Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 120(27): e2301549120, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37364114

ABSTRACT

Modern infectious disease outbreaks often involve changes in host tropism, the preferential adaptation of pathogens to specific hosts. The Lyme disease-causing bacterium Borrelia burgdorferi (Bb) is an ideal model to investigate the molecular mechanisms of host tropism, because different variants of these tick-transmitted bacteria are distinctly maintained in rodents or bird reservoir hosts. To survive in hosts and escape complement-mediated immune clearance, Bb produces the outer surface protein CspZ that binds the complement inhibitor factor H (FH) to facilitate bacterial dissemination in vertebrates. Despite high sequence conservation, CspZ variants differ in human FH-binding ability. Together with the FH polymorphisms between vertebrate hosts, these findings suggest that minor sequence variation in this bacterial outer surface protein may confer dramatic differences in host-specific, FH-binding-mediated infectivity. We tested this hypothesis by determining the crystal structure of the CspZ-human FH complex, and identifying minor variation localized in the FH-binding interface yielding bird and rodent FH-specific binding activity that impacts infectivity. Swapping the divergent region in the FH-binding interface between rodent- and bird-associated CspZ variants alters the ability to promote rodent- and bird-specific early-onset dissemination. We further linked these loops and respective host-specific, complement-dependent phenotypes with distinct CspZ phylogenetic lineages, elucidating evolutionary mechanisms driving host tropism emergence. Our multidisciplinary work provides a novel molecular basis for how a single, short protein motif could greatly modulate pathogen host tropism.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Animals , Humans , Immune Evasion/genetics , Phylogeny , Viral Tropism , Lyme Disease/microbiology , Bacterial Proteins/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Complement System Proteins/genetics , Membrane Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 119(13): e2117770119, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35312359

ABSTRACT

Spirochetal pathogens, such as the causative agent of Lyme disease, Borrelia burgdorferi sensu lato, encode an abundance of lipoproteins; however, due in part to their evolutionary distance from more well-studied bacteria, such as Proteobacteria and Firmicutes, few spirochetal lipoproteins have assigned functions. Indeed, B. burgdorferi devotes almost 8% of its genome to lipoprotein genes and interacts with its environment primarily through the production of at least 80 surface-exposed lipoproteins throughout its tick vector­vertebrate host lifecycle. Several B. burgdorferi lipoproteins have been shown to serve roles in cellular adherence or immune evasion, but the functions for most B. burgdorferi surface lipoproteins remain unknown. In this study, we developed a B. burgdorferi lipoproteome screening platform utilizing intact spirochetes that enables the identification of previously unrecognized host interactions. As spirochetal survival in the bloodstream is essential for dissemination, we targeted our screen to C1, the first component of the classical (antibody-initiated) complement pathway. We identified two high-affinity C1 interactions by the paralogous lipoproteins, ElpB and ElpQ (also termed ErpB and ErpQ, respectively). Using biochemical, microbiological, and biophysical approaches, we demonstrate that ElpB and ElpQ bind the activated forms of the C1 proteases, C1r and C1s, and represent a distinct mechanistic class of C1 inhibitors that protect the spirochete from antibody-mediated complement killing. In addition to identifying a mode of complement inhibition, our study establishes a lipoproteome screening methodology as a discovery platform for identifying direct host­pathogen interactions that are central to the pathogenesis of spirochetes, such as the Lyme disease agent.


