1
|
Alcón-Chino MET, Bonoldi VLN, Pereira RMR, Gazeta GS, Carvalho JPRS, Napoleão-Pêgo P, Durans AM, Souza ALA, De-Simone SG. New Epitopes for the Serodiagnosis of Human Borreliosis. Microorganisms 2024; 12:2212. [PMID: 39597601 PMCID: PMC11596413 DOI: 10.3390/microorganisms12112212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Lyme disease, a zoonotic infection caused by the bacterium Borrelia burgdorferi, is transmitted to humans through the bites of infected ticks. Its diagnosis primarily relies on serological methods; however, the existing borreliosis techniques have shown a variable sensitivity and specificity. Our study aimed to map IgG epitopes from five outer membrane proteins (Omp) from B. burgdorferi [Filament flagellar 41kD (PI1089), flagellar hook-associated protein (Q44767), Flagellar hook k2 protein (O51173), Putative Omp BURGA03 (Q44849), and 31 kDa OspA (P0CL66)] lipoprotein to find specific epitopes for the development of accurate diagnosis methods. Using the spot synthesis technique, a library of 380 peptides was constructed to identify linear B cell epitopes recognized by human IgG in response to specific B. burgdorferi-associated proteins. The reactivity of this epitope when chemically synthesized was then evaluated using ELISA with a panel of the patient's sera. Cross-reactivity was assessed through data bank access and in vitro analysis. Among the 19 epitopes identified, four were selected for further investigation based on their signal intensity, secondary structure, and peptide matching. Validation was performed using ELISA, and ROC curve analysis demonstrated a sensitivity of ≥85.71%, specificity of ≥92.31, accuracy of ≥90.7, and AUC value of ≥0.91 for all peptides. Our cross-reactivity analysis demonstrated that the Burg/02/huG, Burg/03/huG, and Burg/12/huG peptides were not reactive to antibodies from patients with Leptospirosis and syphilis compared to those from the B. burgdorferi group. These peptides indicated an excellent performance in distinguishing between B. burgdorferi-infected and non-infected individuals and exhibited a neglected reactivity to antibodies in sera from patients with Leptospirosis and syphilis. These peptides are promising targets for recombinant development, potentially leading to more accurate serological tests and vaccines.
Collapse
Affiliation(s)
- Mônica E. T. Alcón-Chino
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), FIOCRUZ, Rio de Janeiro 21040-900, RJ, Brazil; (M.E.T.A.-C.); (J.P.R.S.C.); (P.N.-P.); (A.M.D.)
- Post-Graduation Program in Science and Biotechnology, Department of Molecular and Cellular Biology, Biology Institute, Federal Fluminense University, Niterói 22040-036, RJ, Brazil
| | - Virgínia L. N. Bonoldi
- Clinical Hospital, Faculty of Medicine, São Paulo University, São Paulo 05403-000, SP, Brazil;
| | - Rosa M. R. Pereira
- Faculty of Medicine, São Paulo University, São Paulo 01246-903, SP, Brazil;
| | - Gilberto S. Gazeta
- Laboratory of Ticks and Other Wingless Arthropods-National Reference for Vectors of Rickettsioses, Instituto Oswaldo Cruz-IOC, FIOCRUZ, Rio de Janeiro 21041-250, RJ, Brazil;
| | - João P. R. S. Carvalho
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), FIOCRUZ, Rio de Janeiro 21040-900, RJ, Brazil; (M.E.T.A.-C.); (J.P.R.S.C.); (P.N.-P.); (A.M.D.)
- Post-Graduation Program in Science and Biotechnology, Department of Molecular and Cellular Biology, Biology Institute, Federal Fluminense University, Niterói 22040-036, RJ, Brazil
| | - Paloma Napoleão-Pêgo
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), FIOCRUZ, Rio de Janeiro 21040-900, RJ, Brazil; (M.E.T.A.-C.); (J.P.R.S.C.); (P.N.-P.); (A.M.D.)
| | - Andressa M. Durans
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), FIOCRUZ, Rio de Janeiro 21040-900, RJ, Brazil; (M.E.T.A.-C.); (J.P.R.S.C.); (P.N.-P.); (A.M.D.)
| | - André L. A. Souza
- Multidisciplinary Biochemistry Teaching Laboratory, UNIG, Nova Iguaçu 26260-045, RJ, Brazil;
| | - Salvatore G. De-Simone
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), FIOCRUZ, Rio de Janeiro 21040-900, RJ, Brazil; (M.E.T.A.-C.); (J.P.R.S.C.); (P.N.-P.); (A.M.D.)
- Post-Graduation Program in Science and Biotechnology, Department of Molecular and Cellular Biology, Biology Institute, Federal Fluminense University, Niterói 22040-036, RJ, Brazil
- Laboratory of Epidemiology and Molecular Systematics, Oswaldo Cruz Institut, FIOCRUZ, Rio de Janeiro 21040-900, RJ, Brazil
| |
Collapse
|
2
|
Thomas S, Schulz AM, Leong JM, Zeczycki TN, Garcia BL. The molecular determinants of classical pathway complement inhibition by OspEF-related proteins of Borrelia burgdorferi. J Biol Chem 2024; 300:107236. [PMID: 38552741 PMCID: PMC11066524 DOI: 10.1016/j.jbc.2024.107236] [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: 01/26/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/04/2024] Open
Abstract
The complement system serves as the first line of defense against invading pathogens by promoting opsonophagocytosis and bacteriolysis. Antibody-dependent activation of complement occurs through the classical pathway and relies on the activity of initiating complement proteases of the C1 complex, C1r and C1s. The causative agent of Lyme disease, Borrelia burgdorferi, expresses two paralogous outer surface lipoproteins of the OspEF-related protein family, ElpB and ElpQ, that act as specific inhibitors of classical pathway activation. We have previously shown that ElpB and ElpQ bind directly to C1r and C1s with high affinity and specifically inhibit C2 and C4 cleavage by C1s. To further understand how these novel protease inhibitors function, we carried out a series of hydrogen-deuterium exchange mass spectrometry (HDX-MS) experiments using ElpQ and full-length activated C1s as a model of Elp-protease interaction. Comparison of HDX-MS profiles between unbound ElpQ and the ElpQ/C1s complex revealed a putative C1s-binding site on ElpQ. HDX-MS-guided, site-directed ElpQ mutants were generated and tested for direct binding to C1r and C1s using surface plasmon resonance. Several residues within the C-terminal region of ElpQ were identified as important for protease binding, including a single conserved tyrosine residue that was required for ElpQ- and ElpB-mediated complement inhibition. Collectively, our study identifies key molecular determinants for classical pathway protease recognition by Elp proteins. This investigation improves our understanding of the unique complement inhibitory mechanism employed by Elp proteins which serve as part of a sophisticated complement evasion system present in Lyme disease spirochetes.
Collapse
Affiliation(s)
- Sheila Thomas
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Anna M Schulz
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - John M Leong
- Department of Molecular Biology and Microbiology, Tufts School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - Tonya N Zeczycki
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Brandon L Garcia
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.
| |
Collapse
|
3
|
Koloski CW, Hurry G, Foley-Eby A, Adam H, Goldstein S, Zvionow P, Detmer SE, Voordouw MJ. Male C57BL/6J mice have higher presence and abundance of Borrelia burgdorferi in their ventral skin compared to female mice. Ticks Tick Borne Dis 2024; 15:102308. [PMID: 38215632 DOI: 10.1016/j.ttbdis.2024.102308] [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: 09/18/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/14/2024]
Abstract
Borrelia burgdorferi is a tick-borne spirochete that causes Lyme disease in humans. The host immune system controls the abundance of the spirochete in the host tissues. Recent work with immunocompetent Mus musculus mice strain C3H/HeJ found that males had a higher tissue infection prevalence and spirochete load compared to females. The purpose of this study was to determine whether host sex and acquired immunity interact to influence the prevalence and abundance of spirochetes in the tissues of the commonly used mouse strain C57BL/6. Wildtype (WT) mice and their SCID counterparts (C57BL/6) were experimentally infected with B. burgdorferi via tick bite. Ear biopsies were sampled at weeks 4, 8, and 12 post-infection (PI) and five tissues (left ear, ventral skin, heart, tibiotarsal joint of left hind leg, and liver) were collected at necropsy (16 weeks PI). The mean spirochete load in the tissues of the SCID mice was 260.4x higher compared to the WT mice. In WT mice, the infection prevalence in the ventral skin was significantly higher in males (40.0 %) compared to females (0.0 %), and the spirochete load in the rear tibiotarsal joint was significantly higher (4.3x) in males compared to females. In SCID mice, the spirochete load in the ventral skin was 200.0x higher in males compared to females, but there were no significant sex-specific difference in spirochete load in the other tissues (left ear, heart, tibiotarsal joint, or liver). Thus, the absence of acquired immunity greatly amplified the spirochete load in the ventral skin of male mice. It is important to note that the observed sex-specific differences in laboratory mice cannot be extrapolated to humans. Future studies should investigate the mechanisms underlying the male bias in the abundance of B. burgdorferi in the mouse skin.
Collapse
Affiliation(s)
- Cody W Koloski
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Georgia Hurry
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Alexandra Foley-Eby
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Hesham Adam
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Savannah Goldstein
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Pini Zvionow
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Susan E Detmer
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Maarten J Voordouw
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada.
| |
Collapse
|
4
|
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: 0] [Impact Index Per Article: 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
|
5
|
Grąźlewska W, Holec-Gąsior L. Antibody Cross-Reactivity in Serodiagnosis of Lyme Disease. Antibodies (Basel) 2023; 12:63. [PMID: 37873860 PMCID: PMC10594444 DOI: 10.3390/antib12040063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/25/2023] Open
Abstract
Lyme disease is a tick-borne disease caused by spirochetes belonging to the Borrelia burgdorferi sensu lato complex. The disease is characterized by a varied course; therefore, the basis for diagnosis is laboratory methods. Currently, a two-tiered serological test is recommended, using an ELISA as a screening test and a Western blot as a confirmatory test. This approach was introduced due to the relatively high number of false-positive results obtained when using an ELISA alone. However, even this approach has not entirely solved the problem of false-positive results caused by cross-reactive antibodies. Many highly immunogenic B. burgdorferi s.l. proteins are recognized nonspecifically by antibodies directed against other pathogens. This also applies to antigens, such as OspC, BmpA, VlsE, and FlaB, i.e., those commonly used in serodiagnostic assays. Cross-reactions can be caused by both bacterial (relapsing fever Borrelia, Treponema pallidum) and viral (Epstein-Baar virus, Cytomegalovirus) infections. Additionally, a rheumatoid factor has also been shown to nonspecifically recognize B. burgdorferi s.l. proteins, resulting in false-positive results. Therefore, it is necessary to carefully interpret the results of serodiagnostic tests so as to avoid overdiagnosis of Lyme disease, which causes unnecessary implementations of strong antibiotic therapies and delays in the correct diagnosis.
