1
|
Pfeifle A, Anderson-Duvall R, Tamming LA, Zhang W, Thulasi Raman SN, Gravel C, Wu J, Coatsworth H, Voordouw MJ, Zhang X, Johnston MJW, Chen W, Sauve S, Wang L, Li X. Borrelia burgdorferi Strain-Specific Differences in Mouse Infectivity and Pathology. Pathogens 2025; 14:352. [PMID: 40333117 PMCID: PMC12029986 DOI: 10.3390/pathogens14040352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 05/09/2025] Open
Abstract
Lyme disease (LD), caused by infection with the tick-borne bacteria, Borrelia burgdorferi, is associated with a wide array of symptoms in human patients. Variations in clinical manifestations are thought to be influenced by genetic differences among B. burgdorferi strains. In this study, we evaluated the infectivity, tissue bacterial load, pathology, and immunogenicity of five strains of B. burgdorferi sensu stricto (297 Ah130, Bb16-54, B31-A3, Bb16-126, JD1) in female C3H/HeN mice at three infectious doses (104, 105, 106 spirochetes). We found that strains Bb16-126 and JD1 were the most infectious, resulting in 100% infection across all the tested doses. Strain Bb16-126 caused the highest bacterial burden in the heart tissue and significant carditis, whereas JD1 exhibited the lowest spirochete load in the heart and minimal carditis. In comparison, strain B31-A3 demonstrated the highest abundance in the tibiotarsal joint. Infection with all the strains induced severe lymph node hyperplasia, with JD1 producing the greatest increase in cellularity. Using a diagnostic C6 peptide ELISA, all the strains induced significant anti-C6 IgM and IgG antibody titers at 14 days post-infection; however, strain B31-A3 elicited the highest anti-C6 IgM titers. Our findings demonstrate the importance of strain diversity in shaping B. burgdorferi pathogenesis in a mouse model and provide insights for developing strain-specific diagnostic, therapeutic, and vaccine strategies.
Collapse
Affiliation(s)
- Annabelle Pfeifle
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON K1A 0K9, Canada; (A.P.); (R.A.-D.); (L.A.T.); (W.Z.); (S.N.T.R.); (C.G.); (J.W.); (X.Z.); (M.J.W.J.); (S.S.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Rose Anderson-Duvall
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON K1A 0K9, Canada; (A.P.); (R.A.-D.); (L.A.T.); (W.Z.); (S.N.T.R.); (C.G.); (J.W.); (X.Z.); (M.J.W.J.); (S.S.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Levi A. Tamming
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON K1A 0K9, Canada; (A.P.); (R.A.-D.); (L.A.T.); (W.Z.); (S.N.T.R.); (C.G.); (J.W.); (X.Z.); (M.J.W.J.); (S.S.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Wanyue Zhang
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON K1A 0K9, Canada; (A.P.); (R.A.-D.); (L.A.T.); (W.Z.); (S.N.T.R.); (C.G.); (J.W.); (X.Z.); (M.J.W.J.); (S.S.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Sathya N. Thulasi Raman
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON K1A 0K9, Canada; (A.P.); (R.A.-D.); (L.A.T.); (W.Z.); (S.N.T.R.); (C.G.); (J.W.); (X.Z.); (M.J.W.J.); (S.S.)
| | - Caroline Gravel
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON K1A 0K9, Canada; (A.P.); (R.A.-D.); (L.A.T.); (W.Z.); (S.N.T.R.); (C.G.); (J.W.); (X.Z.); (M.J.W.J.); (S.S.)
| | - Jianguo Wu
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON K1A 0K9, Canada; (A.P.); (R.A.-D.); (L.A.T.); (W.Z.); (S.N.T.R.); (C.G.); (J.W.); (X.Z.); (M.J.W.J.); (S.S.)
| | - Heather Coatsworth
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3M4, Canada;
| | - Maarten J. Voordouw
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Xu Zhang
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON K1A 0K9, Canada; (A.P.); (R.A.-D.); (L.A.T.); (W.Z.); (S.N.T.R.); (C.G.); (J.W.); (X.Z.); (M.J.W.J.); (S.S.)
- School of Pharmaceutical Sciences, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Michael J. W. Johnston
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON K1A 0K9, Canada; (A.P.); (R.A.-D.); (L.A.T.); (W.Z.); (S.N.T.R.); (C.G.); (J.W.); (X.Z.); (M.J.W.J.); (S.S.)
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center, National Research Council of Canada, Ottawa, ON K1N 1J1, Canada;
| | - Simon Sauve
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON K1A 0K9, Canada; (A.P.); (R.A.-D.); (L.A.T.); (W.Z.); (S.N.T.R.); (C.G.); (J.W.); (X.Z.); (M.J.W.J.); (S.S.)
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Xuguang Li
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON K1A 0K9, Canada; (A.P.); (R.A.-D.); (L.A.T.); (W.Z.); (S.N.T.R.); (C.G.); (J.W.); (X.Z.); (M.J.W.J.); (S.S.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| |
Collapse
|
2
|
Bonin JL, Torres SR, Marcinkiewicz AL, Duhamel GE, Yang X, Pal U, DiSpirito JM, Nowak TA, Lin YP, MacNamara KC. Impact of E. muris infection on B. burgdorferi-induced joint pathology in mice. Front Immunol 2024; 15:1430419. [PMID: 39229265 PMCID: PMC11368855 DOI: 10.3389/fimmu.2024.1430419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Tick-borne infections are increasing in the United States and around the world. The most common tick-borne disease in the United States is Lyme disease caused by infection with the spirochete Borrelia burgdorferi (Bb), and pathogenesis varies from subclinical to severe. Bb infection is transmitted by Ixodes ticks, which can carry multiple other microbial pathogens, including Ehrlichia species. To address how the simultaneous inoculation of a distinct pathogen impacted the course of Bb-induced disease, we used C57BL/6 (B6) mice which are susceptible to Bb infection but develop only mild joint pathology. While infection of B6 mice with Bb alone resulted in minimal inflammatory responses, mice co-infected with both Bb and the obligate intracellular pathogen Ehrlichia muris (Em) displayed hematologic changes, inflammatory cytokine production, and emergency myelopoiesis similar to what was observed in mice infected only with Em. Moreover, infection of B6 mice with Bb alone resulted in no detectable joint inflammation, whereas mice co-infected with both Em and Bb exhibited significant inflammation of the ankle joint. Our findings support the concept that co-infection with Ehrlichia can exacerbate inflammation, resulting in more severe Bb-induced disease.
Collapse
Affiliation(s)
- Jesse L. Bonin
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Steven R. Torres
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Ashley L. Marcinkiewicz
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, United States
| | - Gerald E. Duhamel
- New York State Animal Health Diagnostic Center and Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Xiuli Yang
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, United States
| | - Utpal Pal
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, United States
| | - Julia M. DiSpirito
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Tristan A. Nowak
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, United States
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, United States
| | - Yi-Pin Lin
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, United States
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, United States
| | - Katherine C. MacNamara
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| |
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
|
Petnicki-Ocwieja T, McCarthy JE, Powale U, Langston PK, Helble JD, Hu LT. Borrelia burgdorferi initiates early transcriptional re-programming in macrophages that supports long-term suppression of inflammation. PLoS Pathog 2023; 19:e1011886. [PMID: 38157387 PMCID: PMC10783791 DOI: 10.1371/journal.ppat.1011886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 01/11/2024] [Accepted: 12/06/2023] [Indexed: 01/03/2024] Open
Abstract
Borrelia burgdorferi (Bb), the causative agent of Lyme disease, establishes a long-term infection and leads to disease manifestations that are the result of host immune responses to the pathogen. Inflammatory manifestations resolve spontaneously despite continued bacterial presence, suggesting inflammatory cells become less responsive over time. This is mimicked by in vitro repeated stimulations, resulting in tolerance, a phenotypic subset of innate immune memory. We performed comparative transcriptional analysis of macrophages in acute and memory states and identified sets of Tolerized, Hyper-Induced, Secondary-Induced and Hyper-Suppressed genes resulting from memory induction, revealing previously unexplored networks of genes affected by cellular re-programming. Tolerized gene families included inflammatory mediators and interferon related genes as would be predicted by the attenuation of inflammation over time. To better understand how cells mediate inflammatory hypo-responsiveness, we focused on genes that could mediate maintenance of suppression, such as Hyper-Induced genes which are up-regulated in memory states. These genes were notably enriched in stress pathways regulated by anti-inflammatory modulators. We examined one of the most highly expressed negative regulators of immune pathways during primary stimulation, Aconitate decarboxylase 1 (Acod1), and tested its effects during in vivo infection with Bb. As predicted by our in vitro model, we show its inflammation-suppressive downstream effects are sustained during in vivo long-term infection with Bb, with a specific role in Lyme carditis.
Collapse
Affiliation(s)
- Tanja Petnicki-Ocwieja
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Julie E. McCarthy
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Urmila Powale
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, Massachusetts, United States of America
| | - P. Kent Langston
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, Massachusetts, United States of America
| | - Jennifer D. Helble
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Linden T. Hu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
5
|
Gaber AM, Mandric I, Nitirahardjo C, Piontkivska H, Hillhouse AE, Threadgill DW, Zelikovsky A, Rogovskyy AS. Comparative transcriptome analysis of Peromyscus leucopus and C3H mice infected with the Lyme disease pathogen. Front Cell Infect Microbiol 2023; 13:1115350. [PMID: 37113133 PMCID: PMC10126474 DOI: 10.3389/fcimb.2023.1115350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Lyme disease (LD), the most prevalent tick-borne disease of humans in the Northern Hemisphere, is caused by the spirochetal bacterium of Borreliella burgdorferi (Bb) sensu lato complex. In nature, Bb spirochetes are continuously transmitted between Ixodes ticks and mammalian or avian reservoir hosts. Peromyscus leucopus mice are considered the primary mammalian reservoir of Bb in the United States. Earlier studies demonstrated that experimentally infected P. leucopus mice do not develop disease. In contrast, C3H mice, a widely used laboratory strain of Mus musculus in the LD field, develop severe Lyme arthritis. To date, the exact tolerance mechanism of P. leucopus mice to Bb-induced infection remains unknown. To address this knowledge gap, the present study has compared spleen transcriptomes of P. leucopus and C3H/HeJ mice infected with Bb strain 297 with those of their respective uninfected controls. Overall, the data showed that the spleen transcriptome of Bb-infected P. leucopus mice was much more quiescent compared to that of the infected C3H mice. To date, the current investigation is one of the few that have examined the transcriptome response of natural reservoir hosts to Borreliella infection. Although the experimental design of this study significantly differed from those of two previous investigations, the collective results of the current and published studies have consistently demonstrated very limited transcriptomic responses of different reservoir hosts to the persistent infection of LD pathogens. Importance The bacterium Borreliella burgdorferi (Bb) causes Lyme disease, which is one of the emerging and highly debilitating human diseases in countries of the Northern Hemisphere. In nature, Bb spirochetes are maintained between hard ticks of Ixodes spp. and mammals or birds. In the United States, the white-footed mouse, Peromyscus leucopus, is one of the main Bb reservoirs. In contrast to humans and laboratory mice (e.g., C3H mice), white-footed mice rarely develop clinical signs (disease) despite being (persistently) infected with Bb. How the white-footed mouse tolerates Bb infection is the question that the present study has attempted to address. Comparisons of genetic responses between Bb-infected and uninfected mice demonstrated that, during a long-term Bb infection, C3H mice reacted much stronger, whereas P. leucopus mice were relatively unresponsive.