Subject(s)
Bacterial Proteins , Borrelia burgdorferi , Complement C1q , Immune Evasion , Lipoproteins , Lyme Disease , Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Complement C1q/immunology , Humans , Immunoglobulins/immunology , Lipoproteins/immunology , Lyme Disease/immunology , Lyme Disease/microbiology , Proteome/immunology
3.
PLoS Pathog ; 17(7): e1009801, 2021 07.
Article in English | MEDLINE | ID: mdl-34324600

ABSTRACT

Pathogens possess the ability to adapt and survive in some host species but not in others-an ecological trait known as host tropism. Transmitted through ticks and carried mainly by mammals and birds, the Lyme disease (LD) bacterium is a well-suited model to study such tropism. Three main causative agents of LD, Borrelia burgdorferi, B. afzelii, and B. garinii, vary in host ranges through mechanisms eluding characterization. By feeding ticks infected with different Borrelia species, utilizing feeding chambers and live mice and quail, we found species-level differences in bacterial transmission. These differences localize on the tick blood meal, and specifically complement, a defense in vertebrate blood, and a polymorphic bacterial protein, CspA, which inactivates complement by binding to a host complement inhibitor, Factor H (FH). CspA selectively confers bacterial transmission to vertebrates that produce FH capable of allele-specific recognition. CspA is the only member of the Pfam54 gene family to exhibit host-specific FH-binding. Phylogenetic analyses revealed convergent evolution as the driver of such uniqueness, and that FH-binding likely emerged during the last glacial maximum. Our results identify a determinant of host tropism in Lyme disease infection, thus defining an evolutionary mechanism that shapes host-pathogen associations.


Subject(s)
Bacterial Proteins/genetics , Borrelia burgdorferi/growth & development , Lyme Disease/immunology , Lyme Disease/transmission , Viral Tropism/physiology , Animals , Bacterial Proteins/metabolism , Biological Evolution , Borrelia burgdorferi/genetics , Borrelia burgdorferi/immunology , Complement Factor H/metabolism , Host-Pathogen Interactions/physiology , Humans , Immune Evasion/physiology , Mice , Quail , Species Specificity , Ticks
4.
Appl Environ Microbiol ; 88(5): e0155521, 2022 03 08.
Article in English | MEDLINE | ID: mdl-34986011

ABSTRACT

Lyme borreliosis is the most common vector-borne disease in the Northern Hemisphere, caused by spirochetes belonging to the Borrelia burgdorferi sensu lato species complex, which are transmitted by ixodid ticks. B. burgdorferi sensu lato species produce a family of proteins on the linear plasmid 54 (PFam54), some of which confer the functions of cell adhesion and inactivation of complement, the first line of host defense. However, the impact of PFam54 in promoting B. burgdorferi sensu lato pathogenesis remains unclear because of the hurdles to simultaneously knock out all PFam54 proteins in a spirochete. Here, we describe two Borrelia bavariensis strains, PBN and PNi, isolated from patients naturally lacking PFam54 but maintaining the rest of the genome with greater than 95% identity to the reference B. bavariensis strain, PBi. We found that PBN and PNi less efficiently survive in human serum than PBi. Such defects were restored by introducing two B. bavariensis PFam54 recombinant proteins, BGA66 and BGA71, confirming the role of these proteins in providing complement evasion of B. bavariensis. Further, we found that all three strains remain detectable in various murine tissues 21 days post-subcutaneous infection, supporting the nonessential role of B. bavariensis PFam54 in promoting spirochete persistence. This study identified and utilized isolates deficient in PFam54 to associate the defects with the absence of these proteins, building the foundation to further study the role of each PFam54 protein in contributing to B. burgdorferi sensu lato pathogenesis. IMPORTANCE To establish infections, Lyme borreliae utilize various means to overcome the host's immune system. Proteins encoded by the PFam54 gene array play a role in spirochete survival in vitro and in vivo. Moreover, this gene array has been described in all currently available Lyme borreliae genomes. By investigating the first two Borrelia bavariensis isolates naturally lacking the entire PFam54 gene array, we showed that both patient isolates display an increased susceptibility to human serum, which can be rescued in the presence of two PFam54 recombinant proteins. However, both isolates remain infectious to mice after intradermal inoculation, suggesting the nonessential role of PFam54 during the long-term, but may differ slightly in the colonization of specific tissues. Furthermore, these isolates show high genomic similarity to type strain PBi (>95%) and could be used in future studies investigating the role of each PFam54 protein in Lyme borreliosis pathogenesis.