Collapse
Affiliation(s)
| | - Lucyna Holec-Gąsior
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland;
| |
Collapse
|
6
|
Genné D, Jiricka W, Sarr A, Voordouw MJ. Tick-to-host transmission differs between Borrelia afzelii strains. Microbiol Spectr 2023; 11:e0167523. [PMID: 37676027 PMCID: PMC10580945 DOI: 10.1128/spectrum.01675-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/03/2023] [Indexed: 09/08/2023] Open
Abstract
Many vector-borne pathogens establish multiple-strain infections in the vertebrate host and the arthropod vector. Multiple-strain infections in the host influence strain acquisition by naive vectors. Whether multiple-strain infections in the vector influence strain-specific transmission to naive hosts remains unknown. The spirochete, Borrelia afzelii, causes Lyme borreliosis and multiple-strain infections are common in both the tick vector and vertebrate host. Our study used two B. afzelii strains: Fin-Jyv-A3 and NE4049. Donor mice were infected with Fin-Jyv-A3 alone, NE4049 alone, or with both strains. Larval ticks fed on donor mice and molted into nymphal ticks infected with either strain or both strains. These nymphs were fed on test mice to determine whether multiple-strain infections in the nymph influence nymph-to-host transmission (NHT). Multiple-strain infection in the donor mice reduced the acquisition of both strains by ticks by 23%. Thus, a substantial fraction of infected nymphs from the multiple strain treatment were infected with the "wrong" competitor strain rather than the "right" focal strain. As a result, nymphs from the multiple strain treatment were 46% less likely to infect the test mice with the focal strain compared to nymphs from the single strain treatment. However, multiple-strain infection in the nymphal tick had no effect on the NHT of either strain. The nymphal spirochete load of Fin-Jyv-A3 was 1.9 times higher compared to NE4049. NHT of Fin-Jyv-A3 (79%) was 1.5 times higher compared to NE4049 (53%). Our study suggests that B. afzelii strains with higher nymphal spirochete loads have higher NHT. IMPORTANCE For many vector-borne pathogens, multiple-strain infections in the vertebrate host or arthropod vector are common. Multiple-strain infections in the host reduce strain acquisition by feeding vectors. Whether multiple-strain infections in the vector influence strain transmission to the host remains unknown. In our study, we used two strains of the tick-borne spirochete Borrelia afzelii, which causes Lyme borreliosis, to investigate whether multiple-strain infections in the nymphal tick influenced nymph-to-host transmission (NHT) of strains. Multiple-strain infections in mice reduced the acquisition of both B. afzelii strains by nymphal ticks. As a result, nymphs from the multiple strain treatment were less likely to infect naive test mice with the focal strain. Multiple-strain infection in the nymphal ticks did not influence the NHT of either strain. The strain with the higher bacterial abundance in the nymph had higher NHT. Our study suggests that pathogen abundance in the arthropod vector is important for vector-to-host transmission.
Collapse
Affiliation(s)
- Dolores Genné
- Laboratory of Ecology and Evolution of Parasites, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | - Whitney Jiricka
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Anouk Sarr
- Laboratory of Ecology and Evolution of Parasites, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | - Maarten J. Voordouw
- Laboratory of Ecology and Evolution of Parasites, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
7
|
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: 0] [Impact Index Per Article: 0] [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
|
8
|
Willems R, Verhaeghe N, Perronne C, Borgermans L, Annemans L. Cost of illness in patients with post-treatment Lyme disease syndrome in Belgium. Eur J Public Health 2023; 33:668-674. [PMID: 36972275 PMCID: PMC10393486 DOI: 10.1093/eurpub/ckad045] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND A proportion of patients with Lyme borreliosis (LB) report long-term persisting signs and symptoms, even after recommended antibiotic treatment, which is termed post-treatment Lyme disease syndrome (PTLDS). Consensus on guidance regarding diagnosis and treatment is currently lacking. Consequently, patients suffer and are left searching for answers, negatively impacting their quality of life and healthcare expenditure. Yet, health economic data on PTLDS remain scarce. The aim of this article is therefore to assess the cost-of-illness related to PTLDS, including the patient perspective. METHODS PTLDS patients (N = 187) with confirmed diagnosis of LB were recruited by a patient organization. Patients completed a self-reported questionnaire on LB-related healthcare utilization, absence from work and unemployment. Unit costs (reference year 2018) were obtained from national databases and published literature. Mean costs and uncertainty intervals were calculated via bootstrapping. Data were extrapolated to the Belgian population. Generalized linear models were used to determine associated covariates with total direct costs and out-of-pocket expenditures. RESULTS Mean annual direct costs amounted to €4618 (95% CI €4070-5152), of which 49.5% were out-of-pocket expenditures. Mean annual indirect costs amounted to €36 081 (€31 312-40 923). Direct and indirect costs at the population level were estimated at €19.4 and 151.5 million, respectively. A sickness or disability benefit as source of income was associated with higher direct and out-of-pocket costs. CONCLUSIONS The economic burden associated with PTLDS on patients and society is substantial, with patients consuming large amounts of non-reimbursed healthcare resources. Guidance on adequate diagnosis and treatment of PTLDS is needed.
Collapse
Affiliation(s)
- Ruben Willems
- Department of Public Health and Primary Care, Interuniversity Centre for Health Economics Research (I-CHER), Ghent University, Gent, Belgium
| | - Nick Verhaeghe
- Department of Public Health and Primary Care, Interuniversity Centre for Health Economics Research (I-CHER), Ghent University, Gent, Belgium
| | - Christian Perronne
- Infectious Diseases Department, University Hospital Raymond Poincaré, APHP, Université de Versailles Saint-Quentin-Paris Saclay, Garches, France
| | - Liesbeth Borgermans
- Department of Public Health and Primary Care, Interuniversity Centre for Health Economics Research (I-CHER), Ghent University, Gent, Belgium
| | - Lieven Annemans
- Department of Public Health and Primary Care, Interuniversity Centre for Health Economics Research (I-CHER), Ghent University, Gent, Belgium
| |
Collapse
|
9
|
Host Cell Binding Mediated by Leptospira interrogans Adhesins. Int J Mol Sci 2022; 23:ijms232415550. [PMID: 36555188 PMCID: PMC9779477 DOI: 10.3390/ijms232415550] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Leptospirosis is a neglected infectious disease with global impact on both humans and animals. The increase in urban development without sanitation planning is one of the main reasons for the disease spreading. The symptoms are similar to those of flu-like diseases, such as dengue, yellow fever, and malaria, which can result in a misleading clinical diagnosis. The characterization of host-pathogen interactions is important in the development of new vaccines, treatments, and diagnostics. However, the pathogenesis of leptospirosis is not well understood, and many gaps remain to be addressed. Here, we aimed to determine if Leptospira strains, virulent, culture-attenuated, and saprophytic, and the major outer membrane proteins OmpL37, OmpL1, LipL21, LipL41, and LipL46 are able to adhere to different endothelial, epithelial and fibroblast cell lines in vitro. We showed that virulent leptospires robustly bind to all cells compared to the culture-attenuated and saprophytic lines. The recombinant proteins exhibited certain adhesion, but only OmpL1 and LipL41 were able to bind to several cell lines, either in monolayer or in cell suspension. Blocking OmpL1 with polyclonal antibodies caused a decrease in bacterial binding to cells, contrasting with an increase observed when anti-LipL41 antibodies were used. The adhesion of OmpL1 to HMEC-1 and EA.hy926 was inhibited when cells were pre-incubated with collagen IV, suggesting that both compete for the same cell receptor. We present here for the first time the interaction of five leptospiral outer membrane proteins with several cell lines, and we conclude that LipL41 and OmpL1 may have an impact on leptospiral adhesion to mammalian cells and may mediate the colonization process in leptospiral pathogenesis.
Collapse
|
10
|
Curtis MW, Fierros CH, Hahn BL, Surdel MC, Kessler J, Anderson PN, Vandewalle-Capo M, Bonde M, Zhu J, Bergström S, Coburn J. Identification of amino acid domains of Borrelia burgdorferi P66 that are surface exposed and important for localization, oligomerization, and porin function of the protein. Front Cell Infect Microbiol 2022; 12:991689. [PMID: 36211976 PMCID: PMC9539438 DOI: 10.3389/fcimb.2022.991689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/31/2022] [Indexed: 12/01/2022] Open
Abstract
P66, a bifunctional integral outer membrane protein, is necessary for Borrelia burgdorferi to establish initial infection and to disseminate in mice. The integrin binding function of P66 facilitates extravasation and dissemination, but the role of its porin function during murine infection has not been investigated. A limitation to studying P66 porin function during mammalian infection has been the lack of structural information for P66. In this study, we experimentally characterized specific domains of P66 with regard to structure and function. First, we aligned the amino acid sequences of P66 from Lyme disease-causing Borrelia and relapsing fever-causing Borrelia to identify conserved and unique domains between these disease-causing clades. Then, we examined whether specific domains of P66 are exposed on the surface of the bacteria by introducing c-Myc epitope tags into each domain of interest. The c-Myc epitope tag inserted C-terminally to E33 (highly conserved domain), to T187 (integrin binding region domain and a non-conserved domain), and to E334 (non-conserved domain) were all detected on the surface of Borrelia burgdorferi. The c-Myc epitope tag inserted C-terminally to E33 and D303 in conserved domains disrupted P66 oligomerization and porin function. In a murine model of infection, the E33 and D303 mutants exhibited decreased infectivity and dissemination. Taken together, these results suggest the importance of these conserved domains, and potentially P66 porin function, in vivo.