Collapse
Affiliation(s)
- Alhussien M. Gaber
- Department of Veterinary Pathobiology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Igor Mandric
- Department of Computer Science, Georgia State University, Atlanta, GA, United States
| | - Caroline Nitirahardjo
- Department of Biological Sciences, and School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Helen Piontkivska
- Department of Biological Sciences, and School of Biomedical Sciences, Kent State University, Kent, OH, United States
- Brain Health Research Institute, Kent State University, Kent, OH, United States
| | - Andrew E. Hillhouse
- Texas A&M Institute for Genomics Sciences and Society, Texas A&M University, College Station, TX, United States
| | - David W. Threadgill
- Texas A&M Institute for Genomics Sciences and Society, Texas A&M University, College Station, TX, United States
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Texas A&M University, College Station, TX, United States
| | - Alex Zelikovsky
- Department of Computer Science, Georgia State University, Atlanta, GA, United States
| | - Artem S. Rogovskyy
- Department of Veterinary Pathobiology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
6
|
Jackson CD, Hilliard KA, Brown CR. 12/15-lipoxygenase activity promotes efficient inflammation resolution in a murine model of Lyme arthritis. Front Immunol 2023; 14:1144172. [PMID: 37143678 PMCID: PMC10151577 DOI: 10.3389/fimmu.2023.1144172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/22/2023] [Indexed: 05/06/2023] Open
Abstract
Infection of C3H/HeJ (C3H) mice with Borrelia burgdorferi results in the development of a robust inflammatory arthritis that peaks around 3-4 weeks post-infection and then spontaneously resolves over the next few weeks. Mice lacking cyclooxygenase (COX)-2 or 5-lipoxygenase (5-LO) activity develop arthritis similar to wild-type mice but display delayed or prolonged joint resolution. Since 12/15-lipoxygenase (12/15-LO) activity is generally down-stream of both COX-2 and 5-LO activity and results in the production of pro-resolution lipids such as lipoxins and resolvins among others, we investigated the impact of 12/15-LO deficiency on the resolution of Lyme arthritis in mice on a C3H background. We found the expression of Alox15 (12/15-LO gene) peaked around 4-weeks post-infection in C3H mice suggesting a role for 12/15-LO in mediating arthritis resolution. A deficiency in 12/15-LO resulted in exacerbated ankle swelling and arthritis severity during the resolution phase without compromising anti-Borrelia antibody production and spirochete clearance. However, clearance of inflammatory cells was impeded. Therapeutic treatment of B. burgdorferi-infected C3H mice with lipoxin A4 (LXA4) near the peak of disease resulted in significantly decreased ankle swelling and a switch of joint macrophages to a resolving phenotype but did not directly impact arthritis severity. These results demonstrate that 12/15-LO lipid metabolites are important components of inflammatory arthritis resolution in murine Lyme arthritis and may be a therapeutic target for treatment of joint edema and pain for Lyme arthritis patients without compromising spirochete clearance.
Collapse
|
7
|
Hamilton PT, Maluenda E, Sarr A, Belli A, Hurry G, Duron O, Plantard O, Voordouw MJ. Borrelia afzelii Infection in the Rodent Host Has Dramatic Effects on the Bacterial Microbiome of Ixodes ricinus Ticks. Appl Environ Microbiol 2021; 87:e0064121. [PMID: 34191531 PMCID: PMC8388833 DOI: 10.1128/aem.00641-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/24/2021] [Indexed: 12/21/2022] Open
Abstract
The microbiome of blood-sucking arthropods can shape their competence to acquire and maintain infections with vector-borne pathogens. We used a controlled study to investigate the interactions between Borrelia afzelii, which causes Lyme borreliosis in Europe, and the bacterial microbiome of Ixodes ricinus, its primary tick vector. We applied a surface sterilization treatment to I. ricinus eggs to produce dysbiosed tick larvae that had a low bacterial abundance and a changed bacterial microbiome compared to those of the control larvae. Dysbiosed and control larvae fed on B. afzelii-infected mice and uninfected control mice, and the engorged larvae were left to molt into nymphs. The nymphs were tested for B. afzelii infection, and their bacterial microbiome underwent 16S rRNA amplicon sequencing. Surprisingly, larval dysbiosis had no effect on the vector competence of I. ricinus for B. afzelii, as the nymphal infection prevalence and the nymphal spirochete load were the same between the dysbiosed group and the control group. The strong effect of egg surface sterilization on the tick bacterial microbiome largely disappeared once the larvae molted into nymphs. The most important determinant of the bacterial microbiome of I. ricinus nymphs was the B. afzelii infection status of the mouse on which the nymphs had fed as larvae. Nymphs that had taken their larval blood meal from an infected mouse had a less abundant but more diverse bacterial microbiome than the control nymphs. Our study demonstrates that vector-borne infections in the vertebrate host shape the microbiome of the arthropod vector. IMPORTANCE Many blood-sucking arthropods transmit pathogens that cause infectious disease. For example, Ixodes ricinus ticks transmit the bacterium Borrelia afzelii, which causes Lyme disease in humans. Ticks also have a microbiome, which can influence their ability to acquire and transmit tick-borne pathogens such as B. afzelii. We sterilized I. ricinus eggs with bleach, and the tick larvae that hatched from these eggs had a dramatically reduced and changed bacterial microbiome compared to that of control larvae. These larvae fed on B. afzelii-infected mice, and the resultant nymphs were tested for B. afzelii and for their bacterial microbiome. We found that our manipulation of the bacterial microbiome had no effect on the ability of the tick larvae to acquire and maintain populations of B. afzelii. In contrast, we found that B. afzelii infection had dramatic effects on the bacterial microbiome of I. ricinus nymphs. Our study demonstrates that infections in the vertebrate host can shape the tick microbiome.
Collapse
Affiliation(s)
| | - Elodie Maluenda
- Laboratory of Ecology and Evolution of Parasites, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | - Anouk Sarr
- Laboratory of Ecology and Evolution of Parasites, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | - Alessandro Belli
- Laboratory of Ecology and Epidemiology of Parasites, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | - Georgia Hurry
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Olivier Duron
- Centre of Research in Ecology and Evolution of Diseases (CREES), Montpellier, France
- MIVEGEC (Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle), Centre National de la Recherche Scientifique (CNRS), Institut pour la Recherche et le Développement (IRD), Université Montpellier (UM), Montpellier, France
| | | | - 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
|
8
|
Cunha Neves JA, Roseira J, Tavares de Sousa H, Machado R. Exploring the Ways of “The Great Imitator”: A Case Report of Syphilitic Hepatitis. GE-PORTUGUESE JOURNAL OF GASTROENTEROLOGY 2021; 29:280-283. [PMID: 35979246 PMCID: PMC9274993 DOI: 10.1159/000516944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/22/2021] [Indexed: 11/19/2022]
Abstract
Introduction Syphilis is a chronic infection caused by Treponema pallidum. Manifestations of this disease are vast, and syphilitic hepatitis is a rarely depicted form of secondary syphilis. Case Presentation We report the case of a 63-year-old man with worsening jaundice, maculopapular rash and perianal discomfort. Proctological examination with anoscopy revealed a perianal gray/white area with millimetric pale granules along the anal canal. Liver function tests showed a mixed pattern. Venereal Disease Research Laboratory, T. pallidum hemagglutination assay and IgM fluorescent treponemal antibody absorbance were positive. The patient was successfully treated with a single dose of penicillin G. Discussion/Conclusion Syphilitic hepatitis is scarcely reported in the literature. Secondary syphilis with mild hepatitis rarely leads to hepatic cytolysis and jaundice. Many signs of secondary syphilis including syphilitic hepatitis may be linked to immune responses initiated during early infection. Over the past decades, evidence has emerged on the importance of innate and adaptive cellular immune responses in the immunopathogenesis of syphilis. This report raises awareness to a clinical entity that should be considered in patients at risk for sexually transmitted diseases, who present with intestinal discomfort or liver dysfunction, as it is a treatable and fully reversible condition.
Collapse
Affiliation(s)
- João A. Cunha Neves
- Department of Gastroenterology, Centro Hospitalar Universitário do Algarve, Portimão, Portugal
- *João A. Cunha Neves,
| | - Joana Roseira
- Department of Gastroenterology, Centro Hospitalar Universitário do Algarve, Portimão, Portugal
| | - Helena Tavares de Sousa
- Department of Gastroenterology, Centro Hospitalar Universitário do Algarve, Portimão, Portugal
- Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
| | - Rui Machado
- Department of Infectious Diseases, Centro Hospitalar Universitário do Algarve, Portimão, Portugal
| |
Collapse
|
9
|
Akoolo L, Djokic V, Rocha SC, Parveen N. Pathogenesis of Borrelia burgdorferi and Babesia microti in TLR4-Competent and TLR4-dysfunctional C3H mice. Cell Microbiol 2021; 23:e13350. [PMID: 33938125 PMCID: PMC8459286 DOI: 10.1111/cmi.13350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 12/28/2022]
Abstract
Toll‐like receptors (TLRs) are a class of membrane‐spanning proteins of host cells. TLR2 and TLR4 are displayed on the surface of macrophages, neutrophils and dendritic cells and recognise structurally conserved microbial signatures defined as Pathogen associated molecular patterns (PAMPs). C3H mice are susceptible to tick‐borne pathogens; Lyme disease causing Borrelia burgdorferi that manifests arthritis and carditis and Apicomplexan protozoan, Babesia microti (Bm) that causes significant parasitemia associated with erythrocytopenia and haemoglobinuria. B. burgdorferi lacks typical TLR4 ligand lipopolysaccharides (LPS) and Bm TLR ligand(s) remain unknown. Only Borrelia lipoproteins that signal through TLR2 are established as PAMPs of these pathogens for TLR2/TLR4. Infection of C3H mice with each pathogen individually resulted in increase in the percentage of splenic B, T and FcR+ cells while their co‐infection significantly diminished levels of these cells and caused increased B. burgdorferi burden in the specific organs. The most pronounced inflammatory arthritis was observed in co‐infected C3H/HeJ mice. Parasitemia levels and kinetics of resolution of Bm in both mice strains were not significantly different. Transfected HEK293 cells showed pronounced signalling by B. burgdorferi through TLR2 and to some extent by TLR4 while Bm and infected erythrocytes did not show any response confirming our results in mice.
Collapse
Affiliation(s)
- Lavoisier Akoolo
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Vitomir Djokic
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Sandra C Rocha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Nikhat Parveen
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
10
|
Thompson D, Watt JA, Brissette CA. Host transcriptome response to Borrelia burgdorferi sensu lato. Ticks Tick Borne Dis 2020; 12:101638. [PMID: 33360384 DOI: 10.1016/j.ttbdis.2020.101638] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022]
Abstract
The host immune response to infection is a well-coordinated system of innate and adaptive immune cells working in concert to prevent the colonization and dissemination of a pathogen. While this typically leads to a beneficial outcome and the suppression of disease pathogenesis, the Lyme borreliosis bacterium, Borrelia burgdorferi sensu lato, can elicit an immune profile that leads to a deleterious state. As B. burgdorferi s.l. produces no known toxins, it is suggested that the immune and inflammatory response of the host are responsible for the manifestation of symptoms, including flu-like symptoms, musculoskeletal pain, and cognitive disorders. The past several years has seen a substantial increase in the use of microarray and sequencing technologies to investigate the transcriptome response induced by B. burgdorferi s.l., thus enabling researchers to identify key factors and pathways underlying the pathophysiology of Lyme borreliosis. In this review we present the major host transcriptional outcomes induced by the bacterium across several studies and discuss the overarching theme of the host inflammatory and immune response, and how it influences the pathology of Lyme borreliosis.
Collapse
Affiliation(s)
- Derick Thompson
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, United States.
| | - John A Watt
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, United States.
| | - Catherine A Brissette
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, United States.
| |
Collapse
|
11
|
Skovsbo Clausen A, Ørbæk M, Renee Pedersen R, Oestrup Jensen P, Lebech AM, Kjaer A. 64Cu-DOTATATE Positron Emission Tomography (PET) of Borrelia Burgdorferi Infection: In Vivo Imaging of Macrophages in Experimental Model of Lyme Arthritis. Diagnostics (Basel) 2020; 10:diagnostics10100790. [PMID: 33036200 PMCID: PMC7601205 DOI: 10.3390/diagnostics10100790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages play a key role in the inflammatory response in Lyme arthritis (LA) and could be a target for diagnosing and monitoring active Borrelia burgdorferi sensu lato (Bb) infection. Therefore, we evaluated the potential of macrophage imaging using 64Cu-DOTATATE PET/CT for detection of Bb activity in a murine model of LA. LA was established in C3H/HeNRj mice infected with Bb B31 strain ML23 pBBE22luc. Bioluminescence imaging was performed to detect migration of spirochetes and inflammatory phagocytes to the joints. Three weeks post-infection 64Cu-DOTATATE PET/CT imaging was performed at an early (3 h) and late (48 h) time point. Plasma levels of a systemic macrophage marker in plasma CD163 were measured. 64Cu-DOTATATE uptake in infected joints was increased at the early (p < 0.0001) and late time points (p = 0.0005) compared with uptake in non-infected controls. No significant difference in plasma levels of CD163 was measured. 64Cu-DOTATATE PET allows for in vivo detection and quantification of LA locally in the joints through non-invasive visualization of macrophages. In contrast, measurement of a systemic macrophage marker in plasma, CD163, did not allow to detect disease. We suggest that 64Cu-DOTATATE PET could become a valuable diagnostic tool for in situ detection of Bb infection-related inflammation.
