1
|
Brangulis K, Sürth V, Marcinkiewicz AL, Akopjana I, Kazaks A, Bogans J, Huber A, Lin YP, Kraiczy P. CspZ variant-specific interaction with Factor H incorporates a metal site to support Lyme borreliae complement evasion. J Biol Chem 2024:108083. [PMID: 39675703 DOI: 10.1016/j.jbc.2024.108083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024] Open
Abstract
Polymorphic microbial immune evasion proteins dictate the pathogen species- or strain-specific virulence. Metals can impact how microbial proteins confer host-pathogen interactions, but whether this activity can be allelically variable is unclear. Here, we investigate the polymorphic CspZ protein of Lyme disease (LD) spirochete bacteria to assess the role of metals in protein-protein interaction. CspZ facilitates evasion of the complement system, the first-line of immune defense through binding to the complement regulator Factor H (FH). By obtaining a high-resolution co-crystal CspZ-FH structure, we identified a zinc coordinating the binding of FH SCR6-7 domains to a Glu65 on a loop from CspZ of B. burgdorferi B31. However, zinc is dispensable for human FH binding for CspZ orthologs with a different loop orientation and/or lacking this glutamate. Phylogenetic analysis of all known human FH binding CspZ variants further grouped the proteins into three unique lineages correlating with loop sequences. This suggests multiple FH-binding mechanisms evolved through LD spirochete-host interactions. Overall, this multidisciplinary work elucidates how the allelically-specific immune evasion role of metals is impacted by microbial protein polymorphisms.
Collapse
Affiliation(s)
- Kalvis Brangulis
- Latvian Biomedical Research and Study Centre, Riga, Latvia; Department of Human Physiology and Biochemistry, Riga Stradins University, Riga, Latvia.
| | - Valerie Sürth
- Goethe University Frankfurt, University Hospital of Frankfurt, Institute of Medical Microbiology and Infection Control, Frankfurt, Germany
| | - Ashley L Marcinkiewicz
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA; Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | - Inara Akopjana
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Andris Kazaks
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Janis Bogans
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Alisa Huber
- Goethe University Frankfurt, University Hospital of Frankfurt, Institute of Medical Microbiology and Infection Control, Frankfurt, Germany
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA; Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA; Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA.
| | - Peter Kraiczy
- Goethe University Frankfurt, University Hospital of Frankfurt, Institute of Medical Microbiology and Infection Control, Frankfurt, Germany.
| |
Collapse
|
2
|
Brangulis K, Malfetano J, Marcinkiewicz AL, Wang A, Chen YL, Lee J, Liu Z, Yang X, Strych U, Bottazzi ME, Pal U, Hsieh CL, Chen WH, Lin YP. Mechanistic insights into structure-based design of a Lyme disease subunit vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619738. [PMID: 39554036 PMCID: PMC11565809 DOI: 10.1101/2024.10.23.619738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The quality of protective immunity plays a critical role in modulating vaccine efficacy, with native antigens often not able to trigger sufficiently strong immune responses for pathogen killing. This warrants creation of structure-based vaccine design, leveraging high-resolution antigen structures for mutagenesis to improve protein stability and efficient immunization strategies. Here, we investigated the mechanisms underlying structure-based vaccine design using CspZ-YA, a vaccine antigen from Borrelia burgdorferi, the bacteria causing Lyme disease (LD), the most common vector-borne disease in the Northern Hemisphere. Compared to wild-type CspZ-YA, we found CspZ-YAI183Y and CspZ-YAC187S required lower immunization frequency to protect mice from LD-associated manifestations and bacterial colonization. We observed indistinguishable human and mouse antigenicity between wild-type and mutant CspZ-YA proteins after native infection or active immunization. This supports our newly generated, high-resolution structures of CspZ-YAI183Y and CspZ-YAC187S, showing no altered surface epitopes after mutagenesis. However, CspZ-YAI183Y and CspZ-YAC187S favored the interactions between helices H and I, consistent with their elevated thermostability. Such findings are further strengthened by increasing ability of protective CspZ-YA monoclonal antibodies in binding to CspZ-YA at a physiological temperature (37°C). Overall, this study demonstrated enhanced intramolecular interactions improved long-term stability of antigens while maintaining protective epitopes, providing a mechanism for structure-based vaccine design. These findings can ultimately be extended to other vaccine antigens against newly emerging pathogens for the improvement of protective immunity.
Collapse
Affiliation(s)
| | - Jill Malfetano
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
| | - Ashley L. Marcinkiewicz
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, Grafton, MA, USA
| | - Alan Wang
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
| | - Yi-Lin Chen
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Jungsoon Lee
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Zhuyun Liu
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Xiuli Yang
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, United States
| | - Ulrich Strych
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Maria-Elena Bottazzi
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
- Department of Biology, Baylor University, Waco, TX, United States
| | - Utpal Pal
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, United States
| | - Ching-Lin Hsieh
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Wen-Hsiang Chen
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, Grafton, MA, USA
- Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA
| |
Collapse
|
3
|
Cramer NA, Socarras KM, Earl J, Ehrlich GD, Marconi RT. Borreliella burgdorferi factor H-binding proteins are not required for serum resistance and infection in mammals. Infect Immun 2024; 92:e0052923. [PMID: 38289123 PMCID: PMC10929407 DOI: 10.1128/iai.00529-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 03/13/2024] Open
Abstract
The causative agent of Lyme disease (LD), Borreliella burgdorferi, binds factor H (FH) and other complement regulatory proteins to its surface. B. burgdorferi B31 (type strain) encodes five FH-binding proteins (FHBPs): CspZ, CspA, and the OspE paralogs OspEBBN38, OspEBBL39, and OspEBBP38. This study assessed potential correlations between the production of individual FHBPs, FH-binding ability, and serum resistance using a panel of infectious B. burgdorferi clonal populations recovered from dogs. FHBP production was assessed in cultivated spirochetes and by antibody responses in naturally infected humans, dogs, and eastern coyotes (wild canids). FH binding specificity and sensitivity to dog and human serum were also assessed and compared. No correlation was observed between the production of individual FHBPs and FH binding with serum resistance, and CspA was determined to not be produced in animals. Notably, one or more clones isolated from dogs lacked CspZ or the OspE proteins (a finding confirmed by genome sequence determination) and did not bind FH derived from canines. The data presented do not support a correlation between FH binding and the production of individual FHBPs with serum resistance and infectivity. In addition, the limited number and polymorphic nature of cp32s in B. burgdorferi clone DRI85A that were identified through genome sequencing suggest no strict requirement for a defined set of these replicons for infectivity. This study reveals that the immune evasion mechanisms employed by B. burgdorferi are diverse, complex, and yet to be fully defined.
Collapse
Affiliation(s)
- Nicholas A. Cramer
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Kalya M. Socarras
- Department of Microbiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Joshua Earl
- Department of Microbiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Garth D. Ehrlich
- Department of Microbiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Richard T. Marconi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| |
Collapse
|
4
|
Outer surface protein E (OspE) mediates Borrelia burgdorferi sensu stricto strain-specific complement evasion in the eastern fence lizard, Sceloporus undulatus. Ticks Tick Borne Dis 2023; 14:102081. [PMID: 36403322 DOI: 10.1016/j.ttbdis.2022.102081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022]
Abstract
In North America, Lyme disease is primarily caused by the spirochetal bacterium Borrelia burgdorferi sensu stricto (Bb), which is transmitted between multiple vertebrate hosts and ixodid ticks, and is a model commonly used to study host-pathogen interactions. While Bb is consistently observed in its mammalian and avian reservoirs, the bacterium is rarely isolated from North American reptiles. Two closely related lizard species, the eastern fence lizard (Sceloporus undulatus) and the western fence lizard (Sceloporus occidentalis), are examples of reptiles parasitized by Ixodes ticks. Vertebrates are known to generate complement as an innate defense mechanism, which can be activated before Bb disseminate to distal tissues. Complement from western fence lizards has proven lethal against one Bb strain, implying the role of complement in making those lizards unable to serve as hosts to Bb. However, Bb DNA is occasionally identified in distal tissues of field-collected eastern fence lizards, suggesting some Bb strains may overcome complement-mediated clearance in these lizards. These findings raise questions regarding the role of complement and its impact on Bb interactions with North American lizards. In this study, we found Bb seropositivity in a small population of wild-caught eastern fence lizards and observed Bb strain-specific survivability in lizard sera. We also found that a Bb outer surface protein, OspE, from Bb strains viable in sera, promotes lizard serum survivability and binds to a complement inhibitor, factor H, from eastern fence lizards. Our data thus identify bacterial and host determinants of eastern fence lizard complement evasion, providing insights into the role of complement influencing Bb interactions with North American lizards.