Collapse
Affiliation(s)
- Michael W. Curtis
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Christa H. Fierros
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Beth L. Hahn
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Matthew C. Surdel
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Julie Kessler
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Phillip N. Anderson
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Marine Vandewalle-Capo
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
| | - Mari Bonde
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Jieqing Zhu
- Blood Research Institute, Versiti, Milwaukee, WI, United States
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sven Bergström
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
| | - Jenifer Coburn
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
11
|
Lin YP, Tufts DM, Combs M, Dupuis AP, Marcinkiewicz AL, Hirsbrunner AD, Diaz AJ, Stout JL, Blom AM, Strle K, Davis AD, Kramer LD, Kolokotronis SO, Diuk-Wasser MA. Cellular and immunological mechanisms influence host-adapted phenotypes in a vector-borne microparasite. Proc Biol Sci 2022; 289:20212087. [PMID: 35193398 PMCID: PMC8864362 DOI: 10.1098/rspb.2021.2087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/18/2022] [Indexed: 01/15/2023] Open
Abstract
Predicting pathogen emergence and spillover risk requires understanding the determinants of a pathogens' host range and the traits involved in host competence. While host competence is often considered a fixed species-specific trait, it may be variable if pathogens diversify across hosts. Balancing selection can lead to maintenance of pathogen polymorphisms (multiple-niche-polymorphism; MNP). The causative agent of Lyme disease, Borrelia burgdorferi (Bb), provides a model to study the evolution of host adaptation, as some Bb strains defined by their outer surface protein C (ospC) genotype, are widespread in white-footed mice and others are associated with non-rodent vertebrates (e.g. birds). To identify the mechanisms underlying potential strain × host adaptation, we infected American robins and white-footed mice, with three Bb strains of different ospC genotypes. Bb burdens varied by strain in a host-dependent fashion, and strain persistence in hosts largely corresponded to Bb survival at early infection stages and with transmission to larvae (i.e. fitness). Early survival phenotypes are associated with cell adhesion, complement evasion and/or inflammatory and antibody-mediated removal of Bb, suggesting directional selective pressure for host adaptation and the potential role of MNP in maintaining OspC diversity. Our findings will guide future investigations to inform eco-evolutionary models of host adaptation for microparasites.
Collapse
Affiliation(s)
- Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
- Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA
| | - Danielle M. Tufts
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, NY, USA
- Infectious Diseases and Microbiology Department, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew Combs
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, NY, USA
| | - Alan P. Dupuis
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
| | | | | | - Alexander J. Diaz
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
| | - Jessica L. Stout
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
| | - Anna M. Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmo, Sweden
| | - Klemen Strle
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
- Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA
| | - April D. Davis
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
| | - Laura D. Kramer
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
- Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA
| | - Sergios-Orestis Kolokotronis
- Department of Epidemiology and Biostatistics, School of Public Health, College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Institute for Genomic Health, College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Division of Infectious Diseases, Department of Medicine, College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Maria A. Diuk-Wasser
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, NY, USA
| |
Collapse
|
12
|
Nanomechanical mechanisms of Lyme disease spirochete motility enhancement in extracellular matrix. Commun Biol 2021; 4:268. [PMID: 33649506 PMCID: PMC7921401 DOI: 10.1038/s42003-021-01783-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 02/01/2021] [Indexed: 01/31/2023] Open
Abstract
As opposed to pathogens passively circulating in the body fluids of their host, pathogenic species within the Spirochetes phylum are able to actively coordinate their movement in the host to cause systemic infections. Based on the unique morphology and high motility of spirochetes, we hypothesized that their surface adhesive molecules might be suitably adapted to aid in their dissemination strategies. Designing a system that mimics natural environmental signals, which many spirochetes face during their infectious cycle, we observed that a subset of their surface proteins, particularly Decorin binding protein (Dbp) A/B, can strongly enhance the motility of spirochetes in the extracellular matrix of the host. Using single-molecule force spectroscopy, we disentangled the mechanistic details of DbpA/B and decorin/laminin interactions. Our results show that spirochetes are able to leverage a wide variety of adhesion strategies through force-tuning transient molecular binding to extracellular matrix components, which concertedly enhance spirochetal dissemination through the host.
Collapse
|
13
|
Pietikäinen A, Åstrand M, Cuellar J, Glader O, Elovaara H, Rouhiainen M, Salo J, Furihata T, Salminen TA, Hytönen J. Conserved lysine residues in decorin binding proteins of Borrelia garinii are critical in adhesion to human brain microvascular endothelial cells. Mol Microbiol 2021; 115:1395-1409. [PMID: 33512032 DOI: 10.1111/mmi.14687] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 11/28/2022]
Abstract
Lyme borreliosis is a tick-borne disease caused by Borrelia burgdorferi sensu lato spirochetes (Lyme borreliae). When the disease affects the central nervous system, it is referred to as neuroborreliosis. In Europe, neuroborreliosis is most often caused by Borrelia garinii. Although it is known that in the host Lyme borreliae spread from the tick bite site to distant tissues via the blood vasculature, the adherence of Lyme borreliae to human brain microvascular endothelial cells has not been studied before. Decorin binding proteins are adhesins expressed on Lyme borreliae. They mediate the adhesion of Lyme borreliae to decorin and biglycan, and the lysine residues located in the binding site of decorin binding proteins are important to the binding activity. In this study, we show that lysine residues located in the canonical binding site can also be found in decorin binding proteins of Borrelia garinii, and that these lysines contribute to biglycan and decorin binding. Most importantly, we show that the lysine residues are crucial for the binding of Lyme borreliae to decorin and biglycan expressing human brain microvascular endothelial cells, which in turn suggests that they are involved in the pathogenesis of neuroborreliosis.
Collapse
Affiliation(s)
- Annukka Pietikäinen
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland.,Laboratory Division, Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Mia Åstrand
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,National Doctoral Programme in Informational and Structural Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Julia Cuellar
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Otto Glader
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland.,Doctoral Programme in Clinical Research, Turku, Finland
| | - Heli Elovaara
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Meri Rouhiainen
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland.,Doctoral Programme in Clinical Research, Turku, Finland
| | - Jemiina Salo
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Tomomi Furihata
- Laboratory of Clinical Pharmacy and Experimental Therapeutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Tiina A Salminen
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Jukka Hytönen
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland.,Laboratory Division, Clinical Microbiology, Turku University Hospital, Turku, Finland
| |
Collapse
|
14
|
Coburn J, Garcia B, Hu LT, Jewett MW, Kraiczy P, Norris SJ, Skare J. Lyme Disease Pathogenesis. Curr Issues Mol Biol 2020; 42:473-518. [PMID: 33353871 DOI: 10.21775/cimb.042.473] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lyme disease Borrelia are obligately parasitic, tick- transmitted, invasive, persistent bacterial pathogens that cause disease in humans and non-reservoir vertebrates primarily through the induction of inflammation. During transmission from the infected tick, the bacteria undergo significant changes in gene expression, resulting in adaptation to the mammalian environment. The organisms multiply and spread locally and induce inflammatory responses that, in humans, result in clinical signs and symptoms. Borrelia virulence involves a multiplicity of mechanisms for dissemination and colonization of multiple tissues and evasion of host immune responses. Most of the tissue damage, which is seen in non-reservoir hosts, appears to result from host inflammatory reactions, despite the low numbers of bacteria in affected sites. This host response to the Lyme disease Borrelia can cause neurologic, cardiovascular, arthritic, and dermatologic manifestations during the disseminated and persistent stages of infection. The mechanisms by which a paucity of organisms (in comparison to many other infectious diseases) can cause varied and in some cases profound inflammation and symptoms remains mysterious but are the subjects of diverse ongoing investigations. In this review, we provide an overview of virulence mechanisms and determinants for which roles have been demonstrated in vivo, primarily in mouse models of infection.
Collapse
Affiliation(s)
- Jenifer Coburn
- Center For Infectious Disease Research, Medical College of Wisconsin, 8701 Watertown Plank Rd., TBRC C3980, Milwaukee, WI 53226, USA
| | - Brandon Garcia
- Department of Microbiology and Immunology, East Carolina University, Brody School of Medicine, Greenville, NC 27858, USA
| | - Linden T Hu
- Department of Molecular Biology and Microbiology, Vice Dean of Research, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA 02111, USA
| | - Mollie W Jewett
- Immunity and Pathogenesis Division Head, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 6900 Lake Nona Blvd. Orlando, FL 32827, USA
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital Frankfurt, Goethe University Frankfurt, Paul-Ehrlich-Str. 40, 60596 Frankfurt, Germany
| | - Steven J Norris
- Department of Pathology and Laboratory Medicine, University of Texas Medical School at Houston, P.O. Box 20708, Houston, TX 77225, USA
| | - Jon Skare
- Professor and Associate Head, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX 77807, USA
| |
Collapse
|
15
|
Primus S, Rocha SC, Giacani L, Parveen N. Identification and Functional Assessment of the First Placental Adhesin of Treponema pallidum That May Play Critical Role in Congenital Syphilis. Front Microbiol 2020; 11:621654. [PMID: 33408711 PMCID: PMC7779807 DOI: 10.3389/fmicb.2020.621654] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/27/2020] [Indexed: 11/25/2022] Open
Abstract
Syphilis is a global, re-emerging sexually transmitted infection and congenital syphilis remains a major cause of adverse pregnancy outcomes due to bacterial infection in developing nations with a high rate of fetus loss. The molecular mechanisms involved in pathogenesis of the causative agent, Treponema pallidum subsp. pallidum remain poorly understood due to the difficulties of working with this pathogen, including the inability to grow it in pure culture. To reduce the spread of syphilis, we must first increase our knowledge of the virulence factors of T. pallidum and their contribution to syphilis manifestations. Tp0954 was predicted to be a surface lipoprotein of T. pallidum. Therefore, we experimentally demonstrated that Tp0954 is indeed a surface protein and further investigated its role in mediating bacterial attachment to various mammalian host cells. We found that expression of Tp0954 in a poorly adherent, but physiologically related derivative strain of the Lyme disease causing spirochete Borrelia burgdorferi B314 strain promotes its binding to epithelial as well as non-epithelial cells including glioma and placental cell lines. We also found that Tp0954 expression facilitates binding of this strain to purified dermatan sulfate and heparin, and also that bacterial binding to mammalian cell lines is mediated by the presence of heparan sulfate and dermatan sulfate in the extracellular matrix of the specific cell lines. These results suggest that Tp0954 may be involved not only in initiating T. pallidum infection by colonizing skin epithelium, but it may also contribute to disseminated infection and colonization of distal tissues. Significantly, we found that Tp0954 promotes binding to the human placental choriocarcinoma BeWo cell line, which is of trophoblastic endocrine cell type, as well as human placental tissue sections, suggesting its role in placental colonization and possible contribution to transplacental transmission of T. pallidum. Altogether, these novel findings offer an important step toward unraveling syphilis pathogenesis, including placental colonization and T. pallidum vertical transmission from mother to fetus during pregnancy.