Collapse
Affiliation(s)
- Anne Skovsbo Clausen
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, DK-2100 Copenhagen, Denmark;
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Mathilde Ørbæk
- Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; (M.Ø.); (A.-M.L.)
| | - Regitze Renee Pedersen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark; (R.R.P.); (P.O.J.)
| | - Peter Oestrup Jensen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark; (R.R.P.); (P.O.J.)
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark
- Center for Rheumatology and Spine Diseases, Institute for Inflammation Research, Copenhagen University Hospital, Rigshospitalet, DK-2100, Copenhagen, Denmark
| | - Anne-Mette Lebech
- Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; (M.Ø.); (A.-M.L.)
- Institute of Clinical Medicine, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, DK-2100 Copenhagen, Denmark;
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Correspondence: ; Tel.: +45-35327504
| |
Collapse
|
12
|
Thompson D, Sorenson J, Greenmyer J, Brissette CA, Watt JA. The Lyme disease bacterium, Borrelia burgdorferi, stimulates an inflammatory response in human choroid plexus epithelial cells. PLoS One 2020; 15:e0234993. [PMID: 32645014 PMCID: PMC7347220 DOI: 10.1371/journal.pone.0234993] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/05/2020] [Indexed: 11/19/2022] Open
Abstract
The main functions of the choroid plexus (CP) are the production of cerebral spinal fluid (CSF), the formation of the blood-CSF barrier, and regulation of immune response. This barrier allows for the exchange of specific nutrients, waste, and peripheral immune cells between the blood stream and CSF. Borrelia burgdorferi (Bb), the causative bacteria of Lyme disease, is associated with neurological complications including meningitis-indeed, Bb has been isolated from the CSF of patients. While it is accepted that B. burgdorferi can enter the central nervous system (CNS) of patients, it is unknown how the bacteria crosses this barrier and how the pathogenesis of the disease leads to the observed symptoms in patients. We hypothesize that during infection Borrelia burgdorferi will induce an immune response conducive to the chemotaxis of immune cells and subsequently lead to a pro-inflammatory state with the CNS parenchyma. Primary human choroid plexus epithelial cells were grown in culture and infected with B. burgdorferi strain B31 MI-16 for 48 hours. RNA was isolated and used for RNA sequencing and RT-qPCR validation. Secreted proteins in the supernatant were analyzed via ELISA. Transcriptome analysis based on RNA sequencing determined a total of 160 upregulated genes and 98 downregulated genes. Pathway and biological process analysis determined a significant upregulation in immune and inflammatory genes specifically in chemokine and interferon related pathways. Further analysis revealed downregulation in genes related to cell to cell junctions including tight and adherens junctions. These results were validated via RT-qPCR. Protein analysis of secreted factors showed an increase in inflammatory chemokines, corresponding to our transcriptome analysis. These data further demonstrate the role of the CP in the modulation of the immune response in a disease state and give insight into the mechanisms by which Borrelia burgdorferi may disseminate into, and act upon, the CNS. Future experiments aim to detail the impact of B. burgdorferi on the blood-CSF-barrier (BCSFB) integrity and inflammatory response within animal models.
Collapse
Affiliation(s)
- Derick Thompson
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Jordyn Sorenson
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Jacob Greenmyer
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Catherine A. Brissette
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - John A. Watt
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| |
Collapse
|
13
|
Zhong X, Lundberg M, Råberg L. Comparison of spleen transcriptomes of two wild rodent species reveals differences in the immune response against Borrelia afzelii. Ecol Evol 2020; 10:6421-6434. [PMID: 32724523 PMCID: PMC7381583 DOI: 10.1002/ece3.6377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/23/2020] [Accepted: 04/22/2020] [Indexed: 01/24/2023] Open
Abstract
Different host species often differ considerably in susceptibility to a given pathogen, but the causes of such differences are rarely known. The natural hosts of the tick-transmitted bacterium Borrelia afzelii, which is one of causative agents of Lyme borreliosis in humans, include a variety of small mammals like voles and mice. Previous studies have shown that B. afzelii-infected bank voles (Myodes glareolus) have about ten times higher bacterial load than infected yellow-necked mice (Apodemus flavicollis), indicating that these two species differ in resistance. In this study, we compared the immune response to B. afzelii infection in these host species by using RNA sequencing to quantify gene expression in spleen. Gene set enrichment analysis (GSEA) showed that several immune pathways were down-regulated in infected animals in both bank voles and yellow-necked mice. Moreover, IFNα response was up-regulated in B. afzelii-infected yellow-necked mice, while IL6 signaling and the complement pathway were down-regulated in infected bank voles; differences in regulation of these three pathways between bank voles and yellow-necked mice could thus contribute to the difference in resistance to B. afzelii between the species. This study provides knowledge of gene expression induced by a zoonotic pathogen in its natural host, and possible species-specific regulation of immune responses associated with resistance.
Collapse
Affiliation(s)
| | | | - Lars Råberg
- Department of BiologyLund UniversityLundSweden
| |
Collapse
|
14
|
Hilliard KA, Blaho VA, Jackson CD, Brown CR. Leukotriene B4 receptor BLT1 signaling is critical for neutrophil apoptosis and resolution of experimental Lyme arthritis. FASEB J 2019; 34:2840-2852. [PMID: 31908031 DOI: 10.1096/fj.201902014r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/04/2019] [Accepted: 12/10/2019] [Indexed: 11/11/2022]
Abstract
Eicosanoids are powerful mediators of inflammation and are known to drive both the progression and regression of arthritis. We previously reported the infection of C3H 5-lipoxygenase (LO)-deficient mice with Borrelia burgdorferi results in prolonged nonresolving Lyme arthritis. Here we define the role of the 5-LO metabolite leukotriene (LT)B4 and its high-affinity receptor, BLT1, in this response. C3H and C3H BLT1-/- mice were infected with B. burgdorferi and arthritis progression was monitored by ankle swelling over time. Similar to 5-LO-/- mice, BLT1-/- mice developed nonresolving Lyme arthritis characterized by increased neutrophils in the joint at later time points than WT mice, but with fewer apoptotic (caspase-3+ ) neutrophils. In vitro, BLT1-/- neutrophils were defective in their ability to undergo apoptosis due to the lack of LTB4 -mediated down-regulation of cAMP, subsequent failure to induce Death-Inducing Signaling Complex (DISC) components, and decreased FasL and CD36 expression. Inhibition of adenylyl cyclase with SQ 22,536 restored BLT1-/- BMN apoptosis, FasL and CD36 expression, and clearance by macrophages. We conclude that LTB4/BLT1 signaling has an unexpected critical role in mediating neutrophil apoptosis via the down-regulation of cAMP. Loss of BLT1 signaling led to defective clearance of neutrophils from the inflamed joint and failed arthritis resolution.
Collapse
Affiliation(s)
- Kinsey A Hilliard
- Department of Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Victoria A Blaho
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - Christa D Jackson
- Department of Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Charles R Brown
- Department of Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
15
|
Maternal Antibodies Provide Bank Voles with Strain-Specific Protection against Infection by the Lyme Disease Pathogen. Appl Environ Microbiol 2019; 85:AEM.01887-19. [PMID: 31540991 DOI: 10.1128/aem.01887-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023] Open
Abstract
Multistrain microbial pathogens often induce strain-specific antibody responses in their vertebrate hosts. Mothers can transmit antibodies to their offspring, which can provide short-term, strain-specific protection against infection. Few experimental studies have investigated this phenomenon for multiple strains of zoonotic pathogens occurring in wildlife reservoir hosts. The tick-borne bacterium Borrelia afzelii causes Lyme disease in Europe and consists of multiple strains that cycle between the tick vector (Ixodes ricinus) and vertebrate hosts, such as the bank vole (Myodes glareolus). We used a controlled experiment to show that female bank voles infected with B. afzelii via tick bite transmit protective antibodies to their offspring. To test the specificity of protection, the offspring were challenged using a natural tick bite challenge with either the maternal strain to which the mothers had been exposed or a different strain. The maternal antibodies protected the offspring against a homologous infectious challenge but not against a heterologous infectious challenge. The offspring from the uninfected control mothers were equally susceptible to both strains. Borrelia outer surface protein C (OspC) is an antigen that is known to induce strain-specific immunity. Maternal antibodies in the offspring reacted more strongly with homologous than with heterologous recombinant OspC, but other antigens may also mediate strain-specific immunity. Our study shows that maternal antibodies provide strain-specific protection against B. afzelii in an ecologically important rodent reservoir host. The transmission of maternal antibodies may have important consequences for the epidemiology of multistrain pathogens in nature.IMPORTANCE Many microbial pathogen populations consist of multiple strains that induce strain-specific antibody responses in their vertebrate hosts. Females can transmit these antibodies to their offspring, thereby providing them with short-term strain-specific protection against microbial pathogens. We investigated this phenomenon using multiple strains of the tick-borne microbial pathogen Borrelia afzelii and its natural rodent reservoir host, the bank vole, as a model system. We found that female bank voles infected with B. afzelii transmitted to their offspring maternal antibodies that provided highly efficient but strain-specific protection against a natural tick bite challenge. The transgenerational transfer of antibodies could be a mechanism that maintains the high strain diversity of this tick-borne pathogen in nature.
Collapse
|
16
|
Colonization and pathology of Borrelia afzelii in its natural hosts. Ticks Tick Borne Dis 2019; 10:822-827. [PMID: 31005618 DOI: 10.1016/j.ttbdis.2019.03.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 03/12/2019] [Accepted: 03/24/2019] [Indexed: 01/01/2023]
Abstract
Studies of Borrelia burgdorferi sensu lato in laboratory mice and humans have shown that spirochaetes disseminate from the site of infection (skin) to internal tissues, and cause various pathological effects. However, less is known about colonization and pathology of Lyme borreliosis spirochaetes in their natural hosts. In the present study, we assessed the colonization and manifestations during B. afzelii infection in reservoir hosts (yellow-necked mouse, Apodemus flavicollis; bank vole, Myodes glareolus; common shrew, Sorex araneus) infected in the wild. The infection prevalence and bacterial load was measured in skin (ear), joints and heart by quantitative PCR, and pathology in infected joints was evaluated by histology. The prevalence of B. afzelii was higher in skin than in joints and heart, but most animals that were positive in skin were also positive in internal tissues, and there was no difference between species in tissue-specific prevalence. Thus, spirochaetes disseminated from skin to other tissues in a similar way in all species. The bacterial load varied among host species and among different tissues within the same host species. In the case of skin and joints, bank voles and common shrews had higher bacterial loads than yellow-necked mice. In hearts, voles had higher bacterial loads than shrews and mice. Histological analyses showed no inflammation in joints of infected animals when compared to controls. We conclude that B. afzelii disseminates to internal tissues in natural hosts, but that levels of colonization vary between both species and tissues. There is as yet little evidence for pathological effects in natural hosts.
Collapse
|
17
|
Hilliard KA, Brown CR. Treatment of Borrelia burgdorferi-Infected Mice with Apoptotic Cells Attenuates Lyme Arthritis via PPAR-γ. THE JOURNAL OF IMMUNOLOGY 2019; 202:1798-1806. [PMID: 30700583 DOI: 10.4049/jimmunol.1801179] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/02/2019] [Indexed: 12/25/2022]
Abstract
Infection of mice with Borrelia burgdorferi causes an inflammatory arthritis that peaks 3-4 wk postinfection and then spontaneously resolves. Although the recruitment of neutrophils is known to drive the development of arthritis, mechanisms of disease resolution remain unclear. Efficient clearance of apoptotic cells (AC) is likely an important component of arthritis resolution. In this article, we show the number of AC increases in the joints of B. burgdorferi-infected mice around day 21 postinfection and peaks around day 28. Injection of AC directly into the ankles of B. burgdorferi-infected mice limited ankle swelling but had no effect on spirochete clearance or arthritis severity scores. In vitro, addition of AC to bone marrow macrophage cultures decreased B. burgdorferi-induced TNF-α and KC and increased IL-10. In addition, phagocytosis of B. burgdorferi and neutrophil migration to LTB4 were inhibited by AC. Exogenous AC caused an increase in peroxisome proliferator-activated receptor-γ (PPAR-γ) expression both in vitro and in vivo during B. burgdorferi infection. The PPAR-γ agonist rosiglitazone elicited similar changes in macrophage cytokine production and neutrophil migration as exogenous AC. Addition of the PPAR-γ antagonist GW 9662 abrogated the effects of AC in vitro. Injection of rosiglitazone directly into the tibiotarsal joints of B. burgdorferi-infected mice decreased ankle swelling and immune cell recruitment, similar to the injection of AC. These results suggest that clearance of AC plays a role in the resolution of inflammation during experimental Lyme arthritis through the activation of PPAR-γ. PPAR-γ agonists, such as rosiglitazone, may therefore be effective treatments for inducing arthritis resolution.