Collapse
|
5
|
Huang J, Chen J, Xie Y, Liu Z. Subversion of the immune response of human pathogenic spirochetes. J Clin Lab Anal 2022; 36:e24414. [PMID: 35403248 PMCID: PMC9102653 DOI: 10.1002/jcla.24414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/21/2022] [Accepted: 03/28/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Jielite Huang
- Department of Clinical Laboratory The Second Affiliated Hospital, Hengyang Medical School, University of South China Hengyang China
| | - Jinlin Chen
- Department of Clinical Laboratory The Second Affiliated Hospital, Hengyang Medical School, University of South China Hengyang China
| | - Yafeng Xie
- Department of Clinical Laboratory The Second Affiliated Hospital, Hengyang Medical School, University of South China Hengyang China
- Institution of Pathogenic Biology Medical College Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study University of South China Hengyang China
| | - Zhuoran Liu
- Department of Clinical Laboratory The Second Affiliated Hospital, Hengyang Medical School, University of South China Hengyang China
| |
Collapse
|
6
|
Pereira MJ, Wager B, Garrigues RJ, Gerlach E, Quinn JD, Dowdell AS, Osburne MS, Zückert WR, Kraiczy P, Garcia BL, Leong JM. Lipoproteome screening of the Lyme disease agent identifies inhibitors of antibody-mediated complement killing. Proc Natl Acad Sci U S A 2022; 119:e2117770119. [PMID: 35312359 PMCID: PMC9060444 DOI: 10.1073/pnas.2117770119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/09/2022] [Indexed: 12/17/2022] Open
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 vector–vertebrate 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 host–pathogen interactions that are central to the pathogenesis of spirochetes, such as the Lyme disease agent.
Collapse
Affiliation(s)
- Michael J. Pereira
- Department of Molecular Biology and Microbiology, Tufts School of Medicine, Tufts University, Boston, MA 02155
| | - Beau Wager
- Department of Molecular Biology and Microbiology, Tufts School of Medicine, Tufts University, Boston, MA 02155
| | - Ryan J. Garrigues
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858
| | - Eva Gerlach
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Goethe University Frankfurt, D-60596 Frankfurt, Germany
| | - Joshua D. Quinn
- Department of Molecular Biology and Microbiology, Tufts School of Medicine, Tufts University, Boston, MA 02155
| | - Alexander S. Dowdell
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Marcia S. Osburne
- Department of Molecular Biology and Microbiology, Tufts School of Medicine, Tufts University, Boston, MA 02155
| | - Wolfram R. Zückert
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, KS 66103
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Goethe University Frankfurt, D-60596 Frankfurt, Germany
| | - Brandon L. Garcia
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts School of Medicine, Tufts University, Boston, MA 02155
| |
Collapse
|
7
|
Utilizing Two Borrelia bavariensis Isolates Naturally Lacking the PFam54 Gene Array To Elucidate the Roles of PFam54-Encoded Proteins. Appl Environ Microbiol 2022; 88:e0155521. [PMID: 34986011 DOI: 10.1128/aem.01555-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
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.
Collapse
|
8
|
Strnad M, Rego ROM. The need to unravel the twisted nature of the Borrelia burgdorferi sensu lato complex across Europe. MICROBIOLOGY-SGM 2021; 166:428-435. [PMID: 32125267 DOI: 10.1099/mic.0.000899] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Lyme borreliosis is a vector-borne infection caused by bacteria under the Borrelia burgdorferi sensu lato complex, both in Europe and North America. Differential gene expression at different times throughout its infectious cycle allows the spirochete to survive very diverse environments within different mammalian hosts as well as the tick vector. To date, the vast majority of data about spirochetal proteins and their functions are from genetic studies carried out on North American strains of a single species, i.e. B. burgdorferi sensu stricto. The whole-genome sequences recently obtained for several European species/strains make it feasible to adapt and use genetic techniques to study inherent differences between them. This review highlights the crucial need to undertake independent studies of genospecies within Europe, given their varying genetic content and pathogenic potential, and differences in clinical manifestation.
Collapse
Affiliation(s)
- Martin Strnad
- Faculty of Science, University of South Bohemia, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.,Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branisovska 31, 37005, Ceske Budejovice, Czech Republic
| | - Ryan O M Rego
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branisovska 31, 37005, Ceske Budejovice, Czech Republic
| |
Collapse
|
9
|
Coburn J, Garcia B, Hu LT, Jewett MW, Kraiczy P, Norris SJ, Skare J. Lyme Disease Pathogenesis. Curr Issues Mol Biol 2020; 42:473-518. [PMID: 33353871 DOI: 10.21775/cimb.042.473] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
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.
Collapse
Affiliation(s)
- Jenifer Coburn
- Center For Infectious Disease Research, Medical College of Wisconsin, 8701 Watertown Plank Rd., TBRC C3980, Milwaukee, WI 53226, USA
| | - Brandon Garcia
- Department of Microbiology and Immunology, East Carolina University, Brody School of Medicine, Greenville, NC 27858, USA
| | - Linden T Hu
- Department of Molecular Biology and Microbiology, Vice Dean of Research, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA 02111, USA
| | - Mollie W Jewett
- Immunity and Pathogenesis Division Head, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 6900 Lake Nona Blvd. Orlando, FL 32827, USA
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital Frankfurt, Goethe University Frankfurt, Paul-Ehrlich-Str. 40, 60596 Frankfurt, Germany
| | - Steven J Norris
- Department of Pathology and Laboratory Medicine, University of Texas Medical School at Houston, P.O. Box 20708, Houston, TX 77225, USA
| | - Jon Skare
- Professor and Associate Head, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX 77807, USA
| |
Collapse
|
10
|
Skare JT, Garcia BL. Complement Evasion by Lyme Disease Spirochetes. Trends Microbiol 2020; 28:889-899. [PMID: 32482556 DOI: 10.1016/j.tim.2020.05.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/26/2020] [Accepted: 05/05/2020] [Indexed: 01/12/2023]
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.
Collapse
Affiliation(s)
- Jon T Skare
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan/College Station, TX, USA.
| | - Brandon L Garcia
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
11
|
The Factor H-Binding Site of CspZ as a Protective Target against Multistrain, Tick-Transmitted Lyme Disease. Infect Immun 2020; 88:IAI.00956-19. [PMID: 32122944 DOI: 10.1128/iai.00956-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/27/2020] [Indexed: 01/13/2023] Open
Abstract
The spirochete Borrelia burgdorferi sensu 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.
Collapse
|
12
|
Lin YP, Frye AM, Nowak TA, Kraiczy P. New Insights Into CRASP-Mediated Complement Evasion in the Lyme Disease Enzootic Cycle. Front Cell Infect Microbiol 2020; 10:1. [PMID: 32083019 PMCID: PMC7002432 DOI: 10.3389/fcimb.2020.00001] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 01/06/2020] [Indexed: 12/23/2022] Open
Abstract
Lyme disease (LD), which is caused by genospecies of the Borrelia burgdorferi sensu lato complex, is the most common vector-borne disease in the Northern hemisphere. Spirochetes are transmitted by Ixodes ticks and maintained in diverse vertebrate animal hosts. Following tick bite, spirochetes initially establish a localized infection in the skin. However, they may also disseminate hematogenously to several distal sites, including heart, joints, or the CNS. Because they need to survive in diverse microenvironments, from tick vector to mammalian hosts, spirochetes have developed multiple strategies to combat the numerous host defense mechanisms. One of these strategies includes the production of a number of complement-regulator acquiring surface proteins (CRASPs) which encompass CspA, CspZ, and OspE paralogs to blunt the complement pathway. These proteins are capable of preventing complement activation on the spirochete surface by binding to complement regulator Factor H. The genes encoding these CRASPs differ in their expression patterns during the tick-to-host infection cycle, implying that these proteins may exhibit different functions during infection. This review summarizes the recent published reports which investigated the roles that each of these molecules plays in conferring tick-borne transmission and dissemination in vertebrate hosts. These findings offer novel mechanistic insights into LD pathobiology and may facilitate the identification of new targets for preventive strategies against Lyme borreliosis.