Subject(s)
Borrelia burgdorferi Group , Borrelia , Ixodes , Lyme Disease , Animals , Borrelia/genetics , Borrelia burgdorferi Group/genetics , Humans , Mice , Plasmids , Spirochaetales
5.
Parasitol Res ; 121(3): 781-803, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35122516

ABSTRACT

Beside mosquitoes, ticks are well-known vectors of different human pathogens. In the Northern Hemisphere, Lyme borreliosis (Eurasia, LB) or Lyme disease (North America, LD) is the most commonly occurring vector-borne infectious disease caused by bacteria of the genus Borrelia which are transmitted by hard ticks of the genus Ixodes. The reported incidence of LB in Europe is about 22.6 cases per 100,000 inhabitants annually with a broad range depending on the geographical area analyzed. However, the epidemiological data are largely incomplete, because LB is not notifiable in all European countries. Furthermore, not only differ reporting procedures between countries, there is also variation in case definitions and diagnostic procedures. Lyme borreliosis is caused by several species of the Borrelia (B.) burgdorferi sensu lato (s.l.) complex which are maintained in complex networks including ixodid ticks and different reservoir hosts. Vector and host influence each other and are affected by multiple factors including climate that have a major impact on their habitats and ecology. To classify factors that influence the risk of transmission of B. burgdorferi s.l. to their different vertebrate hosts as well as to humans, we briefly summarize the current knowledge about the pathogens including their astonishing ability to overcome various host immune responses, regarding the main vector in Europe Ixodes ricinus, and the disease caused by borreliae. The research shows, that a higher standardization of case definition, diagnostic procedures, and standardized, long-term surveillance systems across Europe is necessary to improve clinical and epidemiological data.


Subject(s)
Borrelia burgdorferi , Ixodes , Lyme Disease , Animals , Europe/epidemiology , Humans , Ixodes/microbiology , Mosquito Vectors
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.
Infect Immun ; 88(5)2020 04 20.
Article in English | MEDLINE | ID: mdl-32122944

ABSTRACT

The spirochete Borrelia burgdorferisensu lato is the causative agent of Lyme disease (LD). The spirochetes produce the CspZ protein that binds to a complement regulator, factor H (FH). Such binding downregulates activation of host complement to facilitate spirochete evasion of complement killing. However, vaccination with CspZ does not protect against LD infection. In this study, we demonstrated that immunization with CspZ-YA, a CspZ mutant protein with no FH-binding activity, protected mice from infection by several spirochete genotypes introduced via tick feeding. We found that the sera from CspZ-YA-vaccinated mice more efficiently eliminated spirochetes and blocked CspZ FH-binding activity than sera from CspZ-immunized mice. We also found that vaccination with CspZ, but not CspZ-YA, triggered the production of anti-FH antibodies, justifying CspZ-YA as an LD vaccine candidate. The mechanistic and efficacy information derived from this study provides insights into the development of a CspZ-based LD vaccine.


Subject(s)
Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Complement Factor H/immunology , Lyme Disease/immunology , Ticks/microbiology , Animals , Antibodies/immunology , Binding Sites/immunology , Complement System Proteins/immunology , Female , Humans , Lyme Disease Vaccines/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H
8.
PLoS Pathog ; 14(5): e1007106, 2018 05.
Article in English | MEDLINE | ID: mdl-29813137