Collapse
Affiliation(s)
- Kinsey A Hilliard
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211
| | - Charles R Brown
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211
| |
Collapse
|
18
|
Induction of Interleukin 10 by Borrelia burgdorferi Is Regulated by the Action of CD14-Dependent p38 Mitogen-Activated Protein Kinase and cAMP-Mediated Chromatin Remodeling. Infect Immun 2018; 86:IAI.00781-17. [PMID: 29311239 DOI: 10.1128/iai.00781-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/31/2017] [Indexed: 12/22/2022] Open
Abstract
Host genotype influences the severity of murine Lyme borreliosis, caused by the spirochetal bacterium Borrelia burgdorferi C57BL/6 (B6) mice develop mild Lyme arthritis, whereas C3H/HeN (C3H) mice develop severe Lyme arthritis. Differential expression of interleukin 10 (IL-10) has long been associated with mouse strain differences in Lyme pathogenesis; however, the underlying mechanism(s) of this genotype-specific IL-10 regulation remained elusive. Herein we reveal a cAMP-mediated mechanism of IL-10 regulation in B6 macrophages that is substantially diminished in C3H macrophages. Under cAMP and CD14-p38 mitogen-activated protein kinase (MAPK) signaling, B6 macrophages stimulated with B. burgdorferi produce increased amounts of IL-10 and decreased levels of arthritogenic cytokines, including tumor necrosis factor (TNF). cAMP relaxes chromatin, while p38 increases binding of the transcription factors signal transducer and activator of transcription 3 (STAT3) and specific protein 1 (SP1) to the IL-10 promoter, leading to increased IL-10 production in B6 bone marrow-derived monocytes (BMDMs). Conversely, macrophages derived from arthritis-susceptible C3H mice possess significantly less endogenous cAMP, produce less IL-10, and thus are ill equipped to mitigate the damaging consequences of B. burgdorferi-induced TNF. Intriguingly, an altered balance between anti-inflammatory and proinflammatory cytokines and CD14-dependent regulatory mechanisms also is operative in primary human peripheral blood-derived monocytes, providing potential insight into the clinical spectrum of human Lyme disease. In line with this notion, we have demonstrated that cAMP-enhancing drugs increase IL-10 production in myeloid cells, thus curtailing inflammation associated with murine Lyme borreliosis. Discovery of novel treatments or repurposing of FDA-approved cAMP-modulating medications may be a promising avenue for treatment of patients with adverse clinical outcomes, including certain post-Lyme complications, in whom dysregulated immune responses may play a role.
Collapse
|
19
|
Embers ME, Hasenkampf NR, Jacobs MB, Tardo AC, Doyle-Meyers LA, Philipp MT, Hodzic E. Variable manifestations, diverse seroreactivity and post-treatment persistence in non-human primates exposed to Borrelia burgdorferi by tick feeding. PLoS One 2017; 12:e0189071. [PMID: 29236732 PMCID: PMC5728523 DOI: 10.1371/journal.pone.0189071] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/17/2017] [Indexed: 01/21/2023] Open
Abstract
The efficacy and accepted regimen of antibiotic treatment for Lyme disease has been a point of significant contention among physicians and patients. While experimental studies in animals have offered evidence of post-treatment persistence of Borrelia burgdorferi, variations in methodology, detection methods and limitations of the models have led to some uncertainty with respect to translation of these results to human infection. With all stages of clinical Lyme disease having previously been described in nonhuman primates, this animal model was selected in order to most closely mimic human infection and response to treatment. Rhesus macaques were inoculated with B. burgdorferi by tick bite and a portion were treated with recommended doses of doxycycline for 28 days at four months post-inoculation. Signs of infection, clinical pathology, and antibody responses to a set of five antigens were monitored throughout the ~1.2 year study. Persistence of B. burgdorferi was evaluated using xenodiagnosis, bioassays in mice, multiple methods of molecular detection, immunostaining with polyclonal and monoclonal antibodies and an in vivo culture system. Our results demonstrate host-dependent signs of infection and variation in antibody responses. In addition, we observed evidence of persistent, intact, metabolically-active B. burgdorferi after antibiotic treatment of disseminated infection and showed that persistence may not be reflected by maintenance of specific antibody production by the host.
Collapse
Affiliation(s)
- Monica E. Embers
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
- * E-mail:
| | - Nicole R. Hasenkampf
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | - Mary B. Jacobs
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | - Amanda C. Tardo
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | - Lara A. Doyle-Meyers
- Division of Veterinary Medicine, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | - Mario T. Philipp
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States of America
| | - Emir Hodzic
- Center for Comparative Medicine, Schools of Medicine and Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| |
Collapse
|
20
|
Christodoulides A, Boyadjian A, Kelesidis T. Spirochetal Lipoproteins and Immune Evasion. Front Immunol 2017; 8:364. [PMID: 28424696 PMCID: PMC5372817 DOI: 10.3389/fimmu.2017.00364] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 03/14/2017] [Indexed: 12/28/2022] Open
Abstract
Spirochetes are a major threat to public health. However, the exact pathogenesis of spirochetal diseases remains unclear. Spirochetes express lipoproteins that often determine the cross talk between the host and spirochetes. Lipoproteins are pro-inflammatory, modulatory of immune responses, and enable the spirochetes to evade the immune system. In this article, we review the modulatory effects of spirochetal lipoproteins related to immune evasion. Understanding lipoprotein-induced immunomodulation will aid in elucidating innate pathogenesis processes and subsequent adaptive mechanisms potentially relevant to spirochetal disease vaccine development and treatment.
Collapse
Affiliation(s)
- Alexei Christodoulides
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Ani Boyadjian
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Theodoros Kelesidis
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
21
|
Shi Y, Xu L, Tang J, Fang L, Ma S, Ma X, Nie J, Pi X, Qiu A, Zhuang S, Liu N. Inhibition of HDAC6 protects against rhabdomyolysis-induced acute kidney injury. Am J Physiol Renal Physiol 2017; 312:F502-F515. [PMID: 28052874 DOI: 10.1152/ajprenal.00546.2016] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/29/2016] [Accepted: 12/29/2016] [Indexed: 12/18/2022] Open
Abstract
Histone deacetylase 6 (HDAC6) inhibition has been reported to protect against ischemic stroke and prolong survival after sepsis in animal models. However, it remains unknown whether HDAC6 inhibition offers a renoprotective effect after acute kidney injury (AKI). In this study, we examined the effect of tubastatin A (TA), a highly selective inhibitor of HDAC6, on AKI in a murine model of glycerol (GL) injection-induced rhabdomyolysis. Following GL injection, the mice developed severe acute tubular injury as indicated by renal dysfunction; expression of neutrophil gelatinase-associated lipocalin (NGAL), an injury marker of renal tubules; and an increase of TdT-mediated dUTP nick-end labeling (TUNEL)-positive tubular cells. These changes were companied by increased HDAC6 expression in the cytoplasm of renal tubular cells. Administration of TA significantly reduced serum creatinine and blood urea nitrogen levels as well as attenuated renal tubular damage in injured kidneys. HDAC6 inhibition also resulted in decreased expression of NGAL, reduced apoptotic cell, and inactivated caspase-3 in the kidney after acute injury. Moreover, injury to the kidney increased phosphorylation of nuclear factor (NF)-κB and expression of multiple cytokines/chemokines including tumor necrotic factor-α and interleukin-6 and monocyte chemoattractant protein-1, as well as macrophage infiltration. Treatment with TA attenuated all those responses. Finally, HDAC6 inhibition reduced the level of oxidative stress by suppressing malondialdehyde (MDA) and preserving expression of superoxide dismutase (SOD) in the injured kidney. Collectively, these data indicate that HDAC6 contributes to the pathogenesis of rhabdomyolysis-induced AKI and suggest that HDAC6 inhibitors have therapeutic potential for AKI treatment.
Collapse
Affiliation(s)
- Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liuqing Xu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinhua Tang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lu Fang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuchen Ma
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyan Ma
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing Nie
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaoling Pi
- Department of Internal Medicine, Pudong New District Gongli Hospital, Shanghai, China
| | - Andong Qiu
- School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai, China; and
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medicine, Rhode Island Hospital and Brown University School of Medicine, Providence, Rhode Island
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China;
| |
Collapse
|
22
|
Russell TM, Tang X, Goldstein JM, Bagarozzi D, Johnson BJB. The salt-sensitive structure and zinc inhibition of Borrelia burgdorferi protease BbHtrA. Mol Microbiol 2015; 99:586-96. [PMID: 26480895 DOI: 10.1111/mmi.13251] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2015] [Indexed: 11/28/2022]
Abstract
HtrA serine proteases are highly conserved and essential ATP-independent proteases with chaperone activity. Bacteria express a variable number of HtrA homologues that contribute to the virulence and pathogenicity of bacterial pathogens. Lyme disease spirochetes possess a single HtrA protease homologue, Borrelia burgdorferi HtrA (BbHtrA). Previous studies established that, like the human orthologue HtrA1, BbHtrA is proteolytically active against numerous extracellular proteins in vitro. In this study, we utilized size exclusion chromatography and blue native polyacrylamide gel electrophoresis (BN-PAGE) to demonstrate BbHtrA oligomeric structures that were substrate independent and salt sensitive. Examination of the influence of transition metals on the activity of BbHtrA revealed that this protease is inhibited by Zn(2+) > Cu(2+) > Mn(2+). Extending this analysis to two other HtrA proteases, E. coli DegP and HtrA1, revealed that all three HtrA proteases were reversibly inhibited by ZnCl2 at all micro molar concentrations examined. Commercial inhibitors for HtrA proteases are not available and physiologic HtrA inhibitors are unknown. Our observation of conserved zinc inhibition of HtrA proteases will facilitate structural and functional studies of additional members of this important class of proteases.
Collapse
Affiliation(s)
- Theresa M Russell
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Fort Collins, CO, USA
| | - Xiaoling Tang
- Division of Scientific Resources, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Atlanta, GA, USA
| | - Jason M Goldstein
- Division of Scientific Resources, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Atlanta, GA, USA
| | - Dennis Bagarozzi
- Division of Scientific Resources, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Atlanta, GA, USA
| | - Barbara J B Johnson
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Fort Collins, CO, USA
| |
Collapse
|
23
|
Jung S, Schickel JN, Kern A, Knapp AM, Eftekhari P, Da Silva S, Jaulhac B, Brink R, Soulas-Sprauel P, Pasquali JL, Martin T, Korganow AS. Chronic bacterial infection activates autoreactive B cells and induces isotype switching and autoantigen-driven mutations. Eur J Immunol 2015; 46:131-46. [PMID: 26474536 DOI: 10.1002/eji.201545810] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/25/2015] [Accepted: 10/13/2015] [Indexed: 01/05/2023]
Abstract
The links between infections and the development of B-cell-mediated autoimmune diseases are still unclear. In particular, it has been suggested that infection-induced stimulation of innate immune sensors can engage low affinity autoreactive B lymphocytes to mature and produce mutated IgG pathogenic autoantibodies. To test this hypothesis, we established a new knock-in mouse model in which autoreactive B cells could be committed to an affinity maturation process. We show that a chronic bacterial infection allows the activation of such B cells and the production of nonmutated IgM autoantibodies. Moreover, in the constitutive presence of their soluble antigen, some autoreactive clones are able to acquire a germinal center phenotype, to induce Aicda gene expression and to introduce somatic mutations in the IgG heavy chain variable region on amino acids forming direct contacts with the autoantigen. Paradoxically, only lower affinity variants are detected, which strongly suggests that higher affinity autoantibodies secreting B cells are counterselected. For the first time, we demonstrate in vivo that a noncross-reactive infectious agent can activate and induce autoreactive B cells to isotype switching and autoantigen-driven mutations, but on a nonautoimmune background, tolerance mechanisms prevent the formation of consequently dangerous autoimmunity.
