1
|
Hillman C, Theriault H, Dmitriev A, Hansra S, Rosa PA, Wachter J. Borrelia burgdorferi lacking all cp32 prophage plasmids retains full infectivity in mice. EMBO Rep 2025; 26:1997-2012. [PMID: 40108404 PMCID: PMC12018966 DOI: 10.1038/s44319-025-00378-9] [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: 04/04/2023] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 03/22/2025] Open
Abstract
The causative agent of Lyme disease, Borrelia burgdorferi, contains a unique, segmented genome comprising multiple linear and circular plasmids. To date, the genomes of over 63 sequenced Lyme disease Borrelia carry one or more 32 kbp circular plasmids (cp32) or cp32-like elements. The cp32 plasmids are endogenous prophages and encode, among other elements, a family of surface exposed lipoproteins termed OspEF-related proteins. These lipoproteins are synthesized during mammalian infection and are considered important components of the spirochete's adaptive response to the vertebrate host. Here, we detail the construction and infectivity of the first described B. burgdorferi strain lacking all cp32 plasmids. Despite their universal presence, our findings indicate that B. burgdorferi does not require any cp32 plasmids to complete the experimental mouse-tick-mouse infectious cycle and a total lack of cp32s does not impair spirochete infectivity.
Collapse
Affiliation(s)
- Chad Hillman
- Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Hannah Theriault
- Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Department of Biomedical Sciences, School of Public Health, State University of New York, Albany, NY, 12144, USA
- The Arbovirus Laboratory, New York State Department of Health, Wadsworth Center, Slingerlands, NY, 12159, USA
| | - Anton Dmitriev
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Satyender Hansra
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Patricia A Rosa
- Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jenny Wachter
- Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
2
|
Cantero P, Ehret-Sabatier L, Lenormand C, Hansmann Y, Sauleau E, Zilliox L, Westermann B, Jaulhac B, Mutter D, Barthel C, Perdu-Alloy P, Martinot M, Lipsker D, Boulanger N. Detection of Borrelia burgdorferi sensu lato by proteomics: a complementary diagnosis tool on erythema migrans biopsies. Clin Microbiol Infect 2025; 31:78-86. [PMID: 39454756 DOI: 10.1016/j.cmi.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
OBJECTIVES We have developed targeted proteomics in the context of Lyme borreliosis (LM) as a new direct diagnostic tool for detecting Borrelia proteins in the skin of patients with erythema migrans. If satisfactory, this proteomic technique could be used in addition to culture and/or PCR for disseminated infections where Borrelia detection is essential to demonstrate active infection. In these infections, the diagnosis is indirect and relies mainly on serology. METHODS We recruited 46 patients with LM and 11 controls and collected two skin biopsies from each patient. One biopsy was used for Borrelia burgdorferi sensu lato PCR and culture and the other one was for targeted mass-spectrometry-based proteomics. Six markers of infection were selected for proteomics: Outer surface protein C (OspC), flagellin, enolase, lipoprotein gi|365823350, decorin binding protein A, and glyceraldehyde-3-phosphate dehydrogenase. RESULTS Culturing Borrelia from the biopsies increased the sensitivity of the methods. Among the patients included for analysis, 61% (28 patients), 61% (28), and 46% (21) were detected as positive by proteomics, PCR, and culture, respectively. PCR and proteomics were complementary. OspC and flagellin were the most frequently detected protein markers of infection by proteomics, which in some patients, detected up to nine peptides for the flagellin. DISCUSSION It is possible to identify bacterial makers from the skin by proteomics. Our approach can be used to diagnose tick-borne diseases such as LM. TRIAL REGISTRATION clinicaltrials.gov identifier: NCT02414789.
Collapse
Affiliation(s)
- Paola Cantero
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, Centre National de la Recherche Scientifique, Institut Pluridisciplinaire Hubert Curien, Unité Mixte de Recherche, Strasbourg, France; Infrastructure Nationale de Protéomique ProFI, Strasbourg, France
| | - Laurence Ehret-Sabatier
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, Centre National de la Recherche Scientifique, Institut Pluridisciplinaire Hubert Curien, Unité Mixte de Recherche, Strasbourg, France; Infrastructure Nationale de Protéomique ProFI, Strasbourg, France
| | - Cédric Lenormand
- Faculté de Médecine, Université de Strasbourg et Clinique Dermatologique, Hôpital Universitaire de Strasbourg, Strasbourg, France; UR3073-Pathogen-Host- Arthropod Vectors Interactions-Group Borrelia, Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Yves Hansmann
- UR3073-Pathogen-Host- Arthropod Vectors Interactions-Group Borrelia, Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France; Service de Maladies Infectieuses, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Erik Sauleau
- Groupe Méthode en Recherche Clinique, Pôle Santé Publique, Hôpitaux Universitaires de Strasbourg et ICube Unité Mixte de Recherche, Université de Strasbourg/Centre National de la Recherche Scientifique, Strasbourg, France
| | - Laurence Zilliox
- French National Reference Center for Borrelia, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Benoit Westermann
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, Centre National de la Recherche Scientifique, Institut Pluridisciplinaire Hubert Curien, Unité Mixte de Recherche, Strasbourg, France; Infrastructure Nationale de Protéomique ProFI, Strasbourg, France
| | - Benoit Jaulhac
- UR3073-Pathogen-Host- Arthropod Vectors Interactions-Group Borrelia, Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France; French National Reference Center for Borrelia, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Didier Mutter
- Service de Chirurgie Digestive et Endocrinienne, Hôpitaux Universitaires de Strasbourg, Strasbourg France
| | - Cathy Barthel
- UR3073-Pathogen-Host- Arthropod Vectors Interactions-Group Borrelia, Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Pauline Perdu-Alloy
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, Centre National de la Recherche Scientifique, Institut Pluridisciplinaire Hubert Curien, Unité Mixte de Recherche, Strasbourg, France; Infrastructure Nationale de Protéomique ProFI, Strasbourg, France
| | - Martin Martinot
- Service de Maladies Infectieuses, Hôpital de Colmar, Colmar, France
| | - Dan Lipsker
- Faculté de Médecine, Université de Strasbourg et Clinique Dermatologique, Hôpital Universitaire de Strasbourg, Strasbourg, France
| | - Nathalie Boulanger
- UR3073-Pathogen-Host- Arthropod Vectors Interactions-Group Borrelia, Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France; French National Reference Center for Borrelia, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
| |
Collapse
|
3
|
Strnad M, Rudenko N, Rego RO. Pathogenicity and virulence of Borrelia burgdorferi. Virulence 2023; 14:2265015. [PMID: 37814488 PMCID: PMC10566445 DOI: 10.1080/21505594.2023.2265015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 09/25/2023] [Indexed: 10/11/2023] Open
Abstract
Infection with Borrelia burgdorferi often triggers pathophysiologic perturbations that are further augmented by the inflammatory responses of the host, resulting in the severe clinical conditions of Lyme disease. While our apprehension of the spatial and temporal integration of the virulence determinants during the enzootic cycle of B. burgdorferi is constantly being improved, there is still much to be discovered. Many of the novel virulence strategies discussed in this review are undetermined. Lyme disease spirochaetes must surmount numerous molecular and mechanical obstacles in order to establish a disseminated infection in a vertebrate host. These barriers include borrelial relocation from the midgut of the feeding tick to its body cavity and further to the salivary glands, deposition to the skin, haematogenous dissemination, extravasation from blood circulation system, evasion of the host immune responses, localization to protective niches, and establishment of local as well as distal infection in multiple tissues and organs. Here, the various well-defined but also possible novel strategies and virulence mechanisms used by B. burgdorferi to evade obstacles laid out by the tick vector and usually the mammalian host during colonization and infection are reviewed.
Collapse
Affiliation(s)
- Martin Strnad
- Biology Centre CAS, Institute of Parasitology, České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská, Czech Republic
| | - Natalie Rudenko
- Biology Centre CAS, Institute of Parasitology, České Budějovice, Czech Republic
| | - Ryan O.M. Rego
- Biology Centre CAS, Institute of Parasitology, České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská, Czech Republic
| |
Collapse
|
4
|
Tan X, Castellanos M, Chaconas G. Choreography of Lyme Disease Spirochete Adhesins To Promote Vascular Escape. Microbiol Spectr 2023; 11:e0125423. [PMID: 37255427 PMCID: PMC10434219 DOI: 10.1128/spectrum.01254-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/12/2023] [Indexed: 06/01/2023] Open
Abstract
The Lyme disease spirochete Borrelia burgdorferi sensu lato can cause a multitude of clinical manifestations because of its ability to disseminate into any organ system via migration through soft tissue, the lymphatic system, and the circulatory system. The latter is believed to constitute the predominant pathway for dissemination to distal sites from the inoculating tick bite. In spite of its importance, the hematogenous dissemination process remains largely uncharacterized, particularly due to difficulties studying this process in a living host and the lack of an in vitro system that recapitulates animal infection. In the current work, we provide the first information regarding the stage of the vascular transmigration pathway where three important adhesins function during invasion of mouse knee joint peripheral tissue from postcapillary venules. Using intravital imaging coupled with genetic experiments employing sequential double infection, we show a complex temporal choreography of P66, decorin binding proteins (DbpA/B), and outer surface protein C (OspC) at discrete steps along the pathway of vascular escape, underscoring the importance of B. burgdorferi adhesins in hematogenous dissemination in the mouse knee joint and the complexity of vascular transmigration by a disseminating pathogen. IMPORTANCE Lyme disease is caused by the spirochete Borrelia burgdorferi, which is transmitted by a bite from an infected tick. Disease development involves a complex series of host-pathogen interactions as well as dissemination of the infecting organisms to sites distal to the original tick bite. The predominant pathway for this is believed to be hematogenous dissemination. The mechanism by which the spirochetes escape circulation is unknown. Here, using intravital microscopy, where the Lyme spirochete can be observed in a living mouse, we have studied the stage in the vascular escape process where each of three surface adhesins functions to facilitate escape of the spirochete from postcapillary venules to invade mouse knee joint peripheral tissue. A complex pattern of involvement at various locations in the multistage process is described using a unique experimental approach that is applicable to other disseminating pathogens.
Collapse
Affiliation(s)
- Xi Tan
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Mildred Castellanos
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - George Chaconas
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology & Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
5
|
Flynn CD, Sandomierski M, Kim K, Lewis J, Lloyd V, Ignaszak A. Electrochemical Detection of Borrelia burgdorferi Using a Biomimetic Flow Cell System. ACS MEASUREMENT SCIENCE AU 2023; 3:208-216. [PMID: 37360035 PMCID: PMC10288608 DOI: 10.1021/acsmeasuresciau.3c00004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/11/2023] [Accepted: 03/13/2023] [Indexed: 06/28/2023]
Abstract
Lyme disease, caused by infection with pathogenic Borrelia bacteria, has emerged as a pervasive illness throughout North America and many other regions of the world in recent years, owing in part to climate-mediated habitat expansion of the tick vectors. Standard diagnostic testing has remained largely unchanged over the past several decades and is indirect, relying on detection of antibodies against the Borrelia pathogen, rather than detection of the pathogen itself. The development of new rapid, point-of-care tests for Lyme disease that directly detects the pathogen could drastically improve patient health by enabling faster and more frequent testing that could better inform patient treatment. Here, we describe a proof-of-concept electrochemical sensing approach to the detection of the Lyme disease-causing bacteria, which utilizes a biomimetic electrode to interact with the Borrelia bacteria that induce impedance alterations. In addition, the catch-bond mechanism between bacterial BBK32 protein and human fibronectin protein, which exhibits improved bond strength with increased tensile force, is tested within an electrochemical injection flow-cell to achieve Borrelia detection under shear stress.