ABSTRACT

Borrelia burgdorferi sensu lato (Bbsl), the causative agent of Lyme disease, establishes an initial infection in the host's skin following a tick bite, and then disseminates to distant organs, leading to multisystem manifestations. Tick-to-vertebrate host transmission requires that Bbsl survives during blood feeding. Complement is an important innate host defense in blood and interstitial fluid. Bbsl produces a polymorphic surface protein, CspA, that binds to a complement regulator, Factor H (FH) to block complement activation in vitro. However, the role that CspA plays in the Bbsl enzootic cycle remains unclear. In this study, we demonstrated that different CspA variants promote spirochete binding to FH to inactivate complement and promote serum resistance in a host-specific manner. Utilizing a tick-to-mouse transmission model, we observed that a cspA-knockout B. burgdorferi is eliminated from nymphal ticks in the first 24 hours of feeding and is unable to be transmitted to naïve mice. Conversely, ectopically producing CspA derived from B. burgdorferi or B. afzelii, but not B. garinii in a cspA-knockout strain restored spirochete survival in fed nymphs and tick-to-mouse transmission. Furthermore, a CspA point mutant, CspA-L246D that was defective in FH-binding, failed to survive in fed nymphs and at the inoculation site or bloodstream in mice. We also allowed those spirochete-infected nymphs to feed on C3-/- mice that lacked functional complement. The cspA-knockout B. burgdorferi or this mutant strain complemented with cspA variants or cspA-L246D was found at similar levels as wild type B. burgdorferi in the fed nymphs and mouse tissues. These novel findings suggest that the FH-binding activity of CspA protects spirochetes from complement-mediated killing in fed nymphal ticks, which ultimately allows Bbsl transmission to mammalian hosts.


Subject(s)
Arachnid Vectors/microbiology , Bacterial Proteins/metabolism , Borrelia burgdorferi Group/physiology , Complement Factor H/metabolism , Ixodes/microbiology , Lyme Disease/transmission , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi Group/immunology , Complement Factor H/genetics , Complement System Proteins/metabolism , Coturnix , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flagellin/genetics , Flagellin/metabolism , Flow Cytometry , Horses , Humans , Lyme Disease/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Nymph/microbiology , Polymorphism, Genetic , Species Specificity , Surface Plasmon Resonance
9.
Cell Microbiol ; 21(2): e12998, 2019 02.
Article in English | MEDLINE | ID: mdl-30571845

ABSTRACT

Lyme disease, caused by the spirochete Borrelia burgdorferi, is the most common vector-borne disease in the United States and Europe. The spirochetes are transmitted from mammalian and avian reservoir hosts to humans via ticks. Following tick bites, spirochetes colonize the host skin and then disseminate haematogenously to various organs, a process that requires this pathogen to evade host complement, an innate immune defence system. CspZ, a spirochete surface protein, facilitates resistance to complement-mediated killing in vitro by binding to the complement regulator, factor H (FH). Low expression levels of CspZ in spirochetes cultivated in vitro or during initiation of infection in vivo have been a major hurdle in delineating the role of this protein in pathogenesis. Here, we show that treatment of B. burgdorferi with human blood induces CspZ production and enhances resistance to complement. By contrast, a cspZ-deficient mutant and a strain that expressed an FH-nonbinding CspZ variant were impaired in their ability to cause bacteraemia and colonize tissues of mice or quail; virulence of these mutants was however restored in complement C3-deficient mice. These novel findings suggest that FH binding to CspZ facilitates B. burgdorferi complement evasion in vivo and promotes systemic infection in vertebrate hosts.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/immunology , Complement C3/immunology , Lyme Disease/immunology , Membrane Proteins/metabolism , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi/pathogenicity , Complement C3/genetics , Complement Factor H/immunology , Complement Factor H/metabolism , Coturnix , Humans , Ixodes/microbiology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
10.
Med Microbiol Immunol ; 209(3): 277-299, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31784893

ABSTRACT

The capacity of pathogenic microorganisms to adhere to host cells and avoid clearance by the host immune system is the initial and most decisive step leading to infections. Bacteria have developed different strategies to attach to diverse host surface structures. One important strategy is the adhesion to extracellular matrix (ECM) proteins (e.g., collagen, fibronectin, laminin) that are highly abundant in connective tissue and basement membranes. Gram-negative bacteria express variable outer membrane proteins (adhesins) to attach to the host and to initiate the process of infection. Understanding the underlying molecular mechanisms of bacterial adhesion is a prerequisite for targeting this interaction by "anti-ligands" to prevent colonization or infection of the host. Future development of such "anti-ligands" (specifically interfering with bacteria-host matrix interactions) might result in the development of a new class of anti-infective drugs for the therapy of infections caused by multidrug-resistant Gram-negative bacteria. This review summarizes our current knowledge about the manifold interactions of adhesins expressed by Gram-negative bacteria with ECM proteins and the use of this information for the generation of novel therapeutic antivirulence strategies.