Collapse
Affiliation(s)
- Sophie Jung
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/ Laboratory of Excellence Medalis, Molecular and Cellular Biology Institute (IBMC), Strasbourg, France.,"Pôle de Médecine et de Chirurgie Bucco-Dentaires", University Hospital and Biological Sciences, Faculty of Dentistry, University of Strasbourg, Strasbourg, France
| | - Jean-Nicolas Schickel
- Department of Immunobiology, Yale University School of Medicine, Connecticut, New Haven, CT, USA
| | - Aurélie Kern
- EA 7290, "Early Bacterial Virulence", Bacteriology Institute, Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Anne-Marie Knapp
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/ Laboratory of Excellence Medalis, Molecular and Cellular Biology Institute (IBMC), Strasbourg, France
| | - Pierre Eftekhari
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/ Laboratory of Excellence Medalis, Molecular and Cellular Biology Institute (IBMC), Strasbourg, France
| | - Sylvia Da Silva
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/ Laboratory of Excellence Medalis, Molecular and Cellular Biology Institute (IBMC), Strasbourg, France
| | - Benoît Jaulhac
- EA 7290, "Early Bacterial Virulence", Bacteriology Institute, Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Robert Brink
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia.,St. Vincent's Clinical School, University of New South Wales, Darlinghurst, Sydney, Australia
| | - Pauline Soulas-Sprauel
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/ Laboratory of Excellence Medalis, Molecular and Cellular Biology Institute (IBMC), Strasbourg, France.,Faculty of Pharmacy, University of Strasbourg, Illkirch-Graffenstaden, France.,Department of Clinical Immunology, University Hospital, Strasbourg, France
| | - Jean-Louis Pasquali
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/ Laboratory of Excellence Medalis, Molecular and Cellular Biology Institute (IBMC), Strasbourg, France.,Department of Clinical Immunology, University Hospital, Strasbourg, France.,Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Thierry Martin
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/ Laboratory of Excellence Medalis, Molecular and Cellular Biology Institute (IBMC), Strasbourg, France.,Department of Clinical Immunology, University Hospital, Strasbourg, France.,Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Anne-Sophie Korganow
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/ Laboratory of Excellence Medalis, Molecular and Cellular Biology Institute (IBMC), Strasbourg, France.,Department of Clinical Immunology, University Hospital, Strasbourg, France.,Faculty of Medicine, University of Strasbourg, Strasbourg, France
| |
Collapse
|
24
|
Outer surface protein OspC is an antiphagocytic factor that protects Borrelia burgdorferi from phagocytosis by macrophages. Infect Immun 2015; 83:4848-60. [PMID: 26438793 DOI: 10.1128/iai.01215-15] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/27/2015] [Indexed: 12/15/2022] Open
Abstract
Outer surface protein C (OspC) is one of the major lipoproteins expressed on the surface of Borrelia burgdorferi during tick feeding and the early phase of mammalian infection. OspC is required for B. burgdorferi to establish infection in both immunocompetent and SCID mice and has been proposed to facilitate evasion of innate immune defenses. However, the exact biological function of OspC remains elusive. In this study, we showed that the ospC-deficient spirochete could not establish infection in NOD-scid IL2rγ(null) mice that lack B cells, T cells, NK cells, and lytic complement. The ospC mutant also could not establish infection in anti-Ly6G-treated SCID and C3H/HeN mice (depletion of neutrophils). However, depletion of mononuclear phagocytes at the skin site of inoculation in SCID and C3H/HeN mice allowed the ospC mutant to establish infection in vivo. In phagocyte-depleted mice, the ospC mutant was able to colonize the joints and triggered neutrophilia during dissemination. Furthermore, we found that phagocytosis of green fluorescent protein (GFP)-expressing ospC mutant spirochetes by murine peritoneal macrophages and human THP-1 macrophage-like cells, but not in PMN-HL60, was significantly higher than parental wild-type B. burgdorferi strains, suggesting that OspC has an antiphagocytic property. In addition, overproduction of OspC in spirochetes also decreased the uptake of spirochetes by murine peritoneal macrophages. Together, our findings provide evidence that mononuclear phagocytes play a key role in clearance of the ospC mutant and that OspC promotes spirochetes' evasion of macrophages during early Lyme borreliosis.
Collapse
|
25
|
A short-term Borrelia burgdorferi infection model identifies tissue tropisms and bloodstream survival conferred by adhesion proteins. Infect Immun 2015; 83:3184-94. [PMID: 26015482 DOI: 10.1128/iai.00349-15] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/22/2015] [Indexed: 12/18/2022] Open
Abstract
Borrelia burgdorferi, the causative agent of Lyme disease in the United States, is able to persist in the joint, heart, skin, and central nervous system for the lifetime of its mammalian host. Borrelia species achieve dissemination to distal sites in part by entry into and travel within the bloodstream. Much work has been performed in vitro describing the roles of many B. burgdorferi outer surface proteins in adhesion to host cell surface proteins and extracellular matrix components, although the biological relevance of these interactions is only beginning to be explored in vivo. A need exists in the field for an in vivo model to define the biological roles of B. burgdorferi adhesins in tissue-specific vascular interactions. We have developed an in vivo model of vascular interaction of B. burgdorferi in which the bacteria are injected intravenously and allowed to circulate for 1 h. This model has shown that the fibronectin binding protein BB0347 has a tropism for joint tissue. We also have shown an importance of the integrin binding protein, P66, in binding to vasculature of the ear and heart. This model also revealed unexpected roles for Borrelia adhesins BBK32 and OspC in bacterial burdens in the bloodstream. The intravenous inoculation model of short-term infection provides new insights into critical B. burgdorferi interactions with the host required for initial survival and tissue colonization.
Collapse
|
26
|
Infection of Interleukin 17 Receptor A-Deficient C3H Mice with Borrelia burgdorferi Does Not Affect Their Development of Lyme Arthritis and Carditis. Infect Immun 2015; 83:2882-8. [PMID: 25939508 DOI: 10.1128/iai.00533-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 04/24/2015] [Indexed: 12/12/2022] Open
Abstract
Recently, a number of studies have reported the presence of interleukin 17 (IL-17) in patients with Lyme disease, and several murine studies have suggested a role for this cytokine in the development of Lyme arthritis. However, the role of IL-17 has not been studied using the experimental Lyme borreliosis model of infection of C3H mice with Borrelia burgdorferi. In the current study, we investigated the role of IL-17 in the development of experimental Lyme borreliosis by infecting C3H mice devoid of the common IL-17 receptor A subunit (IL-17RA) and thus deficient in most IL-17 signaling. Infection of both C3H and C3H IL-17RA(-/-) mice led to the production of high levels of IL-17 in the serum, low levels in the heart tissue, and no detectable IL-17 in the joint tissue. The development and severity of arthritis and carditis in the C3H IL-17RA(-/-) mice were similar to what was seen in wild-type C3H mice. In addition, development of antiborrelia antibodies and clearance of spirochetes from tissues were similar for the two mouse strains. These results demonstrate a limited role for IL-17 signaling through IL-17RA in the development of disease following infection of C3H mice with B. burgdorferi.
Collapse
|
27
|
Macrophage Polarization during Murine Lyme Borreliosis. Infect Immun 2015; 83:2627-35. [PMID: 25870230 DOI: 10.1128/iai.00369-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 04/09/2015] [Indexed: 12/14/2022] Open
Abstract
Infection of C3H mice with Borrelia burgdorferi, the causative agent of Lyme disease, reliably produces an infectious arthritis and carditis that peak around 3 weeks postinfection and then spontaneously resolve. Macrophage polarization has been suggested to drive inflammation, the clearance of bacteria, and tissue repair and resolution in a variety of infectious disease models. During Lyme disease it is clear that macrophages are capable of clearing Borrelia spirochetes and exhausted neutrophils; however, the role of macrophage phenotype in disease development or resolution has not been studied. Using classical (NOS2) and alternative (CD206) macrophage subset-specific markers, we determined the phenotype of F4/80(+) macrophages within the joints and heart throughout the infection time course. Within the joint, CD206(+) macrophages dominated throughout the course of infection, and NOS2(+) macrophage numbers became elevated only during the peak of inflammation. We also found dual NOS2(+) CD206(+) macrophages which increased during resolution. In contrast to findings for the ankle joints, numbers of NOS2(+) and CD206(+) macrophages in the heart were similar at the peak of inflammation. 5-Lipoxygenase-deficient (5-LOX(-/-)) mice, which display a failure of Lyme arthritis resolution, recruited fewer F4/80(+) cells to the infected joints and heart, but macrophage subset populations were unchanged. These results highlight differences in the inflammatory infiltrates during Lyme arthritis and carditis and demonstrate the coexistence of multiple macrophage subsets within a single inflammatory site.
Collapse
|
28
|
Abstract
The Lyme disease spirochete, Borrelia burgdorferi, controls protein expression patterns during its tick-mammal infection cycle. Earlier studies demonstrated that B. burgdorferi synthesizes 4,5-dihydroxy-2,3-pentanedione (autoinducer-2 [AI-2]) and responds to AI-2 by measurably changing production of several infection-associated proteins. luxS mutants, which are unable to produce AI-2, exhibit altered production of several proteins. B. burgdorferi cannot utilize the other product of LuxS, homocysteine, indicating that phenotypes of luxS mutants are not due to the absence of that molecule. Although a previous study found that a luxS mutant was capable of infecting mice, a critical caveat to those results is that bacterial loads were not quantified. To more precisely determine whether LuxS serves a role in mammalian infection, mice were simultaneously inoculated with congenic wild-type and luxS strains, and bacterial numbers were assessed using quantitative PCR. The wild-type bacteria substantially outcompeted the mutants, suggesting that LuxS performs a significant function during mammalian infection. These data also provide further evidence that nonquantitative infection studies do not necessarily provide conclusive results and that regulatory factors may not make all-or-none, black-or-white contributions to infectivity.
Collapse
|
29
|
Kelesidis T. The Cross-Talk between Spirochetal Lipoproteins and Immunity. Front Immunol 2014; 5:310. [PMID: 25071771 PMCID: PMC4075078 DOI: 10.3389/fimmu.2014.00310] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 06/17/2014] [Indexed: 12/11/2022] Open
Abstract
Spirochetal diseases such as syphilis, Lyme disease, and leptospirosis are major threats to public health. However, the immunopathogenesis of these diseases has not been fully elucidated. Spirochetes interact with the host through various structural components such as lipopolysaccharides (LPS), surface lipoproteins, and glycolipids. Although spirochetal antigens such as LPS and glycolipids may contribute to the inflammatory response during spirochetal infections, spirochetes such as Treponema pallidum and Borrelia burgdorferi lack LPS. Lipoproteins are most abundant proteins that are expressed in all spirochetes and often determine how spirochetes interact with their environment. Lipoproteins are pro-inflammatory, may regulate responses from both innate and adaptive immunity and enable the spirochetes to adhere to the host or the tick midgut or to evade the immune system. However, most of the spirochetal lipoproteins have unknown function. Herein, the immunomodulatory effects of spirochetal lipoproteins are reviewed and are grouped into two main categories: effects related to immune evasion and effects related to immune activation. Understanding lipoprotein-induced immunomodulation will aid in elucidating innate immunopathogenesis processes and subsequent adaptive mechanisms potentially relevant to spirochetal disease vaccine development and to inflammatory events associated with spirochetal diseases.
Collapse
Affiliation(s)
- Theodoros Kelesidis
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles , Los Angeles, CA , USA
| |
Collapse
|
30
|
Cervantes JL, Hawley KL, Benjamin SJ, Weinerman B, Luu SM, Salazar JC. Phagosomal TLR signaling upon Borrelia burgdorferi infection. Front Cell Infect Microbiol 2014; 4:55. [PMID: 24904837 PMCID: PMC4033037 DOI: 10.3389/fcimb.2014.00055] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 04/09/2014] [Indexed: 12/31/2022] Open
Abstract
Internalization and degradation of live Bb within phagosomal compartments of monocytes, macrophages and dendritic cells (DCs), allows for the release of lipoproteins, nucleic acids and other microbial products, triggering a broad and robust inflammatory response. Toll-like receptors (TLRs) are key players in the recognition of spirochetal ligands from whole viable organisms (i.e., vita-PAMPs). Herein we will review the role of endosomal TLRs in the response to the Lyme disease spirochete.