Collapse
Affiliation(s)
- Connor D. Flynn
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Department
of Chemistry, University of Toronto, Toronto, ON M5S 3G8, Canada
- Department
of Chemistry, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| | - Mariusz Sandomierski
- Department
of Chemistry, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
- Institute
of Chemical Technology and Engineering, Poznan University of Technology, ul. Berdychowo 4, 60-965 Poznań, Poland
| | - Kelly Kim
- Department
of Chemistry, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| | - Julie Lewis
- Department
of Biology, Mount Allison University, Sackville, NB E4L 1E2, Canada
| | - Vett Lloyd
- Department
of Biology, Mount Allison University, Sackville, NB E4L 1E2, Canada
| | - Anna Ignaszak
- Department
of Chemistry, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| |
Collapse
|
6
|
Carnazzo MC, Scholin C, Shweta FNU, Calvin AD. Lyme disease presenting as complete heart block in a young man: Case report and review of pathogenesis. IDCases 2023; 32:e01799. [PMID: 37234726 PMCID: PMC10205533 DOI: 10.1016/j.idcr.2023.e01799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Lyme carditis is a serious complication of Lyme disease, the most common vector-borne infection in both the United States and Northern Europe. It is a rare manifestation of Lyme disease that primarily affects young adults with a marked 3:1 male-to-female predominance. The presentation of Lyme carditis is heterogenous and often non-specific, although the most common clinical manifestation is AV block, which can be acute in onset and can rapidly progress to complete heart block. We discuss the case of a young adult male with complete heart block as a complication of Lyme infection, presenting with two episodes of syncope without prodromal symptoms months after tick bites. There are several pathogen, host and environmental factors that can play an important role in the epidemiology and pathogenesis of this serious condition that is reversible if treated in a timely manner. It is important for clinicians to be familiar with the presentation and treatment of this infection that is now being observed in a wider geographic distribution so as to avoid serious long-term complications and unnecessary permanent pacemaking implantation.
Collapse
Affiliation(s)
- Maria Chiara Carnazzo
- Post-graduate School of Emergency Medicine, Faculty of Medicine and Surgery, University of Modena and Reggio Emilia, Modena, MO 41125, Italy
| | - Celine Scholin
- Medical College of Wisconsin - Central Wisconsin, Wausau, WI 54401, United States
| | - FNU Shweta
- Department of Infectious Disease, Mayo Clinic Health System, Eau Claire, WI 54703, United States
| | - Andrew D. Calvin
- Department of Cardiovascular Medicine, Mayo Clinic Health System, Eau Claire, WI 54703, United States
| |
Collapse
|
7
|
Wu F, Shen K, Xie YI, Wang H, Sun Y, Wang Q. Gene Expression Profiling in Human Brain Microvascular Endothelial Cells in Response to Treponema pallidum Subspecies pallidum. AN ACAD BRAS CIENC 2020; 92:e20191234. [PMID: 33331441 DOI: 10.1590/0001-3765202020191234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/27/2020] [Indexed: 11/22/2022] Open
Abstract
Neurosyphilis (NS) is a neurological disorder caused by Treponema pallidum subspecies pallidum (T. pallidum), but how T. pallidum attach to and cross the blood-brain barrier (BBB) and how BBB response to this bacteria remain unclear. To explore how the human brain microvascular endothelial cells (HBMECs) response to T. pallidum, the Agilent SurePrint G3 Human Gene Expression 8×60K microarray was used. The results revealed that 249 genes were differentially expressed in HBMECs infected with T. pallidum. In particular, genes encoding proteins involved in bacterial adhesion, endothelial cell activation and immune response were regulated by T. pallidum. Furthermore, Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were performed to determine the biological functions of differentially expressed genes. In summary, T. pallidum changes the gene expression profile in HBMECs, and differentially expressed genes are associated with widespread biological and pathophysiological functions. Above all, this is the first paper reporting the effects of T. pallidum on HBMECs. These data develop a new platform for further molecular experiments on the pathogenesis of NS.
Collapse
Affiliation(s)
- Fan Wu
- Department of Dermatology, Sir Run Run Hospital, Nanjing Medical University, No. 109 Longmian Road, Molin District, Nanjing, 211100, China
| | - Kangjie Shen
- The First Clinical Medical College of Nanjing Medical University, No. 818 Tianyuandong Road, Molin District, Nanjing, 211100, China
| | - Y I Xie
- The First Clinical Medical College of Nanjing Medical University, No. 818 Tianyuandong Road, Molin District, Nanjing, 211100, China
| | - Hongye Wang
- The First Clinical Medical College of Nanjing Medical University, No. 818 Tianyuandong Road, Molin District, Nanjing, 211100, China
| | - Yifan Sun
- The First Clinical Medical College of Nanjing Medical University, No. 818 Tianyuandong Road, Molin District, Nanjing, 211100, China
| | - Qianqiu Wang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 12 Jiangwangmiao Road, Xuanwuhu District, Nanjing, 210042, China
| |
Collapse
|
8
|
Abstract
Lyme disease (Lyme borreliosis) is a tick-borne, zoonosis of adults and children caused by genospecies of the Borrelia burgdorferi sensu lato complex. The ailment, widespread throughout the Northern Hemisphere, continues to increase globally due to multiple environmental factors, coupled with increased incursion of humans into habitats that harbor the spirochete. B. burgdorferi sensu lato is transmitted by ticks from the Ixodes ricinus complex. In North America, B. burgdorferi causes nearly all infections; in Europe, B. afzelii and B. garinii are most associated with human disease. The spirochete's unusual fragmented genome encodes a plethora of differentially expressed outer surface lipoproteins that play a seminal role in the bacterium's ability to sustain itself within its enzootic cycle and cause disease when transmitted to its incidental human host. Tissue damage and symptomatology (i.e., clinical manifestations) result from the inflammatory response elicited by the bacterium and its constituents. The deposition of spirochetes into human dermal tissue generates a local inflammatory response that manifests as erythema migrans (EM), the hallmark skin lesion. If treated appropriately and early, the prognosis is excellent. However, in untreated patients, the disease may present with a wide range of clinical manifestations, most commonly involving the central nervous system, joints, or heart. A small percentage (~10%) of patients may go on to develop a poorly defined fibromyalgia-like illness, post-treatment Lyme disease (PTLD) unresponsive to prolonged antimicrobial therapy. Below we integrate current knowledge regarding the ecologic, epidemiologic, microbiologic, and immunologic facets of Lyme disease into a conceptual framework that sheds light on the disorder that healthcare providers encounter.
Collapse
Affiliation(s)
- Justin D. Radolf
- Department of Medicine, UConn Health, Farmington, CT 06030, USA
- Department of Pediatrics, UConn Health, Farmington, CT 06030, USA
- Departments of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
- Departments of Molecular Biology and Biophysics, UConn Health, Farmington, CT 06030, USA
- Department of Immunology, UConn Health, Farmington, CT 06030, USA
| | - Klemen Strle
- Division of Infectious Diseases, Wadsworth Center, NY Department of Health, Albany NY, 12208, USA
| | - Jacob E. Lemieux
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Franc Strle
- Department of Infectious Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
9
|
Lin YP, Yu Y, Marcinkiewicz AL, Lederman P, Hart TM, Zhang F, Linhardt RJ. Non-anticoagulant Heparin as a Pre-exposure Prophylaxis Prevents Lyme Disease Infection. ACS Infect Dis 2020; 6:503-514. [PMID: 31961652 DOI: 10.1021/acsinfecdis.9b00425] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lyme disease (LD) is caused by the spirochete Borrelia burgdorferi sensu lato (Bbsl). After transmission to humans by ticks, Bbsl spreads to multiple organs, leading to arthritis, carditis, and neuroborreliosis. No effective prophylaxis against human LD prior to tick exposure is currently available. Thus, a pre-exposure prophylaxis (PrEP) against LD is needed. The establishment of LD bacteria at diverse sites is dictated partly by the binding of Bbsl to proteoglycans (PGs) and glycosaminoglycans (GAGs) in tissues. The drug heparin is structurally similar to these GAGs and inhibits Bbsl attachment to PGs, GAGs, cells, and tissues, suggesting its potential to prevent LD. However, the anticoagulant activity of heparin often results in hemorrhage, hampering the development of this compound as LD PrEP. We have previously synthesized a non-anticoagulant version of heparin (NACH), which was verified for safety in mice and humans. Here, we showed that NACH blocks Bbsl attachment to PGs, GAGs, and mammalian cells. We also found that treating mice with NACH prior to the exposure of ticks carrying Bbsl followed by continuous administration of this compound prevents tissue colonization by Bbsl. Furthermore, NACH-treated mice develop greater levels of IgG and IgM against Bbsl at early stages of infection, suggesting that the upregulation of antibody immune responses may be one of the mechanisms for NACH-mediated LD prevention. This is one of the first studies examining the ability of a heparin-based compound to prevent LD prior to tick exposure. The information presented might also be extended to prevent other infectious diseases agents.
Collapse
Affiliation(s)
- Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, 120 New Scotland Avenue, Albany, New York 12208, United States
- Department of Biomedical Sciences, State University of New York at Albany, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Yanlei Yu
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York 12180, United States
| | - Ashley L. Marcinkiewicz
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, 120 New Scotland Avenue, Albany, New York 12208, United States
| | - Patricia Lederman
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, 120 New Scotland Avenue, Albany, New York 12208, United States
| | - Thomas M. Hart
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, 120 New Scotland Avenue, Albany, New York 12208, United States
- Department of Biological Science, State University of New York at Albany, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York 12180, United States
| | - Robert J. Linhardt
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York 12180, United States
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York 12180, United States
- Departments of Biology and Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York 12180, United States
| |
Collapse
|
10
|
Bernard Q, Grillon A, Lenormand C, Ehret-Sabatier L, Boulanger N. Skin Interface, a Key Player for Borrelia Multiplication and Persistence in Lyme Borreliosis. Trends Parasitol 2020; 36:304-314. [PMID: 32007396 DOI: 10.1016/j.pt.2019.12.017] [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: 09/01/2019] [Revised: 11/27/2019] [Accepted: 12/25/2019] [Indexed: 01/01/2023]
Abstract
The skin plays a key role in vector-borne diseases because it is the site where the arthropod coinoculates pathogens and its saliva. Lyme borreliosis, particularly well investigated in this context, is a multisystemic infectious disease caused by Borrelia burgdorferi sensu lato and transmitted by the hard tick Ixodes. Numerous in vitro studies were conducted to better understand the role of specific skin cells and tick saliva in host defense, vector feeding, and pathogen transmission. The skin was also evidenced in various animal models as the site of bacterial multiplication and persistence. We present the achievements in this field as well as the gaps that impede comprehensive knowledge of the disease pathophysiology and the development of efficient diagnostic tools and vaccines in humans.
Collapse
Affiliation(s)
- Quentin Bernard
- Fédération de Médecine Translationnelle de Strasbourg, Institut de Bactériologie, Université de Strasbourg, VBP EA7290, F-67000 Strasbourg, France
| | - Antoine Grillon
- Fédération de Médecine Translationnelle de Strasbourg, Institut de Bactériologie, Université de Strasbourg, VBP EA7290, F-67000 Strasbourg, France
| | - Cédric Lenormand
- Fédération de Médecine Translationnelle de Strasbourg, Institut de Bactériologie, Université de Strasbourg, VBP EA7290, F-67000 Strasbourg, France; Clinique Dermatologique, Hôpital Universitaire de Strasbourg, Strasbourg, France
| | - Laurence Ehret-Sabatier
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, F-67000 Strasbourg, France
| | - Nathalie Boulanger
- Fédération de Médecine Translationnelle de Strasbourg, Institut de Bactériologie, Université de Strasbourg, VBP EA7290, F-67000 Strasbourg, France; French National Reference Center on Lyme Borreliosis, CHRU, F-67000 Strasbourg, France.
| |
Collapse
|
11
|
Bamm VV, Ko JT, Mainprize IL, Sanderson VP, Wills MKB. Lyme Disease Frontiers: Reconciling Borrelia Biology and Clinical Conundrums. Pathogens 2019; 8:E299. [PMID: 31888245 PMCID: PMC6963551 DOI: 10.3390/pathogens8040299] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/06/2019] [Accepted: 12/12/2019] [Indexed: 12/18/2022] Open
Abstract
Lyme disease is a complex tick-borne zoonosis that poses an escalating public health threat in several parts of the world, despite sophisticated healthcare infrastructure and decades of effort to address the problem. Concepts like the true burden of the illness, from incidence rates to longstanding consequences of infection, and optimal case management, also remain shrouded in controversy. At the heart of this multidisciplinary issue are the causative spirochetal pathogens belonging to the Borrelia Lyme complex. Their unusual physiology and versatile lifestyle have challenged microbiologists, and may also hold the key to unlocking mysteries of the disease. The goal of this review is therefore to integrate established and emerging concepts of Borrelia biology and pathogenesis, and position them in the broader context of biomedical research and clinical practice. We begin by considering the conventions around diagnosing and characterizing Lyme disease that have served as a conceptual framework for the discipline. We then explore virulence from the perspective of both host (genetic and environmental predispositions) and pathogen (serotypes, dissemination, and immune modulation), as well as considering antimicrobial strategies (lab methodology, resistance, persistence, and clinical application), and borrelial adaptations of hypothesized medical significance (phenotypic plasticity or pleomorphy).