Subject(s)
Adhesins, Bacterial/physiology , Bacterial Adhesion , Extracellular Matrix Proteins/physiology , Fibronectins/physiology , Gram-Negative Bacteria/physiology , Host Microbial Interactions , Gram-Negative Bacteria/pathogenicity , Humans
11.
Mol Microbiol ; 99(2): 407-24, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26434356

ABSTRACT

Borrelia (B.) bavariensis exhibits a marked tropism for nervous tissues and frequently causes neurological manifestations in humans. The molecular mechanism by which B. bavariensis overcomes innate immunity, in particular, complement remains elusive. In contrast to other serum-resistant spirochetes, none of the B. bavariensis isolates investigated bound complement regulators of the alternative (AP) and classical pathway (CP) or proteolytically inactivated complement components. Focusing on outer surface proteins BGA66 and BGA71, we demonstrated that both molecules either inhibit AP, CP and terminal pathway (TP) activation, or block activation of the CP and TP respectively. Both molecules bind complement components C7, C8 and C9, and thereby prevent assembly of the terminal complement complex. This inhibitory activity was confirmed by the introduction of the BGA66 and BGA71 encoding genes into a serum-sensitive B. garinii strain. Transformed spirochetes producing either BGA66 or BGA71 overcome complement-mediated killing, thus indicating that both proteins independently facilitate serum resistance of B. bavariensis. The generation of C-terminally truncated proteins as well as a chimeric BGA71 protein lead to the localization of the complement-interacting binding site within the N-terminus. Collectively, our data reveal a novel immune evasion strategy of B. bavariensis that is directed against the activation of the TP.


Subject(s)
Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Complement System Proteins/immunology , Lyme Disease/immunology , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/immunology , Humans , Lyme Disease/microbiology , Mice
12.
J Infect Dis ; 213(9): 1388-99, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26681776

ABSTRACT

Acinetobacter baumannii is an emerging opportunistic pathogen, responsible for up to 10% of gram-negative, nosocomial infections. The global increase of multidrug-resistant and pan-resistant Acinetobacter isolates presents clinicians with formidable challenges. To establish a persistent infection,A. baumannii must overcome the detrimental effects of complement as the first line of defense against invading microorganisms. However, the immune evasion principles underlying serum resistance inA. baumannii remain elusive. Here, we identified a novel plasminogen-binding protein, termed CipA. Bound plasminogen, upon conversion to active plasmin, degraded fibrinogen and complement C3b and contributed to serum resistance. Furthermore, CipA directly inhibited the alternative pathway of complement in vitro, irrespective of its ability to bind plasminogen. A CipA-deficient mutant was efficiently killed by human serum and showed a defect in the penetration of endothelial monolayers, demonstrating that CipA is a novel multifunctional protein that contributes to the pathogenesis ofA. baumannii.