Collapse
Affiliation(s)
- Yi-Pin Lin
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, United States
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, NY, United States
| | - Amber M. Frye
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, United States
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, NY, United States
| | - Tristan A. Nowak
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, United States
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, NY, United States
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
13
|
Walter L, Sürth V, Röttgerding F, Zipfel PF, Fritz-Wolf K, Kraiczy P. Elucidating the Immune Evasion Mechanisms of Borrelia mayonii, the Causative Agent of Lyme Disease. Front Immunol 2019; 10:2722. [PMID: 31849943 PMCID: PMC6902028 DOI: 10.3389/fimmu.2019.02722] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 11/06/2019] [Indexed: 01/24/2023] Open
Abstract
Borrelia (B.) mayonii sp. nov. has recently been reported as a novel human pathogenic spirochete causing Lyme disease (LD) in North America. Previous data reveal a higher spirochaetemia in the blood compared to patients infected by LD spirochetes belonging to the B. burgdorferi sensu lato complex, suggesting that this novel genospecies must exploit strategies to overcome innate immunity, in particular complement. To elucidate the molecular mechanisms of immune evasion, we utilized various methodologies to phenotypically characterize B. mayonii and to identify determinants involved in the interaction with complement. Employing serum bactericidal assays, we demonstrated that B. mayonii resists complement-mediated killing. To further elucidate the role of the key regulators of the alternative pathway (AP), factor H (FH), and FH-like protein 1 (FHL-1) in immune evasion of B. mayonii, serum adsorption experiments were conducted. The data revealed that viable spirochetes recruit both regulators from human serum and FH retained its factor I-mediated C3b-inactivating activity when bound to the bacterial cells. In addition, two prominent FH-binding proteins of approximately 30 and 18 kDa were detected in B. mayonii strain MN14-1420. Bioinformatics identified a gene, exhibiting 60% identity at the DNA level to the cspA encoding gene of B. burgdorferi. Following PCR amplification, the gene product was produced as a His-tagged protein. The CspA-orthologous protein of B. mayonii interacted with FH and FHL-1, and both bound regulators promoted inactivation of C3b in the presence of factor I. Additionally, the CspA ortholog counteracted complement activation by inhibiting the alternative and terminal but not the classical and Lectin pathways, respectively. Increasing concentrations of CspA of B. mayonii also strongly affected C9 polymerization, terminating the formation of the membrane attack complex. To assess the role of CspA of B. mayonii in facilitating serum resistance, a gain-of-function strain was generated, harboring a shuttle vector allowing expression of the CspA encoding gene under its native promotor. Spirochetes producing the native protein on the cell surface overcame complement-mediated killing, indicating that CspA facilitates serum resistance of B. mayonii. In conclusion, here we describe the molecular mechanism utilized by B. mayonii to resists complement-mediated killing by capturing human immune regulators.
Collapse
Affiliation(s)
- Lea Walter
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Valerie Sürth
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Florian Röttgerding
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Peter F Zipfel
- Department of Infection Biology, Leibniz-Institute for Natural Products Research and Infection Biology, Jena, Germany.,Friedrich Schiller University, Jena, Germany
| | - Karin Fritz-Wolf
- Max Planck Institute for Medical Research, Heidelberg, Germany.,Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University Giessen, Giessen, Germany
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
14
|
Federizon J, Lin YP, Lovell JF. Antigen Engineering Approaches for Lyme Disease Vaccines. Bioconjug Chem 2019; 30:1259-1272. [PMID: 30987418 DOI: 10.1021/acs.bioconjchem.9b00167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Increasing rates of Lyme disease necessitate preventive measures such as immunization to mitigate the risk of contracting the disease. At present, there is no human Lyme disease vaccine available on the market. Since the withdrawal of the first and only licensed Lyme disease vaccine based on lipidated recombinant OspA, vaccine and antigen research has aimed to overcome its risks and shortcomings. Replacement of the putative cross-reactive T-cell epitope in OspA via mutation or chimerism addresses the potential risk of autoimmunity. Multivalent approaches in Lyme disease vaccines have been pursued to address sequence heterogeneity of Lyme borreliae antigens and to induce a repertoire of functional antibodies necessary for efficient heterologous protection. This Review summarizes recent antigen engineering strategies that have paved the way for the development of next generation vaccines against Lyme disease, some of which have reached clinical testing. Bioconjugation methods that incorporate antigens to self-assembling nanoparticles for immune response potentiation are also discussed.
Collapse
Affiliation(s)
- Jasmin Federizon
- Department of Biomedical Engineering , University at Buffalo, State University of New York , Buffalo , New York 14260 , United States
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health , Albany , New York 12208 , United States.,Department of Biomedical Sciences , State University of New York at Albany , Albany , New York 12222 , United States
| | - Jonathan F Lovell
- Department of Biomedical Engineering , University at Buffalo, State University of New York , Buffalo , New York 14260 , United States
| |
Collapse
|
15
|
Feng H, Fang F, Yuan L, Xiao M, Yang XY, Huang Y. Downregulated expression of CFHL1 is associated with unfavorable prognosis in postoperative patients with hepatocellular carcinoma. Exp Ther Med 2019; 17:4073-4079. [PMID: 31007744 PMCID: PMC6469037 DOI: 10.3892/etm.2019.7455] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 02/15/2019] [Indexed: 12/13/2022] Open
Abstract
Complement factor H-related protein 1 (CFHL1) was recently reported to be a potential biomarker in several types of cancer. CFHL1, however, has not been found to be of prognostic value in hepatocellular carcinoma (HCC) to date. In the present study, the expression levels of CFHL1 were evaluated in 8 pairs fresh frozen tissue samples using western blotting. Furthermore, the expression level of CFHL1 was evaluated in 76 pairs of formalin-fixed, paraffin-embedded (FFPE) HCC and peritumoral tissues (expression pattern cohort), and 278 FFPE HCC tissues (prognostic cohort) using tissue microarray-based immunohistochemistry. The Kaplan-Meier method, Cox regression proportional hazard model and receiver operating characteristic curve analysis were used to evaluate prognostic factors. The expression level of CFHL1 was reduced in HCC tissues in 67.1% (51/76) of the cases compared with the corresponding peritumoral tissues. Survival analyses indicated that patients with HCC with low CFHL1 expression had a worse time-to-recurrence (TTR) and overall survival (OS) compared with those with high CFHL1 expression in the prognostic cohort (P=0.002 for OS and P=0.017 for TTR). Both univariate and multivariate analyses indicated that CHFL1 was an independent prognostic factor for TTR and OS (P=0.017 and P=0.002, respectively). In addition, The Cancer Genome Atlas (TCGA) and Human Protein Atlas were used for further validation. Furthermore, a prognostic model included tumor size, tumor number, liver cirrhosis and CFHL1 expression was evaluated. The results of the present study demonstrated that CFHL1 was downregulated in HCC and its level was associated with patient prognosis; therefore, CFHL1 is a potential prognostic marker for HCC.
Collapse
Affiliation(s)
- Hao Feng
- Department of Gynecology and Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, P.R. China
| | - Fang Fang
- Department of Radiotherapy, Navy Medical University, Shanghai 200433, P.R. China
| | - Lei Yuan
- Department of Biliary Tract Surgery, Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Mingjia Xiao
- Department of Biliary Tract Surgery, Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Xiao-Yu Yang
- Department of Biliary Tract Surgery, Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Yao Huang
- Department of Biliary Tract Surgery, Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| |
Collapse
|
16
|
Tufts DM, Hart TM, Chen GF, Kolokotronis SO, Diuk-Wasser MA, Lin YP. Outer surface protein polymorphisms linked to host-spirochete association in Lyme borreliae. Mol Microbiol 2019; 111:868-882. [PMID: 30666741 DOI: 10.1111/mmi.14209] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2019] [Indexed: 12/15/2022]
Abstract
Lyme borreliosis is caused by multiple species of the spirochete bacteria Borrelia burgdorferi sensu lato. The spirochetes are transmitted by ticks to vertebrate hosts, including small- and medium-sized mammals, birds, reptiles, and humans. Strain-to-strain variation in host-specific infectivity has been documented, but the molecular basis that drives this differentiation is still unclear. Spirochetes possess the ability to evade host immune responses and colonize host tissues to establish infection in vertebrate hosts. In turn, hosts have developed distinct levels of immune responses when invaded by different species/strains of Lyme borreliae. Similarly, the ability of Lyme borreliae to colonize host tissues varies among different spirochete species/strains. One potential mechanism that drives this strain-to-strain variation of immune evasion and colonization is the polymorphic outer surface proteins produced by Lyme borreliae. In this review, we summarize research on strain-to-strain variation in host competence and discuss the evidence that supports the role of spirochete-produced protein polymorphisms in driving this variation in host specialization. Such information will provide greater insights into the adaptive mechanisms driving host and Lyme borreliae association, which will lead to the development of interventions to block pathogen spread and eventually reduce Lyme borreliosis health burden.