Collapse
Affiliation(s)
- Jorge L Cervantes
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA ; Division of Infectious Diseases, Connecticut Children's Medical Center Hartford, CT, USA
| | - Kelly L Hawley
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA ; Division of Infectious Diseases, Connecticut Children's Medical Center Hartford, CT, USA
| | - Sarah J Benjamin
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA
| | - Bennett Weinerman
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA
| | - Stephanie M Luu
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center Farmington, CT, USA
| | - Juan C Salazar
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA ; Division of Infectious Diseases, Connecticut Children's Medical Center Hartford, CT, USA ; Department of Immunology, University of Connecticut Health Center Farmington, CT, USA
| |
Collapse
|
31
|
Campfield BT, Nolder CL, Marinov A, Bushnell D, Davis A, Spychala C, Hirsch R, Nowalk AJ. Follistatin-like protein 1 is a critical mediator of experimental Lyme arthritis and the humoral response to Borrelia burgdorferi infection. Microb Pathog 2014; 73:70-9. [PMID: 24768929 DOI: 10.1016/j.micpath.2014.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 04/10/2014] [Accepted: 04/15/2014] [Indexed: 12/19/2022]
Abstract
Follistatin-like protein 1 (FSTL-1) has recently been described as a critical mediator of CIA and a marker of disease activity. Lyme arthritis, caused by Borrelia burgdorferi, shares similarities with autoimmune arthritis and the experimental murine model collagen-induced arthritis (CIA). Because FSTL-1 is important in CIA and autoimmune arthritides, and Lyme arthritis shares similarities with CIA, we hypothesized that FSTL-1 may be an important mediator of Lyme arthritis. We demonstrate for the first time that FSTL-1 is induced by B. burgdorferi infection and is required for the development of Lyme arthritis in a murine model, utilizing a gene insertion to generate FSTL-1 hypomorphic mice. Using qPCR and qRT-PCR, we found that despite similar early infectious burden, FSTL-1 hypomorphic mice have improved spirochetal clearance in the face of attenuated arthritis and inflammatory cytokine production. Further, FSTL-1 mediates pathogen-specific antibody production and antigen recognition when assessed by ELISA and one- and two-dimensional immunoblotting. This study is the first to describe a role for FSTL-1 in the development of Lyme arthritis and anti-Borrelia response, and the first to demonstrate a role for FSTL-1 in response to infection, highlighting the potential for FSTL-1 as a target in the treatment of B. burgdorferi infection.
Collapse
Affiliation(s)
- Brian T Campfield
- Department of Pediatrics, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Christi L Nolder
- Department of Pediatrics, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Anthony Marinov
- Department of Pediatrics, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Daniel Bushnell
- Department of Pediatrics, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Amy Davis
- Department of Pathology, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Caressa Spychala
- Department of Pediatrics, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Raphael Hirsch
- Department of Pediatrics, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Andrew J Nowalk
- Department of Pediatrics, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| |
Collapse
|
32
|
Chung Y, Zhang N, Wooten RM. Borrelia burgdorferi elicited-IL-10 suppresses the production of inflammatory mediators, phagocytosis, and expression of co-stimulatory receptors by murine macrophages and/or dendritic cells. PLoS One 2013; 8:e84980. [PMID: 24367705 PMCID: PMC3868605 DOI: 10.1371/journal.pone.0084980] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 11/27/2013] [Indexed: 11/18/2022] Open
Abstract
Borrelia burgdorferi (Bb) is a tick-borne spirochete that is the causative agent for Lyme disease. Our previous studies indicate that virulent Bb can potently enhance IL-10 production by macrophages (MØs) and that blocking IL-10 production significantly enhances bacterial clearance. We hypothesize that skin-associated APC types, such as MØs and dendritic cells (DCs) are potent producers of IL-10 in response to Bb, which may act in autocrine fashion to suppress APC responses critical for efficient Bb clearance. Our goal is to delineate which APC immune functions are dysregulated by Bb-elicited IL-10 using a murine model of Lyme disease. Our in vitro studies indicated that both APCs rapidly produce IL-10 upon exposure to Bb, that these levels inversely correlate with the production of many Lyme-relevant proinflammatory cytokines and chemokines, and that APCs derived from IL-10(-/-) mice produced greater amounts of these proinflammatory mediators than wild-type APCs. Phagocytosis assays determined that Bb-elicited IL-10 levels can diminish Bb uptake and trafficking by MØs, suppresses ROS production, but does not affect NO production; Bb-elicited IL-10 had little effect on phagocytosis, ROS, and NO production by DCs. In general, Bb exposure caused little-to-no upregulation of several critical surface co-stimulatory markers by MØs and DCs, however eliminating Bb-elicited IL-10 allowed a significant upregulation in many of these co-stimulatory receptors. These data indicate that IL-10 elicited from Bb-stimulated MØs and DCs results in decreased production of proinflammatory mediators and co-stimulatory molecules, and suppress phagocytosis-associated events that are important for mediating both innate and adaptive immune responses by APCs.
Collapse
Affiliation(s)
- Yutein Chung
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine, Toledo, Ohio, United States of America
| | - Nan Zhang
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine, Toledo, Ohio, United States of America
| | - R. Mark Wooten
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine, Toledo, Ohio, United States of America
- * E-mail:
| |
Collapse
|
33
|
Presence of Arp specifically contributes to joint tissue edema associated with early-onset Lyme arthritis. Infect Immun 2013; 82:43-51. [PMID: 24101694 DOI: 10.1128/iai.01061-13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Antiserum to the Borrelia burgdorferi arthritis-related protein, Arp, has been shown to prevent or reduce arthritis in immunodeficient mice. To directly investigate the requirement for this lipoprotein in the generation of Lyme arthritis, we utilized targeted deletion to generate a B. burgdorferi clone that lacked only the arp gene locus. Infection of Lyme disease-susceptible C3H/HeN mice with the arp deletion mutant demonstrated significantly reduced tibiotarsal joint swelling during the first 6 weeks of infection compared to a wild-type control. The severity of joint swelling was restored to wild-type levels in mice infected with an arp mutant clone complemented in cis. Interestingly, the reduced swelling of joint tissues exhibited by mice infected with the arp deletion mutant did not directly correspond to reduced underlying arthritis. Histopathology data at 2 weeks postinfection showed some reduction in arthritis severity caused by the arp mutant clone; however, by 8 weeks, no significant difference was observed between joint tissues infected by the wild-type or arp mutant clones. The spirochete load in the joint tissues of mice infected with the arp mutant was found to be greater than that exhibited by the wild-type control. Our findings demonstrate that this lipoprotein contributes to the generation of early-onset joint swelling and suggests that arp expression has a negative secondary effect on total spirochete numbers in joint tissues.
Collapse
|
34
|
Russell TM, Delorey MJ, Johnson BJB. Borrelia burgdorferi BbHtrA degrades host ECM proteins and stimulates release of inflammatory cytokines in vitro. Mol Microbiol 2013; 90:241-51. [PMID: 23980719 DOI: 10.1111/mmi.12377] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2013] [Indexed: 01/08/2023]
Abstract
The Lyme disease spirochaete, Borrelia burgdorferi, causes damage to diverse host tissues and induces inflammation but the mechanisms of injury are poorly understood. We recently reported that a surface-exposed B. burgdorferi protease, which is expressed during human disease and is conserved within the major Lyme disease spirochaete species, degrades the extracellular matrix proteoglycan, aggrecan. Here we demonstrate that BbHtrA also degrades fibronectin and numerous proteoglycans found in skin, joints and neural tissues. BbHtrA degradation of fibronectin released known pro-inflammatory fibronectin fragments FnIII(13-14) and Fnf-29, which may amplify the inflammatory processes triggered by the presence of the bacteria. When this hypothesis was tested directly by exposing chondrocytes to BbHtrA in vitro, inflammatory cytokines (sICAM-1 and IL-6) and chemokines (CXCL1, CCL1, CCL2 and CCL5) that are hallmarks of Lyme disease were induced. These results provide the first evidence that, by utilizing BbHtrA, B. burgdorferi may actively participate in its dissemination and in the tissue damage and inflammation observed in Lyme disease.
Collapse
Affiliation(s)
- Theresa M Russell
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, USA
| | | | | |
Collapse
|
35
|
Gautam A, Dixit S, Embers M, Gautam R, Philipp MT, Singh SR, Morici L, Dennis VA. Different patterns of expression and of IL-10 modulation of inflammatory mediators from macrophages of Lyme disease-resistant and -susceptible mice. PLoS One 2012; 7:e43860. [PMID: 23024745 PMCID: PMC3443101 DOI: 10.1371/journal.pone.0043860] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 07/26/2012] [Indexed: 12/04/2022] Open
Abstract
C57BL/6J (C57) mice develop mild arthritis (Lyme disease-resistant) whereas C3H/HeN (C3H) mice develop severe arthritis (Lyme disease-susceptible) after infection with the spirochete Borrelia burgdorferi. We hypothesized that susceptibility and resistance to Lyme disease, as modeled in mice, is associated with early induction and regulation of inflammatory mediators by innate immune cells after their exposure to live B. burgdorferi spirochetes. Here, we employed multiplex ELISA and qRT-PCR to investigate quantitative differences in the levels of cytokines and chemokines produced by bone marrow-derived macrophages from C57 and C3H mice after these cells were exposed ex vivo to live spirochetes or spirochetal lipoprotein. Upon stimulation, the production of both cytokines and chemokines was up-regulated in macrophages from both mouse strains. Interestingly, however, our results uncovered two distinct patterns of spirochete- and lipoprotein-inducible inflammatory mediators displayed by mouse macrophages, such that the magnitude of the chemokine up-regulation was larger in C57 cells than it was in C3H cells, for most chemokines. Conversely, cytokine up-regulation was more intense in C3H cells. Gene transcript analyses showed that the displayed patterns of inflammatory mediators were associated with a TLR2/TLR1 transcript imbalance: C3H macrophages expressed higher TLR2 transcript levels as compared to those expressed by C57 macrophages. Exogenous IL-10 dampened production of inflammatory mediators, especially those elicited by lipoprotein stimulation. Neutralization of endogenously produced IL-10 increased production of inflammatory mediators, notably by macrophages of C57 mice, which also displayed more IL-10 than C3H macrophages. The distinct patterns of pro-inflammatory mediator production, along with TLR2/TLR1 expression, and regulation in macrophages from Lyme disease-resistant and -susceptible mice suggests itself as a blueprint to further investigate differential pathogenesis of Lyme disease.
Collapse
Affiliation(s)
- Aarti Gautam
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana, United States of America
| | - Saurabh Dixit
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana, United States of America
- Center for Nanobiotechnology Research, Alabama State University, Montgomery, Alabama, United States of America
| | - Monica Embers
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana, United States of America
| | - Rajeev Gautam
- Division of Microbiology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana, United States of America
| | - Mario T. Philipp
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana, United States of America
| | - Shree R. Singh
- Center for Nanobiotechnology Research, Alabama State University, Montgomery, Alabama, United States of America
| | - Lisa Morici
- Department of Microbiology and Immunology, Tulane University, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Vida A. Dennis
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana, United States of America
- Center for Nanobiotechnology Research, Alabama State University, Montgomery, Alabama, United States of America
| |
Collapse
|
36
|
The DBA/1 strain is a novel mouse model for experimental Borrelia burgdorferi infection. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:1567-73. [PMID: 22855391 DOI: 10.1128/cvi.00251-12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Lyme arthritis, caused by Borrelia burgdorferi, has similarities to rheumatoid arthritis and its experimental murine model, collagen-induced arthritis (CIA). Currently, no common strain exists for examination of arthritis models of Lyme arthritis and CIA, which are typically studied in C3H/HeJ and DBA/1 mice, respectively. The aim of this study was to define the characteristics of Borrelia burgdorferi infection and arthritis in the DBA/1 murine strain. Murine Lyme arthritis was induced in C3H/HeJ and DBA/1 mice by subcutaneous infection with B. burgdorferi. Tibiotarsal joints were measured during infection, and mice were sacrificed for histologic, microbiologic, and serologic analysis on days 14 and 42 postinfection. All bladder cultures obtained from C3H/HeJ and DBA/1 mice at 14 days postinfection grew Borrelia. There was no significant difference in spirochetal burdens in hearts and tibiotarsal joints at days 14 and 42 postinfection. Tibiotarsal joint swelling and histologic scoring were not significantly different between the two strains. Serologic analysis revealed increased IgG2a production in C3H/HeJ mice compared to DBA/1 mice. Analysis of 2-dimensional immunoblots revealed several specific antigens (LA7, BBA03, BBA64, BBA73, OspA, and VlsE) which were not recognized by DBA/1 sera. We conclude that the DBA/1 murine strain is a suitable model for the study of Lyme arthritis and experimental B. burgdorferi infection, allowing direct comparison between Lyme arthritis and collagen-induced arthritis. The specificity of the humoral immune response differs between the two strains, further study of which may reveal important findings about how individual strains respond to B. burgdorferi infection.