Subject(s)
Acinetobacter baumannii/metabolism , Acinetobacter baumannii/pathogenicity , Bacterial Proteins/metabolism , Complement System Proteins/metabolism , Plasminogen/metabolism , Acinetobacter Infections/microbiology , Acinetobacter baumannii/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Membrane/chemistry , Complement System Proteins/chemistry , Humans , Plasminogen/chemistry , Protein Binding
13.
J Cell Mol Med ; 19(10): 2507-11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26152778

ABSTRACT

Cytokine networks initiated by means of innate immunity are regarded as a major determinant of host defence in response to acute infection by bacteria including Borrelia burgdorferi. Herein, we demonstrate that interferon (IFN)-α, either endogenously produced after exposure of cells to toll-like receptor-9-activating CpG oligonucleotides or provided as recombinant cytokine, weakens activation of the anti-bacterial interleukin (IL)-1/IL-22 axis in human peripheral blood mononuclear cells exposed to viable B. burgdorferi. As IFN-α has been related to pathological dissemination of the spirochaete, data suggest an immunoregulatory role of type I IFN in this context that is able to significantly modify cytokine profiles thereby possibly determining early course of B. burgdorferi infection.


Subject(s)
Borrelia burgdorferi/physiology , Interferon-alpha/pharmacology , Interleukins/biosynthesis , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/microbiology , Borrelia burgdorferi/drug effects , Humans , Leukocytes, Mononuclear/drug effects , Oligodeoxyribonucleotides/pharmacology , Interleukin-22
14.
Mol Med ; 21: 26-37, 2015 Feb 23.
Article in English | MEDLINE | ID: mdl-25730773

ABSTRACT

In a first genome-wide association study (GWAS) approach to anti-Borrelia seropositivity, we identified two significant single nucleotide polymorphisms (SNPs) (rs17850869, P = 4.17E-09; rs41289586, P = 7.18E-08). Both markers, located on chromosomes 16 and 3, respectively, are within or close to genes previously connected to spinocerebellar ataxia. The risk SNP rs41289586 represents a missense variant (R263H) of anoctamin 10 (ANO10), a member of a protein family encoding Cl(-) channels and phospholipid scramblases. ANO10 augments volume-regulated Cl(-) currents (IHypo) in Xenopus oocytes, HEK293 cells, lymphocytes and macrophages and controls volume regulation by enhancing regulatory volume decrease (RVD). ANO10 supports migration of macrophages and phagocytosis of spirochetes. The R263H variant is inhibitory on IHypo, RVD and intracellular Ca(2+) signals, which may delay spirochete clearance, thereby sensitizing adaptive immunity. Our data demonstrate for the first time that ANO10 has a central role in innate immune defense against Borrelia infection.


Subject(s)
Borrelia Infections/genetics , Borrelia Infections/immunology , Borrelia/immunology , Genetic Variation , Macrophages/metabolism , Membrane Proteins/genetics , Open Reading Frames , Animals , Anoctamins , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Borrelia Infections/epidemiology , Borrelia Infections/microbiology , Case-Control Studies , Cell Line , Cell Size , Gene Expression , Genome-Wide Association Study , HEK293 Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Macrophages/pathology , Mental Disorders/genetics , Mental Disorders/microbiology , Oocytes , Phenotype , Polymorphism, Single Nucleotide , Seroepidemiologic Studies
15.
J Biol Chem ; 288(35): 25229-25243, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23861404