Collapse
Affiliation(s)
- Danielle M Tufts
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, NY, USA
| | - Thomas M Hart
- Department of Biological Sciences, University at Albany, Albany, NY, USA.,Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Grace F Chen
- Department of Biology, Misericordia University, Dallas, PA, USA
| | - Sergios-Orestis Kolokotronis
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Maria A Diuk-Wasser
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, NY, USA
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA.,Department of Biomedical Sciences, University at Albany, Albany, NY, USA
| |
Collapse
|
17
|
Mühleip JJ, Lin YP, Kraiczy P. Further Insights Into the Interaction of Human and Animal Complement Regulator Factor H With Viable Lyme Disease Spirochetes. Front Vet Sci 2019; 5:346. [PMID: 30766876 PMCID: PMC6365980 DOI: 10.3389/fvets.2018.00346] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/28/2018] [Indexed: 12/19/2022] Open
Abstract
Spirochetes belonging to the Borrelia (B.) burgdorferi sensu lato (s.l.) complex differ in their ability to establish infection and to survive in diverse vertebrate hosts. Association with and adaption to various hosts most likely correlates with the spirochetes' ability to acquire complement regulator factor H (FH) to overcome the host's innate immune response. Here we assessed binding of serum FH from human and various animals including bovine, cat, chicken, dog, horse, mouse, rabbit, and rat to viable B. burgdorferi sensu stricto (s.s.), B. afzelii, B. garinii, B. spielmanii, B. valaisiana, and B. lusitaniae. Spirochetes ectopically producing CspA orthologs of B. burgdorferi s.s., B. afzelii, and B. spielmanii, CspZ, ErpC, and ErpP, respectively, were also investigated. Our comparative analysis using viable bacterial cells revealed a striking heterogeneity among Lyme disease spirochetes regarding their FH-binding patterns that almost mirrors the serum susceptibility of the respective borrelial genospecies. Moreover, native CspA from B. burgdorferi s.s., B. afzelii, and B. spielmanii as well as CspZ were identified as key ligands of FH from human, horse, and rat origin while ErpP appears to bind dog and mouse FH and to a lesser extent human FH. By contrast, ErpC did not bind FH from human as well as from animal origin. These findings indicate a strong restriction of distinct borrelial proteins toward binding of polymorphic FH of various vertebrate hosts.
Collapse
Affiliation(s)
- Jovana Jasmin Mühleip
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| | - Yi-Pin Lin
- Department of Biomedical Science, State University of New York at Albany, Albany, NY, United States.,Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, NY, United States
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| |
Collapse
|
18
|
Marcinkiewicz AL, Dupuis AP, Zamba-Campero M, Nowak N, Kraiczy P, Ram S, Kramer LD, Lin YP. Blood treatment of Lyme borreliae demonstrates the mechanism of CspZ-mediated complement evasion to promote systemic infection in vertebrate hosts. Cell Microbiol 2019; 21:e12998. [PMID: 30571845 DOI: 10.1111/cmi.12998] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 11/30/2022]
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.
Collapse
Affiliation(s)
- Ashley L Marcinkiewicz
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Alan P Dupuis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Maxime Zamba-Campero
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Nancy Nowak
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Laura D Kramer
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA.,Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, USA
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA.,Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, USA
| |
Collapse
|
19
|
Bigelmayr S, Koenigs A, Kraiczy P. Inter- and intraspecies-specific adhesion of Lyme borreliae to human keratinocytes. Ticks Tick Borne Dis 2019; 10:207-212. [DOI: 10.1016/j.ttbdis.2018.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/12/2018] [Accepted: 10/24/2018] [Indexed: 12/11/2022]
|
20
|
Vechtova P, Sterbova J, Sterba J, Vancova M, Rego ROM, Selinger M, Strnad M, Golovchenko M, Rudenko N, Grubhoffer L. A bite so sweet: the glycobiology interface of tick-host-pathogen interactions. Parasit Vectors 2018; 11:594. [PMID: 30428923 PMCID: PMC6236881 DOI: 10.1186/s13071-018-3062-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 08/14/2018] [Indexed: 11/10/2022] Open
Abstract
Vector-borne diseases constitute 17% of all infectious diseases in the world; among the blood-feeding arthropods, ticks transmit the highest number of pathogens. Understanding the interactions between the tick vector, the mammalian host and the pathogens circulating between them is the basis for the successful development of vaccines against ticks or the tick-transmitted pathogens as well as for the development of specific treatments against tick-borne infections. A lot of effort has been put into transcriptomic and proteomic analyses; however, the protein-carbohydrate interactions and the overall glycobiology of ticks and tick-borne pathogens has not been given the importance or priority deserved. Novel (bio)analytical techniques and their availability have immensely increased the possibilities in glycobiology research and thus novel information in the glycobiology of ticks and tick-borne pathogens is being generated at a faster pace each year. This review brings a comprehensive summary of the knowledge on both the glycosylated proteins and the glycan-binding proteins of the ticks as well as the tick-transmitted pathogens, with emphasis on the interactions allowing the infection of both the ticks and the hosts by various bacteria and tick-borne encephalitis virus.
Collapse
Affiliation(s)
- Pavlina Vechtova
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic. .,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic.
| | - Jarmila Sterbova
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Jan Sterba
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Marie Vancova
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Ryan O M Rego
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Martin Selinger
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Martin Strnad
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Maryna Golovchenko
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic
| | - Nataliia Rudenko
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic
| | - Libor Grubhoffer
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| |
Collapse
|
21
|
Marcinkiewicz AL, Lieknina I, Kotelovica S, Yang X, Kraiczy P, Pal U, Lin YP, Tars K. Eliminating Factor H-Binding Activity of Borrelia burgdorferi CspZ Combined with Virus-Like Particle Conjugation Enhances Its Efficacy as a Lyme Disease Vaccine. Front Immunol 2018; 9:181. [PMID: 29472926 PMCID: PMC5809437 DOI: 10.3389/fimmu.2018.00181] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 01/22/2018] [Indexed: 12/18/2022] Open
Abstract
The spirochete Borrelia burgdorferi is the causative agent of Lyme disease, the most common tick-borne disease in the US and Europe. No potent human vaccine is currently available. The innate immune complement system is vital to host defense against pathogens, as complement activation on the surface of spirochetes results in bacterial killing. Complement system is inhibited by the complement regulator factor H (FH). To escape killing, B. burgdorferi produces an outer surface protein CspZ that binds FH to inhibit complement activation on the cell surface. Immunization with CspZ alone does not protect mice from infection, which we speculate is because FH-binding cloaks potentially protective epitopes. We modified CspZ by conjugating to virus-like particles (VLP-CspZ) and eliminating FH binding (modified VLP-CspZ) to increase immunogenicity. We observed greater bactericidal antibody titers in mice vaccinated with modified VLP-CspZ: A serum dilution of 1:395 (modified VLP-CspZ) vs 1:143 (VLP-CspZ) yielded 50% borreliacidal activity. Immunizing mice with modified VLP-CspZ cleared spirochete infection, as did passive transfer of elicited antibodies. This work developed a novel Lyme disease vaccine candidate by conjugating CspZ to VLP and eliminating FH-binding ability. Such a strategy of conjugating an antigen to a VLP and eliminating binding to the target ligand can serve as a general model for developing vaccines against other bacterial infectious agents.
Collapse
Affiliation(s)
- Ashley L. Marcinkiewicz
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, United States
| | - Ilva Lieknina
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | - Xiuli Yang
- Department of Veterinary Medicine, Virginia–Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, College Park, MD, United States
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | - Utpal Pal
- Department of Veterinary Medicine, Virginia–Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, College Park, MD, United States
| | - Yi-Pin Lin
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, United States
- Department of Biomedical Science, State University of New York at Albany, Albany, NY, United States
| | - Kaspars Tars
- Latvian Biomedical Research and Study Centre, Riga, Latvia
- Faculty of Biology, University of Latvia, Riga, Latvia
| |
Collapse
|
22
|
Caine JA, Lin YP, Kessler JR, Sato H, Leong JM, Coburn J. Borrelia burgdorferi outer surface protein C (OspC) binds complement component C4b and confers bloodstream survival. Cell Microbiol 2017; 19:10.1111/cmi.12786. [PMID: 28873507 PMCID: PMC5680108 DOI: 10.1111/cmi.12786] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 01/15/2023]
Abstract
Borrelia burgdorferi (Bb) is the causative agent of Lyme disease in the United States, a disease that can result in carditis, and chronic and debilitating arthritis and/or neurologic symptoms if left untreated. Bb survives in the midgut of the Ixodes scapularis tick, or within tissues of immunocompetent hosts. In the early stages of infection, the bacteria are present in the bloodstream where they must resist clearance by the innate immune system of the host. We have found a novel role for outer surface protein C (OspC) from B. burgdorferi and B. garinii in interactions with the complement component C4b and bloodstream survival in vivo. Our data show that OspC inhibits the classical and lectin complement pathways and competes with complement protein C2 for C4b binding. Resistance to complement is important for maintenance of the lifecycle of Bb, enabling survival of the pathogen within the host as well as in the midgut of a feeding tick when ospC expression is induced.
Collapse
Affiliation(s)
- Jennifer A. Caine
- Department of Microbiology and Immunology, Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, WI
| | - Yi-Pin Lin
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY
| | - Julie R. Kessler
- Department of Microbiology and Immunology, Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, WI
| | - Hiromi Sato
- Department of Microbiology and Immunology, Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, WI
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
| | - Jenifer Coburn
- Department of Microbiology and Immunology, Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, WI
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
23
|
Röttgerding F, Wagemakers A, Koetsveld J, Fingerle V, Kirschfink M, Hovius JW, Zipfel PF, Wallich R, Kraiczy P. Immune evasion of Borrelia miyamotoi: CbiA, a novel outer surface protein exhibiting complement binding and inactivating properties. Sci Rep 2017; 7:303. [PMID: 28331202 PMCID: PMC5428533 DOI: 10.1038/s41598-017-00412-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/27/2017] [Indexed: 12/13/2022] Open
Abstract
Borrelia (B.) miyamotoi, an emerging tick-borne relapsing fever spirochete, resists complement-mediated killing. To decipher the molecular principles of immune evasion, we sought to identify determinants contributing to complement resistance. Employing bioinformatics, we identified a gene encoding for a putative Factor H-binding protein, termed CbiA (complement binding and inhibitory protein A). Functional analyses revealed that CbiA interacted with complement regulator Factor H (FH), C3, C3b, C4b, C5, and C9. Upon binding to CbiA, FH retained its cofactor activity for Factor I-mediated inactivation of C3b. The Factor H-binding site within CbiA was mapped to domain 20 whereby the C-terminus of CbiA was involved in FH binding. Additionally, CbiA directly inhibited the activation of the classical pathway and the assembly of the terminal complement complex. Of importance, CbiA displayed inhibitory activity when ectopically produced in serum-sensitive B. garinii G1, rendering this surrogate strain resistant to human serum. In addition, long-term in vitro cultivation lead to an incremental loss of the cbiA gene accompanied by an increase in serum susceptibility. In conclusion, our data revealed a dual strategy of B. miyamotoi to efficiently evade complement via CbiA, which possesses complement binding and inhibitory activities.
Collapse
Affiliation(s)
- Florian Röttgerding
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| | - Alex Wagemakers
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Joris Koetsveld
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Volker Fingerle
- National Reference Center for Borrelia, Oberschleißheim, Germany
| | | | - Joppe W Hovius
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany.,Friedrich Schiller University, Jena, Germany
| | - Reinhard Wallich
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany.
| |
Collapse
|
24
|
Marcinkiewicz AL, Kraiczy P, Lin YP. There Is a Method to the Madness: Strategies to Study Host Complement Evasion by Lyme Disease and Relapsing Fever Spirochetes. Front Microbiol 2017; 8:328. [PMID: 28303129 PMCID: PMC5332432 DOI: 10.3389/fmicb.2017.00328] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/16/2017] [Indexed: 01/04/2023] Open
Abstract
Lyme disease and relapsing fever are caused by various Borrelia species. Lyme disease borreliae, the most common vector-borne pathogens in both the U.S. and Europe, are transmitted by Ixodes ticks and disseminate from the site of tick bites to tissues leading to erythema migrans skin rash, arthritis, carditis, and neuroborreliosis. Relapsing fever borreliae, carried by ticks and lice, trigger reoccurring fever episodes. Following transmission, spirochetes survive in the blood to induce bacteremia at the early stages of infection, which is thought to promote evasion of the host complement system. The complement system acts as an important innate immune defense mechanism in humans and vertebrates. Upon activation, the cleaved complement components form complexes on the pathogen surface to eventually promote bacteriolysis. The complement system is negatively modulated by a number of functionally diverse regulators to avoid tissue damage. To evade and inhibit the complement system, spirochetes are capable of binding complement components and regulators. Complement inhibition results in bacterial survival in serum (serum resistance) and is thought to promote bloodstream survival, which facilitates spirochete dissemination and disease manifestations. In this review, we discuss current methodologies to elucidate the mechanisms of Borrelia spp. that promote serum resistance and bloodstream survival, as well as novel methods to study factors responsible for bloodstream survival of Lyme disease borreliae that can be applied to relapsing fever borreliae. Understanding the mechanisms these pathogens utilize to evade the complement system will ultimately aid in the development of novel therapeutic strategies and disease prevention to improve human health.
Collapse
Affiliation(s)
- Ashley L Marcinkiewicz
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health Albany, NY, USA
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt am Main Frankfurt am Main, Germany
| | - Yi-Pin Lin
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health Albany, NY, USA
| |
Collapse
|
25
|
Kingry LC, Batra D, Replogle A, Rowe LA, Pritt BS, Petersen JM. Whole Genome Sequence and Comparative Genomics of the Novel Lyme Borreliosis Causing Pathogen, Borrelia mayonii. PLoS One 2016; 11:e0168994. [PMID: 28030649 PMCID: PMC5193363 DOI: 10.1371/journal.pone.0168994] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/09/2016] [Indexed: 12/11/2022] Open
Abstract
Borrelia mayonii, a Borrelia burgdorferi sensu lato (Bbsl) genospecies, was recently identified as a cause of Lyme borreliosis (LB) among patients from the upper midwestern United States. By microscopy and PCR, spirochete/genome loads in infected patients were estimated at 105 to 106 per milliliter of blood. Here, we present the full chromosome and plasmid sequences of two B. mayonii isolates, MN14-1420 and MN14-1539, cultured from blood of two of these patients. Whole genome sequencing and assembly was conducted using PacBio long read sequencing (Pacific Biosciences RSII instrument) followed by hierarchical genome-assembly process (HGAP). The B. mayonii genome is ~1.31 Mbp in size (26.9% average GC content) and is comprised of a linear chromosome, 8 linear and 7 circular plasmids. Consistent with its taxonomic designation as a new Bbsl genospecies, the B. mayonii linear chromosome shares only 93.83% average nucleotide identity with other genospecies. Both B. mayonii genomes contain plasmids similar to B. burgdorferi sensu stricto lp54, lp36, lp28-3, lp28-4, lp25, lp17, lp5, 5 cp32s, cp26, and cp9. The vls locus present on lp28-10 of B. mayonii MN14-1420 is remarkably long, being comprised of 24 silent vls cassettes. Genetic differences between the two B. mayonii genomes are limited and include 15 single nucleotide variations as well as 7 fewer silent vls cassettes and a lack of the lp5 plasmid in MN14-1539. Notably, 68 homologs to proteins present in B. burgdorferi sensu stricto appear to be lacking from the B. mayonii genomes. These include the complement inhibitor, CspZ (BB_H06), the fibronectin binding protein, BB_K32, as well as multiple lipoproteins and proteins of unknown function. This study shows the utility of long read sequencing for full genome assembly of Bbsl genomes, identifies putative genome regions of B. mayonii that may be linked to clinical manifestation or tissue tropism, and provides a valuable resource for pathogenicity, diagnostic and vaccine studies.
Collapse
Affiliation(s)
- Luke C. Kingry
- Division of Vector-Borne Diseases, Bacterial Diseases Branch, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
| | - Dhwani Batra
- Division of Scientific Resources, Biotechnology Core Facility Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Adam Replogle
- Division of Vector-Borne Diseases, Bacterial Diseases Branch, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
| | - Lori A. Rowe
- Division of Scientific Resources, Biotechnology Core Facility Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Bobbi S. Pritt
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jeannine M. Petersen
- Division of Vector-Borne Diseases, Bacterial Diseases Branch, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
26
|
Kraiczy P. Hide and Seek: How Lyme Disease Spirochetes Overcome Complement Attack. Front Immunol 2016; 7:385. [PMID: 27725820 PMCID: PMC5036304 DOI: 10.3389/fimmu.2016.00385] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/13/2016] [Indexed: 11/15/2022] Open
Abstract
Overcoming the first line of the innate immune system is a general hallmark of pathogenic microbes to avoid recognition and to enter the human host. In particular, spirochetes belonging to the Borrelia burgdorferi sensu lato complex have developed various means to counter the immune response and to successfully survive in diverse host environments for a prolonged period of time. In regard to complement resistance, Borrelia utilize a plethora of immune evasion strategies involves capturing of host-derived complement regulators, terminating complement activation as well as shedding of cell-destroying complement complexes to manipulate and to expeditiously inhibit human complement. Owing to their mode of action, the interacting surface-exposed proteins identified among B. burgdorferi sensu stricto (s.s.), Borrelia afzelii, Borrelia spielmanii, and Borrelia bavariensis can be classified into at least two major categories, namely, molecules that directly interfere with distinct complement components including BBK32, CspA, BGA66, BGA71, and a CD59-like protein or molecules, which indirectly counteract complement activation by binding various complement regulators such as Factor H, Factor H-like protein 1 (FHL-1), Factor H-related proteins FHR-1, FHR-2, or C4Bp. The latter group of genetically and structurally unrelated proteins has been collectively referred to as “complement regulator-acquiring surface proteins” and consists of CspA, CspZ, ErpA, ErpC, ErpP, and the as yet unidentified protein p43. This review focuses on the current knowledge of immune evasion mechanisms exhibited by Lyme disease spirochetes and highlights the role of complement-interfering, infection-associated molecules playing an important part in these processes. Deciphering the immune evasion strategies may provide novel avenues for improved diagnostic approaches and therapeutic interventions.
Collapse
Affiliation(s)
- Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt , Frankfurt am Main , Germany
| |
Collapse
|
27
|
Travelling between Two Worlds: Complement as a Gatekeeper for an Expanded Host Range of Lyme Disease Spirochetes. Vet Sci 2016; 3:vetsci3020012. [PMID: 29056721 PMCID: PMC5644625 DOI: 10.3390/vetsci3020012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/03/2016] [Accepted: 06/06/2016] [Indexed: 01/21/2023] Open
Abstract
Evading innate immunity is a prerequisite for pathogenic microorganisms in order to survive in their respective hosts. Concerning Lyme disease spirochetes belonging to the Borrelia (B.) burgdorferi sensu lato group, a broad range of diverse vertebrates serve as reservoir or even as incidental hosts, including humans. The capability to infect multiple hosts implies that spirochetes have developed sophisticated means to counter the destructive effects of complement of humans and various animals. While the means by which spirochetes overcome the hosts immune defense are far from being completely understood, there is a growing body of evidence suggesting that binding of the key regulator of the alternative pathway, Factor H, plays a pivotal role for immune evasion and that Factor H is an important determinant of host specificity. This review covers (i) the contribution of complement in host-specificity and transmissibility of Lyme disease spirochetes; (ii) the involvement of borrelial-derived determinants to host specificity; (iii) the interplay of human and animal Factor H with complement-acquiring surface proteins of diverse borrelial species; and (iv) the potential role of additional animal complement proteins in the immune evasion of spirochetes.
Collapse
|
28
|
Hammerschmidt C, Klevenhaus Y, Koenigs A, Hallström T, Fingerle V, Skerka C, Pos KM, Zipfel PF, Wallich R, Kraiczy P. BGA66 and BGA71 facilitate complement resistance of Borrelia bavariensis by inhibiting assembly of the membrane attack complex. Mol Microbiol 2015; 99:407-24. [PMID: 26434356 DOI: 10.1111/mmi.13239] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2015] [Indexed: 01/09/2023]
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.
Collapse
Affiliation(s)
- Claudia Hammerschmidt
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| | - Yvonne Klevenhaus
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| | - Arno Koenigs
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| | - Teresia Hallström
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Volker Fingerle
- National Reference Center for Borrelia, Oberschleißheim, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Klaas Martinus Pos
- Institute of Biochemistry, Goethe University of Frankfurt, Frankfurt, Germany
| | - Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany.,Friedrich Schiller University, Jena, Germany
| | - Reinhard Wallich
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| |
Collapse
|
29
|
Vieira ML, Nascimento ALTO. Interaction of spirochetes with the host fibrinolytic system and potential roles in pathogenesis. Crit Rev Microbiol 2015; 42:573-87. [PMID: 25914944 DOI: 10.3109/1040841x.2014.972336] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
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.
Collapse
|
30
|
The relapsing fever spirochete Borrelia miyamotoi resists complement-mediated killing by human serum. Ticks Tick Borne Dis 2014; 5:898-901. [DOI: 10.1016/j.ttbdis.2014.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/10/2014] [Indexed: 11/22/2022]
|
31
|
Brangulis K, Petrovskis I, Kazaks A, Bogans J, Otikovs M, Jaudzems K, Ranka R, Tars K. Structural characterization of CspZ, a complement regulator factor H and FHL-1 binding protein from Borrelia burgdorferi. FEBS J 2014; 281:2613-22. [PMID: 24702793 DOI: 10.1111/febs.12808] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 03/29/2014] [Accepted: 04/04/2014] [Indexed: 01/10/2023]
Abstract
Borrelia burgdorferi is the causative agent of Lyme disease and is found in two different types of hosts in nature - Ixodes ticks and various mammalian organisms. To initiate disease and survive in mammalian host organisms, B. burgdorferi must be able to transfer to a new host, proliferate, attach to different tissue and resist the immune response. To resist the host's immune response, B. burgdorferi produces at least five different outer surface proteins that can bind complement regulator factor H (CFH) and/or factor H-like protein 1 (CFHL-1). The crystal structures of two uniquely folded complement binding proteins, which belong to two distinct gene families and are not found in other bacteria, have been previously described. The crystal structure of the CFH and CFHL-1 binding protein CspZ (also known as BbCRASP-2 or BBH06) from B. burgdorferi, which belongs to a third gene family, is reported in this study. The structure reveals that the overall fold is different from the known structures of the other complement binding proteins in B. burgdorferi or other bacteria; this structure does not resemble the fold of any known protein deposited in the Protein Data Bank. The N-terminal part of the CspZ protein forms a four-helix bundle and has features similar to the FAT domain (focal adhesion targeting domain) and a related domain found in the vinculin/α-catenin family. By combining our findings from the crystal structure of CspZ with previous mutagenesis studies, we have identified a likely binding surface on CspZ for CFH and CFHL-1.
Collapse
Affiliation(s)
- Kalvis Brangulis
- Latvian Biomedical Research and Study Centre, Riga, Latvia; Riga Stradins University, Latvia
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Versatile roles of CspA orthologs in complement inactivation of serum-resistant Lyme disease spirochetes. Infect Immun 2013; 82:380-92. [PMID: 24191298 DOI: 10.1128/iai.01094-13] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
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.
Collapse
|
33
|
Schwab J, Hammerschmidt C, Richter D, Skerka C, Matuschka FR, Wallich R, Zipfel PF, Kraiczy P. Borrelia valaisiana resist complement-mediated killing independently of the recruitment of immune regulators and inactivation of complement components. PLoS One 2013; 8:e53659. [PMID: 23320099 PMCID: PMC3539980 DOI: 10.1371/journal.pone.0053659] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 12/04/2012] [Indexed: 11/30/2022] Open
Abstract
Spirochetes belonging to the Borrelia (B.) burgdorferi sensu lato complex differ in their resistance to complement-mediated killing, particularly in regard to human serum. In the present study, we elucidate the serum and complement susceptibility of B. valaisiana, a genospecies with the potential to cause Lyme disease in Europe as well as in Asia. Among the investigated isolates, growth of ZWU3 Ny3 was not affected while growth of VS116 and Bv9 was strongly inhibited in the presence of 50% human serum. Analyzing complement activation, complement components C3, C4 and C6 were deposited on the surface of isolates VS116 and Bv9, and similarly the membrane attack complex was formed on their surface. In contrast, no surface-deposited components and no aberrations in cell morphology were detected for serum-resistant ZWU3 Ny3. While further investigating the protective role of bound complement regulators in mediating complement resistance, we discovered that none of the B. valaisiana isolates analyzed bound complement regulators Factor H, Factor H-like protein 1, C4b binding protein or C1 esterase inhibitor. In addition, B. valaisiana also lacked intrinsic proteolytic activity to degrade complement components C3, C3b, C4, C4b, and C5. Taken together, these findings suggest that certain B. valaisiana isolates differ in their capability to resist complement-mediating killing by human serum. The molecular mechanism utilized by B. valaisiana to inhibit bacteriolysis appears not to involve binding of the key host complement regulators of the alternative, classical, and lectin pathways as already known for serum-resistant Lyme disease or relapsing fever borreliae.
Collapse
Affiliation(s)
- Jasmin Schwab
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| | - Claudia Hammerschmidt
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| | - Dania Richter
- Abteilung Parasitologie, Institut für Pathologie, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Franz-Rainer Matuschka
- Abteilung Parasitologie, Institut für Pathologie, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Reinhard Wallich
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Peter F. Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
- Friedrich Schiller University, Jena, Germany
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
- * E-mail:
| |
Collapse
|
34
|
Complement evasion by Borrelia burgdorferi: it takes three to tango. Trends Parasitol 2013; 29:119-28. [PMID: 23298533 DOI: 10.1016/j.pt.2012.12.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 11/30/2012] [Accepted: 12/04/2012] [Indexed: 01/03/2023]
Abstract
The complement system is one of the major innate defense mechanisms Borrelia burgdorferi sensu lato has to overcome to establish an infection of mammalian hosts and to cause Lyme borreliosis in humans. Borrelia prevents complement-mediated killing during host colonization through (i) recruitment of host complement regulators by Borrelia, (ii) evasion mechanisms by Borrelia itself, and (iii) exploitation of tick proteins by Borrelia. These interactions with complement can be host species-specific. This review provides an overview of interactions between Borrelia, tick, and host leading to evasion of complement-mediated killing.
Collapse
|
35
|
Kraiczy P, Stevenson B. Complement regulator-acquiring surface proteins of Borrelia burgdorferi: Structure, function and regulation of gene expression. Ticks Tick Borne Dis 2012; 4:26-34. [PMID: 23219363 DOI: 10.1016/j.ttbdis.2012.10.039] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 10/15/2012] [Accepted: 10/16/2012] [Indexed: 12/27/2022]
Abstract
Borrelia burgdorferi, the etiological agent of Lyme disease, exploits an array of strategies to establish infection and to overcome host innate and adaptive immune responses. One key borrelial immune escape mechanism involves the inactivation of host complement attack through acquisition of human immune regulators factor H (CFH), factor H-like protein 1 (FHL1), factor H-related protein 1 (CFHR1), CFHR2, and/or CFHR5. Binding of these host proteins is primarily mediated by bacterial surface-exposed proteins that have been collectively referred to as complement regulator-acquiring surface proteins, or CRASPs. Different strains of B. burgdorferi produce as many as 5 different CRASP molecules that comprise 3 distinct, genetically unrelated groups. Depending on bacterial genetic composition, different combinations of these proteins can be found on the borrelial outer surface. The 3 groups differ in their gene location, gene regulatory mechanisms, expression patterns during the tick-mammal infection cycle, protein sequence and structure as well as binding affinity for complement regulators and other serum proteins. These attributes influence the proteins' abilities to contribute to complement resistance of this emerging human pathogen. In this review, we focus on the current knowledge on structure, function, and gene regulation of these B. burgdorferi infection-associated proteins.
Collapse
Affiliation(s)
- Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, Frankfurt University Hospital, Paul-Ehrlich-Strasse 40, 6 Frankfurt, Germany.
| | | |
Collapse
|
36
|
Contribution of the infection-associated complement regulator-acquiring surface protein 4 (ErpC) to complement resistance of Borrelia burgdorferi. Clin Dev Immunol 2012; 2012:349657. [PMID: 22400034 PMCID: PMC3287035 DOI: 10.1155/2012/349657] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 10/05/2011] [Indexed: 11/17/2022]
Abstract
Borrelia burgdorferi evades complement-mediated killing by interacting with complement regulators through distinct complement regulator-acquiring surface proteins (CRASPs). Here, we extend our analyses to the contribution of CRASP-4 in mediating complement resistance of B. burgdorferi and its interaction with human complement regulators. CRASP-4 (also known as ErpC) was immobilized onto magnetic beads and used to capture proteins from human serum. Following Western blotting, factor H (CFH), CFH-related protein 1 (CFHR1), CFHR2, and CFHR5 were identified as ligands of CRASP-4. To analyze the impact of native CRASP-4 on mediating survival of serum-sensitive cells in human serum, a B. garinii strain was generated that ectopically expresses CRASP-4. CRASP-4-producing bacteria bound CFHR1, CFHR2, and CFHR5 but not CFH. In addition, transformed spirochetes deposited significant amounts of lethal complement components on their surface and were susceptible to human serum, thus indicating that CRASP-4 plays a subordinate role in complement resistance of B. burgdorferi.
Collapse
|
37
|
Siegel C, Hallström T, Skerka C, Eberhardt H, Uzonyi B, Beckhaus T, Karas M, Wallich R, Stevenson B, Zipfel PF, Kraiczy P. Complement factor H-related proteins CFHR2 and CFHR5 represent novel ligands for the infection-associated CRASP proteins of Borrelia burgdorferi. PLoS One 2010; 5:e13519. [PMID: 20975954 PMCID: PMC2958145 DOI: 10.1371/journal.pone.0013519] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Accepted: 09/21/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND One virulence property of Borrelia burgdorferi is its resistance to innate immunity, in particular to complement-mediated killing. Serum-resistant B. burgdorferi express up to five distinct complement regulator-acquiring surface proteins (CRASP) which interact with complement regulator factor H (CFH) and factor H-like protein 1 (FHL1) or factor H-related protein 1 (CFHR1). In the present study we elucidate the role of the infection-associated CRASP-3 and CRASP-5 protein to serve as ligands for additional complement regulatory proteins as well as for complement resistance of B. burgdorferi. METHODOLOGY/PRINCIPAL FINDINGS To elucidate whether CRASP-5 and CRASP-3 interact with various human proteins, both borrelial proteins were immobilized on magnetic beads. Following incubation with human serum, bound proteins were eluted and separated by Glycine-SDS-PAGE. In addition to CFH and CFHR1, complement regulators CFHR2 and CFHR5 were identified as novel ligands for both borrelial proteins by employing MALDI-TOF. To further assess the contributions of CRASP-3 and CRASP-5 to complement resistance, a serum-sensitive B. garinii strain G1 which lacks all CFH-binding proteins was used as a valuable model for functional analyses. Both CRASPs expressed on the B. garinii outer surface bound CFH as well as CFHR1 and CFHR2 in ELISA. In contrast, live B. garinii bound CFHR1, CFHR2, and CFHR5 and only miniscute amounts of CFH as demonstrated by serum adsorption assays and FACS analyses. Further functional analysis revealed that upon NHS incubation, CRASP-3 or CRASP-5 expressing borreliae were killed by complement. CONCLUSIONS/SIGNIFICANCE In the absence of CFH and the presence of CFHR1, CFHR2 and CFHR5, assembly and integration of the membrane attack complex was not efficiently inhibited indicating that CFH in co-operation with CFHR1, CFHR2 and CFHR5 supports complement evasion of B. burgdorferi.
Collapse
Affiliation(s)
- Corinna Siegel
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt/Main, Germany
| | - Teresia Hallström
- Department of Infection Biology, Leibniz-Institute for Natural Products Research and Infection Biology, Jena, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz-Institute for Natural Products Research and Infection Biology, Jena, Germany
| | - Hannes Eberhardt
- Department of Infection Biology, Leibniz-Institute for Natural Products Research and Infection Biology, Jena, Germany
| | - Barbara Uzonyi
- Department of Infection Biology, Leibniz-Institute for Natural Products Research and Infection Biology, Jena, Germany
| | - Tobias Beckhaus
- Institute of Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Frankfurt/Main, Germany
| | - Michael Karas
- Institute of Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Frankfurt/Main, Germany
| | - Reinhard Wallich
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Brian Stevenson
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky, United States of America
| | - Peter F. Zipfel
- Department of Infection Biology, Leibniz-Institute for Natural Products Research and Infection Biology, Jena, Germany
- Friedrich Schiller University, Jena, Germany
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt/Main, Germany
- * E-mail:
| |
Collapse
|
38
|
Inadequate binding of immune regulator factor H is associated with sensitivity of Borrelia lusitaniae to human complement. Infect Immun 2010; 78:4467-76. [PMID: 20823202 DOI: 10.1128/iai.00138-10] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Spirochetes belonging to the Borrelia burgdorferi sensu lato complex differ in resistance to complement-mediated killing by human serum. Here, we characterize complement sensitivity of a panel of B. lusitaniae isolates derived from ticks collected in Germany and Portugal as well as one patient-derived isolate, PoHL. All isolates are highly susceptible to complement-mediated lysis in human serum and activate complement predominantly by the alternative pathway, leading to an increased deposition of complement components C3, C6, and the terminal complement complex. Interestingly, serum-sensitive B. lusitaniae isolates were able to bind immune regulator factor H (CFH), and some strains also bound CFH-related protein 1 (CFHR1) and CFHR2. Moreover, CFH bound to the surface of B. lusitaniae was inefficient in mediating C3b conversion. Furthermore, the identification and characterization of a potential CFH-binding protein, OspE, revealed that this molecule possesses a significantly reduced binding capacity for CFH compared to that of CFH-binding OspE paralogs expressed by various serum-resistant Borrelia species. This finding suggests that a reduced binding capability of CFH is associated with an increased serum sensitivity of B. lusitaniae to human complement.