Collapse
|
37
|
Iliopoulou BP, Huber BT. Emergence of chronic Lyme arthritis: putting the breaks on CD28 costimulation. Immunopharmacol Immunotoxicol 2010; 31:180-5. [PMID: 18792834 DOI: 10.1080/08923970802391459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Lyme disease is a debilitating infection that is caused upon a bite of Borrelia burgdorferi (Bb)-infected ticks. One of the most prominent clinical manifestations is the development of chronic Lyme arthritis. Months after Bb infection, approximately 60% of untreated Lyme patients experience intermittent arthritic attacks that may last for years. The use of the CD28(-/-) mouse in Bb infection has helped to shed light into the mechanisms that govern this inflammatory process, which seems to be tightly regulated. In this current review, the effect of immunoregulation, as well as CD28 deficiency in the development of chronic Lyme arthritis is discussed.
Collapse
|
38
|
Mullooly C, Higgins SP. Secondary syphilis: the classical triad of skin rash, mucosal ulceration and lymphadenopathy. Int J STD AIDS 2010; 21:537-45. [DOI: 10.1258/ijsa.2010.010243] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
After years of declining incidence in many developed countries, syphilis infection has re-emerged as a major public health problem in the past decade. The secondary stage of syphilis epitomizes the capacity of the infection to present in myriad ways. The skin, lymph glands and mucosal membranes are the most commonly affected tissues.
Collapse
Affiliation(s)
- C Mullooly
- Department of Genitourinary Medicine, North Manchester General Hospital, Crumpsall, Manchester M8 5RB, UK
| | - S P Higgins
- Department of Genitourinary Medicine, North Manchester General Hospital, Crumpsall, Manchester M8 5RB, UK
| |
Collapse
|
39
|
CD14 signaling restrains chronic inflammation through induction of p38-MAPK/SOCS-dependent tolerance. PLoS Pathog 2009; 5:e1000687. [PMID: 20011115 PMCID: PMC2781632 DOI: 10.1371/journal.ppat.1000687] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Accepted: 11/10/2009] [Indexed: 11/19/2022] Open
Abstract
Current thinking emphasizes the primacy of CD14 in facilitating recognition of microbes by certain TLRs to initiate pro-inflammatory signaling events and the importance of p38-MAPK in augmenting such responses. Herein, this paradigm is challenged by demonstrating that recognition of live Borrelia burgdorferi not only triggers an inflammatory response in the absence of CD14, but one that is, in part, a consequence of altered PI3K/AKT/p38-MAPK signaling and impaired negative regulation of TLR2. CD14 deficiency results in increased localization of PI3K to lipid rafts, hyperphosphorylation of AKT, and reduced activation of p38. Such aberrant signaling leads to decreased negative regulation by SOCS1, SOCS3, and CIS, thereby compromising the induction of tolerance in macrophages and engendering more severe and persistent inflammatory responses to B. burgdorferi. Importantly, these altered signaling events and the higher cytokine production observed can be mimicked through shRNA and pharmacological inhibition of p38 activity in CD14-expressing macrophages. Perturbation of this CD14/p38-MAPK-dependent immune regulation may underlie development of infectious chronic inflammatory syndromes. Macrophages express CD14 which partners with Toll-like receptor 2/1 to recognize bacterial lipoproteins such as those of Borrelia burgdorferi, the causative agent of Lyme disease. In vitro evidence demonstrates that blocking CD14 recognition of bacterial components ablates innate host cell inflammatory responses. Similarly, blocking downstream p38 kinase activity dampens the cellular response to these same microbial stimuli. This body of work underpins two well-established paradigms which cite the primacy of CD14 in facilitating TLR recognition of microbes to initiate proinflammatory signaling events and the importance of p38 in augmenting such responses. However, contrary to these paradigms, our prior study using a mouse model of Lyme disease demonstrated an association between CD14 deficiency, increased bacterial burden, and more severe and persistent disease. Herein, we provide a mechanistic explanation for this unanticipated host immune response implicating impaired negative regulation of inflammatory signaling pathways as an underlying cause. Consequent to impaired negative regulation the host becomes “intolerant” of continued exposure to bacteria and thus mounts a perpetual inflammatory response to their presence. An intriguing question raised by these findings is whether individual differences in the severity and clinical course of infection might reflect the susceptibility of the patient's innate immune system to tolerization.
Collapse
|
40
|
Miller JC, Ma Y, Bian J, Sheehan KC, Zachary JF, Weis JH, Schreiber RD, Weis JJ. A critical role for type I IFN in arthritis development following Borrelia burgdorferi infection of mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:8492-503. [PMID: 19050267 PMCID: PMC3024833 DOI: 10.4049/jimmunol.181.12.8492] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gene expression analysis previously revealed a robust IFN-responsive gene induction profile that was selectively up-regulated in Borrelia burgdorferi-infected C3H mice at 1 wk postinfection. This profile was correlated with arthritis development, as it was absent from infected, mildly arthritic C57BL/6 mice. In this report we now demonstrate that profile induction in infected C3H scid mice occurs independently of B or T lymphocyte infiltration in the joint tissue. Additionally, type I IFN receptor-blocking Abs, but not anti-IFN-gamma Abs, dramatically reduced arthritis, revealing a critical but previously unappreciated role for type I IFN in Lyme arthritis development. Certain examined IFN-inducible transcripts were also significantly diminished within joint tissue of mice treated with anti-IFNAR1, whereas expression of other IFN-responsive genes was more markedly altered by anti-IFN-gamma treatment. These data indicate that induction of the entire IFN profile is not necessary for arthritis development. These findings further tie early type I IFN induction to Lyme arthritis development, a connection not previously made. Bone marrow-derived macrophages readily induced IFN-responsive genes following B. burgdorferi stimulation, and this expression required a functional type I IFN receptor. Strikingly, induction of these genes was independent of TLRs 2,4, and 9 and of the adapter molecule MyD88. These data demonstrate that the extracellular pathogen B. burgdorferi uses a previously unidentified receptor and a pathway traditionally associated with viruses and intracellular bacteria to initiate transcription of type I IFN and IFN-responsive genes and to initiate arthritis development.
Collapse
Affiliation(s)
| | - Ying Ma
- Department of Pathology, University of Utah, Salt Lake City, UT 84112
| | - Jiantao Bian
- Department of Pathology, University of Utah, Salt Lake City, UT 84112
| | - Kathleen C.F. Sheehan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - James F. Zachary
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802
| | - John H. Weis
- Department of Pathology, University of Utah, Salt Lake City, UT 84112
| | - Robert D. Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Janis J. Weis
- Department of Pathology, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
41
|
NKT cells prevent chronic joint inflammation after infection with Borrelia burgdorferi. Proc Natl Acad Sci U S A 2008; 105:19863-8. [PMID: 19060201 DOI: 10.1073/pnas.0810519105] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Borrelia burgdorferi is the etiologic agent of Lyme disease, a multisystem inflammatory disorder that principally targets the skin, joints, heart, and nervous system. The role of T lymphocytes in the development of chronic inflammation resulting from B. burgdorferi infection has been controversial. We previously showed that natural killer T (NKT) cells with an invariant (i) TCR alpha chain (iNKT cells) recognize glycolipids from B. burgdorferi, but did not establish an in vivo role for iNKT cells in Lyme disease pathogenesis. Here, we evaluate the importance of iNKT cells for host defense against these pathogenic spirochetes by using Valpha14i NKT cell-deficient (Jalpha18(-/-)) BALB/c mice. On tick inoculation with B. burgdorferi, Jalpha18(-/-) mice exhibited more severe and prolonged arthritis as well as a reduced ability to clear spirochetes from infected tissues. Valpha14i NKT cell deficiency also resulted in increased production of antibodies directed against both B. burgdorferi protein antigens and borrelial diacylglycerols; the latter finding demonstrates that anti-glycolipid antibody production does not require cognate help from Valpha14i NKT cells. Valpha14i NKT cells in infected wild-type mice expressed surface activation markers and produced IFNgamma in vivo after infection, suggesting a participatory role for this unique population in cellular immunity. Our data are consistent with the hypothesis that the antigen-specific activation of Valpha14i NKT cells is important for the prevention of persistent joint inflammation and spirochete clearance, and they counter the long-standing notion that humoral rather than cellular immunity is sufficient to facilitate Lyme disease resolution.
Collapse
|
42
|
Iliopoulou BP, Alroy J, Huber BT. Persistent arthritis in Borrelia burgdorferi-infected HLA-DR4-positive CD28-negative mice post-antibiotic treatment. ARTHRITIS AND RHEUMATISM 2008; 58:3892-901. [PMID: 19035513 PMCID: PMC2775539 DOI: 10.1002/art.24028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE The immunologic events that lead to persistent joint inflammation in certain patients with Lyme arthritis post-antibiotic treatment have been elusive so far. The prevalence of this condition is highest in individuals with rheumatoid arthritis-associated HLA-DR alleles. This study was undertaken to generate a murine model with persistent arthritis post-antibiotic treatment. METHODS We have previously shown that CD28(-/-) mice develop intermittent monarticular Lyme arthritis that is responsive to antibiotics. Since there seems to be a link in humans between persistent arthritic manifestations post-antibiotic treatment and the HLA-DR4 allele, we generated DR4+/+CD28(-/-)MHCII(-/-) mice, infected them with Borrelia burgdorferi, and subsequently treated them with antibiotics. RESULTS Thirty-eight percent of the B burgdorferi-infected DR4+/+CD28(-/-)MHCII(-/-) mice, but none of the B burgdorferi-infected CD28(-/-)MHCII(-/-) mice, remained arthritic post-antibiotic treatment. A significant fraction (36%) of these mice, but none of the mice in which arthritis resolved, had serum antibodies to outer surface protein A of B burgdorferi. After abrogation of active B burgdorferi infection, the inflammatory reaction in mice with persistent joint inflammation was restricted to the joints, since their draining lymph nodes were no longer enlarged. Increased CD20 and interferon-gamma messenger RNA expression in the inflamed joints of these mice suggested a possible role of B cells and inflammatory cytokines in the pathogenesis of persistent arthritis post-antibiotic treatment. CONCLUSION The establishment of this murine model allows, for the first time, the elucidation of the immunologic events that lead to persistent Lyme arthritis post-antibiotic therapy in genetically susceptible individuals.
Collapse
MESH Headings
- Animals
- Anti-Bacterial Agents/pharmacology
- Antibodies, Bacterial/blood
- Antigens, CD20/genetics
- Antigens, Surface/immunology
- Arthritis, Infectious/epidemiology
- Arthritis, Infectious/immunology
- Arthritis, Infectious/microbiology
- B-Lymphocytes/immunology
- B-Lymphocytes/microbiology
- Bacterial Outer Membrane Proteins/immunology
- Bacterial Vaccines/immunology
- Borrelia burgdorferi
- CD28 Antigens/genetics
- Disease Models, Animal
- Female
- Genes, MHC Class II/genetics
- HLA-DR4 Antigen/genetics
- Humans
- Interferon-gamma/genetics
- Lipoproteins/immunology
- Lyme Disease/complications
- Lyme Disease/drug therapy
- Lyme Disease/immunology
- Lymph Nodes/immunology
- Male
- Mice
- Mice, Inbred Strains
- Mice, Transgenic
- RNA, Messenger/metabolism
- Seroepidemiologic Studies
Collapse
|
43
|
Adenoviral delivery of interleukin-10 fails to attenuate experimental Lyme disease. Infect Immun 2008; 76:5500-7. [PMID: 18824530 DOI: 10.1128/iai.00808-08] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Production of interleukin-10 (IL-10) by C57BL/6 mice following infection with Borrelia burgdorferi has been proposed as a mechanism whereby resistance to the development of experimental Lyme arthritis is maintained. In the current study, we sought to determine the role of IL-10 during infection of arthritis- and carditis-susceptible C3H mice. Infection of C3H IL-10(-/-) mice led to increased joint swelling and arthritis severity scores over those of wild-type C3H mice. Measurement of B. burgdorferi numbers in joints or disseminated tissues indicated a more efficient clearance of spirochetes in the absence of IL-10, similar to that reported in C57BL/6 IL-10(-/-) mice. However, in contrast to previous in vitro work, infection of C3H IL-10(-/-) mice led to decreased in vivo expression of the cytokines KC, IL-1beta, IL-4, and IL-12p70 in the infected joints. Finally, adenoviral expression of IL-10 in the infected joints of C3H mice was unable to modulate the development of severe Lyme arthritis and had no effect on spirochete clearance or Borrelia-specific antibody production. Development of Lyme carditis appeared to be independent of modulation by IL-10. These results suggest that IL-10 limits the development of joint inflammation in both arthritis-resistant and -susceptible mouse strains infected with B. burgdorferi and that increased IL-10 production cannot rescue genetic susceptibility to development of pathology in this model.
Collapse
|
44
|
Miller JC, Ma Y, Crandall H, Wang X, Weis JJ. Gene expression profiling provides insights into the pathways involved in inflammatory arthritis development: murine model of Lyme disease. Exp Mol Pathol 2008; 85:20-7. [PMID: 18462718 PMCID: PMC2565650 DOI: 10.1016/j.yexmp.2008.03.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Accepted: 03/03/2008] [Indexed: 01/01/2023]
Abstract
The spirochete Borrelia burgdorferi, the etiologic agent of Lyme disease, causes severe subacute arthritis in susceptible inbred mouse strains, such as C3H/HeN, but only mild arthritis in resistant strains such as C57BL/6. The degree of Lyme arthritis severity is controlled in part by host genetics and several quantitative trait loci have been identified which contribute to this regulation. In addition, the anti-inflammatory cytokine IL-10 assumes an important role in the control of arthritis in C57BL/6 mice. However, the identification of genes and signaling pathways that dictate arthritis severity has remained elusive. In an attempt to elucidate such genes and pathways, the power of microarray analysis was combined with information gleaned from gene manipulation models. As a result of this approach, two novel gene profiles were identified: an IFN-inducible profile in arthritis-susceptible C3H and IL-10(-/-) mice, and an epidermal/differentiation profile in C57BL/6 mice. Application of this information to TLR2(-/-) mice, which also develop severe arthritis, indicated that they also upregulated IFN-responsive genes. These results provided new insight into the regulation of Lyme arthritis development and illustrated the utility of combining gene expression analyses with genetically manipulated mouse models in unraveling mechanisms underlying specific disease processes.
Collapse
Affiliation(s)
| | - Ying Ma
- Department of Pathology, University of Utah, Salt Lake City, Utah 84112
| | - Hillary Crandall
- Department of Pathology, University of Utah, Salt Lake City, Utah 84112
| | - Xiaohui Wang
- Department of Pathology, University of Utah, Salt Lake City, Utah 84112
| | - Janis J. Weis
- Department of Pathology, University of Utah, Salt Lake City, Utah 84112
| |
Collapse
|
45
|
Tsuchiya K, Komori M, Zheng QY, Ferrieri P, Lin J. Interleukin-10 is an essential modulator of mucoid metaplasia in a mouse otitis media model. Ann Otol Rhinol Laryngol 2008; 117:630-6. [PMID: 18771082 PMCID: PMC2692729 DOI: 10.1177/000348940811700814] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Inflammatory cytokines are involved in the development of mucous cell metaplasia and hyperplasia (MCM) in otitis media (OM). However, which cytokines play an essential role in the MCM of OM is not clear at the moment. METHODS In this study, we hypothesized that interleukin-10 (IL-10) played an indispensable role in the MCM of bacterial OM, and we used IL-10 knockout mice to test this hypothesis. RESULTS In wild-type mice, both Streptococcus pneumoniae and Haemophilus influenzae triggered the development of MCM in the middle ear mucosa. In IL-10 knockout mice, the number of goblet cells and mucin-producing cells in the middle ear was significantly reduced after bacterial middle ear infection compared with that in wild-type mice. CONCLUSIONS We conclude that IL-10 plays an essential role in the MCM of bacterial OM. Interleukin-10 is a potential target for the treatment of MCM in OM.
Collapse
Affiliation(s)
- Katsuyuki Tsuchiya
- University of Minnesota Otitis Media Research Center, Department of Otolaryngology, University of Minnesota School of Medicine, Minneapolis, MN
| | - Masahiro Komori
- University of Minnesota Otitis Media Research Center, Department of Otolaryngology, University of Minnesota School of Medicine, Minneapolis, MN
| | - Qing Yin Zheng
- The Department of Otolaryngology-HNS, Case Western Reserve University, Cleveland, OH
| | - Patricia Ferrieri
- University of Minnesota Otitis Media Research Center, Department of Infectious Disease Divition of Pediatrics, University of Minnesota School of Medicine, Minneapolis, MN
| | - Jizhen Lin
- University of Minnesota Otitis Media Research Center, Department of Otolaryngology, University of Minnesota School of Medicine, Minneapolis, MN
| |
Collapse
|
46
|
Wang G, Petzke MM, Iyer R, Wu H, Schwartz I. Pattern of proinflammatory cytokine induction in RAW264.7 mouse macrophages is identical for virulent and attenuated Borrelia burgdorferi. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 180:8306-15. [PMID: 18523297 PMCID: PMC2614377 DOI: 10.4049/jimmunol.180.12.8306] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Lyme disease pathogenesis results from a complex interaction between Borrelia burgdorferi and the host immune system. The intensity and nature of the inflammatory response of host immune cells to B. burgdorferi may be a determining factor in disease progression. Gene array analysis was used to examine the expression of genes encoding cytokines, chemokines, and related factors in the joint tissue of infected C3H/HeJ mice and in a murine macrophage-like cell line in response to a disseminating or attenuated clinical isolate of B. burgdorferi. Both isolates elicited a robust proinflammatory response in RAW264.7 cells characterized by an increase in transcript levels of genes encoding CC and CXC chemokines, proinflammatory cytokines, and TNF superfamily members. Transcription of genes encoding IL-1beta, IL-6, MCP-1, MIP-1alpha, CXCR4, and TLR2 induced in RAW264.7 cells by either live or heat-killed spirochetes did not differ significantly at any time point over a 24-h period, nor was there a difference in the protein levels of IL-10, TNF-alpha, IL-6, and IL-12p70 in culture supernatants. Thus, induction of host macrophage expression of proinflammatory mediators by host macrophages does not contribute to the differential pathogenicity of different B. burgdorferi strains.
Collapse
Affiliation(s)
- Guiqing Wang
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595
| | - Mary M. Petzke
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595
| | - Radha Iyer
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595
| | - Hongyan Wu
- Department of Medicine, New York Medical College, Valhalla, NY 10595
| | - Ira Schwartz
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595
| |
Collapse
|
47
|
Abstract
The spirochete Borrelia burgdorferi is a tick-borne obligate parasite whose normal reservoir is a variety of small mammals. Although infection of these natural hosts does not lead to disease, infection of humans can result in Lyme disease as a consequence of the human immunopathologic response to B burgdorferi. Consistent with the pathogenesis of Lyme disease, bacterial products that allow B burgdorferi to replicate and survive seem to be primarily what is required for the bacterium to cause disease in a susceptible host. This article describes the basic biology of B burgdorferi and reviews some of the bacterial components required for infection of and survival in the mammalian and tick hosts.
Collapse
Affiliation(s)
- Kit Tilly
- Laboratory of Zoonotic Pathogens, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, 903 South Fourth St., Hamilton, Montana 59840, Ph. 406-363-9239, FAX 406-375-9681,
| | - Patricia A. Rosa
- Laboratory of Zoonotic Pathogens, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, 903 South Fourth St., Hamilton, Montana 59840, Ph. 406-363-9209, FAX 406-375-9681,
| | - Philip E. Stewart
- Laboratory of Zoonotic Pathogens, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, 903 South Fourth St., Hamilton, Montana 59840, Ph. 406-363-9393, FAX 406-375-9681,
| |
Collapse
|
48
|
Lenz LL, Andrews-Polymenis HL. Silencing the alarm: insights into the interaction between host and pathogen. Conference on Microbial Pathogenesis: Mechanisms of Infectious Disease. EMBO Rep 2008; 9:27-32. [PMID: 18084188 PMCID: PMC2246619 DOI: 10.1038/sj.embor.7401127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Accepted: 10/22/2007] [Indexed: 11/08/2022] Open
Affiliation(s)
- Laurel L Lenz
- Integrated Department of Immunology, National Jewish Medical and Research Center, Goodman Building, K510, 1400 Jackson Street, Denver, Colorado 80206, USA
| | - Helene L Andrews-Polymenis
- Department of Microbial and Molecular Pathogenesis, Texas A&M University System Health Science Center, College of Medicine, 407 Reynolds Medical Building, College Station, Texas 77843-1114, USA
| |
Collapse
|
49
|
Cruz AR, Moore MW, La Vake CJ, Eggers CH, Salazar JC, Radolf JD. Phagocytosis of Borrelia burgdorferi, the Lyme disease spirochete, potentiates innate immune activation and induces apoptosis in human monocytes. Infect Immun 2008; 76:56-70. [PMID: 17938216 PMCID: PMC2223637 DOI: 10.1128/iai.01039-07] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Revised: 09/07/2007] [Accepted: 10/08/2007] [Indexed: 01/07/2023] Open
Abstract
We have previously demonstrated that phagocytosed Borrelia burgdorferi induces activation programs in human peripheral blood mononuclear cells that differ qualitatively and quantitatively from those evoked by equivalent lipoprotein-rich lysates. Here we report that ingested B. burgdorferi induces significantly greater transcription of proinflammatory cytokine genes than do lysates and that live B. burgdorferi, but not B. burgdorferi lysate, is avidly internalized by monocytes, where the bacteria are completely degraded within phagolysosomes. In the course of these experiments, we discovered that live B. burgdorferi also induced a dose-dependent decrease in monocytes but not a decrease in dendritic cells or T cells and that the monocyte population displayed morphological and biochemical hallmarks of apoptosis. Particularly noteworthy was the finding that apoptotic changes occurred predominantly in monocytes that had internalized spirochetes. Abrogation of phagocytosis with cytochalasin D prevented the death response. Heat-killed B. burgdorferi, which was internalized as well as live organisms, induced a similar degree of apoptosis of monocytes but markedly less cytokine production. Surprisingly, opsonophagocytosis of Treponema pallidum did not elicit a discernible cell death response. Our combined results demonstrate that B. burgdorferi confined to phagolysosomes is a potent inducer of cytosolic signals that result in (i) production of NF-kappaB-dependent cytokines, (ii) assembly of the inflammasome and activation of caspase-1, and (iii) induction of programmed cell death. We propose that inflammation and apoptosis represent mutually reinforcing components of the immunologic arsenal that the host mobilizes to defend itself against infection with Lyme disease spirochetes.
Collapse
Affiliation(s)
- Adriana R Cruz
- Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-3715, USA
| | | | | | | | | | | |
Collapse
|
50
|
Viable Borrelia burgdorferi enhances interleukin-10 production and suppresses activation of murine macrophages. Infect Immun 2007; 76:1153-62. [PMID: 18086805 DOI: 10.1128/iai.01404-07] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Although it is capable of eliciting strong innate and adaptive immune responses, Borrelia burgdorferi often evades immune clearance through largely unknown mechanisms. Our previous studies determined that infected interlukin-10-/- (IL-10-/-) mice show significantly lower B. burgdorferi levels than wild-type (B6) mice and that IL-10 inhibits innate immune responses critical for controlling B. burgdorferi infection. To determine whether virulent B. burgdorferi preferentially enhances IL-10 production, we developed an in vitro coculture medium (RPMI.B) in which both B. burgdorferi and primary macrophages (Mphis) remain viable. B. burgdorferi grew at similar rates and was able to regulate expression of immunoreactive proteins with similar kinetics in RPMI.B and in traditional BSK medium; in contrast, B. burgdorferi cultured in conventional tissue culture medium (RPMI) rapidly lost viability. Coculture of viable B. burgdorferi in RPMI.B with Mphis resulted in more rapid and significant increases in IL-10 transcripts and secreted proteins than coculture with nonviable B. burgdorferi in RPMI, which corresponded with decreased production of proinflammatory cytokines. Addition of live B. burgdorferi to Mphis in RPMI.B also elicited substantially higher IL-10 levels than heat-killed bacteria elicited, confirming that increased IL-10 production was not inherent to coculture in RPMI.B. Transfer of supernatants from B. burgdorferi-stimulated Mphis into naïve Mphi cultures resulted in suppressed activation upon subsequent stimulation with different bacterial agonists, and this suppression was obviated by IL-10-specific antibody. In vivo analyses determined that murine skin samples exhibited substantial upregulation of IL-10 within 24 h of injection of B. burgdorferi. Together, these results suggest that viable B. burgdorferi can suppress early Mphi responses during infection by causing increased release of IL-10.
Collapse
|