Collapse
Affiliation(s)
| | | | | | | | - Melanie K. B. Wills
- G. Magnotta Lyme Disease Research Lab, Molecular and Cellular Biology, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; (V.V.B.); (J.T.K.); (I.L.M.); (V.P.S.)
| |
Collapse
|
12
|
Vaca DJ, Thibau A, Schütz M, Kraiczy P, Happonen L, Malmström J, Kempf VAJ. Interaction with the host: the role of fibronectin and extracellular matrix proteins in the adhesion of Gram-negative bacteria. Med Microbiol Immunol 2019; 209:277-299. [PMID: 31784893 PMCID: PMC7248048 DOI: 10.1007/s00430-019-00644-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/14/2019] [Indexed: 01/03/2023]
Abstract
The capacity of pathogenic microorganisms to adhere to host cells and avoid clearance by the host immune system is the initial and most decisive step leading to infections. Bacteria have developed different strategies to attach to diverse host surface structures. One important strategy is the adhesion to extracellular matrix (ECM) proteins (e.g., collagen, fibronectin, laminin) that are highly abundant in connective tissue and basement membranes. Gram-negative bacteria express variable outer membrane proteins (adhesins) to attach to the host and to initiate the process of infection. Understanding the underlying molecular mechanisms of bacterial adhesion is a prerequisite for targeting this interaction by “anti-ligands” to prevent colonization or infection of the host. Future development of such “anti-ligands” (specifically interfering with bacteria-host matrix interactions) might result in the development of a new class of anti-infective drugs for the therapy of infections caused by multidrug-resistant Gram-negative bacteria. This review summarizes our current knowledge about the manifold interactions of adhesins expressed by Gram-negative bacteria with ECM proteins and the use of this information for the generation of novel therapeutic antivirulence strategies.
Collapse
Affiliation(s)
- Diana J Vaca
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University Frankfurt am Main, Paul-Ehrlich-Str. 40, 60596, Frankfurt, Germany
| | - Arno Thibau
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University Frankfurt am Main, Paul-Ehrlich-Str. 40, 60596, Frankfurt, Germany
| | - Monika Schütz
- Institute for Medical Microbiology and Infection Control, University Hospital, Eberhard Karls-University, Tübingen, Germany
| | - Peter Kraiczy
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University Frankfurt am Main, Paul-Ehrlich-Str. 40, 60596, Frankfurt, Germany
| | - Lotta Happonen
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Volkhard A J Kempf
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University Frankfurt am Main, Paul-Ehrlich-Str. 40, 60596, Frankfurt, Germany.
| |
Collapse
|
13
|
Caimano MJ, Groshong AM, Belperron A, Mao J, Hawley KL, Luthra A, Graham DE, Earnhart CG, Marconi RT, Bockenstedt LK, Blevins JS, Radolf JD. The RpoS Gatekeeper in Borrelia burgdorferi: An Invariant Regulatory Scheme That Promotes Spirochete Persistence in Reservoir Hosts and Niche Diversity. Front Microbiol 2019; 10:1923. [PMID: 31507550 PMCID: PMC6719511 DOI: 10.3389/fmicb.2019.01923] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/05/2019] [Indexed: 11/28/2022] Open
Abstract
Maintenance of Borrelia burgdorferi within its enzootic cycle requires a complex regulatory pathway involving the alternative σ factors RpoN and RpoS and two ancillary trans-acting factors, BosR and Rrp2. Activation of this pathway occurs within ticks during the nymphal blood meal when RpoS, the effector σ factor, transcribes genes required for tick transmission and mammalian infection. RpoS also exerts a 'gatekeeper' function by repressing σ70-dependent tick phase genes (e.g., ospA, lp6.6). Herein, we undertook a broad examination of RpoS functionality throughout the enzootic cycle, beginning with modeling to confirm that this alternative σ factor is a 'genuine' RpoS homolog. Using a novel dual color reporter system, we established at the single spirochete level that ospA is expressed in nymphal midguts throughout transmission and is not downregulated until spirochetes have been transmitted to a naïve host. Although it is well established that rpoS/RpoS is expressed throughout infection, its requirement for persistent infection has not been demonstrated. Plasmid retention studies using a trans-complemented ΔrpoS mutant demonstrated that (i) RpoS is required for maximal fitness throughout the mammalian phase and (ii) RpoS represses tick phase genes until spirochetes are acquired by a naïve vector. By transposon mutant screening, we established that bba34/oppA5, the only OppA oligopeptide-binding protein controlled by RpoS, is a bona fide persistence gene. Lastly, comparison of the strain 297 and B31 RpoS DMC regulons identified two cohorts of RpoS-regulated genes. The first consists of highly conserved syntenic genes that are similarly regulated by RpoS in both strains and likely required for maintenance of B. burgdorferi sensu stricto strains in the wild. The second includes RpoS-regulated plasmid-encoded variable surface lipoproteins ospC, dbpA and members of the ospE/ospF/elp, mlp, revA, and Pfam54 paralogous gene families, all of which have evolved via inter- and intra-strain recombination. Thus, while the RpoN/RpoS pathway regulates a 'core' group of orthologous genes, diversity within RpoS regulons of different strains could be an important determinant of reservoir host range as well as spirochete virulence.
Collapse
Affiliation(s)
- Melissa J. Caimano
- Department of Medicine, UConn Health, Farmington, CT, United States,Department of Pediatrics, UConn Health, Farmington, CT, United States,Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, United States,*Correspondence: Melissa J. Caimano,
| | | | - Alexia Belperron
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Jialing Mao
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Kelly L. Hawley
- Department of Pediatrics, UConn Health, Farmington, CT, United States,Division of Infectious Diseases and Immunology, Connecticut Children’s Medical Center, Hartford, CT, United States
| | - Amit Luthra
- Department of Medicine, UConn Health, Farmington, CT, United States
| | - Danielle E. Graham
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Christopher G. Earnhart
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, VA, United States
| | - Richard T. Marconi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, VA, United States
| | - Linda K. Bockenstedt
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Jon S. Blevins
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Justin D. Radolf
- Department of Medicine, UConn Health, Farmington, CT, United States,Department of Pediatrics, UConn Health, Farmington, CT, United States,Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, United States,Department of Genetics and Genome Science, UConn Health, Farmington, CT, United States,Department of Immunology, UConn Health, Farmington, CT, United States
| |
Collapse
|
14
|
Cuellar J, Pietikäinen A, Glader O, Liljenbäck H, Söderström M, Hurme S, Salo J, Hytönen J. Borrelia burgdorferi Infection in Biglycan Knockout Mice. J Infect Dis 2019; 220:116-126. [DOI: 10.1093/infdis/jiz050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/28/2019] [Indexed: 12/25/2022] Open
Affiliation(s)
- Julia Cuellar
- Institute of Biomedicine, University of Turku
- Turku Doctoral Programme of Molecular Medicine, Faculty of Medicine, University of Turku
| | - Annukka Pietikäinen
- Institute of Biomedicine, University of Turku
- Turku Doctoral Programme of Molecular Medicine, Faculty of Medicine, University of Turku
| | - Otto Glader
- Institute of Biomedicine, University of Turku
| | - Heidi Liljenbäck
- Turku Center for Disease Modeling, University of Turku
- Turku PET Centre, University of Turku
| | - Mirva Söderström
- Department of Pathology and Forensic Medicine, University of Turku and Turku University Hospital
| | - Saija Hurme
- Department of Biostatistics, University of Turku
| | | | - Jukka Hytönen
- Institute of Biomedicine, University of Turku
- Laboratory Division, Unit of Clinical Microbiology, Turku University Hospital, Turku, Finland
| |
Collapse
|
15
|
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]
|
16
|
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
|
17
|
Bergström S, Normark J. Microbiological features distinguishing Lyme disease and relapsing fever spirochetes. Wien Klin Wochenschr 2018; 130:484-490. [PMID: 30074091 PMCID: PMC6096528 DOI: 10.1007/s00508-018-1368-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 07/13/2018] [Indexed: 12/13/2022]
Abstract
The recent proposal of splitting the genus Borrelia into two genera in the newly formed family of Borreliaceae, i. e. Borrelia and Borreliella has motivated us to reflect upon how these organisms has been characterized and differentiated. This article therefore aims to take a closer look on the biology and virulence attributes of the two suggested genera, i. e. those causing Lyme borreliosis and relapsing fever borreliosis. Both genera have much in common with similar infection biological features. They are both characterized as bacterial zoonoses, transmitted by hematophagous arthropods with almost identical microbiological appearance. Nevertheless, a closer look at the genotypic and phenotypic characteristics clearly reveals several differences that might motivate the suggested split. On the other hand, a change of this well-established classification within the genus Borrelia might impose an economical burden as well as a great confusion in society, including medical and scientific societies as well as the general population.
Collapse
Affiliation(s)
- Sven Bergström
- Department of Molecular Biology, Umeå University, 6K och 6L, Sjukhusområdet, 901 87, Umeå, Sweden. .,Umeå Center for Microbial Research, Umeå University, Umeå, Sweden. .,Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden.
| | - Johan Normark
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden.,Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
| |
Collapse
|
18
|
Borrelia burgdorferi adhere to blood vessels in the dura mater and are associated with increased meningeal T cells during murine disseminated borreliosis. PLoS One 2018; 13:e0196893. [PMID: 29723263 PMCID: PMC5933741 DOI: 10.1371/journal.pone.0196893] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 04/20/2018] [Indexed: 11/19/2022] Open
Abstract
Borrelia burgdorferi, the causative agent of Lyme disease, is a vector-borne bacterial infection that is transmitted through the bite of an infected tick. If not treated with antibiotics during the early stages of infection, disseminated infection can spread to the central nervous system (CNS). In non-human primates (NHPs) it has been demonstrated that the leptomeninges are among the tissues colonized by B. burgdorferi spirochetes. Although the NHP model parallels aspects of human borreliosis, a small rodent model would be ideal to study the trafficking of spirochetes and immune cells into the CNS. Here we show that during early and late disseminated infection, B. burgdorferi infects the meninges of intradermally infected mice, and is associated with concurrent increases in meningeal T cells. We found that the dura mater was consistently culture positive for spirochetes in transcardially perfused mice, independent of the strain of B. burgdorferi used. Within the dura mater, spirochetes were preferentially located in vascular regions, but were also present in perivascular, and extravascular regions, as late as 75 days post-infection. At the same end-point, we observed significant increases in the number of CD3+ T cells within the pia and dura mater, as compared to controls. Flow cytometric analysis of leukocytes isolated from the dura mater revealed that CD3+ cell populations were comprised of both CD4 and CD8 T cells. Overall, our data demonstrate that similarly to infection in peripheral tissues, spirochetes adhere to the dura mater during disseminated infection, and are associated with increases in the number of meningeal T cells. Collectively, our results demonstrate that there are aspects of B. burgdorferi meningeal infection that can be modelled in laboratory mice, suggesting that mice may be useful for elucidating mechanisms of meningeal pathogenesis by B. burgdorferi.