ABSTRACT

The Lyme disease spirochete Borrelia burgdorferi lacks endogenous, surface-exposed proteases. In order to efficiently disseminate throughout the host and penetrate tissue barriers, borreliae rely on recruitment of host proteases, such as plasmin(ogen). Here we report the identification of a novel plasminogen-binding protein, BBA70. Binding of plasminogen is dose-dependent and is affected by ionic strength. The BBA70-plasminogen interaction is mediated by lysine residues, primarily located in a putative C-terminal α-helix of BBA70. These lysine residues appear to interact with the lysine-binding sites in plasminogen kringle domain 4 because a deletion mutant of plasminogen lacking that domain was unable to bind to BBA70. Bound to BBA70, plasminogen activated by urokinase-type plasminogen activator was able to degrade both a synthetic chromogenic substrate and the natural substrate fibrinogen. Furthermore, BBA70-bound plasmin was able to degrade the central complement proteins C3b and C5 and inhibited the bacteriolytic effects of complement. Consistent with these functional activities, BBA70 is located on the borrelial outer surface. Additionally, serological evidence demonstrated that BBA70 is produced during mammalian infection. Taken together, recruitment and activation of plasminogen could play a beneficial role in dissemination of B. burgdorferi in the human host and may possibly aid the spirochete in escaping the defense mechanisms of innate immunity.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Plasminogen/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Borrelia burgdorferi/chemistry , Borrelia burgdorferi/genetics , Borrelia burgdorferi/immunology , Complement C3b/chemistry , Complement C3b/genetics , Complement C3b/immunology , Complement C3b/metabolism , Complement C5/chemistry , Complement C5/genetics , Complement C5/immunology , Complement C5/metabolism , Fibrinolysin/chemistry , Fibrinolysin/genetics , Fibrinolysin/immunology , Fibrinolysin/metabolism , Humans , Immunity, Innate , Lyme Disease/genetics , Lyme Disease/immunology , Lyme Disease/metabolism , Plasminogen/chemistry , Plasminogen/genetics , Plasminogen/immunology , Protein Binding , Protein Structure, Tertiary , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/immunology , Urokinase-Type Plasminogen Activator/metabolism
16.
Infect Immun ; 82(1): 380-92, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24191298

ABSTRACT

CspA of the Lyme disease spirochete Borrelia burgdorferi represents a key molecule in immune evasion, protecting borrelial cells from complement-mediated killing. As previous studies focused almost exclusively on CspA of B. burgdorferi, here we investigate the different binding capacities of CspA orthologs of Borrelia burgdorferi, B. afzelii, and B. spielmanii for complement regulator factor H and plasminogen and their ability to inhibit complement activation by either binding these host-derived plasma proteins or independently by direct interaction with components involved in formation of the lethal, pore-like terminal complement complex. To further examine their function in serum resistance in vivo, a serum-sensitive B. garinii strain was used to generate spirochetes, ectopically producing functional CspA orthologs. Irrespective of their species origin, all three CspA orthologs impart resistance to complement-mediated killing when produced in a serum-sensitive B. garinii surrogate strain. To analyze the inhibitory effect on complement activation and to assess the potential to inactivate C3b by binding of factor H and plasminogen, recombinant CspA orthologs were also investigated. All three CspA orthologs simultaneously bound factor H and plasminogen but differed in regard to their capacity to inactivate C3b via bound plasmin(ogen) and inhibit formation of the terminal complement complex. CspA of B. afzelii binds plasmin(ogen) and inhibits the terminal complement complex more efficiently than CspA of B. burgdorferi and B. spielmanii. Taken together, CspA orthologs of serum-resistant Lyme disease spirochetes act as multifunctional evasion molecules that inhibit complement on two central activation levels, C3b generation and assembly of the terminal complement complex.


Subject(s)
Bacterial Proteins/physiology , Borrelia burgdorferi/physiology , Complement System Proteins/metabolism , Lyme Disease/microbiology , Analysis of Variance , Bacteriolysis/physiology , Blood Bactericidal Activity , Borrelia/physiology , Cells, Cultured , Complement C3b/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Lyme Disease/immunology , Plasminogen/metabolism , Protein Binding/physiology
17.
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
18.
Travel Med Infect Dis ; 59: 102714, 2024.
Article in English | MEDLINE | ID: mdl-38490400

ABSTRACT

BACKGROUND: Tick- and louse-borne relapsing fever are highly-neglected, vector-borne diseases caused by diverse Borrelia species. Presently, there are no data available on the endemicity of tick- and louse-borne relapsing fever spirochetes in Kenya. Here, we present data of a retrospective study on the seroprevalence of louse-borne relapsing fever (LBRF) in northern Kenya. METHODS: A novel immunoassay, recently established for the diagnosis of LBRF was utilized to screen 2005 blood samples collected from individuals with fever without a source in Turkana County, Kenya between May 2009 and November 2010 for anti-LBRF antibodies. RESULTS: Out of the 2005 sera analyzed, 287 samples (14.3 %) were considered anti-LBRF IgG positive. Subsequent analyses revealed that 87 out of 152 sera randomly selected from these 2005 samples were tested positive (57.2 %) for anti-LBRF IgM antibodies. Most of the IgG and IgM positive samples were from individuals living in northern regions of Turkana County. CONCLUSION: Our serological finding provides strong evidence for the occurrence of LBRF in Kenya.