Collapse
Affiliation(s)
- Shekerah Primus
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Sandra C Rocha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Lorenzo Giacani
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Nikhat Parveen
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
16
|
Lin YP, Tan X, Caine JA, Castellanos M, Chaconas G, Coburn J, Leong JM. Strain-specific joint invasion and colonization by Lyme disease spirochetes is promoted by outer surface protein C. PLoS Pathog 2020; 16:e1008516. [PMID: 32413091 PMCID: PMC7255614 DOI: 10.1371/journal.ppat.1008516] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/28/2020] [Accepted: 04/03/2020] [Indexed: 01/06/2023] Open
Abstract
Lyme disease, caused by Borrelia burgdorferi, B. afzelii and B. garinii, is a chronic, multi-systemic infection and the spectrum of tissues affected can vary with the Lyme disease strain. For example, whereas B. garinii infection is associated with neurologic manifestations, B. burgdorferi infection is associated with arthritis. The basis for tissue tropism is poorly understood, but has been long hypothesized to involve strain-specific interactions with host components in the target tissue. OspC (outer surface protein C) is a highly variable outer surface protein required for infectivity, and sequence differences in OspC are associated with variation in tissue invasiveness, but whether OspC directly influences tropism is unknown. We found that OspC binds to the extracellular matrix (ECM) components fibronectin and/or dermatan sulfate in an OspC variant-dependent manner. Murine infection by isogenic B. burgdorferi strains differing only in their ospC coding region revealed that two OspC variants capable of binding dermatan sulfate promoted colonization of all tissues tested, including joints. However, an isogenic strain producing OspC from B. garinii strain PBr, which binds fibronectin but not dermatan sulfate, colonized the skin, heart and bladder, but not joints. Moreover, a strain producing an OspC altered to recognize neither fibronectin nor dermatan sulfate displayed dramatically reduced levels of tissue colonization that were indistinguishable from a strain entirely deficient in OspC. Finally, intravital microscopy revealed that this OspC mutant, in contrast to a strain producing wild type OspC, was defective in promoting joint invasion by B. burgdorferi in living mice. We conclude that OspC functions as an ECM-binding adhesin that is required for joint invasion, and that variation in OspC sequence contributes to strain-specific differences in tissue tropism displayed among Lyme disease spirochetes. Infection by different Lyme disease bacteria is associated with different manifestations, such as cardiac, neurologic, or, in the case of B. burgdorferi, the major cause of Lyme disease in the U.S., joint disease. The basis for these differences is unknown, but likely involve strain-specific interactions with host components in the target tissue. The sequence of the outer surface lipoprotein OspC varies with the strains, and we found that this variation influences the spectrum of host extracellular matrix components recognized. Infection of mice with strains that are identical except for ospC revealed that OspC variants that differ in binding spectrum promote infection of different tissues. A strain producing OspC invaded and colonized the joint in living animals, but an altered OspC protein incapable of binding tissue components did not. Thus, tissue-binding by OspC is critical for infection and joint invasion, and OspC variation directly influences tissue tropism.
Collapse
Affiliation(s)
- Yi-Pin Lin
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, United States of America
| | - Xi Tan
- Departments of Biochemistry & Molecular Biology and Microbiology, Immunology & Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jennifer A. Caine
- Division of Infectious Diseases, and Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Mildred Castellanos
- Departments of Biochemistry & Molecular Biology and Microbiology, Immunology & Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - George Chaconas
- Departments of Biochemistry & Molecular Biology and Microbiology, Immunology & Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jenifer Coburn
- Division of Infectious Diseases, and Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- * E-mail: (JC); (JML)
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail: (JC); (JML)
| |
Collapse
|
17
|
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
|
18
|
Lin YP, Frye AM, Nowak TA, Kraiczy P. New Insights Into CRASP-Mediated Complement Evasion in the Lyme Disease Enzootic Cycle. Front Cell Infect Microbiol 2020; 10:1. [PMID: 32083019 PMCID: PMC7002432 DOI: 10.3389/fcimb.2020.00001] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 01/06/2020] [Indexed: 12/23/2022] Open
Abstract
Lyme disease (LD), which is caused by genospecies of the Borrelia burgdorferi sensu lato complex, is the most common vector-borne disease in the Northern hemisphere. Spirochetes are transmitted by Ixodes ticks and maintained in diverse vertebrate animal hosts. Following tick bite, spirochetes initially establish a localized infection in the skin. However, they may also disseminate hematogenously to several distal sites, including heart, joints, or the CNS. Because they need to survive in diverse microenvironments, from tick vector to mammalian hosts, spirochetes have developed multiple strategies to combat the numerous host defense mechanisms. One of these strategies includes the production of a number of complement-regulator acquiring surface proteins (CRASPs) which encompass CspA, CspZ, and OspE paralogs to blunt the complement pathway. These proteins are capable of preventing complement activation on the spirochete surface by binding to complement regulator Factor H. The genes encoding these CRASPs differ in their expression patterns during the tick-to-host infection cycle, implying that these proteins may exhibit different functions during infection. This review summarizes the recent published reports which investigated the roles that each of these molecules plays in conferring tick-borne transmission and dissemination in vertebrate hosts. These findings offer novel mechanistic insights into LD pathobiology and may facilitate the identification of new targets for preventive strategies against Lyme borreliosis.
Collapse
Affiliation(s)
- Yi-Pin Lin
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, United States
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, NY, United States
| | - Amber M. Frye
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, United States
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, NY, United States
| | - Tristan A. Nowak
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, United States
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, NY, United States
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
19
|
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
|
20
|
Federizon J, Lin YP, Lovell JF. Antigen Engineering Approaches for Lyme Disease Vaccines. Bioconjug Chem 2019; 30:1259-1272. [PMID: 30987418 DOI: 10.1021/acs.bioconjchem.9b00167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Increasing rates of Lyme disease necessitate preventive measures such as immunization to mitigate the risk of contracting the disease. At present, there is no human Lyme disease vaccine available on the market. Since the withdrawal of the first and only licensed Lyme disease vaccine based on lipidated recombinant OspA, vaccine and antigen research has aimed to overcome its risks and shortcomings. Replacement of the putative cross-reactive T-cell epitope in OspA via mutation or chimerism addresses the potential risk of autoimmunity. Multivalent approaches in Lyme disease vaccines have been pursued to address sequence heterogeneity of Lyme borreliae antigens and to induce a repertoire of functional antibodies necessary for efficient heterologous protection. This Review summarizes recent antigen engineering strategies that have paved the way for the development of next generation vaccines against Lyme disease, some of which have reached clinical testing. Bioconjugation methods that incorporate antigens to self-assembling nanoparticles for immune response potentiation are also discussed.
Collapse
Affiliation(s)
- Jasmin Federizon
- Department of Biomedical Engineering , University at Buffalo, State University of New York , Buffalo , New York 14260 , United States
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health , Albany , New York 12208 , United States.,Department of Biomedical Sciences , State University of New York at Albany , Albany , New York 12222 , United States
| | - Jonathan F Lovell
- Department of Biomedical Engineering , University at Buffalo, State University of New York , Buffalo , New York 14260 , United States
| |
Collapse
|
21
|
Tufts DM, Hart TM, Chen GF, Kolokotronis SO, Diuk-Wasser MA, Lin YP. Outer surface protein polymorphisms linked to host-spirochete association in Lyme borreliae. Mol Microbiol 2019; 111:868-882. [PMID: 30666741 DOI: 10.1111/mmi.14209] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2019] [Indexed: 12/15/2022]
Abstract
Lyme borreliosis is caused by multiple species of the spirochete bacteria Borrelia burgdorferi sensu lato. The spirochetes are transmitted by ticks to vertebrate hosts, including small- and medium-sized mammals, birds, reptiles, and humans. Strain-to-strain variation in host-specific infectivity has been documented, but the molecular basis that drives this differentiation is still unclear. Spirochetes possess the ability to evade host immune responses and colonize host tissues to establish infection in vertebrate hosts. In turn, hosts have developed distinct levels of immune responses when invaded by different species/strains of Lyme borreliae. Similarly, the ability of Lyme borreliae to colonize host tissues varies among different spirochete species/strains. One potential mechanism that drives this strain-to-strain variation of immune evasion and colonization is the polymorphic outer surface proteins produced by Lyme borreliae. In this review, we summarize research on strain-to-strain variation in host competence and discuss the evidence that supports the role of spirochete-produced protein polymorphisms in driving this variation in host specialization. Such information will provide greater insights into the adaptive mechanisms driving host and Lyme borreliae association, which will lead to the development of interventions to block pathogen spread and eventually reduce Lyme borreliosis health burden.
Collapse
Affiliation(s)
- Danielle M Tufts
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, NY, USA
| | - Thomas M Hart
- Department of Biological Sciences, University at Albany, Albany, NY, USA.,Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Grace F Chen
- Department of Biology, Misericordia University, Dallas, PA, USA
| | - Sergios-Orestis Kolokotronis
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Maria A Diuk-Wasser
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, NY, USA
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA.,Department of Biomedical Sciences, University at Albany, Albany, NY, USA
| |
Collapse
|
22
|
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]
|
23
|
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
|
24
|
Tylewska-Wierzbanowska S, Rogulska U, Lewandowska G, Chmielewski T. Bactericidal Activity of Octenidine to Various Genospecies of Borrelia burgdorferi, Sensu Lato Spirochetes in Vitro and in Vivo. Pol J Microbiol 2018; 66:259-263. [PMID: 28735308 DOI: 10.5604/01.3001.0010.7874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of our studies was to invent a reliable method for detection of the bactericidal activity of disinfectants against Borrelia burgdorferi in suspension (in vitro) and in cell line cultures (in vivo). In the suspension method, 0.01% octenidine at 20°C and 35°C was bactericidal to Borrelia afzeli; Borrelia garini, B. burgdorferi sensu stricto after 5 minutes treatment. Increase of the temperature to 35°C speed up the bactericidal effect to 1 minute. The bactericidal action of octenidine towards B. burgdorferi spirochetes growing in fibroblasts was less effective and needed a longer time to kill them than in the suspension.