Collapse
|
19
|
The Borrelia burgdorferi Glycosaminoglycan Binding Protein Bgp in the B31 Strain Is Not Essential for Infectivity despite Facilitating Adherence and Tissue Colonization. Infect Immun 2018; 86:IAI.00667-17. [PMID: 29158428 DOI: 10.1128/iai.00667-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/08/2017] [Indexed: 11/20/2022] Open
Abstract
The Lyme disease-causing organism Borrelia burgdorferi is transmitted into the mammalian host by an infected-tick bite. Successful infection relies on the ability of this extracellular pathogen to persist and colonize different tissues. B. burgdorferi encodes a large number of adhesins that are able to interact with host ligands to facilitate adherence and tissue colonization. Multiple glycosaminoglycan binding proteins present in B. burgdorferi offer a degree of redundancy of function during infection, and this highlights the importance of glycosaminoglycans as host cell receptors for spirochete adherence. Of particular interest in this study is Borrelia glycosaminoglycan binding protein (Bgp), which binds to heparin-related glycosaminoglycans. The properties of a bgp transposon mutant and a trans-complemented derivative were compared to those of the wild-type B. burgdorferi in the in vitro binding assays and in infection studies using a C3H/HeJ mouse infection model. We determined that the loss of Bgp impairs spirochete adherence, infectivity, and tissue colonization, resulting in a reduction of inflammatory manifestations of Lyme disease. Although Bgp is not essential for infectivity, it is an important virulence factor of B. burgdorferi that allows adherence and tissue colonization and contributes to disease severity.
Collapse
|
20
|
Lin YP, Li L, Zhang F, Linhardt RJ. Borrelia burgdorferi glycosaminoglycan-binding proteins: a potential target for new therapeutics against Lyme disease. MICROBIOLOGY-SGM 2017; 163:1759-1766. [PMID: 29116038 DOI: 10.1099/mic.0.000571] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The spirochete bacterium Borrelia burgdorferi sensu lato is the causative agent of Lyme disease, the most common vector-borne disease in Europe and the United States. The spirochetes can be transmitted to humans via ticks, and then spread to different tissues, leading to arthritis, carditis and neuroborreliosis. Although antibiotics have commonly been used to treat infected individuals, some treated patients do not respond to antibiotics and experience persistent, long-term arthritis. Thus, there is a need to investigate alternative therapeutics against Lyme disease. The spirochete bacterium colonization is partly attributed to the binding of the bacterial outer-surface proteins to the glycosaminoglycan (GAG) chains of host proteoglycans. Blocking the binding of these proteins to GAGs is a potential strategy to prevent infection. In this review, we have summarized the recent reports of B. burgdorferi sensu lato GAG-binding proteins and discussed the potential use of synthetic and semi-synthetic compounds, including GAG analogues, to block pathogen interaction with GAGs. Such information should motivate the discovery and development of novel GAG analogues as new therapeutics for Lyme disease. New therapeutic approaches should eventually reduce the burden of Lyme disease and improve human health.
Collapse
Affiliation(s)
- Yi-Pin Lin
- Department of Biomedical Science, State University of New York at Albany, Albany, NY, USA.,Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Lingyun Li
- Division of Environmental Health Science, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Robert J Linhardt
- Departments of Biology and Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| |
Collapse
|
21
|
Hyde JA. Borrelia burgdorferi Keeps Moving and Carries on: A Review of Borrelial Dissemination and Invasion. Front Immunol 2017; 8:114. [PMID: 28270812 PMCID: PMC5318424 DOI: 10.3389/fimmu.2017.00114] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/25/2017] [Indexed: 12/21/2022] Open
Abstract
Borrelia burgdorferi is the etiological agent of Lyme disease, a multisystemic, multistage, inflammatory infection resulting in patients experiencing cardiac, neurological, and arthritic complications when not treated with antibiotics shortly after exposure. The spirochetal bacterium transmits through the Ixodes vector colonizing the dermis of a mammalian host prior to hematogenous dissemination and invasion of distal tissues all the while combating the immune response as it traverses through its pathogenic lifecycle. The innate immune response controls the borrelial burden in the dermis, but is unable to clear the infection and thereby prevent progression of disease. Dissemination in the mammalian host requires temporal regulation of virulence determinants to allow for vascular interactions, invasion, and colonization of distal tissues. Virulence determinants and/or adhesins are highly heterogenetic among environmental B. burgdorferi strains with particular genotypes being associated with the ability to disseminate to specific tissues and the severity of disease, but fail to generate cross-protective immunity between borrelial strains. The unique motility of B. burgdorferi rendered by the endoflagella serves a vital function for dissemination and protection from immune recognition. Progress has been made toward understanding the chemotactic regulation coordinating the activity of the two polar localized flagellar motors and their role in borrelial virulence, but this regulation is not yet fully understood. Distinct states of motility allow for dynamic interactions between several B. burgdorferi adhesins and host targets that play roles in transendothelial migration. Transmigration across endothelial and blood-brain barriers allows for the invasion of tissues and elicits localized immune responses. The invasive nature of B. burgdorferi is lacking in proactive mechanisms to modulate disease, such as secretion systems and toxins, but recent work has shown degradation of host extracellular matrices by B. burgdorferi contributes to the invasive capabilities of the pathogen. Additionally, B. burgdorferi may use invasion of eukaryotic cells for immune evasion and protection against environmental stresses. This review provides an overview of B. burgdorferi mechanisms for dissemination and invasion in the mammalian host, which are essential for pathogenesis and the development of persistent infection.
Collapse
Affiliation(s)
- Jenny A Hyde
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center , Bryan, TX , USA
| |
Collapse
|
22
|
Caine JA, Coburn J. Multifunctional and Redundant Roles of Borrelia burgdorferi Outer Surface Proteins in Tissue Adhesion, Colonization, and Complement Evasion. Front Immunol 2016; 7:442. [PMID: 27818662 PMCID: PMC5073149 DOI: 10.3389/fimmu.2016.00442] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 10/07/2016] [Indexed: 12/24/2022] Open
Abstract
Borrelia burgdorferi is the causative agent of Lyme disease in the U.S., with at least 25,000 cases reported to the CDC each year. B. burgdorferi is thought to enter and exit the bloodstream to achieve rapid dissemination to distal tissue sites during infection. Travel through the bloodstream requires evasion of immune surveillance and pathogen clearance in the host, a process at which B. burgdorferi is adept. B. burgdorferi encodes greater than 19 adhesive outer surface proteins many of which have been found to bind to host cells or components of the extracellular matrix. Several others bind to host complement regulatory factors, in vitro. Production of many of these adhesive proteins is tightly regulated by environmental cues, and some have been shown to aid in vascular interactions and tissue colonization, as well as survival in the blood, in vivo. Recent work has described multifaceted and redundant roles of B. burgdorferi outer surface proteins in complement component interactions and tissue targeted adhesion and colonization, distinct from their previously identified in vitro binding capabilities. Recent insights into the multifunctional roles of previously well-characterized outer surface proteins such as BBK32, DbpA, CspA, and OspC have changed the way we think about the surface proteome of these organisms during the tick-mammal life cycle. With the combination of new and old in vivo models and in vitro techniques, the field has identified distinct ligand binding domains on BBK32 and DbpA that afford tissue colonization or blood survival to B. burgdorferi. In this review, we describe the multifunctional and redundant roles of many adhesive outer surface proteins of B. burgdorferi in tissue adhesion, colonization, and bloodstream survival that, together, promote the survival of Borrelia spp. throughout maintenance in their multi-host lifestyle.
Collapse
Affiliation(s)
- Jennifer A. Caine
- Division of Infectious Disease, Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jenifer Coburn
- Division of Infectious Disease, Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
23
|
Gubbiotti MA, Vallet SD, Ricard-Blum S, Iozzo RV. Decorin interacting network: A comprehensive analysis of decorin-binding partners and their versatile functions. Matrix Biol 2016; 55:7-21. [PMID: 27693454 DOI: 10.1016/j.matbio.2016.09.009] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Decorin, a prototype small leucine-rich proteoglycan, regulates a vast array of cellular processes including collagen fibrillogenesis, wound repair, angiostasis, tumor growth, and autophagy. This functional versatility arises from a wide array of decorin/protein interactions also including interactions with its single glycosaminoglycan side chain. The decorin-binding partners encompass numerous categories ranging from extracellular matrix molecules to cell surface receptors to growth factors and enzymes. Despite the diversity of the decorin interacting network, two main roles emerge as prominent themes in decorin function: maintenance of cellular structure and outside-in signaling, culminating in anti-tumorigenic effects. Here we present contemporary knowledge regarding the decorin interacting network and discuss in detail the biological relevance of these pleiotropic interactions, some of which could be targeted by therapeutic interventions.
Collapse
Affiliation(s)
- Maria A Gubbiotti
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sylvain D Vallet
- Pericellular and Extracellular Supramolecular Assemblies, Institute of Molecular and Supramolecular Chemistry and Biochemistry, University Claude Bernard, Lyon, France
| | - Sylvie Ricard-Blum
- Pericellular and Extracellular Supramolecular Assemblies, Institute of Molecular and Supramolecular Chemistry and Biochemistry, University Claude Bernard, Lyon, France
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
24
|
Neal JW, Gasque P. The role of primary infection of Schwann cells in the aetiology of infective inflammatory neuropathies. J Infect 2016; 73:402-418. [PMID: 27546064 DOI: 10.1016/j.jinf.2016.08.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 02/08/2023]
Abstract
Numerous different pathogens are responsible for infective peripheral neuropathies and this is generally the result of the indirect effects of pathogen infection, namely anti pathogen antibodies cross reacting with epitopes on peripheral nerve, auto reactive T cells attacking myelin, circulating immune complexes and complement fixation. Primary infection of Schwann cells (SC) associated with peripheral nerve inflammation is rare requiring pathogens to cross the Blood Peripheral Nerve Barrier (BPNB) evade anti-pathogen innate immune pathways and invade the SC. Spirochetes Borrelia bourgdorferi and Trepomema pallidum are highly invasive, express surface lipo proteins, but despite this SC are rarely infected. However, Trypanosoma cruzi (Chaga's disease) and Mycobacterium leprae. Leprosy are two important causes of peripheral nerve infection and both demonstrate primary infection of SC. This is due to two novel strategies; T. cruzi express a trans-silalidase that mimics host neurotrophic factors and infects SC via tyrosine kinase receptors. M. leprae demonstrates multi receptor SC tropism and subsequent infection promotes nuclear reprogramming and dedifferentiation of host SC into progenitor stem like cells (pSLC) that are vulnerable to M. leprae infection. These two novel pathogen evasion strategies, involving stem cells and receptor mimicry, provide potential therapeutic targets relevant to the prevention of peripheral nerve inflammation by inhibiting primary SC infection.
Collapse
Affiliation(s)
- J W Neal
- Infection and Immunity, Henry Wellcome Building, Cardiff University, Cardiff CF14 4XN, United Kingdom.
| | - P Gasque
- Laboratoire d'Immunologie Clinique et Expérimentale de l'OI (LICE-OI), Centre recherche Immuno-clinique des agents pathogènes de l'OI (CRIC-AP OI) Pôle Biologie Santé, Hôpital Félix Guyon, CHU de la Réunion, Reunion.
| |
Collapse
|
25
|
Kenedy MR, Scott EJ, Shrestha B, Anand A, Iqbal H, Radolf JD, Dyer DW, Akins DR. Consensus computational network analysis for identifying candidate outer membrane proteins from Borrelia spirochetes. BMC Microbiol 2016; 16:141. [PMID: 27400788 PMCID: PMC4939628 DOI: 10.1186/s12866-016-0762-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 07/02/2016] [Indexed: 01/15/2023] Open
Abstract
Background Similar to Gram-negative organisms, Borrelia spirochetes are dual-membrane organisms with both an inner and outer membrane. Although the outer membrane contains integral membrane proteins, few of the borrelial outer membrane proteins (OMPs) have been identified and characterized to date. Therefore, we utilized a consensus computational network analysis to identify novel borrelial OMPs. Results Using a series of computer-based algorithms, we selected all protein-encoding sequences predicted to be OM-localized and/or to form β-barrels in the borrelial OM. Using this system, we identified 41 potential OMPs from B. burgdorferi and characterized three (BB0838, BB0405, and BB0406) to confirm that our computer-based methodology did, in fact, identify borrelial OMPs. Triton X-114 phase partitioning revealed that BB0838 is found in the detergent phase, which would be expected of a membrane protein. Proteolysis assays indicate that BB0838 is partially sensitive to both proteinase K and trypsin, further indicating that BB0838 is surface-exposed. Consistent with a prior study, we also confirmed that BB0405 is surface-exposed and associates with the borrelial OM. Furthermore, we have shown that BB0406, the product of a co-transcribed downstream gene, also encodes a novel, previously uncharacterized borrelial OMP. Interestingly, while BB0406 has several physicochemical properties consistent with it being an OMP, it was found to be resistant to surface proteolysis. Consistent with BB0405 and BB0406 being OMPs, both were found to be capable of incorporating into liposomes and exhibit pore-forming activity, suggesting that both proteins are porins. Lastly, we expanded our computational analysis to identify OMPs from other borrelial organisms, including both Lyme disease and relapsing fever spirochetes. Conclusions Using a consensus computer algorithm, we generated a list of candidate OMPs for both Lyme disease and relapsing fever spirochetes and determined that three of the predicted B. burgdorferi proteins identified were indeed novel borrelial OMPs. The combined studies have identified putative spirochetal OMPs that can now be examined for their roles in virulence, physiology, and disease pathogenesis. Importantly, the studies described in this report provide a framework by which OMPs from any human pathogen with a diderm ultrastructure could be cataloged to identify novel virulence factors and vaccine candidates. Electronic supplementary material The online version of this article (doi:10.1186/s12866-016-0762-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melisha R Kenedy
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Edgar J Scott
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Binu Shrestha
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Arvind Anand
- Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, 06030, USA
| | - Henna Iqbal
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Justin D Radolf
- Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, 06030, USA.,Department of Pediatrics, University of Connecticut Health Center, Farmington, Connecticut, 06030, USA.,Department of Genetics and Genomic Science, University of Connecticut Health Center, Farmington, Connecticut, 06030, USA.,Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, 06030, USA.,Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, Connecticut, 06030, USA
| | - David W Dyer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Darrin R Akins
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA.
| |
Collapse
|
26
|
Muehlenbachs A, Bollweg BC, Schulz TJ, Forrester JD, DeLeon Carnes M, Molins C, Ray GS, Cummings PM, Ritter JM, Blau DM, Andrew TA, Prial M, Ng DL, Prahlow JA, Sanders JH, Shieh WJ, Paddock CD, Schriefer ME, Mead P, Zaki SR. Cardiac Tropism of Borrelia burgdorferi: An Autopsy Study of Sudden Cardiac Death Associated with Lyme Carditis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1195-205. [PMID: 26968341 DOI: 10.1016/j.ajpath.2015.12.027] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/23/2015] [Accepted: 12/15/2015] [Indexed: 12/17/2022]
Abstract
Fatal Lyme carditis caused by the spirochete Borrelia burgdorferi rarely is identified. Here, we describe the pathologic, immunohistochemical, and molecular findings of five case patients. These sudden cardiac deaths associated with Lyme carditis occurred from late summer to fall, ages ranged from young adult to late 40s, and four patients were men. Autopsy tissue samples were evaluated by light microscopy, Warthin-Starry stain, immunohistochemistry, and PCR for B. burgdorferi, and immunohistochemistry for complement components C4d and C9, CD3, CD79a, and decorin. Post-mortem blood was tested by serology. Interstitial lymphocytic pancarditis in a relatively characteristic road map distribution was present in all cases. Cardiomyocyte necrosis was minimal, T cells outnumbered B cells, plasma cells were prominent, and mild fibrosis was present. Spirochetes in the cardiac interstitium associated with collagen fibers and co-localized with decorin. Rare spirochetes were seen in the leptomeninges of two cases by immunohistochemistry. Spirochetes were not seen in other organs examined, and joint tissue was not available for evaluation. Although rare, sudden cardiac death caused by Lyme disease might be an under-recognized entity and is characterized by pancarditis and marked tropism of spirochetes for cardiac tissues.
Collapse
Affiliation(s)
- Atis Muehlenbachs
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia.
| | - Brigid C Bollweg
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | - Joseph D Forrester
- Bacterial Diseases Branch, Division of Vector Borne Infectious Diseases, Centers for Disease Control and Prevention, Ft. Collins, Colorado
| | - Marlene DeLeon Carnes
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Claudia Molins
- Bacterial Diseases Branch, Division of Vector Borne Infectious Diseases, Centers for Disease Control and Prevention, Ft. Collins, Colorado
| | | | | | - Jana M Ritter
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Dianna M Blau
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Thomas A Andrew
- Office of the Chief Medical Examiner, Concord, New Hampshire
| | | | - Dianna L Ng
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Joseph A Prahlow
- The Medical Foundation, South Bend, Indiana; Indiana University School of Medicine-South Bend, South Bend, Indiana
| | - Jeanine H Sanders
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Wun Ju Shieh
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Christopher D Paddock
- Rickettsial Zoonotic Diseases Branch, Division of Vector Borne Infectious Diseases, Atlanta, Georgia
| | - Martin E Schriefer
- Bacterial Diseases Branch, Division of Vector Borne Infectious Diseases, Centers for Disease Control and Prevention, Ft. Collins, Colorado
| | - Paul Mead
- Bacterial Diseases Branch, Division of Vector Borne Infectious Diseases, Centers for Disease Control and Prevention, Ft. Collins, Colorado
| | - Sherif R Zaki
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
27
|
Drecktrah D, Lybecker M, Popitsch N, Rescheneder P, Hall LS, Samuels DS. The Borrelia burgdorferi RelA/SpoT Homolog and Stringent Response Regulate Survival in the Tick Vector and Global Gene Expression during Starvation. PLoS Pathog 2015; 11:e1005160. [PMID: 26371761 PMCID: PMC4570706 DOI: 10.1371/journal.ppat.1005160] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 08/21/2015] [Indexed: 01/09/2023] Open
Abstract
As the Lyme disease bacterium Borrelia burgdorferi traverses its enzootic cycle, alternating between a tick vector and a vertebrate host, the spirochete must adapt and persist in the tick midgut under prolonged nutrient stress between blood meals. In this study, we examined the role of the stringent response in tick persistence and in regulation of gene expression during nutrient limitation. Nutritionally starving B. burgdorferi in vitro increased the levels of guanosine tetraphosphate (ppGpp) and guanosine pentaphosphate (pppGpp), collectively referred to as (p)ppGpp, products of the bifunctional synthetase/hydrolase RelBbu (RelA/SpoT homolog). Conversely, returning B. burgdorferi to a nutrient-rich medium decreased (p)ppGpp levels. B. burgdorferi survival in ticks between the larval and nymph blood meals, and during starvation in vitro, was dependent on RelBbu. Furthermore, normal morphological conversion from a flat-wave shape to a condensed round body (RB) form during starvation was dependent on RelBbu; relBbu mutants more frequently formed RBs, but their membranes were compromised. By differential RNA sequencing analyses, we found that RelBbu regulates an extensive transcriptome, both dependent and independent of nutrient stress. The RelBbu regulon includes the glp operon, which is important for glycerol utilization and persistence in the tick, virulence factors and the late phage operon of the 32-kb circular plasmid (cp32) family. In summary, our data suggest that RelBbu globally modulates transcription in response to nutrient stress by increasing (p)ppGpp levels to facilitate B. burgdorferi persistence in the tick.
Collapse
Affiliation(s)
- Dan Drecktrah
- Division of Biological Sciences, University of Montana, Missoula, Montana, United States of America
| | - Meghan Lybecker
- Department of Biology, University of Colorado, Colorado Springs, Colorado, United States of America
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Niko Popitsch
- Oxford NIHR Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Center for Integrative Bioinformatics Vienna, Max F. Perutz Laboratories, University of Vienna & Medical University of Vienna, Vienna, Austria
| | - Philipp Rescheneder
- Center for Integrative Bioinformatics Vienna, Max F. Perutz Laboratories, University of Vienna & Medical University of Vienna, Vienna, Austria
| | - Laura S. Hall
- Division of Biological Sciences, University of Montana, Missoula, Montana, United States of America
| | - D. Scott Samuels
- Division of Biological Sciences, University of Montana, Missoula, Montana, United States of America
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana, United States of America
| |
Collapse
|
28
|
Feng W, Wang X. Structure of decorin binding protein B from Borrelia burgdorferi and its interactions with glycosaminoglycans. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1823-1832. [PMID: 26275806 DOI: 10.1016/j.bbapap.2015.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/28/2015] [Accepted: 08/09/2015] [Indexed: 11/26/2022]
Abstract
Decorin-binding proteins (DBPs), DBPA and DBPB, are surface lipoproteins on Borrelia burgdorferi, the causative agent of Lyme disease. DBPs bind to the connective tissue proteoglycan decorin and facilitate tissue colonization by the bacterium. Although structural and biochemical properties of DBPA are well understood, little is known about DBPB. In current work, we determined the solution structure of DBPB from strain B31 of B. burgdorferi and characterized its interactions with glycosaminoglycans (GAGs). Our structure shows that DBPB adopts the same topology as DBPA, but possesses a much shorter terminal helix, resulting in a longer unstructured C-terminal tail, which is also rich in basic amino acids. Characterization of DBPB-GAG interactions reveals that, despite similar GAG affinities of DBPA and DBPB, the primary GAG-binding sites in DBPB are different from DBPA. In particular, our results indicate that lysines in the C-terminus of DBPB are vital to DBPB's ability to bind GAGs whereas C-terminal tail for DBPA from strain B31 only plays a minor role in facilitating GAG bindings. Furthermore, the traditional GAG-binding pocket important to DBPA-GAG interactions is only secondary to DBPB's GAG-binding ability.
Collapse
Affiliation(s)
- Wei Feng
- Department of Chemistry & Biochemistry, Arizona State University, Tempe, AZ 85287, USA
| | - Xu Wang
- Department of Chemistry & Biochemistry, Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
29
|
Morgan A, Sepuru KM, Feng W, Rajarathnam K, Wang X. Flexible Linker Modulates Glycosaminoglycan Affinity of Decorin Binding Protein A. Biochemistry 2015. [PMID: 26223367 PMCID: PMC4873102 DOI: 10.1021/acs.biochem.5b00253] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Decorin
binding protein A (DBPA) is a glycosaminoglycan (GAG)-binding
adhesin found on the surface of the bacterium Borrelia
burgdorferi (B. burgdorferi), the causative agent of Lyme disease. DBPA facilitates bacterial
adherence to extracellular matrices of human tissues and is crucial
during the early stage of the infection process. Interestingly, DBPA
from different strains (B31, N40, and PBr) show significant differences
in GAG affinities, but the structural basis for the differences is
not clear. In this study, we show that GAG affinity of N40 DBPA is
modulated in part by flexible segments that control access to the
GAG binding site, such that shortening of the linker leads to higher
GAG affinity when analyzed using ELISA, gel mobility shift assay,
solution NMR, and isothermal titration calorimetry. Our observation
that GAG affinity differences among different B. burgdorferi strains can be attributed to a flexible linker domain regulating
access to the GAG-binding domain is novel. It also provides a rare
example of how neutral amino acids and dynamic segments in GAG binding
proteins can have a large influence on GAG affinity and provides insights
into why the number of basic amino acids in the GAG-binding site may
not be the only factor determining GAG affinity of proteins.