Subject(s)
Antibodies, Bacterial , Borrelia , Immunoglobulin G , Immunoglobulin M , Relapsing Fever , Kenya/epidemiology , Relapsing Fever/epidemiology , Relapsing Fever/diagnosis , Relapsing Fever/microbiology , Relapsing Fever/blood , Humans , Seroepidemiologic Studies , Retrospective Studies , Male , Female , Antibodies, Bacterial/blood , Immunoglobulin G/blood , Borrelia/immunology , Immunoglobulin M/blood , Adult , Animals , Adolescent , Middle Aged , Young Adult , Child , Child, Preschool
19.
Ecol Evol ; 14(5): e11397, 2024 May.
Article in English | MEDLINE | ID: mdl-38779535

ABSTRACT

Lyme borreliosis (LB) is the most common vector-borne disease in the Northern Hemisphere caused by spirochetes belonging to the Borrelia burgdorferi sensu lato (Bbsl) complex. Borrelia spirochetes circulate in obligatory transmission cycles between tick vectors and different vertebrate hosts. To successfully complete this complex transmission cycle, Bbsl encodes for an arsenal of proteins including the PFam54 protein family with known, or proposed, influences to reservoir host and/or vector adaptation. Even so, only fragmentary information is available regarding the naturally occurring level of variation in the PFam54 gene array especially in relation to Eurasian-distributed species. Utilizing whole genome data from isolates (n = 141) originated from three major LB-causing Borrelia species across Eurasia (B. afzelii, B. bavariensis, and B. garinii), we aimed to characterize the diversity of the PFam54 gene array in these isolates to facilitate understanding the evolution of PFam54 paralogs on an intra- and interspecies level. We found an extraordinarily high level of variation in the PFam54 gene array with 39 PFam54 paralogs belonging to 23 orthologous groups including five novel paralogs. Even so, the gene array appears to have remained fairly stable over the evolutionary history of the studied Borrelia species. Interestingly, genes outside Clade IV, which contains genes encoding for proteins associated with Borrelia pathogenesis, more frequently displayed signatures of diversifying selection between clades that differ in hypothesized vector or host species. This could suggest that non-Clade IV paralogs play a more important role in host and/or vector adaptation than previously expected, which would require future lab-based studies to validate.

20.
Article in English | MEDLINE | ID: mdl-23722838

ABSTRACT

Borrelia burgdorferi is a spirochete responsible for Lyme disease, the most commonly occurring vector-borne disease in Europe and North America. The bacterium utilizes a set of proteins, termed complement regulator-acquiring surface proteins (CRASPs), to aid evasion of the human complement system by recruiting and presenting complement regulator factor H on its surface in a manner that mimics host cells. Presented here is the atomic resolution structure of a member of this protein family, ErpC. The structure provides new insights into the mechanism of recruitment of factor H and other factor H-related proteins by acting as a molecular mimic of host glycosaminoglycans. It also describes the architecture of other CRASP proteins belonging to the OspE/F-related paralogous protein family and suggests that they have evolved to bind specific complement proteins, aiding survival of the bacterium in different hosts.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Borrelia burgdorferi , Membrane Proteins/chemistry , Receptors, Cell Surface/chemistry , Bacterial Outer Membrane Proteins/metabolism , Base Sequence , Binding Sites , Complement System Proteins/chemistry , Complement System Proteins/metabolism , Crystallization , Membrane Proteins/metabolism , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Cell Surface/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL