces. Since BBK32-like proteins are expressed in a variety of pathogens we believe that the vascular adhesion mechanisms we have deciphered here may be critical for understanding the dissemination mechanisms of other bacterial pathogens.


Subject(s)
Bacterial Adhesion , Bacterial Proteins/metabolism , Borrelia burgdorferi/physiology , Endothelium, Vascular/metabolism , Fibronectins/metabolism , Glycosaminoglycans/metabolism , Host-Pathogen Interactions , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Borrelia burgdorferi/metabolism , Endothelium, Vascular/microbiology , Lyme Disease/microbiology , Lyme Disease/physiopathology , Mice , Protein Binding
20.
Mol Microbiol ; 83(2): 319-34, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22151008

ABSTRACT

The etiological agent of Lyme disease, Borrelia burgdorferi, is transmitted by ticks of the Ixodes genus and, if untreated, can cause significant morbidity in affected individuals. Recent reports have shown that polyunsaturated fatty acids in the B. burgdorferi cell envelope are potential targets for oxidative damage, which can be lethal. How B. burgdorferi responds to this assault is not known. Herein we report evidence that bb0646 codes for a lipase that is located within the bosR operon and that has specificity for both saturated and polyunsaturated fatty acids. Specifically, strains harbouring mutated copies of the lipase, either in the form of an insertionally inactivated construct or site-directed mutations within the active site, demonstrated attenuated lipolytic and haemolytic phenotypes when compared with the isogenic parent and trans-complements. In vivo analysis showed that while the bb0646 mutant remains infectious, the spirochaetal load is significantly lower than both the isogenic parent and the complemented mutant strains. Taken together, these data demonstrate that BB0646 is a broad substrate specific lipase that contributes to lipolytic and haemolytic activity in vitro and is required for optimal B. burgdorferi infection.


Subject(s)
Borrelia burgdorferi/enzymology , Hemolysin Proteins/metabolism , Lipase/metabolism , Animal Structures/microbiology , Animals , Bacterial Load , Borrelia burgdorferi/genetics , Disease Models, Animal , Fatty Acids/metabolism , Gene Knockout Techniques , Genetic Complementation Test , Hemolysin Proteins/genetics , Lipase/genetics , Lyme Disease/microbiology , Lyme Disease/pathology , Mice , Mutagenesis, Insertional , Mutagenesis, Site-Directed , Mutation, Missense , Operon
SELECTION OF CITATIONS
SEARCH DETAIL
...