Collapse
Affiliation(s)
- Stanisława Tylewska-Wierzbanowska
- National Institute of Public Heath - National Institute of Hygiene, Laboratory of Rickettsia, Chlamydia and Spirochetes, Warsaw, Poland
| | - Urszula Rogulska
- National Institute of Public Heath - National Institute of Hygiene, Laboratory of Rickettsia, Chlamydia and Spirochetes, Warsaw, Poland
| | - Grażyna Lewandowska
- National Institute of Public Heath - National Institute of Hygiene, Laboratory of Rickettsia, Chlamydia and Spirochetes, Warsaw, Poland
| | - Tomasz Chmielewski
- National Institute of Public Heath - National Institute of Hygiene, Laboratory of Rickettsia, Chlamydia and Spirochetes, Warsaw, Poland
| |
Collapse
|
25
|
Saccomano SJ, Hrelic DA. Recognizing and treating Lyme disease. Nurse Pract 2018; 43:13-21. [PMID: 30028768 DOI: 10.1097/01.npr.0000541469.54290.00] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Lyme disease is the most commonly reported vector-borne disease in the United States. After initial antibiotic treatment for patients with Lyme disease, ongoing symptoms that may persist have considerable long-term impact on healthcare costs. Posttreatment Lyme disease syndrome is characterized by a host of chronic symptoms that can leave patients physically and mentally disabled.
Collapse
Affiliation(s)
- Scott J Saccomano
- Scott J. Saccomano is an assistant professor in the College of Health and Human Services at the University of North Carolina, Wilmington, N.C. Debra A. Hrelic is the academic program coordinator for the University of North Carolina, Wilmington, in Wilmington, N.C
| | | |
Collapse
|
26
|
Bernard Q, Thakur M, Smith AA, Kitsou C, Yang X, Pal U. Borrelia burgdorferi protein interactions critical for microbial persistence in mammals. Cell Microbiol 2018; 21:e12885. [PMID: 29934966 PMCID: PMC10082445 DOI: 10.1111/cmi.12885] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 06/11/2018] [Accepted: 06/14/2018] [Indexed: 12/24/2022]
Abstract
Borrelia burgdorferi is the causative agent of Lyme disease that persists in a complex enzootic life cycle, involving Ixodes ticks and vertebrate hosts. The microbe invades ticks and vertebrate hosts in spite of active immune surveillance and potent microbicidal responses, and establishes long-term infection utilising mechanisms that are yet to be unravelled. The pathogen can cause multi-system disorders when transmitted to susceptible mammalian hosts, including in humans. In the past decades, several studies identified a limited number of B. burgdorferi gene-products critical for pathogen persistence, transmission between the vectors and the host, and host-pathogen interactions. This review will focus on the interactions between B. burgdorferi proteins, as well as between microbial proteins and host components, protein and non-protein components, highlighting their roles in pathogen persistence in the mammalian host. A better understanding of the contributions of protein interactions in the microbial virulence and persistence of B. burgdorferi would support development of novel therapeutics against the infection.
Collapse
Affiliation(s)
- Quentin Bernard
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland
| | - Meghna Thakur
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland
| | - Alexis A Smith
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland
| | - Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland
| | - Xiuli Yang
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland
| |
Collapse
|
27
|
Williams SK, Weiner ZP, Gilmore RD. Human neuroglial cells internalize Borrelia burgdorferi by coiling phagocytosis mediated by Daam1. PLoS One 2018; 13:e0197413. [PMID: 29746581 PMCID: PMC5944952 DOI: 10.1371/journal.pone.0197413] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/01/2018] [Indexed: 11/18/2022] Open
Abstract
Borrelia burgdorferi, the agent of Lyme borreliosis, can elude hosts’ innate and adaptive immunity as part of the course of infection. The ability of B. burgdorferi to invade or be internalized by host cells in vitro has been proposed as a mechanism for the pathogen to evade immune responses or antimicrobials. We have previously shown that B. burgdorferi can be internalized by human neuroglial cells. In this study we demonstrate that these cells take up B. burgdorferi via coiling phagocytosis mediated by the formin, Daam1, a process similarly described for human macrophages. Following coincubation with glial cells, B. burgdorferi was enwrapped by Daam1-enriched coiling pseudopods. Coiling of B. burgdorferi was significantly reduced when neuroglial cells were pretreated with anti-Daam1 antibody indicating the requirement for Daam1 for borrelial phagocytosis. Confocal microscopy showed Daam1 colocalizing to the B. burgdorferi surface suggesting interaction with borrelial membrane protein(s). Using the yeast 2-hybrid system for identifying protein-protein binding, we found that the B. burgdorferi surface lipoprotein, BBA66, bound the FH2 subunit domain of Daam1. Recombinant proteins were used to validate binding by ELISA, pull-down, and co-immunoprecipitation. Evidence for native Daam1 and BBA66 interaction was suggested by colocalization of the proteins in the course of borrelial capture by the Daam1-enriched pseudopodia. Additionally, we found a striking reduction in coiling for a BBA66-deficient mutant strain compared to BBA66-expressing strains. These results show that coiling phagocytosis is a mechanism for borrelial internalization by neuroglial cells mediated by Daam1.
Collapse
Affiliation(s)
- Shanna K. Williams
- Bacterial Diseases Branch, Division of Vector Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
| | - Zachary P. Weiner
- Bacterial Diseases Branch, Division of Vector Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
| | - Robert D. Gilmore
- Bacterial Diseases Branch, Division of Vector Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
28
|
Ma XX, Ma P, Chang QY, Liu ZB, Zhang D, Zhou XK, Ma ZR, Cao X. Adaptation ofBorrelia burgdorferito its natural hosts by synonymous codon and amino acid usage. J Basic Microbiol 2018. [DOI: 10.1002/jobm.201700652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Xiao-Xia Ma
- Engineering and Technology Research Center for Animal Cell, Gansu; College of Life Science and Engineering; Northwest Minzu University; Gansu P.R. China
| | - Peng Ma
- Engineering and Technology Research Center for Animal Cell, Gansu; College of Life Science and Engineering; Northwest Minzu University; Gansu P.R. China
| | - Qiu-Yan Chang
- Engineering and Technology Research Center for Animal Cell, Gansu; College of Life Science and Engineering; Northwest Minzu University; Gansu P.R. China
| | - Zhen-Bin Liu
- Engineering and Technology Research Center for Animal Cell, Gansu; College of Life Science and Engineering; Northwest Minzu University; Gansu P.R. China
| | - Derong Zhang
- Engineering and Technology Research Center for Animal Cell, Gansu; College of Life Science and Engineering; Northwest Minzu University; Gansu P.R. China
| | - Xiao-Kai Zhou
- Engineering and Technology Research Center for Animal Cell, Gansu; College of Life Science and Engineering; Northwest Minzu University; Gansu P.R. China
| | - Zhong-Ren Ma
- Engineering and Technology Research Center for Animal Cell, Gansu; College of Life Science and Engineering; Northwest Minzu University; Gansu P.R. China
| | - Xin Cao
- Engineering and Technology Research Center for Animal Cell, Gansu; College of Life Science and Engineering; Northwest Minzu University; Gansu P.R. China
| |
Collapse
|
29
|
Belperron AA, Mao J, Bockenstedt LK. Two Photon Intravital Microscopy of Lyme Borrelia in Mice. Methods Mol Biol 2018; 1690:279-290. [PMID: 29032551 DOI: 10.1007/978-1-4939-7383-5_20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Two-photon intravital microscopy is a powerful tool that allows visualization of cells in intact tissues in a live animal in real time. In recent years, this advanced technology has been applied to understand pathogen-host interactions using fluorescently labeled bacteria. In particular, infectious fluorescent transformants of the Lyme disease spirochete Borrelia burgdorferi, an Ixodes tick-transmitted pathogen, have been imaged by two-photon intravital microscopy to study bacterial motility and interactions of the pathogen with feeding ticks and host tissues. Here, we describe the techniques and equipment used to image mammalian-adapted spirochetes in the skin of living mice in vivo and in joints ex vivo using two-photon intravital microscopy.
Collapse
Affiliation(s)
- Alexia A Belperron
- Department of Internal Medicine/Section of Rheumatology, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Jialing Mao
- Department of Internal Medicine/Section of Rheumatology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Linda K Bockenstedt
- Department of Internal Medicine/Section of Rheumatology, Yale University School of Medicine, New Haven, CT, 06520, USA
| |
Collapse
|
30
|
Lin YP, Leong JM. Characterization of Borrelia burgdorferi Binding to Mammalian Cells and Extracellular Matrix. Methods Mol Biol 2018; 1690:57-67. [PMID: 29032536 DOI: 10.1007/978-1-4939-7383-5_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Lyme disease Borreliae produces outer surface adhesins to confer bacterial attachment to the extracellular matrix (ECM) components on the surface of mammalian cells. Here, we describe protocols to characterize the activity and specificity of these adhesins by flow cytometry or measurement of the binding of radiolabeled spirochetes to immobilized ECM or mammalian cells.
Collapse
Affiliation(s)
- Yi-Pin Lin
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, 120 New Scotland Ave., Albany, NY, 12208, USA.
| | - John M Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA, 02111, USA
| |
Collapse
|
31
|
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
|
32
|
Cabello FC, Godfrey HP, Bugrysheva J, Newman SA. Sleeper cells: the stringent response and persistence in the Borreliella (Borrelia) burgdorferi enzootic cycle. Environ Microbiol 2017; 19:3846-3862. [PMID: 28836724 PMCID: PMC5794220 DOI: 10.1111/1462-2920.13897] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/12/2017] [Accepted: 08/17/2017] [Indexed: 12/27/2022]
Abstract
Infections with tick-transmitted Borreliella (Borrelia) burgdorferi, the cause of Lyme disease, represent an increasingly large public health problem in North America and Europe. The ability of these spirochetes to maintain themselves for extended periods of time in their tick vectors and vertebrate reservoirs is crucial for continuance of the enzootic cycle as well as for the increasing exposure of humans to them. The stringent response mediated by the alarmone (p)ppGpp has been determined to be a master regulator in B. burgdorferi. It modulates the expression of identified and unidentified open reading frames needed to deal with and overcome the many nutritional stresses and other challenges faced by the spirochete in ticks and animal reservoirs. The metabolic and morphologic changes resulting from activation of the stringent response in B. burgdorferi may also be involved in the recently described non-genetic phenotypic phenomenon of tolerance to otherwise lethal doses of antimicrobials and to other antimicrobial activities. It may thus constitute a linchpin in multiple aspects of infections with Lyme disease borrelia, providing a link between the micro-ecological challenges of its enzootic life-cycle and long-term residence in the tissues of its animal reservoirs, with the evolutionary side effect of potential persistence in incidental human hosts.