Collapse
|
39
|
Functional characterization of Borrelia spielmanii outer surface proteins that interact with distinct members of the human factor H protein family and with plasminogen. Infect Immun 2009; 78:39-48. [PMID: 19858303 DOI: 10.1128/iai.00691-09] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Acquisition of complement regulator factor H (CFH) and factor H-like protein 1 (CFHL1) from human serum enables Borrelia spielmanii, one of the etiological agents of Lyme disease, to evade complement-mediated killing by the human host. Up to three distinct complement regulator-acquiring surface proteins (CRASPs) may be expressed by serum-resistant B. spielmanii, each exhibiting an affinity for CFH and/or CFHL1. Here, we describe the functional characterization of the 15-kDa CRASPs of B. spielmanii, members of the polymorphic Erp (OspE/F-related) protein family, that bind two distinct host complement regulators, CFH and factor H-related protein 1 (CFHR1), but not CFHL1. CFH bound to the B. spielmanii CRASPs maintained cofactor activity for factor I-mediated C3b inactivation. Three naturally occurring alleles of this protein bound CFH and CFHR1 while a fourth natural allele could not. Comparative sequence analysis of these protein alleles identified a single amino acid, histidine-79, as playing a significant role in CFH/CFHR1 binding, with substitution by an arginine completely abrogating ligand binding. The mutation of His-79 to Arg did not inhibit binding of plasminogen, another known ligand of this group of borrelial outer-surface proteins.
Collapse
|
40
|
Comparative analysis of the properties and ligand binding characteristics of CspZ, a factor H binding protein, derived from Borrelia burgdorferi isolates of human origin. Infect Immun 2009; 77:4396-405. [PMID: 19620346 DOI: 10.1128/iai.00393-09] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Borrelia burgdorferi CspZ (BBH06/BbCRASP-2) binds the complement regulatory protein factor H (FH) and additional unidentified serum proteins. The goals of this study were to assess the ligand binding capability of CspZ orthologs derived from an extensive panel of human Lyme disease isolates and to further define the molecular basis of the interaction between FH and CspZ. While most B. burgdorferi CspZ orthologs analyzed bound FH, specific, naturally occurring polymorphisms, most of which clustered in a specific loop domain of CspZ, prevented FH binding in some orthologs. Sequence analyses also revealed the existence of CspZ phyletic groups that correlate with FH binding and with the relationships inferred from ribosomal spacer types (RSTs). CspZ type 1 (RST1) and type 3 (RST3) strains bind FH, while CspZ type 2 (RST2) strains do not. Antibody responses to CspZ were also assessed. Anti-CspZ antibodies were detected in mice by week 2 of infection, indicating that there was expression during early-stage infection. Analyses of sera collected from infected mice suggested that CspZ production continued over the course of long-term infection as the antibody titer increased over time. While antibody to CspZ was detected in several human Lyme disease serum samples, the response was not universal, and the titers were generally low. Vaccination studies with mice demonstrated that while CspZ is immunogenic, it does not elicit an antibody that is protective or that inhibits dissemination. The data presented here provide significant new insight into the interaction between CspZ and FH and suggest that there is a correlation between CspZ production and dissemination. However, in spite of its possible contributory role in pathogenesis, the immunological analyses indicated that CspZ is likely to have limited potential as a diagnostic marker and vaccine candidate for Lyme disease.
Collapse
|
41
|
CspA-mediated binding of human factor H inhibits complement deposition and confers serum resistance in Borrelia burgdorferi. Infect Immun 2009; 77:2773-82. [PMID: 19451251 DOI: 10.1128/iai.00318-09] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Borrelia burgdorferi has developed efficient mechanisms for evading the innate immune response during mammalian infection and has been shown to be resistant to the complement-mediated bactericidal activity of human serum. It is well recognized that B. burgdorferi expresses multiple lipoproteins on its surface that bind the human complement inhibitors factor H and factor H-like protein 1 (FH/FHL-1). The binding of FH/FHL-1 on the surface of B. burgdorferi is thought to enhance its ability to evade serum-mediated killing during the acute phase of infection. One of the key B. burgdorferi FH/FHL-1 binding proteins identified thus far was designated CspA. While it is known that CspA binds FH/FHL-1, it is unclear how the interaction between CspA and FH/FHL-1 specifically enhances serum resistance. To better understand how CspA mediates serum resistance in B. burgdorferi, we inactivated cspA in a virulent strain of B. burgdorferi. An affinity ligand blot immunoassay and indirect immunofluorescence revealed that the CspA mutant does not efficiently bind human FH to its surface. Consistent with the lack of FH binding, the CspA mutant was also highly sensitive to killing by human serum. Additionally, the deposition of complement components C3, C6, and C5b-9 was enhanced on the surface of the CspA mutant compared to that of the wild-type strain. The combined data lead us to conclude that the CspA-mediated binding of human FH confers serum resistance by directly inhibiting complement deposition on the surface of B. burgdorferi.
Collapse
|
42
|
Grosskinsky S, Schott M, Brenner C, Cutler SJ, Kraiczy P, Zipfel PF, Simon MM, Wallich R. Borrelia recurrentis employs a novel multifunctional surface protein with anti-complement, anti-opsonic and invasive potential to escape innate immunity. PLoS One 2009; 4:e4858. [PMID: 19308255 PMCID: PMC2654920 DOI: 10.1371/journal.pone.0004858] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Accepted: 02/13/2009] [Indexed: 12/28/2022] Open
Abstract
Borrelia recurrentis, the etiologic agent of louse-borne relapsing fever in humans, has evolved strategies, including antigenic variation, to evade immune defence, thereby causing severe diseases with high mortality rates. Here we identify for the first time a multifunctional surface lipoprotein of B. recurrentis, termed HcpA, and demonstrate that it binds human complement regulators, Factor H, CFHR-1, and simultaneously, the host protease plasminogen. Cell surface bound factor H was found to retain its activity and to confer resistance to complement attack. Moreover, ectopic expression of HcpA in a B. burgdorferi B313 strain, deficient in Factor H binding proteins, protected the transformed spirochetes from complement-mediated killing. Furthermore, HcpA-bound plasminogen/plasmin endows B. recurrentis with the potential to resist opsonization and to degrade extracellular matrix components. Together, the present study underscores the high virulence potential of B. recurrentis. The elucidation of the molecular basis underlying the versatile strategies of B. recurrentis to escape innate immunity and to persist in human tissues, including the brain, may help to understand the pathological processes underlying louse-borne relapsing fever.
Collapse
Affiliation(s)
- Sonja Grosskinsky
- Infectious Immunology, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - Melanie Schott
- Infectious Immunology, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - Christiane Brenner
- Infectious Immunology, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - Sally J. Cutler
- School of Health and Bioscience, University of East London, Stratford, London, United Kingdom
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt/Main, Germany
| | - Peter F. Zipfel
- Department of Infection Biology, Leibniz-Institute for Natural Products Research, Jena, Germany
| | - Markus M. Simon
- Metschnikoff Laboratory, Max-Planck-Institute for Immunobiology, Freiburg, Germany
| | - Reinhard Wallich
- Infectious Immunology, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
43
|
Analysis of a unique interaction between the complement regulatory protein factor H and the periodontal pathogen Treponema denticola. Infect Immun 2009; 77:1417-25. [PMID: 19204088 DOI: 10.1128/iai.01544-08] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Treponema denticola, a spirochete associated with periodontitis, is abundant at the leading edge of subgingival plaque, where it interacts with gingival epithelia. T. denticola produces a number of virulence factors, including dentilisin, a protease which is cytopathic to host cells, and FhbB, a unique T. denticola lipoprotein that binds complement regulatory proteins. Earlier analyses suggested that FhbB specifically bound to factor H (FH)-like protein 1 (FHL-1). However, by using dentilisin-deficient mutants of T. denticola, we found that T. denticola preferentially binds FH and not FHL-1, and that FH is then cleaved by dentilisin to yield an FH subfragment of approximately 50 kDa. FH bound to dentilisin-deficient mutants but was not cleaved and retained its ability to serve as a cofactor for factor I in the cleavage of C3b. To assess the molecular basis of the interaction of FhbB with FH, mutational analyses were conducted. Replacement of specific residues in widely separated domains of FhbB and disruption of a central alpha helix with coiled-coil formation probability attenuated or eliminated FH binding. The data presented here are the first to demonstrate the retention at the cell surface of a proteolytic cleavage product of FH. The precise role of this FH fragment in the host-pathogen interaction remains to be determined.
Collapse
|