Collapse
Affiliation(s)
| | - Krishna Mohan Sepuru
- ‡Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | | | - Krishna Rajarathnam
- ‡Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | | |
Collapse
|
30
|
Structural mechanisms underlying sequence-dependent variations in GAG affinities of decorin binding protein A, a Borrelia burgdorferi adhesin. Biochem J 2015; 467:439-51. [DOI: 10.1042/bj20141201] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Decorin-binding protein A (DBPA) is an important surface adhesin of the bacterium Borrelia burgdorferi, the causative agent of Lyme disease. DBPA facilitates the bacteria's colonization of human tissue by adhering to glycosaminoglycan (GAG), a sulfated polysaccharide. Interestingly, DBPA sequence variation among different strains of Borrelia spirochetes is high, resulting in significant differences in their GAG affinities. However, the structural mechanisms contributing to these differences are unknown. We determined the solution structures of DBPAs from strain N40 of B. burgdorferi and strain PBr of Borrelia garinii, two DBPA variants whose GAG affinities deviate significantly from strain B31, the best characterized version of DBPA. Our structures revealed that significant differences exist between PBr DBPA and B31/N40 DBPAs. In particular, the C-terminus of PBr DBPA, unlike C-termini from B31 and N40 DBPAs, is positioned away from the GAG-binding pocket and the linker between helices one and two of PBr DBPA is highly structured and retracted from the GAG-binding pocket. The repositioning of the C-terminus allowed the formation of an extra GAG-binding epitope in PBr DBPA and the retracted linker gave GAG ligands more access to the GAG-binding epitopes than other DBPAs. Characterization of GAG ligands' interactions with wild-type (WT) PBr and mutants confirmed the importance of the second major GAG-binding epitope and established the fact that the two epitopes are independent of one another and the new epitope is as important to GAG binding as the traditional epitope.
Collapse
|
31
|
Salo J, Jaatinen A, Söderström M, Viljanen MK, Hytönen J. Decorin binding proteins of Borrelia burgdorferi promote arthritis development and joint specific post-treatment DNA persistence in mice. PLoS One 2015; 10:e0121512. [PMID: 25816291 PMCID: PMC4376631 DOI: 10.1371/journal.pone.0121512] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 01/28/2015] [Indexed: 01/01/2023] Open
Abstract
Decorin binding proteins A and B (DbpA and B) of Borrelia burgdorferi are of critical importance for the virulence of the spirochete. The objective of the present study was to further clarify the contribution of DbpA and B to development of arthritis and persistence of B. burgdorferi after antibiotic treatment in a murine model of Lyme borreliosis. With that goal, mice were infected with B. burgdorferi strains expressing either DbpA or DbpB, or both DbpA and B, or with a strain lacking the adhesins. Arthritis development was monitored up to 15 weeks after infection, and bacterial persistence was studied after ceftriaxone and immunosuppressive treatments. Mice infected with the B. burgdorferi strain expressing both DbpA and B developed an early and prominent joint swelling. In contrast, while strains that expressed DbpA or B alone, or the strain that was DbpA and B deficient, were able to colonize mouse joints, they caused only negligible joint manifestations. Ceftriaxone treatment at two or six weeks of infection totally abolished joint swelling, and all ceftriaxone treated mice were B. burgdorferi culture negative. Antibiotic treated mice, which were immunosuppressed by anti-TNF-alpha, remained culture negative. Importantly, among ceftriaxone treated mice, B. burgdorferi DNA was detected by PCR uniformly in joint samples of mice infected with DbpA and B expressing bacteria, while this was not observed in mice infected with the DbpA and B deficient strain. In conclusion, these results show that both DbpA and B adhesins are crucial for early and prominent arthritis development in mice. Also, post-treatment borrelial DNA persistence appears to be dependent on the expression of DbpA and B on B. burgdorferi surface. Results of the immunosuppression studies suggest that the persisting material in the joints of antibiotic treated mice is DNA or DNA containing remnants rather than live bacteria.
Collapse
Affiliation(s)
- Jemiina Salo
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
- Turku Doctoral Programme of Biomedical Sciences, TuBS, Turku, Finland
- * E-mail:
| | - Annukka Jaatinen
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
- Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
| | - Mirva Söderström
- Department of Pathology, Turku University Hospital and University of Turku, Turku, Finland
| | - Matti K. Viljanen
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Jukka Hytönen
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| |
Collapse
|
32
|
Dunn JP, Kenedy MR, Iqbal H, Akins DR. Characterization of the β-barrel assembly machine accessory lipoproteins from Borrelia burgdorferi. BMC Microbiol 2015; 15:70. [PMID: 25887384 PMCID: PMC4377024 DOI: 10.1186/s12866-015-0411-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/12/2015] [Indexed: 12/26/2022] Open
Abstract
Background Like all diderm bacteria studied to date, Borrelia burgdorferi possesses a β-barrel assembly machine (BAM) complex. The bacterial BAM complexes characterized thus far consist of an essential integral outer membrane protein designated BamA and one or more accessory proteins. The accessory proteins are typically lipid-modified proteins anchored to the inner leaflet of the outer membrane through their lipid moieties. We previously identified and characterized the B. burgdorferi BamA protein in detail and more recently identified two lipoproteins encoded by open reading frames bb0324 and bb0028 that associate with the borrelial BamA protein. The role(s) of the BAM accessory lipoproteins in B. burgdorferi is currently unknown. Results Structural modeling of B. burgdorferi BB0028 revealed a distinct β-propeller fold similar to the known structure for the E. coli BAM accessory lipoprotein BamB. Additionally, the structural model for BB0324 was highly similar to the known structure of BamD, which is consistent with the prior finding that BB0324 contains tetratricopeptide repeat regions similar to other BamD orthologs. Consistent with BB0028 and BB0324 being BAM accessory lipoproteins, mutants lacking expression of each protein were found to exhibit altered membrane permeability and enhanced sensitivity to various antimicrobials. Additionally, BB0028 mutants also exhibited significantly impaired in vitro growth. Finally, immunoprecipitation experiments revealed that BB0028 and BB0324 each interact specifically and independently with BamA to form the BAM complex in B. burgdorferi. Conclusions Combined structural studies, functional assays, and co-immunoprecipitation experiments confirmed that BB0028 and BB0324 are the respective BamB and BamD orthologs in B. burgdorferi, and are important in membrane integrity and/or outer membrane protein localization. The borrelial BamB and BamD proteins both interact specifically and independently with BamA to form a tripartite BAM complex in B. burgdorferi. A working model has been developed to further analyze outer membrane biogenesis and outer membrane protein transport in this pathogenic spirochete.
Collapse
Affiliation(s)
- Joshua P Dunn
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Melisha R Kenedy
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Henna Iqbal
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Darrin R Akins
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
33
|
Schüler W, Bunikis I, Weber-Lehman J, Comstedt P, Kutschan-Bunikis S, Stanek G, Huber J, Meinke A, Bergström S, Lundberg U. Complete genome sequence of Borrelia afzelii K78 and comparative genome analysis. PLoS One 2015; 10:e0120548. [PMID: 25798594 PMCID: PMC4370689 DOI: 10.1371/journal.pone.0120548] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/23/2015] [Indexed: 02/04/2023] Open
Abstract
The main Borrelia species causing Lyme borreliosis in Europe and Asia are Borrelia afzelii, B. garinii, B. burgdorferi and B. bavariensis. This is in contrast to the United States, where infections are exclusively caused by B. burgdorferi. Until to date the genome sequences of four B. afzelii strains, of which only two include the numerous plasmids, are available. In order to further assess the genetic diversity of B. afzelii, the most common species in Europe, responsible for the large variety of clinical manifestations of Lyme borreliosis, we have determined the full genome sequence of the B. afzelii strain K78, a clinical isolate from Austria. The K78 genome contains a linear chromosome (905,949 bp) and 13 plasmids (8 linear and 5 circular) together presenting 1,309 open reading frames of which 496 are located on plasmids. With the exception of lp28-8, all linear replicons in their full length including their telomeres have been sequenced. The comparison with the genomes of the four other B. afzelii strains, ACA-1, PKo, HLJ01 and Tom3107, as well as the one of B. burgdorferi strain B31, confirmed a high degree of conservation within the linear chromosome of B. afzelii, whereas plasmid encoded genes showed a much larger diversity. Since some plasmids present in B. burgdorferi are missing in the B. afzelii genomes, the corresponding virulence factors of B. burgdorferi are found in B. afzelii on other unrelated plasmids. In addition, we have identified a species specific region in the circular plasmid, cp26, which could be used for species determination. Different non-coding RNAs have been located on the B. afzelii K78 genome, which have not previously been annotated in any of the published Borrelia genomes.
Collapse
Affiliation(s)
| | - Ignas Bunikis
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | | | | | | | - Gerold Stanek
- Medical University of Vienna, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | | | | | - Sven Bergström
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | | |
Collapse
|
34
|
Wu Q, Guan G, Liu Z, Li Y, Luo J, Yin H. RNA-Seq-based analysis of changes in Borrelia burgdorferi gene expression linked to pathogenicity. Parasit Vectors 2015; 8:155. [PMID: 25886272 PMCID: PMC4395869 DOI: 10.1186/s13071-014-0623-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 12/26/2014] [Indexed: 11/17/2022] Open
Abstract
Background Lyme disease is a global public health problem caused by the spirochaete Borrelia burgdorferi. Our previous studies found differences in disease severity between B. burgdorferi B31- and B. garinii SZ-infected mice. We hypothesized that genes that are differentially expressed between Borrelia isolates encode bacterial factors that contribute to disease diversity. Methods The present study used high-throughput sequencing technology to characterize and compare the transcriptional profiles of B. burgdorferi B31 and B. garinii SZ cultured in vitro. Real-time quantitative RT-PCR was used to validate selected data from RNA-seq experiments. Results A total of 731 genes were differentially expressed between B. burgdorferi B31 and B. garinii SZ isolates, including those encoding lipoproteins and purine transport proteins. The fold difference in expression for B. garinii SZ versus B. burgdorferi B31 ranged from 22.07 to 1.01. Expression of the OspA, OspB and DbpB genes were significantly lower in B. garinii SZ compared to B. burgdorferi B31. Conclusions The results support the hypothesis that global changes in gene expression underlie differences in Borrelia pathogenicity. The findings also provide an empirical basis for studying the mechanism of action of specific genes as well as their potential usefulness for the diagnosis and management of Lyme disease. Electronic supplementary material The online version of this article (doi:10.1186/s13071-014-0623-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiong Wu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Key Laboratory of Grazing Animal Diseases MOA, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, China.
| | - Guiquan Guan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Key Laboratory of Grazing Animal Diseases MOA, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, China.
| | - Zhijie Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Key Laboratory of Grazing Animal Diseases MOA, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, China.
| | - Youquan Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Key Laboratory of Grazing Animal Diseases MOA, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, China.
| | - Jianxun Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Key Laboratory of Grazing Animal Diseases MOA, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, China.
| | - Hong Yin
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Key Laboratory of Grazing Animal Diseases MOA, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, China.
| |
Collapse
|
35
|
Zhi H, Weening EH, Barbu EM, Hyde JA, Höök M, Skare JT. The BBA33 lipoprotein binds collagen and impacts Borrelia burgdorferi pathogenesis. Mol Microbiol 2015; 96:68-83. [PMID: 25560615 DOI: 10.1111/mmi.12921] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2015] [Indexed: 12/14/2022]
Abstract
Borrelia burgdorferi, the etiologic agent of Lyme disease, adapts to the mammalian hosts by differentially expressing several genes in the BosR and Rrp2-RpoN-RpoS dependent pathways, resulting in a distinct protein profile relative to that seen for survival in the Ixodes spp. tick. Previous studies indicate that a putative lipoprotein, BBA33, is produced in an RpoS-dependent manner under conditions that mimic the mammalian component of the borrelial lifecycle. However, the significance and function for BBA33 is not known. Given its linkage to the BosR/Rrp2-RpoN-RpoS regulatory cascade, we hypothesized that BBA33 facilitates B. burgdorferi infection in the mammalian host. The deletion of bba33 eliminated B. burgdorferi infectivity in C3H mice, which was rescued by genetic complementation with intact bba33. With regard to function, a combinatorial peptide approach, coupled with subsequent in vitro binding assays, indicated that BBA33 binds to collagen type VI and, to a lesser extent, collagen type IV. Whole cell binding assays demonstrated BBA33-dependent binding to human collagen type VI. Taken together, these results suggest that BBA33 interacts with collagenous structures and may function as an adhesin in a process that is required to prevent bacterial clearance.
Collapse
Affiliation(s)
- Hui Zhi
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, 77807, USA
| | | | | | | | | | | |
Collapse
|
36
|
Simple objective detection of human lyme disease infection using immuno-PCR and a single recombinant hybrid antigen. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:1094-105. [PMID: 24899074 DOI: 10.1128/cvi.00245-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A serology-based tiered approach has, to date, provided the most effective means of laboratory confirmation of clinically suspected cases of Lyme disease, but it lacks sensitivity in the early stages of disease and is often dependent on subjectively scored immunoblots. We recently demonstrated the use of immuno-PCR (iPCR) for detecting Borrelia burgdorferi antibodies in patient serum samples that were positive for Lyme disease. To better understand the performance of the Lyme disease iPCR assay, the repeatability and variability of the background of the assay across samples from a healthy population (n = 36) were analyzed. Both of these parameters were found to have coefficients of variation of <3%. Using eight antigen-specific iPCR assays and positive call thresholds established for each assay, iPCR IgM and/or IgG diagnosis from Lyme disease patient serum samples (n = 12) demonstrated a strong correlation with that of 2-tier testing. Furthermore, a simplified iPCR approach using a single hybrid antigen and detecting only IgG antibodies confirmed the 2-tier diagnosis in the Lyme disease patient serum samples (n = 12). Validation of the hybrid antigen IgG iPCR assay using a blinded panel of Lyme disease and non-Lyme disease patient serum samples (n = 92) resulted in a sensitivity of 69% (95% confidence interval [CI], 50% to 84%), compared to that of the 2-tier analysis at 59% (95% CI, 41% to 76%), and a specificity of 98% (95% CI, 91% to 100%) compared to that of the 2-tier analysis at 97% (95% CI, 88% to 100%). A single-tier hybrid antigen iPCR assay has the potential to be an improved method for detecting host-generated antibodies against B. burgdorferi.
Collapse
|
37
|
Groshong AM, Blevins JS. Insights into the biology of Borrelia burgdorferi gained through the application of molecular genetics. ADVANCES IN APPLIED MICROBIOLOGY 2014; 86:41-143. [PMID: 24377854 DOI: 10.1016/b978-0-12-800262-9.00002-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Borrelia burgdorferi, the vector-borne bacterium that causes Lyme disease, was first identified in 1982. It is known that much of the pathology associated with Lyme borreliosis is due to the spirochete's ability to infect, colonize, disseminate, and survive within the vertebrate host. Early studies aimed at defining the biological contributions of individual genes during infection and transmission were hindered by the lack of adequate tools and techniques for molecular genetic analysis of the spirochete. The development of genetic manipulation techniques, paired with elucidation and annotation of the B. burgdorferi genome sequence, has led to major advancements in our understanding of the virulence factors and the molecular events associated with Lyme disease. Since the dawn of this genetic era of Lyme research, genes required for vector or host adaptation have garnered significant attention and highlighted the central role that these components play in the enzootic cycle of this pathogen. This chapter covers the progress made in the Borrelia field since the application of mutagenesis techniques and how they have allowed researchers to begin ascribing roles to individual genes. Understanding the complex process of adaptation and survival as the spirochete cycles between the tick vector and vertebrate host will lead to the development of more effective diagnostic tools as well as identification of novel therapeutic and vaccine targets. In this chapter, the Borrelia genes are presented in the context of their general biological roles in global gene regulation, motility, cell processes, immune evasion, and colonization/dissemination.
Collapse
Affiliation(s)
- Ashley M Groshong
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jon S Blevins
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| |
Collapse
|
38
|
Identification of lysine residues in the Borrelia burgdorferi DbpA adhesin required for murine infection. Infect Immun 2014; 82:3186-98. [PMID: 24842928 DOI: 10.1128/iai.02036-14] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Decorin-binding protein A (DbpA) of Borrelia burgdorferi mediates bacterial adhesion to heparin and dermatan sulfate associated with decorin. Lysines K82, K163, and K170 of DbpA are known to be important for in vitro interaction with decorin, and the DbpA structure, initially solved by nuclear magnetic resonance (NMR) spectroscopy, suggests these lysine residues colocalize in a pocket near the C terminus of the protein. In the current study, we solved the structure of DbpA from B. burgdorferi strain 297 using X-ray crystallography and confirmed the existing NMR structural data. In vitro binding experiments confirmed that recombinant DbpA proteins with mutations in K82, K163, or K170 did not bind decorin, which was due to an inability to interact with dermatan sulfate. Most importantly, we determined that the in vitro binding defect observed upon mutation of K82, K163, or K170 in DbpA also led to a defect during infection. The infectivity of B. burgdorferi expressing individual dbpA lysine point mutants was assessed in mice challenged via needle inoculation. Murine infection studies showed that strains expressing dbpA with mutations in K82, K163, and K170 were significantly attenuated and could not be cultured from any tissue. Proper expression and cellular localization of the mutated DbpA proteins were examined, and NMR spectroscopy determined that the mutant DbpA proteins were structurally similar to wild-type DbpA. Taken together, these data showed that lysines K82, K163, and K170 potentiate the binding of DbpA to dermatan sulfate and that an interaction(s) mediated by these lysines is essential for B. burgdorferi murine infection.
Collapse
|
39
|
Brissette CA, Gaultney RA. That's my story, and I'm sticking to it--an update on B. burgdorferi adhesins. Front Cell Infect Microbiol 2014; 4:41. [PMID: 24772392 PMCID: PMC3982108 DOI: 10.3389/fcimb.2014.00041] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 03/18/2014] [Indexed: 11/25/2022] Open
Abstract
Adhesion is the initial event in the establishment of any infection. Borrelia burgdorferi, the etiological agent of Lyme disease, possesses myriad proteins termed adhesins that facilitate contact with its vertebrate hosts. B. burgdorferi adheres to host tissues through interactions with host cells and extracellular matrix, as well as other molecules present in serum and extracellular fluids. These interactions, both general and specific, are critical in the establishment of infection. Modulation of borrelial adhesion to host tissues affects the microorganisms's ability to colonize, disseminate, and persist. In this review, we update the current knowledge on structure, function, and role in pathogenesis of these “sticky” B. burgdorferi infection-associated proteins.
Collapse
Affiliation(s)
- Catherine A Brissette
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences Grand Forks, ND, USA
| | - Robert A Gaultney
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences Grand Forks, ND, USA
| |
Collapse
|
40
|
Morgan A, Wang X. The novel heparin-binding motif in decorin-binding protein A from strain B31 of Borrelia burgdorferi explains the higher binding affinity. Biochemistry 2013; 52:8237-45. [PMID: 24148022 DOI: 10.1021/bi401376u] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Decorin-binding protein A (DBPA), a glycosaminoglycan (GAG)-binding lipoprotein found in Borrelia burgdorferi, is crucial to the transmission of Lyme disease in its earliest stages. Because of its role in the initial transmission of the disease, DBPA is an ideal target for vaccine development. DBPA sequences from different strains also contain considerable heterogeneity, leading to differing affinities for GAGs and proteoglycans among different DBPA sequences. Through biophysical and structural analysis of DBPA from strain B31, we have discovered a novel and important GAG-binding epitope in B31 DBPA. Removal of the epitope greatly attenuated its affinity for DBPA and may explain the differential GAG affinities seen in DBPAs from other strains of B. burgdorferi. Paramagnetic perturbation of the protein with TEMPO-labeled heparin fragments showed bound GAGs are located close to the linker region containing the BXBB motif that plays a significant role in determining the specific affinity and orientation of binding of GAG to DBPA. Thermodynamic contributions of the new motif to GAG binding were also characterized by both nuclear magnetic resonance and isothermal titration calorimetry and compared with those of other DBPA residues previously known to be involved in GAG interactions. These analyses showed the motif is as important as other known binding epitopes. The discovery of the motif offers a possible structural explanation for the previously observed differences in GAG affinities of DBPA variants from different Borrelia strains and improves our understanding of DBPA-GAG interactions.
Collapse
Affiliation(s)
- Ashli Morgan
- Department of Chemistry & Biochemistry, Arizona State University , Tempe, Arizona 85287, United States
| | | |
Collapse
|
41
|
Dynamics of connective-tissue localization during chronic Borrelia burgdorferi infection. J Transl Med 2013; 93:900-10. [PMID: 23797360 PMCID: PMC4139070 DOI: 10.1038/labinvest.2013.81] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 05/28/2013] [Accepted: 05/29/2013] [Indexed: 12/21/2022] Open
Abstract
The etiologic agent of Lyme disease, Borrelia burgdorferi, localizes preferentially in the extracellular matrix during persistence. In chronically infected laboratory mice, there is a direct association between B. burgdorferi and the proteoglycan decorin, which suggests that decorin has a role in defining protective niches for persistent spirochetes. In this study, the tissue colocalization of B. burgdorferi with decorin and the dynamics of borrelial decorin tropism were evaluated during chronic infection. Spirochetes were found to colocalize absolutely with decorin, but not collagen I in chronically infected immunocompetent C3H mice. Passive immunization of infected C3H-scid mice with B. burgdorferi-specific immune serum resulted in the localization of spirochetes in decorin-rich microenvironments, with clearance of spirochetes from decorin-poor microenvironments. In passively immunized C3H-scid mice, tissue spirochete burdens were initially reduced, but increased over time as the B. burgdorferi-specific antibody levels waned. Concurrent repopulation of the previously cleared decorin-poor microenvironments was observed with the rising tissue spirochete burden and declining antibody titer. These findings indicate that the specificity of B. burgdorferi tissue localization during chronic infection is determined by decorin, driven by the borrelia-specific antibody response, and fluctuates with the antibody response.
Collapse
|
42
|
Coburn J, Leong J, Chaconas G. Illuminating the roles of the Borrelia burgdorferi adhesins. Trends Microbiol 2013; 21:372-9. [PMID: 23876218 DOI: 10.1016/j.tim.2013.06.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 06/13/2013] [Accepted: 06/14/2013] [Indexed: 01/13/2023]
Abstract
The Lyme disease spirochetes, Borrelia burgdorferi (sensu lato), must cause persistent, disseminated infection to be maintained in the natural enzootic cycle. In human Lyme disease, spirochetes spread from the site of a tick bite to colonize multiple tissue sites, causing multisystem clinical manifestations. The Lyme spirochetes produce many adhesive surface proteins that collectively recognize diverse host substrates and cell types and are likely to promote dissemination and chronic infection in a variety of tissues. Recent application of state-of-the-art in vivo imaging technologies is illuminating mechanisms of interaction of B. burgdorferi with the host and the importance of multiple adhesins during mammalian infection.
Collapse
Affiliation(s)
- Jenifer Coburn
- Division of Infectious Diseases, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | | | | |
Collapse
|
43
|
Russell TM, Johnson BJB. Lyme disease spirochaetes possess an aggrecan-binding protease with aggrecanase activity. Mol Microbiol 2013; 90:228-40. [PMID: 23710801 DOI: 10.1111/mmi.12276] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2013] [Indexed: 11/30/2022]
Abstract
Connective tissues are the most common area of colonization for the Lyme disease spirochaete Borrelia burgdorferi. Colonization is aided by the interaction between numerous bacterial adhesins with components of the extracellular matrix (ECM). Here we describe a novel interaction between B. burgdorferi and the major ECM proteoglycan found in joints, aggrecan. Using affinity chromatography and mass spectrometry we identify two borrelial aggrecan-binding proteins: the known ECM ligand Bgp (BB0588) and an uncharacterized protease BbHtrA (BB0104). Proteinase K studies demonstrate that BbHtrA is surface exposed. Immunoblots using sera from patients with both early and late Lyme disease establish that BbHtrA is expressed during human disease, immunogenic, and conserved in the three major Lyme disease spirochaete species. Consequences of the interaction between aggrecan and BbHtrA were examined by proteolysis assays. BbHtrA cleaves aggrecan at a site known to destroy aggrecan function and which has been previously observed in the synovial fluid of patients with Lyme arthritis. These data demonstrate that B. burgdorferi possess aggrecan-binding proteins which may provide the organism with additional capability to colonize connective tissues. Moreover, our studies provide the first evidence that B. burgdorferi possess proteolytic activity which may contribute to the pathogenesis of Lyme arthritis.
Collapse
Affiliation(s)
- Theresa M Russell
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, USA
| | | |
Collapse
|
44
|
The early dissemination defect attributed to disruption of decorin-binding proteins is abolished in chronic murine Lyme borreliosis. Infect Immun 2013; 81:1663-73. [PMID: 23460518 DOI: 10.1128/iai.01359-12] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The laboratory mouse model of Lyme disease has revealed that Borrelia burgdorferi differentially expresses numerous outer surface proteins that influence different stages of infection (tick-borne transmission, tissue colonization, dissemination, persistence, and tick acquisition). Deletion of two such outer surface proteins, decorin-binding proteins A and B (DbpA/B), has been documented to decrease infectivity, impede early dissemination, and, possibly, prevent persistence. In this study, DbpA/B-deficient spirochetes were confirmed to exhibit an early dissemination defect in immunocompetent, but not immunodeficient, mice, and the defect was found to resolve with chronicity. Development of disease (arthritis and carditis) was attenuated only in the early stage of infection with DbpA/B-deficient spirochetes in both types of mice. Persistence of the DbpA/B-deficient spirochetes occurred in both immunocompetent and immunodeficient mice in a manner indistinguishable from that of wild-type spirochetes. Dissemination through the lymphatic system was evaluated as an underlying mechanism for the early dissemination defect. At 12 h, 3 days, 7 days, and 14 days postinoculation, DbpA/B-deficient spirochetes were significantly less prevalent and in lower numbers in lymph nodes than wild-type spirochetes. However, in immunodeficient mice, deficiency of DbpA/B did not significantly decrease the prevalence or spirochete numbers in lymph nodes. Complementation of DbpA/B restored a wild-type phenotype. Thus, the results indicated that deficiency of DbpA/B allows the acquired immune response to restrict early dissemination of spirochetes, which appears to be at least partially mediated through the lymphatic system.
Collapse
|
45
|
Chen S, Birk DE. The regulatory roles of small leucine-rich proteoglycans in extracellular matrix assembly. FEBS J 2013; 280:2120-37. [PMID: 23331954 DOI: 10.1111/febs.12136] [Citation(s) in RCA: 283] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/10/2013] [Accepted: 01/15/2013] [Indexed: 12/14/2022]
Abstract
Small leucine-rich proteoglycans (SLRPs) are involved in a variety of biological and pathological processes. This review focuses on their regulatory roles in matrix assembly. SLRPs have protein cores and hypervariable glycosylation with multivalent binding abilities. During development, differential interactions of SLRPs with other molecules result in tissue-specific spatial and temporal distributions. The changing expression patterns play a critical role in the regulation of tissue-specific matrix assembly and therefore tissue function. SLRPs play significant structural roles within extracellular matrices. In addition, they play regulatory roles in collagen fibril growth, fibril organization and extracellular matrix assembly. Moreover, they are involved in mediating cell-matrix interactions. Abnormal SLRP expression and/or structures result in dysfunctional extracellular matrices and pathophysiology. Altered expression of SLRPs has been found in many disease models, and structural deficiency also causes altered matrix assembly. SLRPs regulate assembly of the extracellular matrix, which defines the microenvironment, modulating both the extracellular matrix and cellular functions, with an impact on tissue function.
Collapse
Affiliation(s)
- Shoujun Chen
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612-4799, USA
| | | |
Collapse
|
46
|
Müller KE. Damage of collagen and elastic fibres by borrelia burgdorferi - known and new clinical and histopathological aspects. Open Neurol J 2012; 6:179-86. [PMID: 23986790 PMCID: PMC3751012 DOI: 10.2174/1874205x01206010179] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 01/29/2012] [Accepted: 07/02/2012] [Indexed: 12/25/2022] Open
Abstract
Lyme Borreliosis, or Lyme's disease, manifests itself in numerous skin conditions. Therapeutic intervention should be initiated as soon as a clinical diagnosis of erythema migrans is made. The histopathology of some of the skin conditions associated with Lyme Borreliosis is characterised by structural changes to collagen, and sometimes also elastic fibres. These conditions include morphea, lichen sclerosus et atrophicus and acrodermatitis chronica atrophicans. More recently, further skin conditions have been identified by the new microscopic investigation technique of focus floating microscopy: granuloma annulare, necrobiosis lipoidica, necrobiotic xanthogranuloma, erythema annulare centrifugum, interstitial granulomatous dermatitis, cutaneous sarcoidosis and lymphocytic infiltration; these conditions also sometimes cause changes in the connective tissue. In the case of ligaments and tendons, collagen and elastic fibres predominate structurally. They are also the structures that are targeted by Borrelia. The resultant functional disorders have previously only rarely been associated with Borreliosis in clinical practice. Ligamentopathies and tendinopathies, spontaneous ruptures of tendons after slight strain, dislocation of vertebrae and an accumulation of prolapsed intervertebral discs as well as ossification of tendon insertions can be viewed in this light.
Collapse
Affiliation(s)
- Kurt E Müller
- Medical Practice for Dermatology, Venerology, Occupational Dermatology and Environmental Medicine, Kempten, Bavaria, Germany
| |
Collapse
|
47
|
Wang X. Solution structure of decorin-binding protein A from Borrelia burgdorferi. Biochemistry 2012; 51:8353-62. [PMID: 22985470 DOI: 10.1021/bi3007093] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Decorin-binding protein A (DBPA) is an important lipoprotein from the bacterium Borrelia burgdorferi, the causative agent of Lyme disease. The absence of DBPA drastically reduces the pathogenic potential of the bacterium, and biochemical evidence indicates DBPA's interactions with the glycosaminoglycan (GAG) portion of decorin are crucial to its function. We have determined the solution structure of DBPA and studied DBPA's interactions with various forms of GAGs. DBPA is determined to be a helical bundle protein consisting of five helices held together by a strong hydrophobic core. The structure also possesses a basic patch formed by portions of two helices and two flexible linkers. Low-molecular mass heparin-induced chemical shift perturbations for residues in the region as well as increases in signal intensities of select residues in their presence confirm residues in the pocket are perturbed by heparin binding. Dermatan sulfate fragments, the dominant GAG type found on decorin, were shown to have lower affinity than heparin but are still capable of binding DBPA.
Collapse
Affiliation(s)
- Xu Wang
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
48
|
Gesslbauer B, Poljak A, Handwerker C, Schüler W, Schwendenwein D, Weber C, Lundberg U, Meinke A, Kungl AJ. Comparative membrane proteome analysis of three Borrelia species. Proteomics 2012; 12:845-58. [PMID: 22539435 DOI: 10.1002/pmic.201100211] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The versatility of the surface of Borrelia, the causative agent of Lyme borreliosis, is very important in host-pathogen interactions allowing bacteria to survive in ticks and to persist in a mammalian environment. To identify the surface proteome of Borrelia, we have performed a large comparative proteomic analysis on the three most important pathogenic Borrelia species, namely B. burgdorferi (strain B31), B. afzelii (strain K78), and B. garinii (strain PBi). Isolation of membrane proteins was performed by using three different approaches: (i) a detergent-based fractionation of outer membrane proteins; (ii) a trypsin-based partial shedding of outer cell surface proteins; (iii) biotinylation of membrane proteins and preparation of the biotin-labelled fraction using streptavidin. Proteins derived from the detergent-based fractionation were further sub-fractionated by heparin affinity chromatography since heparin-like molecules play an important role for microbial entry into human cells. All isolated proteins were analysed using either a gel-based liquid chromatography (LC)-MS/MS technique or by two-dimensional (2D)-LC-MS/MS resulting in the identification of 286 unique proteins. Ninety seven of these were found in all three Borrelia species, representing potential targets for a broad coverage vaccine for the prevention of Lyme borreliosis caused by the different Borrelia species.
Collapse
|
49
|
Chan K, Awan M, Barthold SW, Parveen N. Comparative molecular analyses of Borrelia burgdorferi sensu stricto strains B31 and N40D10/E9 and determination of their pathogenicity. BMC Microbiol 2012; 12:157. [PMID: 22846633 PMCID: PMC3511255 DOI: 10.1186/1471-2180-12-157] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 06/11/2012] [Indexed: 11/24/2022] Open
Abstract
Background Lyme disease in the United States is caused primarily by B. burgdorferi sensu stricto while other species are also prevalent in Europe. Genetic techniques have identified several chromosomal and plasmid-borne regulatory and virulence factors involved in Lyme pathogenesis. B31 and N40 are two widely studied strains of B. burgdorferi, which belong to two different 16 S-23 S rRNA spacer types (RST) and outer surface protein C (OspC) allelic groups. However, the presence of several known virulence factors in N40 has not been investigated. This is the first comprehensive study that compared these two strains both in vitro and using the mouse model of infection. Results Phylogenetic analyses predict B31 to be more infectious. However, our studies here indicate that N40D10/E9 is more infectious than the B31 strain at lower doses of inoculation in the susceptible C3H mice. Based-upon a careful analyses of known adhesins of these strains, it is predicted that the absence of a known fibronectin-glycosaminoglycan binding adhesin, bbk32, in the N40 strain could at least partially be responsible for reduction in its binding to Vero cells in vitro. Nevertheless, this difference does not affect the infectivity of N40D10/E9 strain. The genes encoding known regulatory and virulence factors critical for pathogenesis were detected in both strains. Differences in the protein profiles of these B. burgdorferi strains in vitro suggest that the novel, differentially expressed molecules may affect infectivity of B. burgdorferi. Further exacerbation of these molecular differences in vivo could affect the pathogenesis of spirochete strains. Conclusion Based upon the studies here, it can be predicted that N40D10/E9 disseminated infection at lower doses may be enhanced by its lower binding to epithelial cells at the site of inoculation due to the absence of BBK32. We suggest that complete molecular analyses of virulence factors followed by their evaluation using the mouse infection model should form the basis of determining infectivity and pathogenicity of different strains rather than simple phylogenetic group analyses. This study further emphasizes a need to investigate multiple invasive strains of B. burgdorferi to fully appreciate the pathogenic mechanisms that contribute to Lyme disease manifestations.
Collapse
Affiliation(s)
- Kamfai Chan
- Department of Microbiology and Molecular Genetics, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, 225 Warren Street, Newark, NJ 07103-3535, USA
| | | | | | | |
Collapse
|
50
|
Ristow LC, Miller HE, Padmore LJ, Chettri R, Salzman N, Caimano MJ, Rosa PA, Coburn J. The β₃-integrin ligand of Borrelia burgdorferi is critical for infection of mice but not ticks. Mol Microbiol 2012; 85:1105-18. [PMID: 22758390 DOI: 10.1111/j.1365-2958.2012.08160.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
P66 is a Borrelia burgdorferi surface protein with β₃ integrin binding and channel forming activities. In this study, the role of P66 in mammalian and tick infection was examined. B. burgdorferiΔp66 strains were not infectious in wild-type, TLR2⁻/⁻- or MyD88⁻/⁻-deficient mice. Strains with p66 restored to the chromosome restored near wild-type infectivity, while complementation with p66 on a shuttle vector did not restore infectivity. Δp66 mutants are cleared quickly from the site of inoculation, but analyses of cytokine expression and cellular infiltrates at the site of inoculation did not reveal a specific mechanism of clearance. The defect in these mutants cannot be attributed to nutrient limitation or an inability to adapt to the host environment in vivo as Δp66 bacteria were able to survive as well as wild type in dialysis membrane chambers in the rat peritoneum. Δp66 bacteria were able to survive in ticks through the larva to nymph moult, but were non-infectious in mice when delivered by tick bite. Independent lines of evidence do not support any increased susceptibility of the Δp66 strains to factors in mammalian blood. This study is the first to define a B. burgdorferi adhesin as essential for mammalian, but not tick infection.
Collapse
Affiliation(s)
- Laura C Ristow
- Graduate Program in Microbiology, Immunology, and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | | | | | | | | | | |
Collapse
|