Collapse
Affiliation(s)
- Felipe C. Cabello
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Henry P. Godfrey
- Department of Pathology, New York Medical College, Valhalla, NY, USA
| | - Julia Bugrysheva
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Stuart A. Newman
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
33
|
Hsieh CL, Tseng A, He H, Kuo CJ, Wang X, Chang YF. Leptospira Immunoglobulin-Like Protein B Interacts with the 20th Exon of Human Tropoelastin Contributing to Leptospiral Adhesion to Human Lung Cells. Front Cell Infect Microbiol 2017; 7:163. [PMID: 28536676 PMCID: PMC5422739 DOI: 10.3389/fcimb.2017.00163] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/18/2017] [Indexed: 01/21/2023] Open
Abstract
Leptospira immunoglobulin-like protein B (LigB), a surface adhesin, is capable of mediating the attachment of pathogenic leptospira to the host through interaction with various components of the extracellular matrix (ECM). Human tropoelastin (HTE), the building block of elastin, confers resilience and elasticity to lung, and other tissues. Previously identified Ig-like domains of LigB, including LigB4 and LigB12, bind to HTE, which is likely to promote Leptospira adhesion to lung tissue. However, the molecular mechanism that mediates the LigB-HTE interaction is unclear. In this study, the LigB-binding site on HTE was further pinpointed to a N-terminal region of the 20th exon of HTE (HTE20N). Alanine mutants of basic and aromatic residues on HTE20N significantly reduced binding to the LigB. Additionally, HTE-binding site was narrowed down to the first β-sheet of LigB12. On this binding surface, residues F1054, D1061, A1065, and D1066 were critical for the association with HTE. Most importantly, the recombinant HTE truncates could diminish the binding of LigB to human lung fibroblasts (WI-38) by 68%, and could block the association of LigA-expressing L. biflexa to lung cells by 61%. These findings should expand our understanding of leptospiral pathogenesis, particularly in pulmonary manifestations of leptospirosis.
Collapse
Affiliation(s)
- Ching-Lin Hsieh
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell UniversityIthaca, NY, USA
| | - Andrew Tseng
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell UniversityIthaca, NY, USA
| | - Hongxuan He
- National Research Center for Wildlife Borne Diseases, Institute of Zoology, Chinese Academy of SciencesBeijing, China
| | - Chih-Jung Kuo
- Department of Veterinary Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Xuannian Wang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell UniversityIthaca, NY, USA.,Research Center for Biotechnology, Xinxiang UniversityXinxiang, China
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell UniversityIthaca, NY, USA
| |
Collapse
|
34
|
Marcinkiewicz AL, Kraiczy P, Lin YP. There Is a Method to the Madness: Strategies to Study Host Complement Evasion by Lyme Disease and Relapsing Fever Spirochetes. Front Microbiol 2017; 8:328. [PMID: 28303129 PMCID: PMC5332432 DOI: 10.3389/fmicb.2017.00328] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/16/2017] [Indexed: 01/04/2023] Open
Abstract
Lyme disease and relapsing fever are caused by various Borrelia species. Lyme disease borreliae, the most common vector-borne pathogens in both the U.S. and Europe, are transmitted by Ixodes ticks and disseminate from the site of tick bites to tissues leading to erythema migrans skin rash, arthritis, carditis, and neuroborreliosis. Relapsing fever borreliae, carried by ticks and lice, trigger reoccurring fever episodes. Following transmission, spirochetes survive in the blood to induce bacteremia at the early stages of infection, which is thought to promote evasion of the host complement system. The complement system acts as an important innate immune defense mechanism in humans and vertebrates. Upon activation, the cleaved complement components form complexes on the pathogen surface to eventually promote bacteriolysis. The complement system is negatively modulated by a number of functionally diverse regulators to avoid tissue damage. To evade and inhibit the complement system, spirochetes are capable of binding complement components and regulators. Complement inhibition results in bacterial survival in serum (serum resistance) and is thought to promote bloodstream survival, which facilitates spirochete dissemination and disease manifestations. In this review, we discuss current methodologies to elucidate the mechanisms of Borrelia spp. that promote serum resistance and bloodstream survival, as well as novel methods to study factors responsible for bloodstream survival of Lyme disease borreliae that can be applied to relapsing fever borreliae. Understanding the mechanisms these pathogens utilize to evade the complement system will ultimately aid in the development of novel therapeutic strategies and disease prevention to improve human health.
Collapse
Affiliation(s)
- Ashley L Marcinkiewicz
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health Albany, NY, USA
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt am Main Frankfurt am Main, Germany
| | - Yi-Pin Lin
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health Albany, NY, USA
| |
Collapse
|
35
|
Tracy KE, Baumgarth N. Borrelia burgdorferi Manipulates Innate and Adaptive Immunity to Establish Persistence in Rodent Reservoir Hosts. Front Immunol 2017; 8:116. [PMID: 28265270 PMCID: PMC5316537 DOI: 10.3389/fimmu.2017.00116] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 01/25/2017] [Indexed: 01/17/2023] Open
Abstract
Borrelia burgdorferi sensu lato species complex is capable of establishing persistent infections in a wide variety of species, particularly rodents. Infection is asymptomatic or mild in most reservoir host species, indicating successful co-evolution of the pathogen with its natural hosts. However, infected humans and other incidental hosts can develop Lyme disease, a serious inflammatory syndrome characterized by tissue inflammation of joints, heart, muscles, skin, and CNS. Although B. burgdorferi infection induces both innate and adaptive immune responses, they are ultimately ineffective in clearing the infection from reservoir hosts, leading to bacterial persistence. Here, we review some mechanisms by which B. burgdorferi evades the immune system of the rodent host, focusing in particular on the effects of innate immune mechanisms and recent findings suggesting that T-dependent B cell responses are subverted during infection. A better understanding of the mechanisms causing persistence in rodents may help to increase our understanding of the pathogenesis of Lyme disease and ultimately aid in the development of therapies that support effective clearance of the bacterial infection by the host’s immune system.
Collapse
Affiliation(s)
- Karen E Tracy
- Graduate Group in Immunology, University of California Davis, Davis, CA, USA; Center for Comparative Medicine, University of California Davis, Davis, CA, USA
| | - Nicole Baumgarth
- Graduate Group in Immunology, University of California Davis, Davis, CA, USA; Center for Comparative Medicine, University of California Davis, Davis, CA, USA; Department of Pathology, Microbiology and Immunology, University of California Davis, Davis, CA, USA
| |
Collapse
|
36
|
Abstract
Lyme borreliosis is a tick-borne disease that predominantly occurs in temperate regions of the northern hemisphere and is primarily caused by the bacterium Borrelia burgdorferi in North America and Borrelia afzelii or Borrelia garinii in Europe and Asia. Infection usually begins with an expanding skin lesion, known as erythema migrans (referred to as stage 1), which, if untreated, can be followed by early disseminated infection, particularly neurological abnormalities (stage 2), and by late infection, especially arthritis in North America or acrodermatitis chronica atrophicans in Europe (stage 3). However, the disease can present with any of these manifestations. During infection, the bacteria migrate through the host tissues, adhere to certain cells and can evade immune clearance. Yet, these organisms are eventually killed by both innate and adaptive immune responses and most inflammatory manifestations of the infection resolve. Except for patients with erythema migrans, Lyme borreliosis is diagnosed based on a characteristic clinical constellation of signs and symptoms with serological confirmation of infection. All manifestations of the infection can usually be treated with appropriate antibiotic regimens, but the disease can be followed by post-infectious sequelae in some patients. Prevention of Lyme borreliosis primarily involves the avoidance of tick bites by personal protective measures.
Collapse
Affiliation(s)
- Allen C Steere
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114, USA
- Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Franc Strle
- Department of Infectious Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Gary P Wormser
- Division of Infectious Diseases, New York Medical College, Valhalla, New York, USA
| | - Linden T Hu
- Department of Molecular Biology and Microbiology, Tufts Medical Center, Boston, Massachusetts, USA
| | - John A Branda
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joppe W R Hovius
- Center for Experimental and Molecular Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Xin Li
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts, USA
| | - Paul S Mead
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| |
Collapse
|
37
|
A penicillin-binding protein inhibits selection of colistin-resistant, lipooligosaccharide-deficient Acinetobacter baumannii. Proc Natl Acad Sci U S A 2016; 113:E6228-E6237. [PMID: 27681618 DOI: 10.1073/pnas.1611594113] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The Gram-negative bacterial outer membrane fortifies the cell against environmental toxins including antibiotics. Unique glycolipids called lipopolysaccharide/lipooligosaccharide (LPS/LOS) are enriched in the cell-surface monolayer of the outer membrane and promote antimicrobial resistance. Colistin, which targets the lipid A domain of LPS/LOS to lyse the cell, is the last-line treatment for multidrug-resistant Gram-negative infections. Lipid A is essential for the survival of most Gram-negative bacteria, but colistin-resistant Acinetobacter baumannii lacking lipid A were isolated after colistin exposure. Previously, strain ATCC 19606 was the only A. baumannii strain demonstrated to subsist without lipid A. Here, we show that other A. baumannii strains can also survive without lipid A, but some cannot, affording a unique model to study endotoxin essentiality. We assessed the capacity of 15 clinical A. baumannii isolates including 9 recent clinical isolates to develop colistin resistance through inactivation of the lipid A biosynthetic pathway, the products of which assemble the LOS precursor. Our investigation determined that expression of the well-conserved penicillin-binding protein (PBP) 1A, prevented LOS-deficient colony isolation. The glycosyltransferase activity of PBP1A, which aids in the polymerization of the peptidoglycan cell wall, was lethal to LOS-deficient A. baumannii Global transcriptomic analysis of a PBP1A-deficient mutant and four LOS-deficient A. baumannii strains showed a concomitant increase in transcription of lipoproteins and their transporters. Examination of the LOS-deficient A. baumannii cell surface demonstrated that specific lipoproteins were overexpressed and decorated the cell surface, potentially compensating for LOS removal. This work expands our knowledge of lipid A essentiality and elucidates a drug resistance mechanism.
Collapse
|
38
|
Vectors as Epidemiological Sentinels: Patterns of Within-Tick Borrelia burgdorferi Diversity. PLoS Pathog 2016; 12:e1005759. [PMID: 27414806 PMCID: PMC4944968 DOI: 10.1371/journal.ppat.1005759] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/18/2016] [Indexed: 01/13/2023] Open
Abstract
Hosts including humans, other vertebrates, and arthropods, are frequently infected with heterogeneous populations of pathogens. Within-host pathogen diversity has major implications for human health, epidemiology, and pathogen evolution. However, pathogen diversity within-hosts is difficult to characterize and little is known about the levels and sources of within-host diversity maintained in natural populations of disease vectors. Here, we examine genomic variation of the Lyme disease bacteria, Borrelia burgdorferi (Bb), in 98 individual field-collected tick vectors as a model for study of within-host processes. Deep population sequencing reveals extensive and previously undocumented levels of Bb variation: the majority (~70%) of ticks harbor mixed strain infections, which we define as levels Bb diversity pre-existing in a diverse inoculum. Within-tick diversity is thus a sample of the variation present within vertebrate hosts. Within individual ticks, we detect signatures of positive selection. Genes most commonly under positive selection across ticks include those involved in dissemination in vertebrate hosts and evasion of the vertebrate immune complement. By focusing on tick-borne Bb, we show that vectors can serve as epidemiological and evolutionary sentinels: within-vector pathogen diversity can be a useful and unbiased way to survey circulating pathogen diversity and identify evolutionary processes occurring in natural transmission cycles. Lyme disease, caused by a bacteria carried by deer ticks, is the most common vector-borne disease in North America and over 30,000 cases are reported each year in the United States. Ticks may be infected with multiple strains of the Lyme disease bacteria, which differ in transmissibility and the harm they pose to humans. In this study, we collected 98 infected deer ticks from across the United States and southern Canada. We used genetic techniques to investigate the diversity of the Lyme disease bacteria infecting each individual tick. We find that 70% of ticks are infected with multiple strains of the Lyme disease bacteria, indicating that humans may be exposed to and infected with multiple bacterial strains from a single tick bite. We also find evidence that the Lyme disease bacteria is evolving in response to the immune defenses of its natural hosts (including rodents and birds). Our study shows that individual ticks and other disease vectors can be studied as epidemiological sentinels, which reveal the extensive diversity of pathogens circulating in natural disease cycles and how they are evolving.
Collapse
|
39
|
Gagic D, Ciric M, Wen WX, Ng F, Rakonjac J. Exploring the Secretomes of Microbes and Microbial Communities Using Filamentous Phage Display. Front Microbiol 2016; 7:429. [PMID: 27092113 PMCID: PMC4823517 DOI: 10.3389/fmicb.2016.00429] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/17/2016] [Indexed: 01/12/2023] Open
Abstract
Microbial surface and secreted proteins (the secretome) contain a large number of proteins that interact with other microbes, host and/or environment. These proteins are exported by the coordinated activities of the protein secretion machinery present in the cell. A group of bacteriophage, called filamentous phage, have the ability to hijack bacterial protein secretion machinery in order to amplify and assemble via a secretion-like process. This ability has been harnessed in the use of filamentous phage of Escherichia coli in biotechnology applications, including screening large libraries of variants for binding to “bait” of interest, from tissues in vivo to pure proteins or even inorganic substrates. In this review we discuss the roles of secretome proteins in pathogenic and non-pathogenic bacteria and corresponding secretion pathways. We describe the basics of phage display technology and its variants applied to discovery of bacterial proteins that are implicated in colonization of host tissues and pathogenesis, as well as vaccine candidates through filamentous phage display library screening. Secretome selection aided by next-generation sequence analysis was successfully applied for selective display of the secretome at a microbial community scale, the latter revealing the richness of secretome functions of interest and surprising versatility in filamentous phage display of secretome proteins from large number of Gram-negative as well as Gram-positive bacteria and archaea.
Collapse
Affiliation(s)
- Dragana Gagic
- Institute of Fundamental Sciences, Massey UniversityPalmerston North, New Zealand; Animal Science, Grasslands Research Centre, AgResearch Ltd, Palmerston NorthNew Zealand
| | - Milica Ciric
- Institute of Fundamental Sciences, Massey UniversityPalmerston North, New Zealand; Animal Science, Grasslands Research Centre, AgResearch Ltd, Palmerston NorthNew Zealand
| | - Wesley X Wen
- Institute of Fundamental Sciences, Massey University Palmerston North, New Zealand
| | - Filomena Ng
- Animal Science, Grasslands Research Centre, AgResearch Ltd, Palmerston North New Zealand
| | - Jasna Rakonjac
- Institute of Fundamental Sciences, Massey University Palmerston North, New Zealand
| |
Collapse
|
40
|
Kung F, Kaur S, Smith AA, Yang X, Wilder CN, Sharma K, Buyuktanir O, Pal U. A Borrelia burgdorferi Surface-Exposed Transmembrane Protein Lacking Detectable Immune Responses Supports Pathogen Persistence and Constitutes a Vaccine Target. J Infect Dis 2016; 213:1786-95. [PMID: 26747708 DOI: 10.1093/infdis/jiw013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/30/2015] [Indexed: 01/29/2023] Open
Abstract
Borrelia burgdorferi harbors a limited set of transmembrane surface proteins, most of which constitute key targets of humoral immune responses. Here we show that BB0405, a conserved membrane-spanning protein of unknown function, fails to evoke detectable antibody responses despite its extracellular exposure. bb0405 is a member of an operon and ubiquitously expressed throughout the rodent-tick infection cycle. The gene product serves an essential function in vivo, as bb0405-deletion mutants are unable to transmit from ticks and establish infection in mammalian hosts. Despite the lack of BB0405-specific immunoglobulin M or immunoglobulin G antibodies during natural infection, mice immunized with a recombinant version of the protein elicited high-titer and remarkably long-lasting antibody responses, conferring significant host protection against tick-borne infection. Taken together, these studies highlight the essential role of an apparently immune-invisible borrelial transmembrane protein in facilitating infection and its usefulness as a target of protective host immunity blocking the transmission of B. burgdorferi.
Collapse
Affiliation(s)
- Faith Kung
- Department of Veterinary Medicine, University of Maryland-College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park
| | - Simarjot Kaur
- Department of Veterinary Medicine, University of Maryland-College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park
| | - Alexis A Smith
- Department of Veterinary Medicine, University of Maryland-College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park
| | - Xiuli Yang
- Department of Veterinary Medicine, University of Maryland-College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park
| | - Cara N Wilder
- Department of Veterinary Medicine, University of Maryland-College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park
| | - Kavita Sharma
- Department of Veterinary Medicine, University of Maryland-College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park
| | - Ozlem Buyuktanir
- Department of Microbiology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland-College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park
| |
Collapse
|
41
|
Salo J, Pietikäinen A, Söderström M, Auvinen K, Salmi M, Ebady R, Moriarty TJ, Viljanen MK, Hytönen J. Flow-Tolerant Adhesion of a Bacterial Pathogen to Human Endothelial Cells Through Interaction With Biglycan. J Infect Dis 2016; 213:1623-31. [DOI: 10.1093/infdis/jiw003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/29/2015] [Indexed: 11/14/2022] Open
|
42
|
Intravital Imaging of Vascular Transmigration by the Lyme Spirochete: Requirement for the Integrin Binding Residues of the B. burgdorferi P66 Protein. PLoS Pathog 2015; 11:e1005333. [PMID: 26684456 PMCID: PMC4686178 DOI: 10.1371/journal.ppat.1005333] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 11/17/2015] [Indexed: 01/13/2023] Open
Abstract
Vascular extravasation, a key step in systemic infection by hematogenous microbial pathogens, is poorly understood, but has been postulated to encompass features similar to vascular transmigration by leukocytes. The Lyme disease spirochete can cause a variety of clinical manifestations, including arthritis, upon hematogenous dissemination. This pathogen encodes numerous surface adhesive proteins (adhesins) that may promote extravasation, but none have yet been implicated in this process. In this work we report the novel use of intravital microscopy of the peripheral knee vasculature to study transmigration of the Lyme spirochete in living Cd1d-/-mice. In the absence of iNKT cells, major immune modulators in the mouse joint, spirochetes that have extravasated into joint-proximal tissue remain in the local milieu and can be enumerated accurately. We show that BBK32, a fibronectin and glycosaminoglycan adhesin of B. burgdorferi involved in early steps of endothelial adhesion, is not required for extravasation from the peripheral knee vasculature. In contrast, almost no transmigration occurs in the absence of P66, an outer membrane protein that has porin and integrin adhesin functions. Importantly, P66 mutants specifically defective in integrin binding were incapable of promoting extravasation. P66 itself does not promote detectable microvascular interactions, suggesting that vascular adhesion of B. burgdorferi mediated by other adhesins, sets the stage for P66-integrin interactions leading to transmigration. Although integrin-binding proteins with diverse functions are encoded by a variety of bacterial pathogens, P66 is the first to have a documented and direct role in vascular transmigration. The emerging picture of vascular escape by the Lyme spirochete shows similarities, but distinct differences from leukocyte transmigration. Lyme disease is the most common vector-transmitted infection in North America and Europe. Diverse clinical manifestations of Lyme disease result from the dissemination of the spirochetes causing the disease into a variety of tissue sites. Dissemination results from invasion of the vasculature by the bacteria, followed by exit into virtually all tissue types. The mechanism of vascular transmigration by Lyme disease spirochetes remains uncharacterized. Here we describe a novel approach to study transmigration of Lyme disease spirochetes using intravital microscopy of the peripheral knee vasculature in living mice. Our studies have identified an adhesin, P66, and its integrin-binding function as playing important roles in Lyme spirochete transmigration and dissemination.
Collapse
|
43
|
Strnad M, Elsterová J, Schrenková J, Vancová M, Rego ROM, Grubhoffer L, Nebesářová J. Correlative cryo-fluorescence and cryo-scanning electron microscopy as a straightforward tool to study host-pathogen interactions. Sci Rep 2015; 5:18029. [PMID: 26658551 PMCID: PMC4674872 DOI: 10.1038/srep18029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 11/09/2015] [Indexed: 01/04/2023] Open
Abstract
Correlative light and electron microscopy is an imaging technique that enables identification and targeting of fluorescently tagged structures with subsequent imaging at near-to-nanometer resolution. We established a novel correlative cryo-fluorescence microscopy and cryo-scanning electron microscopy workflow, which enables imaging of the studied object of interest very close to its natural state, devoid of artifacts caused for instance by slow chemical fixation. This system was tested by investigating the interaction of the zoonotic bacterium Borrelia burgdorferi with two mammalian cell lines of neural origin in order to broaden our knowledge about the cell-association mechanisms that precedes the entry of the bacteria into the cell. This method appears to be an unprecedentedly fast (<3 hours), straightforward, and reliable solution to study the finer details of pathogen-host cell interactions and provides important insights into the complex and dynamic relationship between a pathogen and a host.
Collapse
Affiliation(s)
- Martin Strnad
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, České Budějovice CZ-37005, Czech Republic.,Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760, České Budějovice CZ-37005, Czech Republic
| | - Jana Elsterová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, České Budějovice CZ-37005, Czech Republic.,Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760, České Budějovice CZ-37005, Czech Republic.,Department of Virology, Veterinary Research Institute, Brno CZ-62100, Czech Republic
| | - Jana Schrenková
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, České Budějovice CZ-37005, Czech Republic.,Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760, České Budějovice CZ-37005, Czech Republic
| | - Marie Vancová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, České Budějovice CZ-37005, Czech Republic.,Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760, České Budějovice CZ-37005, Czech Republic
| | - Ryan O M Rego
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, České Budějovice CZ-37005, Czech Republic
| | - Libor Grubhoffer
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, České Budějovice CZ-37005, Czech Republic.,Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760, České Budějovice CZ-37005, Czech Republic
| | - Jana Nebesářová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, České Budějovice CZ-37005, Czech Republic.,Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760, České Budějovice CZ-37005, Czech Republic.,Faculty of Science, Charles University in Prague, Viničná 1594/7, Praha CZ-12800, Czech Republic
| |
Collapse
|
44
|
Coumou J, Narasimhan S, Trentelman JJ, Wagemakers A, Koetsveld J, Ersoz JI, Oei A, Fikrig E, Hovius JW. Ixodes scapularis dystroglycan-like protein promotes Borrelia burgdorferi migration from the gut. J Mol Med (Berl) 2015; 94:361-70. [PMID: 26594018 PMCID: PMC4803822 DOI: 10.1007/s00109-015-1365-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 10/09/2015] [Accepted: 11/10/2015] [Indexed: 11/30/2022]
Abstract
Abstract The causative agent of Lyme borreliosis, Borrelia burgdorferi, is transmitted by Ixodes ticks. During tick feeding, B. burgdorferi migrates from the tick gut to the salivary glands from where transmission to the host occurs. B. burgdorferi-interacting tick proteins might serve as vaccine targets to thwart B. burgdorferi transmission. A previous screening for B. burgdorferi-interacting Ixodes scapularis gut proteins identified an I. scapularis putative dystroglycan protein (ISCW015049). Here, we describe the ISCW015049’s protein structure and its cellular location in the tick gut in relation to B. burgdorferi migration. Secondly, in vivo B. burgdorferi–tick attachment murine models were performed to study the role of ISCW015049 during B. burgdorferi migration and transmission. In silico analysis confirmed that ISCW015049 is similar to dystroglycan and was named I. scapularis dystroglycan-like protein (ISDLP). Confocal microscopy of gut tissue showed that ISDLP is expressed on the surface of gut cells, is upregulated during tick feeding, and is expressed significantly higher in infected ticks compared to uninfected ticks. Inhibition of ISDLP by RNA interference (RNAi) resulted in lower B. burgdorferi transmission to mice. In conclusion, we have identified a dystroglycan-like protein in I. scapularis gut that can bind to B. burgdorferi and promotes B. burgdorferi migration from the tick gut. Key messages B. burgdorferi exploits tick proteins to orchestrate its transmission to the host. B. burgdorferi is able bind to an I. scapularis dystroglycan-like protein (ISDLP). Inhibition of ISDLP in ticks results in lower B. burgdorferi transmission to mice. ISDLP is a potential target to prevent Lyme borreliosis.
Electronic supplementary material The online version of this article (doi:10.1007/s00109-015-1365-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jeroen Coumou
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| | - Sukanya Narasimhan
- Department of Internal Medicine, Yale University School of Medicine, 06511, New Haven, CT, USA
| | - Jos J Trentelman
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Alex Wagemakers
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Joris Koetsveld
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Jasmin I Ersoz
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Anneke Oei
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Erol Fikrig
- Department of Internal Medicine, Yale University School of Medicine, 06511, New Haven, CT, USA
| | - Joppe W Hovius
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
45
|
Spirochetal motility and chemotaxis in the natural enzootic cycle and development of Lyme disease. Curr Opin Microbiol 2015; 28:106-13. [PMID: 26519910 DOI: 10.1016/j.mib.2015.09.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 09/07/2015] [Indexed: 11/21/2022]
Abstract
Two-thirds of all bacterial genomes sequenced to-date possess an organelle for locomotion, referred to as flagella, periplasmic flagella or type IV pili. These genomes may also contain a chemotaxis-signaling system which governs flagellar rotation, thus leading a coordinated function for motility. Motility and chemotaxis are often crucial for infection or disease process caused by pathogenic bacteria. Although motility-associated genes are well-characterized in some organisms, the highly orchestrated synthesis, regulation, and assembly of periplasmic flagella in spirochetes are just being delineated. Recent advances were fostered by development of unique genetic manipulations in spirochetes coupled with cutting-edge imaging techniques. These contemporary advances in understanding the role of spirochetal motility and chemotaxis in host persistence and disease development are highlighted in this review.
Collapse
|
46
|
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
|
47
|
Yang X, Lin YP, Heselpoth RD, Buyuktanir O, Qin J, Kung F, Nelson DC, Leong JM, Pal U. Middle region of the Borrelia burgdorferi surface-located protein 1 (Lmp1) interacts with host chondroitin-6-sulfate and independently facilitates infection. Cell Microbiol 2015; 18:97-110. [PMID: 26247174 DOI: 10.1111/cmi.12487] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 06/23/2015] [Accepted: 07/10/2015] [Indexed: 11/29/2022]
Abstract
Borrelia burgdorferi surface-located membrane protein 1, also known as Lmp1, has been shown to play critical roles in pathogen evasion of host-acquired immune defences, thereby facilitating persistent infection. Lmp1 possesses three regions representing potentially discrete domains: Lmp1N, Lmp1M and Lmp1C. Because of its insignificant homology to known proteins, how Lmp1 or its specific regions contribute to microbial biology and infection remains enigmatic. Here, we show that distinct from Lmp1N and Lmp1C, Lmp1M is composed of at least 70% alpha helices and completely lacks recognizable beta sheets. The region binds to host glycosaminoglycan chondroitin-6-sulfate molecules and facilitates mammalian cell attachment, suggesting an adhesin function of Lmp1M. Phenotypic analysis of the Lmp1-deficient mutant engineered to produce Lmp1M on the microbial surface suggests that Lmp1M can independently support B. burgdorferi infectivity in murine hosts. Further exploration of functions of Lmp1 distinct regions will shed new light on the intriguing biology and infectivity of spirochetes and help develop novel interventions to combat Lyme disease.
Collapse
Affiliation(s)
- Xiuli Yang
- Department of Veterinary Medicine, University of Maryland, College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park, MD, 20742, USA
| | - Yi-Pin Lin
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA
| | - Ryan D Heselpoth
- Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Ozlem Buyuktanir
- Department of Veterinary Medicine, University of Maryland, College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park, MD, 20742, USA
| | - Jinhong Qin
- Department of Veterinary Medicine, University of Maryland, College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park, MD, 20742, USA
| | - Faith Kung
- Department of Veterinary Medicine, University of Maryland, College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park, MD, 20742, USA
| | - Daniel C Nelson
- Department of Veterinary Medicine, University of Maryland, College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park, MD, 20742, USA.,Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - John M Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park and Virginia-Maryland Regional College of Veterinary Medicine, College Park, MD, 20742, USA
| |
Collapse
|
48
|
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
|
49
|
Borrelia burgdorferi RevA Significantly Affects Pathogenicity and Host Response in the Mouse Model of Lyme Disease. Infect Immun 2015; 83:3675-83. [PMID: 26150536 DOI: 10.1128/iai.00530-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/28/2015] [Indexed: 12/28/2022] Open
Abstract
The Lyme disease spirochete, Borrelia burgdorferi, expresses RevA and numerous outer surface lipoproteins during mammalian infection. As an adhesin that promotes bacterial interaction with fibronectin, RevA is poised to interact with the extracellular matrix of the host. To further define the role(s) of RevA during mammalian infection, we created a mutant that is unable to produce RevA. The mutant was still infectious to mice, although it was significantly less well able to infect cardiac tissues. Complementation of the mutant with a wild-type revA gene restored heart infectivity to wild-type levels. Additionally, revA mutants led to increased evidence of arthritis, with increased fibrotic collagen deposition in tibiotarsal joints. The mutants also induced increased levels of the chemokine CCL2, a monocyte chemoattractant, in serum, and this increase was abolished in the complemented strain. Therefore, while revA is not absolutely essential for infection, deletion of revA had distinct effects on dissemination, arthritis severity, and host response.
Collapse
|
50
|
Abstract
Lyme disease/borreliosis (LD) is a well-known arthropod-transmitted entity in the northern hemisphere. The incidence of LD is reportedly rising throughout the world, although better diagnostic facilities may be contributory. The disease distribution is expanding in Europe, with its presence being now documented at higher altitudes and latitudes. Borrelia burgdorferi sensu lato is the most important genospecies leading to LD, although newer ones continue to be discovered. The variations in clinical spectrum with genospecies involved are an interesting feature. The alteration in gene expression while the organism cycles between two very different hosts is intriguing and has been described. The disease presents in three stages-namely, the early localized, early disseminated, and late stage. Erythema chronicum migrans is the pathognomic early lesion, and its diagnosis is purely clinical; however, laboratory diagnosis is essential for later manifestations. Two-tier serologic testing using an enzyme-linked immunosorbent assay (ELISA) as the first tier and immunoglobulin M (IgM) and IgG immunoblot as the second, if ELISA is positive or equivocal, is the mainstay of diagnosis. Doxycycline is the cornerstone of treatment, whereas parenteral therapy, mainly with ceftriaxone, is indicated in a few specific scenarios.
Collapse
Affiliation(s)
- Virendra N Sehgal
- Dermato-Venereology (Skin/VD) Center, Sehgal Nursing Home, A/6 Panchwati, Delhi, 110 033, India.
| | - Ananta Khurana
- Department of Dermatology, 1 Dr RML Hospital and Postgraduate Institute of Medical Education and research (PGIMER), New Delhi
| |
Collapse
|