1
|
Yang F, Lv J, Huang Y, Ma W, Yang Z. A supramolecular assembly strategy for the treatment of rheumatoid arthritis with ultrasound-augmented inflammatory microenvironment reprograming. Biomaterials 2025; 316:123006. [PMID: 39675142 DOI: 10.1016/j.biomaterials.2024.123006] [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: 09/20/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
As regulators and promotors of joint erosion, pro-inflammatory M1-like macrophages play pivotal roles in the pathogenesis of rheumatoid arthritis (RA). Here, we develop a supramolecular self-assembly (PCSN@MTX) of molybdenum (Mo) based polyoxometalate (POM), β-cyclodextrin (β-CD), and methotrexate (MTX), in which the MTX is loaded by host-guest interaction. PCSN@MTX shows inhibition of synovial M1-like macrophages polarization to alleviate RA. PCSN@MTX has demonstrated ultrasound (US) augmented catalytic behavior in consuming ROS and generating oxygen (O2) with accelerated conversion of Mo5+ to Mo6+ in the POM. In the collagen-induced arthritis mouse model, after systemical administration, the pH-responsive PCSN@MTX shows enhanced accumulation in the acidic joints by in-situ self-assembly. The host-guest complexation between MTX and β-CD is broken via US, achieving an on-demand burst release of MTX. The released MTX and ROS-scavenging synergistically facilitate the M1-to-M2 macrophage phenotype switching, which effectively alleviates RA disease progress under US irradiation. This study provides a paradigm for RA therapy with a promising US-augmented strategy.
Collapse
Affiliation(s)
- Fuhong Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), College of Photonic and Electronic Engineering, Fujian Key Laboratory of Flexible Electronics and Strait Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, 350117, China
| | - Jingqi Lv
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), College of Photonic and Electronic Engineering, Fujian Key Laboratory of Flexible Electronics and Strait Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, 350117, China
| | - Yanli Huang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), College of Photonic and Electronic Engineering, Fujian Key Laboratory of Flexible Electronics and Strait Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, 350117, China
| | - Wen Ma
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), College of Photonic and Electronic Engineering, Fujian Key Laboratory of Flexible Electronics and Strait Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, 350117, China.
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), College of Photonic and Electronic Engineering, Fujian Key Laboratory of Flexible Electronics and Strait Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, 350117, China.
| |
Collapse
|
2
|
McClune ME, Ebohon O, Dressler JM, Davis MM, Tupik JD, Lochhead RB, Booth CJ, Steere AC, Jutras BL. The peptidoglycan of Borrelia burgdorferi can persist in discrete tissues and cause systemic responses consistent with chronic illness. Sci Transl Med 2025; 17:eadr2955. [PMID: 40267217 DOI: 10.1126/scitranslmed.adr2955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 01/27/2025] [Accepted: 03/06/2025] [Indexed: 04/25/2025]
Abstract
Persistent symptoms after an acute infection is an emerging public health concern, but the pathobiology of such conditions is not well understood. One possible scenario involves the persistence of lingering antigen. We have previously reported that patients with postinfectious Lyme arthritis often harbor the peptidoglycan (PG) cell wall of Borrelia burgdorferi, the Lyme disease agent, in the synovial fluid of their inflamed joints after treatment. However, it is not yet known how B. burgdorferi PG persists, in what form, or if it may play a role in other postinfectious complications after Lyme disease. Using a murine model, we developed a real-time in vivo system to track B. burgdorferi PG as a function of cell wall chemistry and validated our findings using both molecular and cellular approaches. Unlike typical bacterial PG, the unique chemical properties of polymeric B. burgdorferi PG drive murine liver accumulation, where the cell wall material persists for weeks. Kupffer cells and hepatocytes phagocytose and retain B. burgdorferi PG and, although liver occupancy coincides with minimal pathology, both organ-specific and secreted protein profiles produced under these conditions bear some similarities to reported proteins enriched in patients with chronic illness after acute infection. Moreover, transcriptomic profiling indicated that B. burgdorferi PG affects energy metabolism in peripheral blood mononuclear cells. Our findings provide mechanistic insights into how a pathogenic molecule can persist after agent clearance, potentially contributing to illness after infection.
Collapse
Affiliation(s)
- Mecaila E McClune
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Human Center for Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Osamudiamen Ebohon
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Human Center for Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Jules M Dressler
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Marisela M Davis
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
| | - Juselyn D Tupik
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
- Department of Biomedical and Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Robert B Lochhead
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Carmen J Booth
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Allen C Steere
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Brandon L Jutras
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Human Center for Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
- Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
3
|
Sze CW, Lynch MJ, Zhang K, Neau DB, Ealick SE, Crane BR, Li C. Lactate dehydrogenase is the Achilles' heel of Lyme disease bacterium Borreliella burgdorferi. mBio 2025; 16:e0372824. [PMID: 40111021 PMCID: PMC11980376 DOI: 10.1128/mbio.03728-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
As a zoonotic pathogen, the Lyme disease bacterium Borreliella burgdorferi has evolved unique metabolic pathways, some of which are specific and essential for its survival and thus present as ideal targets for developing new therapeutics. B. burgdorferi dispenses with the use of thiamin as a cofactor and relies on lactate dehydrogenase (BbLDH) to convert pyruvate to lactate for balancing NADH/NAD+ ratios. This report first demonstrates that BbLDH is a canonical LDH with some unique biochemical and structural features. A loss-of-function study then reveals that BbLDH is essential for B. burgdorferi survival and infectivity, highlighting its therapeutic potential. Drug screening identifies four previously unknown LDH inhibitors with minimal cytotoxicity, two of which inhibit B. burgdorferi growth. This study provides mechanistic insights into the function of BbLDH in the pathophysiology of B. burgdorferi and lays the groundwork for developing genus-specific metabolic inhibitors against B. burgdorferi and potentially other tick-borne pathogens as well. IMPORTANCE Lyme disease (LD) is the most commonly reported tick-borne illness in the U.S. and Europe, and its geographic distribution is continuously expanding worldwide. Though early LD can be treated with antibiotics, chronic LD is recalcitrant to antibiotic treatments and thus requires multiple courses of antibiotic therapy. Currently, there are no human vaccines nor prophylactic antibiotics to prevent LD. As the causative agent of LD, Borreliella burgdorferi has evolved unique metabolic pathways, some of which are specific and essential for its survival and thus present as ideal targets for developing new therapeutics. By using an approach of genetics, biochemistry, structural biology, drug screening, and animal models, this report provides evidence that lactate dehydrogenase can be a potential target for developing genus-specific metabolic inhibitors against B. burgdorferi and potentially other tick-borne pathogens as well.
Collapse
Affiliation(s)
- Ching Wooen Sze
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Michael J. Lynch
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, USA
| | - Kai Zhang
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - David B. Neau
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, USA
| | - Steven E. Ealick
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, USA
| | - Brian R. Crane
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, USA
| | - Chunhao Li
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
4
|
Strle F, Maraspin V, Lotrič-Furlan S, Ogrinc K, Rojko T, Kastrin A, Strle K, Wormser GP, Bogovič P. Lower Frequency of Multiple Erythema Migrans Skin Lesions in Lyme Reinfections, Europe. Emerg Infect Dis 2025; 31:662-668. [PMID: 40133040 PMCID: PMC11950259 DOI: 10.3201/eid3104.241329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025] Open
Abstract
The erythema migrans (EM) skin lesion is the most common clinical manifestation of Lyme borreliosis. Information about EM in Lyme borreliosis reinfection is limited. Of the 12,384 cases with diagnosed EM at an outpatient clinic during 1990-2014 in Slovenia, 1,962 (15.8%) cases occurred in patients who were treated previously for Lyme borreliosis, including 1,849 (94.2%) who had previously had EM. The percentage of reinfected patients who sought care with disseminated Lyme borreliosis at the time of reinfection, as manifested by multiple EM skin lesions, was significantly lower than for EM patients with no history of Lyme borreliosis (5.5% [108/1,962] vs. 7.4% [769/10,427]; p = 0.002). None of the clinical manifestations of Lyme borreliosis in Europe will completely protect against EM developing in patients in the future. The reoccurrence of Lyme borreliosis manifested by multiple EM lesions is significantly less likely than for patients with no history of Lyme borreliosis.
Collapse
|
5
|
Milewski MD, Ecklund K, Lee SW, Coene RP, Sanborn RM, Gossman EC, Miller PE, Heyworth BE, Yen YM. Knee Lyme Arthritis in Pediatric and Adolescent Patients May Be Associated With Meniscal Changes on MRI. JB JS Open Access 2025; 10:e24.00080. [PMID: 40196415 PMCID: PMC11968016 DOI: 10.2106/jbjs.oa.24.00080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
Background Lyme arthritis is a common manifestation of late-stage Lyme disease in pediatric and adolescent patients. Patients with Lyme arthritis typically present with atraumatic knee effusion and may undergo magnetic resonance imaging (MRI) to aid in diagnosis. The incidence of meniscal pathology on MRI in association with Lyme arthritis is unknown. This study aims to evaluate the incidence of meniscal pathology on MRI in young patients with Lyme arthritis. Methods Patients (<18 years old) presenting with a unilateral knee effusion from 2009 to 2019 with a positive Lyme antibody serologic test, MRI within 2 weeks of the positive test, and ultimate diagnosis of Lyme arthritis were included in the study. MRI, which was performed to distinguish Lyme arthritis from other causes of knee effusion, underwent analysis by a pediatric musculoskeletal radiologist. Meniscal signal abnormality was graded as follows: grade 1 = globular, grade 2 = linear nonsurfacing, and grade 3 = surfacing tear. Results Eighty-seven patients (10.6 ± 3.9 years, 71.3% male, 67.8% White) were included. Fourteen (16%) patients had meniscal changes (grade 1: n = 4 [5%]; grade 2: n = 3 [3%]; grade 3: n = 7 [8%]). Thirteen of the 14 patients (93%) with meniscal changes on MRI were treated only with oral antibiotics, with resolution of knee symptoms and return to sports, whereas 1 patient (7%) underwent arthroscopic partial meniscectomy. Conclusions Of the 87 pediatric patients with serologically confirmed Lyme arthritis and MRI of their affected knee, 16% had a coexistent meniscal abnormality on MRI, but only 1 patient overall required surgical treatment related to the meniscus. Physicians should be aware of potential MRI meniscal changes in pediatric and adolescent patients who present with symptomatic knee effusion because of Lyme arthritis. Future research to evaluate the physiologic effects of Lyme arthritis on meniscal tissue is needed. Level of Evidence Level IV Case Series. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Matthew D. Milewski
- Department of Orthopaedic Surgery–Division of Sports Medicine, Boston Children's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Kirsten Ecklund
- Harvard Medical School, Boston, Massachusetts
- Department of Radiology, Boston Children's Hospital, Boston, Massachusetts
| | - Sang Won Lee
- Department of Orthopaedic Surgery–Division of Sports Medicine, Boston Children's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Ryan P. Coene
- Department of Orthopaedic Surgery–Division of Sports Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Ryan M. Sanborn
- Department of Orthopaedic Surgery–Division of Sports Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Emma C. Gossman
- Department of Orthopaedic Surgery–Division of Sports Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Patricia E. Miller
- Department of Orthopaedic Surgery–Division of Sports Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Benton E. Heyworth
- Department of Orthopaedic Surgery–Division of Sports Medicine, Boston Children's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Yi-Meng Yen
- Department of Orthopaedic Surgery–Division of Sports Medicine, Boston Children's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
6
|
Yu Q, Tang X, Hart T, Homer R, Belperron AA, Bockenstedt LK, Ring A, Nakamura A, Fikrig E. Secretory leukocyte protease inhibitor influences periarticular joint inflammation in B. burgdorferi-infected mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.24.625079. [PMID: 39651186 PMCID: PMC11623497 DOI: 10.1101/2024.11.24.625079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Lyme disease, caused by Borrelia burgdorferi, is the most common tick-borne infection in the United States. Arthritis is a major clinical manifestation of infection, and synovial tissue damage has been attributed to the excessive pro-inflammatory responses. The secretory leukocyte protease inhibitor (SLPI) promotes tissue repair and exerts anti-inflammatory effects. The role of SLPI in the development of Lyme arthritis in C57BL/6 mice, which can be infected with B. burgdorferi, but only develop mild joint inflammation, was therefore examined. SLPI-deficient C57BL/6 mice challenged with B. burgdorferi had a higher infection load in the tibiotarsal joints and marked periarticular swelling, compared to infected wild type control mice. The ankle joint tissues of B. burgdorferi-infected SLPI-deficient mice contained significantly higher percentages of infiltrating neutrophils and macrophages. B. burgdorferi-infected SLPI-deficient mice also exhibited elevated serum levels of IL-6, neutrophil elastase, and MMP-8. Moreover, using a recently developed BASEHIT (BActerial Selection to Elucidate Host-microbe Interactions in high Throughput) library, we found that SLPI directly interacts with B. burgdorferi. These data demonstrate the importance of SLPI in suppressing periarticular joint inflammation in Lyme disease.
Collapse
Affiliation(s)
- Qian Yu
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Xiaotian Tang
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Thomas Hart
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Robert Homer
- Department of Pathology, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Alexia A. Belperron
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Linda K. Bockenstedt
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Aaron Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Akira Nakamura
- Divisions of Immunology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
7
|
Miller JB, Yang T, Rebman AW, Cappelli L, Bingham CO, Villegas de Flores MD, Darrah E, Aucott JN. Anti-RA33 Antibodies Are Present in Patients With Lyme Disease. J Clin Rheumatol 2025; 31:65-70. [PMID: 39575669 PMCID: PMC11902579 DOI: 10.1097/rhu.0000000000002176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
BACKGROUND/OBJECTIVE To determine if anti-RA33 antibodies, which can be seen in early forms of inflammatory arthritis, are present in patients with Lyme arthritis (LA). METHODS Anti-RA33 antibodies were tested using a commercially available assay in patients with LA (n = 47) and compared with patients with erythema migrans who returned to health (EM RTH, n = 20) and those with post-treatment Lyme disease (PTLD) (n = 50), characterized by noninflammatory arthralgia, as an observational comparative study utilizing Lyme-exposed patients from various original cohorts. RESULTS We found that anti-RA33 was present in higher proportions of patients with LA (23.4% vs. 0%, p = 0.001) and PTLD (12.0% vs. 0%, p = 0.040) than healthy controls. There was also a trend toward a higher percentage of anti-RA33 positivity in patients with EM RTH versus controls (10.0% vs. 0%, p = 0.080). There were no statistically significant differences among groups of patients with LA, PTLD, and EM RTH ( p ≥ 0.567). There was also no difference in the proportion of patients with antibiotic-responsive LA compared with those with persistent synovitis after antibiotics, termed post-infectious LA, and there were no differences in clinical manifestations, musculoskeletal ultrasound evaluation (synovial hypertrophy, power Doppler, tendinopathy), or patient-reported outcomes based on anti-RA33 status. CONCLUSIONS This is the first study to identify anti-RA33 antibodies in patients with LA, though these antibodies did not identify a unique clinical subset of patients in this cohort. Unexpectedly, we found anti-RA33 antibodies at similar levels in patients with PTLD and EM RTH; further study is needed to determine the relevance of this finding.
Collapse
|
8
|
Freeman-Gallant G, McCarthy K, Yates J, Kulas K, Rudolph MJ, Vance DJ, Mantis NJ. A Refined Human Linear B Cell Epitope Map of Outer Surface Protein C (OspC) From the Lyme Disease Spirochete, Borrelia Burgdorferi. Pathog Immun 2025; 10:159-186. [PMID: 40017585 PMCID: PMC11867186 DOI: 10.20411/pai.v10i1.756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 01/07/2025] [Indexed: 03/01/2025] Open
Abstract
Background A detailed understanding of the human antibody response to outer surface protein C (OspC) of Borrelia burgdorferi has important implications for Lyme disease diagnostics and vaccines. Methods In this report, 13 peptides encompassing 8 reported OspC linear B-cell epitopes from OspC types A, B, and K, including the largely conserved C-terminus (residues 193-210), were evaluated by multiplex immunoassay (MIA) for IgG reactivity with ~700 human serum samples confirmed positive in a 2-tiered Lyme disease diagnostic assay (Bb+) and ~160 post-treatment Lyme disease (PTLD) serum samples. The vmp-like sequence E (VlsE) C6-17 peptide was included as a positive control. Results Serum IgG from Bb+ samples were reactive with 10 of the 13 OspC-derived peptides tested, with the C-terminal peptide (residues 193-210) being the most reactive. Spearman's rank correlation matrices and hierarchical clustering revealed a strong correlation between 193-210 and VlsE C6-17 peptide reactivity but little demonstrable association between 193-210 and the other OspC peptides or recombinant OspC. OspC peptide reactivities (excluding 193-210) were strongly correlated with each other and were disproportionately influenced by a subset of pan-reactive samples. In the PTLD sample set, none of the OspC-derived peptides were significantly reactive over baseline, even though VlsE C6-17 peptide reactivity remained. Conclusions The asynchronous and potentially short-lived serologic response to OspC-derived peptides reveals the complexity of B-cell responses to B. burgdorferi lipoproteins and confounds interpretation of antibody profiles for Lyme disease diagnostics.
Collapse
Affiliation(s)
- Grace Freeman-Gallant
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York
| | - Kathleen McCarthy
- Department of Biomedical Sciences, University at Albany, Albany, New York
| | - Jennifer Yates
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York
- Department of Biomedical Sciences, University at Albany, Albany, New York
| | - Karen Kulas
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York
| | | | - David J. Vance
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York
- Department of Biomedical Sciences, University at Albany, Albany, New York
| | - Nicholas J. Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York
- Department of Biomedical Sciences, University at Albany, Albany, New York
| |
Collapse
|
9
|
Sze CW, Lynch MJ, Zhang K, Neau DB, Ealick SE, Crane BR, Li C. Lactate dehydrogenase is the Achilles' heel of Lyme disease bacterium Borreliella burgdorferi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637162. [PMID: 39974897 PMCID: PMC11839043 DOI: 10.1101/2025.02.07.637162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
As a zoonotic pathogen, the Lyme disease bacterium Borreliella burgdorferi has evolved unique metabolic pathways, some of which are specific and essential for its survival and thus present as ideal targets for developing new therapeutics. B. burgdorferi dispenses with the use of thiamin as a cofactor and relies on lactate dehydrogenase (BbLDH) to convert pyruvate to lactate for balancing NADH/NAD + ratios. This report first demonstrates that BbLDH is a canonical LDH with some unique biochemical and structural features. A loss-of-function study then reveals that BbLDH is essential for B. burgdorferi survival and infectivity, highlighting its therapeutic potential. Drug screening identifies four previously unknown LDH inhibitors with minimal cytotoxicity, two of which inhibit B. burgdorferi growth. This study provides mechanistic insights into the function of BbLDH in the pathophysiology of B. burgdorferi and lays the groundwork for developing genus-specific metabolic inhibitors against B. burgdorferi and potentially other tick-borne pathogens as well.
Collapse
|
10
|
McCausland JW, Kloos ZA, Irnov I, Sonnert ND, Zhou J, Putnick R, Mueller EA, Steere AC, Palm NW, Grimes CL, Jacobs-Wagner C. Bacterial and host enzymes modulate the inflammatory response produced by the peptidoglycan of the Lyme disease agent. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631998. [PMID: 39829805 PMCID: PMC11741416 DOI: 10.1101/2025.01.08.631998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The spirochete Borrelia burgdorferi causes Lyme disease. In some patients, an excessive, dysregulated proinflammatory immune response can develop in joints leading to persistent arthritis. In such patients, persistence of antigenic B. burgdorferi peptidoglycan (PGBb) fragments within joint tissues may contribute to the immunopathogenesis, even after appropriate antibiotic treatment. In live B. burgdorferi cells, the outer membrane shields the polymeric PGBb sacculus from exposure to the immune system. However, unlike most diderm bacteria, B. burgdorferi releases PGBb turnover products into its environment due to the absence of recycling activity. In this study, we identified the released PGBb fragments using a mass spectrometry-based approach. By characterizing the l,d-carboxypeptidase activity of B. burgdorferi protein BB0605 (renamed DacA), we found that PGBb turnover largely occurs at sites of PGBb synthesis. In parallel, we demonstrated that the lytic transglycosylase activity associated with BB0259 (renamed MltS) releases PGBb fragments with 1,6-anhydro bond on their N-acetylmuramyl residues. Stimulation of human cell lines with various synthetic PGBb fragments revealed that 1,6-anhydromuramyl-containing PGBb fragments are poor inducers of a NOD2-dependent immune response relative to their hydrated counterparts. We also showed that the activity of the human N-acetylmuramyl-l-alanine amidase PGLYRP2, which reduces the immunogenicity of PGBb material, is low in joint (synovial) fluids relative to serum. Altogether, our findings suggest that MltS activity helps B. burgdorferi evade PG-based immune detection by NOD2 during growth despite shedding PGBb fragments and that PGBb-induced immunopathology likely results from host sensing of PGBb material from dead (lysed) spirochetes. Additionally, our results suggest the possibility that natural variation in PGLYRP2 activity may contribute to differences in susceptibility to PG-induced inflammation across tissues and individuals.
Collapse
Affiliation(s)
- Joshua W McCausland
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Zachary A Kloos
- Microbiology Program, Yale University, West Haven, Connecticut, USA
| | - Irnov Irnov
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Nicole D Sonnert
- Microbiology Program, Yale University, West Haven, Connecticut, USA
- Department of Immunology, Yale University School of Medicine, New Haven, CT, USA
| | - Junhui Zhou
- Department of Chemistry and Biochemistry, University of Delaware, Neward, DE
| | - Rachel Putnick
- Department of Chemistry and Biochemistry, University of Delaware, Neward, DE
| | - Elizabeth A Mueller
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Alan C Steere
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Noah W Palm
- Department of Immunology, Yale University School of Medicine, New Haven, CT, USA
| | - Catherine L Grimes
- Department of Chemistry and Biochemistry, University of Delaware, Neward, DE
| | - Christine Jacobs-Wagner
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
11
|
Miller AO, Carli AV, Kahlenberg C. Lyme Prosthetic Joint Infection May Be Underappreciated and Can Be Treated Without Surgery: A Case Report. JBJS Case Connect 2025; 15:01709767-202503000-00045. [PMID: 39977543 PMCID: PMC11837956 DOI: 10.2106/jbjs.cc.24.00600] [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] [Indexed: 02/22/2025]
Abstract
CASE A 68-year-old woman with a well-functioning total knee replacement presented with signs and symptoms of acute periprosthetic joint infection (PJI). Lyme serology and synovial fluid PCR were performed due to Borrelia burgdorferi. The patient was treated with oral doxycycline, had prompt resolution of symptoms, and remained asymptomatic 2 years later. CONCLUSION Lyme PJI may be underappreciated as a cause of culture-negative PJI, cannot be diagnosed in routine culture, and can be cured without surgery.
Collapse
|
12
|
Greiter BM, Sidorov S, Osuna E, Seiler M, Relly C, Hackenberg A, Luchsinger I, Cannizzaro E, Martin R, Marchesi M, von Felten S, Egli A, Berger C, Meyer Sauteur PM. Clinical characteristics and serological profiles of Lyme disease in children: a 15-year retrospective cohort study in Switzerland. THE LANCET REGIONAL HEALTH. EUROPE 2025; 48:101143. [PMID: 39736882 PMCID: PMC11683244 DOI: 10.1016/j.lanepe.2024.101143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 01/01/2025]
Abstract
Background Lyme disease (LD) is caused by Borrelia burgdorferi and is the most common tickborne disease in the northern hemisphere. Although classical characteristics of LD are well-known, the diagnosis and treatment are often delayed. Laboratory diagnosis by serological testing is recommended for most LD manifestations. The objective of this study was to describe clinical characteristics and associated serological profiles in children with LD. Methods This retrospective cohort study included children aged 0-18 years, diagnosed with LD according to current guidelines at University Children's Hospital Zurich between January 1, 2006 and December 31, 2020. Two-tier serological testing with the recomWell enzyme-linked immunosorbent assay and recomLine Western blot (MIKROGEN Diagnostik, MIKROGEN GmbH, Neuried, Germany) was performed at the Institute of Medical Microbiology, University of Zurich. Findings In total, 469 children diagnosed with LD were included (median age, 7.9 years); 190 patients (40.5%) with Lyme neuroborreliosis (LNB), 171 (36.5%) patients with skin manifestations (erythema migrans, n = 121; multiple erythema migrans, n = 11; borrelial lymphocytoma, n = 37; and acrodermatitis chronica atrophicans, n = 2), and 108 (23.0%) patients with Lyme arthritis. We observed seasonal variations for patients with skin manifestations and LNB, with high prevalence in May-October, but not for patients with Lyme arthritis. Significant differences between LD manifestation groups were found for age, inflammatory parameters, and specificity and concentration of B. burgdorferi-specific serum antibody responses. We observed distinct patterns of pronounced serum antibody responses against B. burgdorferi antigens in LNB (IgM against VlsE, p41, and OspC) and Lyme arthritis (IgG against p100, VlsE, p58, p41, p39, and p18). Interpretation Our study is one of the largest and most detailed for children with LD. We present unique findings regarding the differences in clinical characteristics and immune responses between various manifestations of LD in children. Funding No specific funding to disclose for this study.
Collapse
Affiliation(s)
- Beat M. Greiter
- Division of Infectious Diseases and Hospital Epidemiology, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Semjon Sidorov
- Division of Infectious Diseases and Hospital Epidemiology, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ester Osuna
- Division of Infectious Diseases and Hospital Epidemiology, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Michelle Seiler
- Emergency Department, University Children's Hospital Zurich, Zurich, Switzerland
| | - Christa Relly
- Division of Infectious Diseases and Hospital Epidemiology, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Annette Hackenberg
- Department of Pediatric Neurology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Isabelle Luchsinger
- Department of Dermatology, Pediatric Skin Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Elvira Cannizzaro
- Department of Rheumatology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Martina Marchesi
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
- Medica-Medical Laboratories, Zurich, Switzerland
| | - Stefanie von Felten
- Department of Biostatistics at Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Adrian Egli
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Christoph Berger
- Division of Infectious Diseases and Hospital Epidemiology, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Patrick M. Meyer Sauteur
- Division of Infectious Diseases and Hospital Epidemiology, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Kumaresan V, Hung CY, Hermann BP, Seshu J. Role of Dual Specificity Phosphatase 1 (DUSP1) in influencing inflammatory pathways in macrophages modulated by Borrelia burgdorferi lipoproteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624562. [PMID: 39605372 PMCID: PMC11601599 DOI: 10.1101/2024.11.20.624562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Borrelia burgdorferi (Bb), the spirochetal agent of Lyme disease, has a large array of lipoproteins that play a significant role in mediating host-pathogen interactions within ticks and vertebrates. Although there is substantial information on the effects of B. burgdorferi lipoproteins (BbLP) on immune modulatory pathways, the application of multi-omics methodologies to decode the transcriptional and proteomic patterns associated with host cell responses induced by lipoproteins in murine bone marrow-derived macrophages (BMDMs) has identified additional effectors and pathways. Single-cell RNA-Seq (scRNA-Seq) performed on BMDMs treated with various concentrations of borrelial lipoproteins revealed macrophage subsets within the BMDMs. Differential expression analysis showed that genes encoding various receptors, type I IFN-stimulated genes, signaling chemokines, and mitochondrial genes are altered in BMDMs in response to lipoproteins. Unbiased proteomics analysis of lysates of BMDMs treated with lipoproteins corroborated several of these findings. Notably, dual specificity phosphatase 1 (Dusp1) gene was upregulated during the early stages of BMDM exposure to BbLP. Pre-treatment with benzylidene-3-cyclohexylamino-1-indanone hydrochloride (BCI), an inhibitor of both DUSP1 and 6 prior to exposure to BbLP, demonstrated that DUSP1 negatively regulates NLRP3-mediated pro-inflammatory signaling and positively regulates the expression of interferon-stimulated genes and those encoding Ccl5, Il1b, and Cd274. Moreover, DUSP1, IkB kinase complex and MyD88 also modulate mitochondrial changes in BMDMs treated with borrelial lipoproteins. These findings advance the potential for exploiting DUSP1 as a therapeutic target to regulate host responses in reservoir hosts to limit survival of B. burgdorferi during its infectious cycle between ticks and mammalian hosts.
Collapse
Affiliation(s)
- Venkatesh Kumaresan
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX-78249
| | - Chiung-Yu Hung
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX-78249
| | - Brian P. Hermann
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX-78249
| | - J. Seshu
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX-78249
| |
Collapse
|
14
|
Marques A. Symptoms after Lyme disease: What's past is prologue. Sci Transl Med 2024; 16:eado2103. [PMID: 39536119 DOI: 10.1126/scitranslmed.ado2103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Protracted fatigue and other symptoms can occur after Lyme disease and other infections, with numerous possible drivers. Studies on posttreatment Lyme disease have been inconclusive, with no confirmed biomarker emerging. Prolonged antibiotic therapy provides no benefit. Thus, a holistic approach toward understanding and treating this complex disease is necessary.
Collapse
Affiliation(s)
- Adriana Marques
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1888 USA
| |
Collapse
|
15
|
Aboulaghras S, Bouyahya A, El Kadri K, Khalid A, Abdalla AN, Hassani R, Lee LH, Bakrim S. Protective and stochastic correlation between infectious diseases and autoimmune disorders. Microb Pathog 2024; 196:106919. [PMID: 39245422 DOI: 10.1016/j.micpath.2024.106919] [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: 08/10/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
A priori, early exposure to a wide range of bacteria, viruses, and parasites appears to fortify and regulate the immune system, potentially reducing the risk of autoimmune diseases. However, improving hygiene conditions in numerous societies has led to a reduction in these microbial exposures, which, according to certain theories, could contribute to an increase in autoimmune diseases. Indeed, molecular mimicry is a key factor triggering immune system reactions; while it seeks pathogens, it can bind to self-molecules, leading to autoimmune diseases associated with microbial infections. On the other hand, a hygiene-based approach aimed at reducing the load of infectious agents through better personal hygiene can be beneficial for such pathologies. This review sheds light on how the evolution of the innate immune system, following the evolution of molecular patterns associated with microbes, contributes to our protection but may also trigger autoimmune diseases linked to microbes. Furthermore, it addresses how hygiene conditions shield us against autoimmune diseases related to microbes but may lead to autoimmune pathologies not associated with microbes.
Collapse
Affiliation(s)
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco.
| | - Kawtar El Kadri
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco.
| | - Asaad Khalid
- Health Research Centre, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia.
| | - Rym Hassani
- Environment and Nature Research Centre, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; Biology Department, University College AlDarb, Jazan University, Jazan 45142, Saudi Arabia.
| | - Learn-Han Lee
- Microbiome Research Group, Research Centre for Life Science and Healthcare, Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute (CBI), University of Nottingham Ningbo China, 315000, Ningbo, China; Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, 47500, Malaysia.
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir, 80000, Morocco.
| |
Collapse
|
16
|
Doskaliuk B, Zimba O. Borrelia burgdorferi and autoimmune mechanisms: implications for mimicry, misdiagnosis, and mismanagement in Lyme disease and autoimmune disorders. Rheumatol Int 2024; 44:2265-2271. [PMID: 38578312 PMCID: PMC11424747 DOI: 10.1007/s00296-024-05580-x] [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: 02/13/2024] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
The genus Borrelia encompasses a diverse group of spirochetes transmitted primarily by ticks, with Borrelia burgdorferi causing Lyme disease, which is prevalent in North America and Europe. Borrelia's structural adaptations and ability to persist in diverse host tissues underscore its pathogenic potential. Beyond traditional infectious responses, Borrelia engages in complex interactions with the host immune system, contributing to autoimmune mechanisms such as molecular mimicry and persistent infections. This intricate interplay manifests in symptoms resembling various autoimmune diseases, including systemic lupus erythematosus, dermatomyositis, local scleroderma, and systemic sclerosis. However, these associations lack a precise explanation, emphasizing the need for further investigation. The cases of misdiagnosis between Lyme borreliosis and autoimmune diseases highlight the critical importance of accurate diagnostics and adherence to guidelines. Understanding Borrelia's impact on immune responses is pivotal for advancing diagnostics and targeted therapeutic interventions in Lyme borreliosis and its potential autoimmune implications.
Collapse
Affiliation(s)
- Bohdana Doskaliuk
- Department of Patophysiology, Ivano-Frankivsk National Medical University, Halytska str. 2, Ivano-Frankivsk, 76000, Ukraine.
| | - Olena Zimba
- Department of Clinical Rheumatology and Immunology, University Hospital in Krakow, Kraków, Poland
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
- Department of Internal Medicine N2, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| |
Collapse
|
17
|
Kim P, Lim SH, Lee JI, Namgung HG, Kim D, Lee EJ. Dysphagia due to Lyme Disease: A Case Report. BRAIN & NEUROREHABILITATION 2024; 17:e25. [PMID: 39649707 PMCID: PMC11621671 DOI: 10.12786/bn.2024.17.e25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/17/2024] [Accepted: 11/27/2024] [Indexed: 12/11/2024] Open
Abstract
Lyme disease is a multisystem infection that can affect the joints, heart, and nervous system when untreated. While it can present with cranial nerve palsy, dysphagia is rarely reported. This case highlights a rare instance of dysphagia in Lyme disease, typically known for neurological symptoms like facial nerve palsy. Despite the absence of erythema migrans or a documented tick bite, the patient's facial palsy, hearing loss, vocal cord paralysis, and dysphagia were attributed to Lyme disease. With the rising prevalence of Lyme disease, similar cases may increase, particularly in endemic regions of North America, Europe, and parts of Asia, emphasizing the need for early diagnosis and treatment in patients with unexplained dysphagia.
Collapse
Affiliation(s)
- Pureum Kim
- Department of Rehabilitation Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seong Hoon Lim
- Department of Rehabilitation Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Institute for Basic Medical Science, Catholic Medical Center, The Catholic University of Korea, Seoul, Korea
| | - Jong In Lee
- Department of Rehabilitation Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ho-Geon Namgung
- Department of Rehabilitation Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Donghwan Kim
- Department of Rehabilitation Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun Ji Lee
- Department of Rehabilitation Medicine, Yeouido St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
18
|
Alçada R, Gonçalves NP, Torres R, Lourenço MH, Flor de Lima B, Guerra Pinto F. Persistent Knee Monoarthritis as a Lyme Disease Presentation: A Diagnostic Challenge - Case Report. Rev Bras Ortop 2024; 59:e149-e154. [PMID: 39735473 PMCID: PMC11679618 DOI: 10.1055/s-0043-1771487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 02/27/2023] [Indexed: 12/31/2024] Open
Abstract
There are several differential diagnoses for knee monoarthritis. We report a patient with recurrent episodes of knee effusion, in which the non-specific clinical condition implied several diagnostic hypotheses, therapeutic inaccuracies, and a delay in implementing adequate treatment. For more than 2 years, the patient underwent different Orthopedics and Rheumatology visits. She received multiple therapies, including a knee arthroscopy with partial meniscectomy with transient improvement of the complaints but not a definitive diagnosis. After collecting synovial fluid samples and successively negative microbiological tests, we established the diagnosis of overlap of septic arthritis by atypical microorganisms isolated from synovial tissue ( Pantoea spp. and Staphylococcus saprophyticus ) and Lyme arthritis. Washing and surgical debridement followed by targeted antibiotic therapy resulted in a transient response due to persistent infection (stage 3). This case demonstrates the need for a multidisciplinary approach to knee monoarthritis.
Collapse
Affiliation(s)
- Rita Alçada
- Serviço de Ortopedia, Hospital de Cascais Dr. José de Almeida, Cascais, Portugal
| | | | - Rita Torres
- Centro Hospitalar Lisboa Ocidental, Lisboa, Portugal
| | | | | | | |
Collapse
|
19
|
Badami GD, Tamburini B, Mohammadnezhad L, Vaz-Rodrigues R, La Barbera L, de la Fuente J, Sireci G. Netosis and trained immunity in tick-borne diseases: a possible pathogenetic role. Cell Immunol 2024; 405-406:104881. [PMID: 39368167 DOI: 10.1016/j.cellimm.2024.104881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Various types of pathogens transmitted by ticks elicit distinct immune responses just like the emerging α-Gal syndrome that is associated with allergic reactions to tick bites. The mechanisms of Neutrophil Extracellular Traps release (called NETosis) and trained immunity in response to tick-borne microbes have not been extensively investigated. In our paper, we explored the intricate interplay of NETosis and trained immunity within the realm of infectious diseases triggered by tick bites and their possible pathogenetic role in autoimmunity. We conducted an extensive literature search to identify studies for this review, considering articles and reviews published in English within the last years. Additionally, we scrutinized the references of all included papers and relevant review articles to ensure comprehensive coverage. We shed light on a plausible correlation between these innate immune responses and their potential implication in certain pathological conditions, with a specific focus on some autoimmune diseases. These findings offer new perspectives for a more profound comprehension of the immunopathogenesis of certain autoimmune-like signs where clinicians should include Tick-Borne Diseases (TBDs) in their differential diagnoses, in those geographical areas of tick infestation.
Collapse
Affiliation(s)
- Giusto Davide Badami
- CLADIBIOR, Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy
| | - Bartolo Tamburini
- CLADIBIOR, Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy; Department of Health Promotion, Mother and Childcare, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90127 Palermo, Italy
| | - Leila Mohammadnezhad
- CLADIBIOR, Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy; Department of Health Promotion, Mother and Childcare, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90127 Palermo, Italy
| | - Rita Vaz-Rodrigues
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13071 Ciudad Real, Spain
| | - Lidia La Barbera
- Department of Health Promotion, Mother and Childcare, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90127 Palermo, Italy
| | - José de la Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13071 Ciudad Real, Spain; Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater OK 74078, USA
| | - Guido Sireci
- CLADIBIOR, Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy.
| |
Collapse
|
20
|
Rudolph MJ, Chen Y, Vorauer C, Vance DJ, Piazza CL, Willsey GG, McCarthy K, Muriuki B, Cavacini LA, Guttman M, Mantis NJ. Structure of a Human Monoclonal Antibody in Complex with Outer Surface Protein C of the Lyme Disease Spirochete, Borreliella burgdorferi. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1234-1243. [PMID: 39240158 DOI: 10.4049/jimmunol.2400247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024]
Abstract
Lyme disease is a tick-borne, multisystem infection caused by the spirochete Borreliella burgdorferi. Although Abs have been implicated in the resolution of Lyme disease, the specific B cell epitopes targeted during human infections remain largely unknown. In this study, we characterized and defined the structural epitope of a patient-derived bactericidal monoclonal IgG (B11) against outer surface protein C (OspC), a homodimeric lipoprotein necessary for B. burgdorferi tick-mediated transmission and early-stage colonization of vertebrate hosts. High-resolution epitope mapping was accomplished through hydrogen deuterium exchange-mass spectrometry and X-ray crystallography. Structural analysis of B11 Fab-OspCA complexes revealed the B11 Fabs associated in a 1:1 stoichiometry with the lateral faces of OspCA homodimers such that the Abs are essentially positioned perpendicular to the spirochete's outer surface. B11's primary contacts reside within the membrane-proximal regions of α-helices 1 and 6 and adjacent loops 5 and 6 in one OspCA monomer. In addition, B11 spans the OspCA dimer interface, engaging opposing α-helix 1', α-helix 2', and loop 2-3' in the second OspCA monomer. The B11-OspCA structure is reminiscent of the recently solved mouse transmission blocking monoclonal IgG B5 in complex with OspCA, indicating a mode of engagement with OspC that is conserved across species. In conclusion, we provide a detailed insight into the interaction between a functional human Ab and an immunodominant Lyme disease Ag long considered an important vaccine candidate.
Collapse
Affiliation(s)
| | - Yang Chen
- New York Structural Biology Center, New York, NY
| | - Clint Vorauer
- Department of Medicinal Chemistry, University of Washington, Seattle, WA
| | - David J Vance
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY
- Department of Biomedical Sciences, University at Albany, Albany, NY
| | - Carol Lyn Piazza
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY
| | - Graham G Willsey
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY
| | | | | | - Lisa A Cavacini
- University of Massachusetts Chan Medical School, Worcester, MA
| | - Miklos Guttman
- Department of Medicinal Chemistry, University of Washington, Seattle, WA
| | - Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY
- Department of Biomedical Sciences, University at Albany, Albany, NY
| |
Collapse
|
21
|
Rouse JR, Danner R, Wahhab A, Pereckas M, Nguyen C, McClune ME, Steere AC, Strle K, Jutras BL, Lochhead RB. HLA-DR-Expressing Fibroblast-Like Synoviocytes Are Inducible Antigen Presenting Cells That Present Autoantigens in Lyme Arthritis. ACR Open Rheumatol 2024; 6:678-689. [PMID: 39073021 PMCID: PMC11471949 DOI: 10.1002/acr2.11710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/09/2024] [Accepted: 06/10/2024] [Indexed: 07/30/2024] Open
Abstract
OBJECTIVE HLA-DR-expressing fibroblast-like synoviocytes (FLS) are a prominent cell type in synovial tissue in chronic inflammatory forms of arthritis. FLS-derived extracellular matrix (ECM) proteins, including fibronectin-1 (FN1), contain immunogenic CD4+ T cell epitopes in patients with postinfectious Lyme arthritis (LA). However, the role of FLS in presentation of these T cell epitopes remains uncertain. METHODS Primary LA FLS and primary murine FLS stimulated with interferon gamma (IFNγ), Borrelia burgdorferi, and/or B burgdorferi peptidoglycan (PG) were assessed for properties associated with antigen presentation. HLA-DR-presented peptides from stimulated LA FLS were identified by immunopeptidomics analysis. OT-II T cells were co-cultured with stimulated murine FLS in the presence of cognate ovalbumin antigen to determine the potential of FLS to act as inducible antigen presenting cells (APCs). RESULTS FLS expressed HLA-DR molecules within inflamed synovial tissue and tendons from patients with postinfectious LA in situ. Major histocompatibility complex (MHC) class II and co-stimulatory molecules were expressed by FLS following in vitro stimulation with IFNγ and B burgdorferi and presented both foreign and self-MHC-II peptides, including an immunogenic T cell epitope derived from Lyme autoantigen FN1. Stimulated FLS induced proliferation of naive OT-II CD4+ T cells that were dependent on OT-II antigen and CD40. Stimulation with B burgdorferi PG enhanced FLS-mediated T cell activation. CONCLUSION MHC-II+ FLS are inducible APCs that can induce CD4+ T cell activation in an antigen- and CD40-dependent manner. Activated FLS can also present ECM-derived Lyme autoantigens, implicating FLS in amplifying tissue-localized autoimmunity in LA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Allen C. Steere
- Massachusetts General Hospital and Harvard Medical SchoolBoston
| | | | | | | |
Collapse
|
22
|
Holers VM, Demoruelle KM, Buckner JH, James EA, Firestein GS, Robinson WH, Steere AC, Zhang F, Norris JM, Kuhn KA, Deane KD. Distinct mucosal endotypes as initiators and drivers of rheumatoid arthritis. Nat Rev Rheumatol 2024; 20:601-613. [PMID: 39251771 DOI: 10.1038/s41584-024-01154-0] [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] [Accepted: 07/30/2024] [Indexed: 09/11/2024]
Abstract
Rheumatoid arthritis (RA) is a potentially devastating autoimmune disease. The great majority of patients with RA are seropositive for anti-citrullinated protein antibodies (ACPAs), rheumatoid factors, or other autoantibodies. The onset of clinically apparent inflammatory arthritis meeting classification criteria (clinical RA) is preceded by ACPA seropositivity for an average of 3-5 years, a period that is designated as 'at-risk' of RA for ACPA-positive individuals who do not display signs of arthritis, or 'pre-RA' for individuals who are known to have progressed to developing clinical RA. Prior studies of individuals at-risk of RA have associated pulmonary mucosal inflammation with local production of ACPAs and rheumatoid factors, leading to development of the 'mucosal origins hypothesis'. Recent work now suggests the presence of multiple distinct mucosal site-specific mechanisms that drive RA evolution. Indicatively, subsets of individuals at-risk of RA and patients with RA harbour a faecal bacterial strain that has exhibited arthritogenic activity in animal models and that favours T helper 17 (TH17) cell responses in patients. Periodontal inflammation and oral microbiota have also been suggested to promote the development of arthritis through breaches in the mucosal barrier. Herein, we argue that mucosal sites and their associated microbial strains can contribute to RA evolution via distinct pathogenic mechanisms, which can be considered causal mucosal endotypes. Future therapies instituted for prevention in the at-risk period, or, perhaps, during clinical RA as therapeutics for active arthritis, will possibly have to address these individual mechanisms as part of precision medicine approaches.
Collapse
Affiliation(s)
- V Michael Holers
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA.
| | | | | | | | - Gary S Firestein
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, CA, USA
| | - William H Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Allen C Steere
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Fan Zhang
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Kristine A Kuhn
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| | - Kevin D Deane
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
23
|
Steere AC, Lemieux JE. Wider recognition and greater understanding of postinfectious, antibiotic-refractory Lyme arthritis. J Clin Invest 2024; 134:e184109. [PMID: 39225104 PMCID: PMC11364397 DOI: 10.1172/jci184109] [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] [Indexed: 09/04/2024] Open
Abstract
Lyme disease, caused by Borrelia burgdorferi (Bb), can progress to Lyme arthritis (LA). While most patients with LA respond successfully to antibiotic therapy, a small percentage fail to improve, a condition known as antibiotic-refractory Lyme arthritis (ARLA). While T cell responses are known to drive ARLA, molecular mechanisms for ARLA remain unknown. In this issue of the JCI, Dirks et al. isolated disease-specific Th cells from patients with ARLA residing in Germany. A distinct TCR-β motif distinguished ARLA from other rheumatic diseases. Notably, the TCR-β motif was linked predominantly to HLA-DRB1*11 or 13 alleles, which differed from alleles in patients from North America. It also mapped primarily to T peripheral helper (Tph) cells, as opposed to classical Th1 cells. These findings provide a roadmap explaining how T cell responses necessary for control of an infection can, despite antibiotic therapy, drive a disadvantageous T cell response, resulting in a postinfectious, inflammatory arthritis.
Collapse
Affiliation(s)
- Allen C. Steere
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology and
| | - Jacob E. Lemieux
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Samaddar S, Rolandelli A, O'Neal AJ, Laukaitis-Yousey HJ, Marnin L, Singh N, Wang X, Butler LR, Rangghran P, Kitsou C, Cabrera Paz FE, Valencia L, R Ferraz C, Munderloh UG, Khoo B, Cull B, Rosche KL, Shaw DK, Oliver J, Narasimhan S, Fikrig E, Pal U, Fiskum GM, Polster BM, Pedra JHF. Bacterial reprogramming of tick metabolism impacts vector fitness and susceptibility to infection. Nat Microbiol 2024; 9:2278-2291. [PMID: 38997520 PMCID: PMC11926704 DOI: 10.1038/s41564-024-01756-0] [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: 05/31/2023] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Arthropod-borne pathogens are responsible for hundreds of millions of infections in humans each year. The blacklegged tick, Ixodes scapularis, is the predominant arthropod vector in the United States and is responsible for transmitting several human pathogens, including the Lyme disease spirochete Borrelia burgdorferi and the obligate intracellular rickettsial bacterium Anaplasma phagocytophilum, which causes human granulocytic anaplasmosis. However, tick metabolic response to microbes and whether metabolite allocation occurs upon infection remain unknown. Here we investigated metabolic reprogramming in the tick ectoparasite I. scapularis and determined that the rickettsial bacterium A. phagocytophilum and the spirochete B. burgdorferi induced glycolysis in tick cells. Surprisingly, the endosymbiont Rickettsia buchneri had a minimal effect on bioenergetics. An unbiased metabolomics approach following A. phagocytophilum infection of tick cells showed alterations in carbohydrate, lipid, nucleotide and protein metabolism, including elevated levels of the pleiotropic metabolite β-aminoisobutyric acid. We manipulated the expression of genes associated with β-aminoisobutyric acid metabolism in I. scapularis, resulting in feeding impairment, diminished survival and reduced bacterial acquisition post haematophagy. Collectively, we discovered that metabolic reprogramming affects interspecies relationships and fitness in the clinically relevant tick I. scapularis.
Collapse
Affiliation(s)
- Sourabh Samaddar
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Agustin Rolandelli
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Anya J O'Neal
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hanna J Laukaitis-Yousey
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Liron Marnin
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Department of Biotechnology, School of Energy Technology, Pandit Deendayal Energy University; Knowledge Corridor, Gandhinagar, India
| | - Xiaowei Wang
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- MP Biomedicals, Solon, OH, USA
| | - L Rainer Butler
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Parisa Rangghran
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Francy E Cabrera Paz
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Luisa Valencia
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Camila R Ferraz
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | | | - Benedict Khoo
- Division of Environmental Health Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Benjamin Cull
- Department of Entomology, University of Minnesota, Saint Paul, MN, USA
| | - Kristin L Rosche
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Dana K Shaw
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Jonathan Oliver
- Division of Environmental Health Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Sukanya Narasimhan
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Erol Fikrig
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Gary M Fiskum
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
25
|
Priya R, Ye M, Raghunanadanan S, Liu Q, Li W, Yu Q, Lou Y, Sintim HO, Yang XF. Borrelia burgdorferi Secretes c-di-AMP as an Extracellular Pathogen-Associated Molecular Pattern to Elicit Type I Interferon Responses in Mammalian Hosts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607721. [PMID: 39185169 PMCID: PMC11343124 DOI: 10.1101/2024.08.13.607721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Borrelia burgdorferi (B. burgdorferi), an extracellular spirochetal pathogen, elicits a type-I interferon (IFN-I) response that contributes to the pathology of Lyme disease, including the development and severity of Lyme arthritis. However, the specific Pathogen-Associated Molecular Patterns (PAMPs) of B. burgdorferi responsible for triggering the IFN-I response are not well understood. Previous studies have identified an unknown, nuclease-resistant component in B. burgdorferi culture supernatants that significantly stimulates the IFN-I response, but its identity remains unknown. In this study, we reveal that B. burgdorferi secretes cyclic-di-adenosine monophosphate (c-di-AMP) as a key extracellular PAMP, inducing the host IFN-I response in macrophages. Using genetically manipulated B. burgdorferi strains, we demonstrate a requirement of c-di-AMP for stimulating IFN-I response by macrophages ex vivo. Additionally, infecting mice with B. burgdorferi alongside exogenous c-di-AMP resulted in a markedly increased IFN-I response in mouse tissues. Furthermore, inactivation or inhibition of the host STING signaling pathway significantly reduced the IFN-I response, indicating that c-di-AMP-induced IFN-I production is STING-dependent. Our findings identify c-di-AMP as a crucial PAMP secreted by B. burgdorferi to elicit the host IFN-I response via activation of STING signaling pathway, suggesting that targeting c-di-AMP production could represent a novel therapeutic strategy against Lyme arthritis.
Collapse
Affiliation(s)
- Raj Priya
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Meiping Ye
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Sajith Raghunanadanan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qiang Liu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
- Wenzhou key laboratory of sanitary microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical UniversityDepartment of Chemistry, 560 Oval Drive, West Lafayette, IN 47907, USA
| | - Wei Li
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qigui Yu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yongliang Lou
- Wenzhou key laboratory of sanitary microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical UniversityDepartment of Chemistry, 560 Oval Drive, West Lafayette, IN 47907, USA
| | - Herman O. Sintim
- Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, West Lafayette, IN 47907, USA
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, 560 Oval Drive, IN 47907, USA
| | - X. Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
26
|
Ehrbar D, Arvikar SL, Sulka KB, Chiumento G, Nelson NLJ, Hernandez SA, Williams MA, Strle F, Steere AC, Strle K. Variants in the Late Cornified Envelope Gene Locus Are Associated With Elevated T-helper 17 Responses in Patients With Postinfectious Lyme Arthritis. J Infect Dis 2024; 230:S40-S50. [PMID: 39140723 PMCID: PMC11322884 DOI: 10.1093/infdis/jiae164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Postinfectious Lyme arthritis (LA) is associated with dysregulated immunity and autoreactive T- and B-cell responses in joints. Here we explored the role of host genetic variation in this outcome. METHODS The frequency of 253 702 single-nucleotide polymorphisms (SNPs) was determined in 147 patients with LA (87 with postinfectious LA and 60 with antibiotic-responsive LA), and for comparison in 90 patients with erythema migrans or the general population (n = 2504). Functional outcome of candidate SNPs was assessed by evaluating their impact on clinical outcome and on immune responses in blood and synovial fluid in patients with LA. RESULTS Six SNPs associated with late cornified envelope (LCE3) genes were present at greater frequency in patients with postinfectious LA compared to those with antibiotic-responsive LA (70% vs 30%; odds ratio, 2; P < .01). These SNPs were associated with heightened levels of inflammatory Th17 cytokines in serum but lower levels of interleukin 27, a regulatory cytokine, implying that they may contribute to dysregulated Th17 immunity in blood. Moreover, in patients with postinfectious LA, the levels of these Th17 mediators correlated directly with autoantibody responses in synovial fluid, providing a possible link between LCE3 SNPs, maladaptive systemic Th17 immunity, and autoreactive responses in joints. CONCLUSIONS Variation in the LCE3 locus, a known genetic risk factor in psoriasis and psoriatic arthritis, is associated with dysregulated systemic Th17 immunity and heightened autoantibody responses in joints. These findings underscore the importance of host genetic predisposition and systemic Th17 immunity in the pathogenesis of postinfectious (antibiotic-refractory) Lyme arthritis.
Collapse
Affiliation(s)
- Dylan Ehrbar
- Department of Biological Sciences, University at Albany
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York
| | - Sheila L Arvikar
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School
| | - Katherine B Sulka
- Department of Immunology, Tufts University Graduate School of Biomedical Sciences
| | - Geena Chiumento
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School
| | - Nicole L J Nelson
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York
| | - Sergio A Hernandez
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts
| | - Morgan A Williams
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts
| | - Franc Strle
- Department of Infectious Diseases, University Medical Center Ljubljana, Slovenia
| | - Allen C Steere
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School
| | - Klemen Strle
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
27
|
Danner R, Prochniak LM, Pereckas M, Rouse JR, Wahhab A, Hackner LG, Lochhead RB. Identification of Major Histocompatibility Complex Class II Epitopes From Lyme Autoantigen Apolipoprotein B-100 and Borrelia burgdorferi Mcp4 in Murine Lyme Arthritis. J Infect Dis 2024; 230:S27-S39. [PMID: 39140726 PMCID: PMC11322890 DOI: 10.1093/infdis/jiae324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND During infection with the Lyme arthritis (LA) pathogen Borrelia burgdorferi, T-cell responses to both host and pathogen are dysregulated, resulting in chronic infection and frequent development of autoimmunity. METHODS To assess CD4+ T-cell epitopes presented during development of LA, we used an unbiased, immunopeptidomics approach to characterize the major histocompatibility complex (MHC) class II immunopeptidome in B burgdorferi-infected C57BL/6 (B6) mice, which develop mild, self-limiting LA, and infected B6 Il10-/- mice, which develop severe, persistent LA at 0, 4, and 16 weeks postinfection (22-23 mice per group). RESULTS Peptides derived from proteins involved in adaptive T- and B-cell responses and cholesterol metabolism, including human Lyme autoantigen apolipoprotein B-100 (apoB-100), were enriched in infected Il10-/- mice; whereas peptides derived from proteins involved in neutrophil extracellular net formation were enriched in infected B6 mice. Presentation of apoB-100 peptides showed evidence of epitope expansion during infection. Of several identified B burgdorferi peptides, only 1, a methyl-accepting chemotaxis protein peptide Mcp4442-462, was immunogenic. CONCLUSIONS ApoB-100, a human Lyme autoantigen, undergoes marked epitope expansion during LA development. The paucity of immunogenic B burgdorferi epitopes supports previous findings suggesting CD4+ T-cell responses are suppressed in murine LA.
Collapse
Affiliation(s)
- Rebecca Danner
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Lauren M Prochniak
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michaela Pereckas
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Joseph R Rouse
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Amanda Wahhab
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Lauren G Hackner
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Robert B Lochhead
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Division of Rheumatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
28
|
Steere AC. Lyme Arthritis: A 50-Year Journey. J Infect Dis 2024; 230:S1-S10. [PMID: 39140724 PMCID: PMC11322885 DOI: 10.1093/infdis/jiae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
Lyme arthritis (LA) was recognized as a separate entity in 1975 because of geographic clustering of children often diagnosed with juvenile rheumatoid arthritis in Lyme, Connecticut. After identification of erythema migrans as a common early feature of the illness, a prospective study of such patients implicated Ixodes scapularis ticks in disease transmission. In 1982, the causative agent, now called Borrelia burgdorferi, was cultured from these ticks and from Lyme disease patients. Subsequently, it was shown that LA could usually be treated successfully with oral antibiotics but sometimes required intravenous antibiotics. Yet, a small percentage of patients developed a dysregulated, proinflammatory immune response leading to persistent postinfectious synovitis with vascular damage, cytotoxic and autoimmune responses, and fibroblast proliferation, a lesion similar to that of rheumatoid arthritis. The message from postinfectious LA for other autoimmune arthritides is that a complex immune response with autoimmune features can begin with a microbial infection.
Collapse
Affiliation(s)
- Allen C Steere
- Center for Immunology and Inflammatory Diseases, Department of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
29
|
Bockenstedt LK, Belperron AA. Insights From Omics in Lyme Disease. J Infect Dis 2024; 230:S18-S26. [PMID: 39140719 DOI: 10.1093/infdis/jiae250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
Lyme disease is a zoonotic infection due to Ixodes tick-transmitted Borrelia burgdorferi sensu lato spirochetes and the most common vector-borne disease in the Northern Hemisphere. Despite nearly 50 years of investigation, the pathogenesis of this infection and its 2 main adverse outcomes-postinfectious Lyme arthritis and posttreatment Lyme disease syndrome-are incompletely understood. Advancement in sequencing and mass spectrometry have led to the rapid expansion of high-throughput omics technologies, including transcriptomics, metabolomics, and proteomics, which are now being applied to human diseases. This review summarizes findings of omics studies conducted on blood and tissue samples of people with acute Lyme disease and its postinfectious outcomes.
Collapse
Affiliation(s)
- Linda K Bockenstedt
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Alexia A Belperron
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
30
|
Dirks J, Fischer J, Klaussner J, Hofmann C, Holl-Wieden A, Buck V, Klemann C, Girschick HJ, Caruana I, Erhard F, Morbach H. Disease-specific T cell receptors maintain pathogenic T helper cell responses in postinfectious Lyme arthritis. J Clin Invest 2024; 134:e179391. [PMID: 38963700 PMCID: PMC11364382 DOI: 10.1172/jci179391] [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: 01/17/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUNDAntibiotic-Refractory Lyme Arthritis (ARLA) involves a complex interplay of T cell responses targeting Borrelia burgdorferi antigens progressing toward autoantigens by epitope spreading. However, the precise molecular mechanisms driving the pathogenic T cell response in ARLA remain unclear. Our aim was to elucidate the molecular program of disease-specific Th cells.METHODSUsing flow cytometry, high-throughput T cell receptor (TCR) sequencing, and scRNA-Seq of CD4+ Th cells isolated from the joints of patients with ARLA living in Europe, we aimed to infer antigen specificity through unbiased analysis of TCR repertoire patterns, identifying surrogate markers for disease-specific TCRs, and connecting TCR specificity to transcriptional patterns.RESULTSPD-1hiHLA-DR+CD4+ effector T cells were clonally expanded within the inflamed joints and persisted throughout disease course. Among these cells, we identified a distinct TCR-β motif restricted to HLA-DRB1*11 or *13 alleles. These alleles, being underrepresented in patients with ARLA living in North America, were unexpectedly prevalent in our European cohort. The identified TCR-β motif served as surrogate marker for a convergent TCR response specific to ARLA, distinguishing it from other rheumatic diseases. In the scRNA-Seq data set, the TCR-β motif particularly mapped to peripheral T helper (TPH) cells displaying signs of sustained proliferation, continuous TCR signaling, and expressing CXCL13 and IFN-γ.CONCLUSIONBy inferring disease-specific TCRs from synovial T cells we identified a convergent TCR response in the joints of patients with ARLA that continuously fueled the expansion of TPH cells expressing a pathogenic cytokine effector program. The identified TCRs will aid in uncovering the major antigen targets of the maladaptive immune response.FUNDINGSupported by the German Research Foundation (DFG) MO 2160/4-1; the Federal Ministry of Education and Research (BMBF; Advanced Clinician Scientist-Program INTERACT; 01EO2108) embedded in the Interdisciplinary Center for Clinical Research (IZKF) of the University Hospital Würzburg; the German Center for Infection Research (DZIF; Clinical Leave Program; TI07.001_007) and the Interdisciplinary Center for Clinical Research (IZKF) Würzburg (Clinician Scientist Program, Z-2/CSP-30).
Collapse
MESH Headings
- Humans
- Lyme Disease/immunology
- Lyme Disease/pathology
- Lyme Disease/genetics
- HLA-DRB1 Chains/genetics
- HLA-DRB1 Chains/immunology
- Female
- Male
- T-Lymphocytes, Helper-Inducer/immunology
- Borrelia burgdorferi/immunology
- Middle Aged
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Adult
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
Collapse
Affiliation(s)
- Johannes Dirks
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Jonas Fischer
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Julia Klaussner
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Christine Hofmann
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Annette Holl-Wieden
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Viktoria Buck
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Christian Klemann
- Department of Pediatric Immunology, Rheumatology, and Infectiology, Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
| | | | - Ignazio Caruana
- Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, Würzburg, Germany
| | - Florian Erhard
- Computational Systems Virology and Bioinformatics, Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
- Faculty for Informatics and Data Science, University of Regensburg, Regensburg, Germany
| | - Henner Morbach
- Pediatric Inflammation Medicine, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
31
|
Koutantou M, Drancourt M, Angelakis E. Prevalence of Lyme Disease and Relapsing Fever Borrelia spp. in Vectors, Animals, and Humans within a One Health Approach in Mediterranean Countries. Pathogens 2024; 13:512. [PMID: 38921809 PMCID: PMC11206712 DOI: 10.3390/pathogens13060512] [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: 05/02/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
The genus Borrelia has been divided into Borreliella spp., which can cause Lyme Disease (LD), and Borrelia spp., which can cause Relapsing Fever (RF). The distribution of genus Borrelia has broadened due to factors such as climate change, alterations in land use, and enhanced human and animal mobility. Consequently, there is an increasing necessity for a One Health strategy to identify the key components in the Borrelia transmission cycle by monitoring the human-animal-environment interactions. The aim of this study is to summarize all accessible data to increase our understanding and provide a comprehensive overview of Borrelia distribution in the Mediterranean region. Databases including PubMed, Google Scholar, and Google were searched to determine the presence of Borreliella and Borrelia spp. in vectors, animals, and humans in countries around the Mediterranean Sea. A total of 3026 were identified and screened and after exclusion of papers that did not fulfill the including criteria, 429 were used. After examination of the available literature, it was revealed that various species associated with LD and RF are prevalent in vectors, animals, and humans in Mediterranean countries and should be monitored in order to effectively manage and prevent potential infections.
Collapse
Affiliation(s)
- Myrto Koutantou
- Diagnostic Department and Public Health Laboratories, Hellenic Pasteur Institute, 11521 Athens, Greece
| | | | - Emmanouil Angelakis
- Diagnostic Department and Public Health Laboratories, Hellenic Pasteur Institute, 11521 Athens, Greece
| |
Collapse
|
32
|
Rudolph MJ, Chen Y, Vorauer C, Vance DJ, Piazza CL, Willsey GG, McCarthy K, Muriuki B, Cavacini LA, Guttman M, Mantis NJ. Structure of a human monoclonal antibody in complex with Outer surface protein C (OspC) of the Lyme disease spirochete, Borreliella burgdorferi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591597. [PMID: 38746285 PMCID: PMC11092446 DOI: 10.1101/2024.04.29.591597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Lyme disease is a tick-borne, multisystem infection caused by the spirochete, Borreliella burgdorferi . Although antibodies have been implicated in the resolution of Lyme disease, the specific B cell epitopes targeted during human infections remain largely unknown. In this study, we characterized and defined the structural epitope of a patient-derived bactericidal monoclonal IgG ("B11") against Outer surface protein C (OspC), a homodimeric lipoprotein necessary for B. burgdorferi tick-mediated transmission and early-stage colonization of vertebrate hosts. High-resolution epitope mapping was accomplished through hydrogen deuterium exchange-mass spectrometry (HDX-MS) and X-ray crystallography. Structural analysis of B11 Fab-OspC A complexes revealed the B11 Fabs associated in a 1:1 stoichiometry with the lateral faces of OspC A homodimers such that the antibodies are essentially positioned perpendicular to the spirochete's outer surface. B11's primary contacts reside within the membrane proximal regions of α-helices 1 and 6 and adjacent loops 5 and 6 in one OspC A monomer. In addition, B11 spans the OspC A dimer interface, engaging opposing α-helix 1', α-helix 2', and loop 2-3' in the second OspC A monomer. The B11-OspC A structure is reminiscent of the recently solved mouse transmission blocking monoclonal IgG B5 in complex with OspC A , indicating a mode of engagement with OspC that is conserved across species. In conclusion, we provide the first detailed insight into the interaction between a functional human antibody and an immunodominant Lyme disease antigen long considered an important vaccine target.
Collapse
|
33
|
Baker H, Amaral JK, Schoen RT. Management of postinfectious inflammatory arthritis. Curr Opin Rheumatol 2024; 36:155-162. [PMID: 38411201 DOI: 10.1097/bor.0000000000001009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
PURPOSE OF REVIEW Postinfectious inflammatory arthritis can result from various pathogens, including bacteria, viruses, fungi, and parasites. Prompt identification and treatment of acute infection is vital, but some cases progress to chronic arthritis despite successful treatment of infection. Postinfectious inflammatory arthritis varies from mild, self-limited arthralgia to severe, refractory arthritis, necessitating ongoing disease-modifying treatment. This review explores the spectrum of postinfectious inflammatory arthritis to provide insights into effective management. RECENT FINDINGS Research continues regarding the benefit of antimicrobial therapy, beyond treatment of the acute infection, to diminish the severity of postinfectious inflammatory arthritis. Following treatment of acute infection, most cases are self-limited so treatment is symptomatic. However, a difficult-to-predict fraction of cases develop chronic postinfectious inflammatory arthritis that can be challenging to manage. Recently, as more biologic, and targeted synthetic DMARDs have become available, treatment options have expanded. SUMMARY In this article, we use the term 'postinfectious inflammatory arthritis' rather than 'reactive arthritis' because it describes a broader spectrum of diseases and emphasizes the common pathogenesis of a postinfectious inflammatory process. We summarize the conventional therapies and recent management developments for the most frequently encountered postinfectious inflammatory arthritides.
Collapse
Affiliation(s)
- Hailey Baker
- Section of Rheumatology, Allergy, and Immunology, Yale School of Medicine
| | - J Kennedy Amaral
- Institute of Diagnostic Medicine of Cariri, Juazeiro do Norte, Ceará, Brazil
| | - Robert T Schoen
- Section of Rheumatology, Allergy, and Immunology, Yale School of Medicine
| |
Collapse
|
34
|
van de Schoor FR, Baarsma ME, Gauw SA, Ursinus J, Vrijmoeth HD, Ter Hofstede HJM, Tulen AD, Harms MG, Wong A, van den Wijngaard CC, Joosten LAB, Hovius JW, Kullberg BJ. Evaluation and 1-year follow-up of patients presenting at a Lyme borreliosis expertise centre: a prospective cohort study with validated questionnaires. Eur J Clin Microbiol Infect Dis 2024; 43:937-946. [PMID: 38492058 PMCID: PMC11108889 DOI: 10.1007/s10096-024-04770-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/31/2024] [Indexed: 03/18/2024]
Abstract
OBJECTIVES To describe the course of symptoms reported by patients with symptoms attributed to Lyme borreliosis (LB) without being subsequently diagnosed with LB. METHODS We performed a prospective cohort study with patients presenting at the outpatient clinic of two clinical LB centres. The primary outcome was the prevalence of persistent symptoms, which were defined as clinically relevant fatigue (CIS, subscale fatigue), pain (SF-36, subscale bodily pain), and cognitive impairment (CFQ) for ≥ 6 months and onset < 6 months over the first year of follow-up. Outcomes were compared with a longitudinal cohort of confirmed LB patients and a general population cohort. Prevalences were standardised to the distribution of pre-defined confounders in the confirmed LB cohort. RESULTS Participants (n = 123) reported mostly fatigue, arthralgia, myalgia, and paraesthesia as symptoms. The primary outcome could be determined for 74.8% (92/123) of participants. The standardised prevalence of persistent symptoms in our participants was 58.6%, which was higher than in patients with confirmed LB at baseline (27.2%, p < 0.0001) and the population cohort (21.2%, p < 0.0001). Participants reported overall improvement of fatigue (p < 0.0001) and pain (p < 0.0001) but not for cognitive impairment (p = 0.062) during the follow-up, though symptom severity at the end of follow-up remained greater compared to confirmed LB patients (various comparisons p < 0.05). CONCLUSION Patients with symptoms attributed to LB who present at clinical LB centres without physician-confirmed LB more often report persistent symptoms and report more severe symptoms compared to confirmed LB patients and a population cohort.
Collapse
Affiliation(s)
- F R van de Schoor
- Radboudumc, Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - M E Baarsma
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - S A Gauw
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - J Ursinus
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - H D Vrijmoeth
- Radboudumc, Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - H J M Ter Hofstede
- Radboudumc, Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - A D Tulen
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - M G Harms
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - A Wong
- Department of Statistics, Informatics and Modeling, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - C C van den Wijngaard
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - L A B Joosten
- Radboudumc, Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - J W Hovius
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Infection and Immunology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| | - B J Kullberg
- Radboudumc, Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
35
|
Horowitz RI, Fallon J, Freeman PR. Combining Double-Dose and High-Dose Pulsed Dapsone Combination Therapy for Chronic Lyme Disease/Post-Treatment Lyme Disease Syndrome and Co-Infections, Including Bartonella: A Report of 3 Cases and a Literature Review. Microorganisms 2024; 12:909. [PMID: 38792737 PMCID: PMC11124288 DOI: 10.3390/microorganisms12050909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/03/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Three patients with relapsing and remitting borreliosis, babesiosis, and bartonellosis, despite extended anti-infective therapy, were prescribed double-dose dapsone combination therapy (DDDCT) for 8 weeks, followed by one or several two-week courses of pulsed high-dose dapsone combination therapy (HDDCT). We discuss these patients' cases to illustrate three important variables required for long-term remission. First, diagnosing and treating active co-infections, including Babesia and Bartonella were important. Babesia required rotations of multiple anti-malarial drug combinations and herbal therapies, and Bartonella required one or several 6-day HDDCT pulses to achieve clinical remission. Second, all prior oral, intramuscular (IM), and/or intravenous (IV) antibiotics used for chronic Lyme disease (CLD)/post-treatment Lyme disease syndrome (PTLDS), irrespective of the length of administration, were inferior in efficacy to short-term pulsed biofilm/persister drug combination therapy i.e., dapsone, rifampin, methylene blue, and pyrazinamide, which improved resistant fatigue, pain, headaches, insomnia, and neuropsychiatric symptoms. Lastly, addressing multiple factors on the 16-point multiple systemic infectious disease syndrome (MSIDS) model was important in achieving remission. In conclusion, DDDCT with one or several 6-7-day pulses of HDDCT, while addressing abnormalities on the 16-point MSIDS map, could represent a novel effective clinical and anti-infective strategy in CLD/PTLDS and associated co-infections including Bartonella.
Collapse
Affiliation(s)
- Richard I. Horowitz
- New York State Department of Health Tick-Borne Working Group, Albany, NY 12224, USA
- Hudson Valley Healing Arts Center, Hyde Park, NY 12538, USA; (J.F.); (P.R.F.)
| | - John Fallon
- Hudson Valley Healing Arts Center, Hyde Park, NY 12538, USA; (J.F.); (P.R.F.)
| | - Phyllis R. Freeman
- Hudson Valley Healing Arts Center, Hyde Park, NY 12538, USA; (J.F.); (P.R.F.)
| |
Collapse
|
36
|
Vance DJ, Basir S, Piazza CL, Willsey GG, Haque HME, Tremblay JM, Rudolph MJ, Muriuki B, Cavacini L, Weis DD, Shoemaker CB, Mantis NJ. Single-domain antibodies reveal unique borrelicidal epitopes on the Lyme disease vaccine antigen, outer surface protein A (OspA). Infect Immun 2024; 92:e0008424. [PMID: 38470113 PMCID: PMC11003225 DOI: 10.1128/iai.00084-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/13/2024] Open
Abstract
Camelid-derived, single-domain antibodies (VHHs) have proven to be extremely powerful tools in defining the antigenic landscape of immunologically heterogeneous surface proteins. In this report, we generated a phage-displayed VHH library directed against the candidate Lyme disease vaccine antigen, outer surface protein A (OspA). Two alpacas were immunized with recombinant OspA serotype 1 from Borrelia burgdorferi sensu stricto strain B31, in combination with the canine vaccine RECOMBITEK Lyme containing lipidated OspA. The phage library was subjected to two rounds of affinity enrichment ("panning") against recombinant OspA, yielding 21 unique VHHs within two epitope bins, as determined through competition enzyme linked immunosorbent assays (ELISAs) with a panel of OspA-specific human monoclonal antibodies. Epitope refinement was conducted by hydrogen exchange-mass spectrometry. Six of the monovalent VHHs were expressed as human IgG1-Fc fusion proteins and shown to have functional properties associated with protective human monoclonal antibodies, including B. burgdorferi agglutination, outer membrane damage, and complement-dependent borreliacidal activity. The VHHs displayed unique reactivity profiles with the seven OspA serotypes associated with B. burgdorferi genospecies in the United States and Europe consistent with there being unique epitopes across OspA serotypes that should be considered when designing and evaluating multivalent Lyme disease vaccines.
Collapse
Affiliation(s)
- David J. Vance
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, USA
- Department of Biomedical Sciences, University at Albany, Albany, New York, USA
| | - Saiful Basir
- Department of Biomedical Sciences, University at Albany, Albany, New York, USA
| | - Carol Lyn Piazza
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, USA
| | - Graham G. Willsey
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, USA
| | | | - Jacque M. Tremblay
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | | | - Beatrice Muriuki
- Department of Medicine, University of Massachusetts Chan School of Medicine, Worcester, Massachusetts, USA
| | - Lisa Cavacini
- Department of Medicine, University of Massachusetts Chan School of Medicine, Worcester, Massachusetts, USA
| | - David D. Weis
- Department of Chemistry, The University of Kansas, Lawrence, Kansas, USA
| | - Charles B. Shoemaker
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Nicholas J. Mantis
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, USA
- Department of Biomedical Sciences, University at Albany, Albany, New York, USA
| |
Collapse
|
37
|
Vrijmoeth HD, Ursinus J, Botey-Bataller J, Kuijpers Y, Chu X, van de Schoor FR, Scicluna BP, Xu CJ, Netea MG, Kullberg BJ, van den Wijngaard CC, Li Y, Hovius JW, Joosten LAB. Genome-wide analyses in Lyme borreliosis: identification of a genetic variant associated with disease susceptibility and its immunological implications. BMC Infect Dis 2024; 24:337. [PMID: 38515037 PMCID: PMC10956190 DOI: 10.1186/s12879-024-09217-z] [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: 08/22/2023] [Accepted: 03/12/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Genetic variation underly inter-individual variation in host immune responses to infectious diseases, and may affect susceptibility or the course of signs and symptoms. METHODS We performed genome-wide association studies in a prospective cohort of 1138 patients with physician-confirmed Lyme borreliosis (LB), the most common tick-borne disease in the Northern hemisphere caused by the bacterium Borrelia burgdorferi sensu lato. Genome-wide variants in LB patients-divided into a discovery and validation cohort-were compared to two healthy cohorts. Additionally, ex vivo monocyte-derived cytokine responses of peripheral blood mononuclear cells to several stimuli including Borrelia burgdorferi were performed in both LB patient and healthy control samples, as were stimulation experiments using mechanistic/mammalian target of rapamycin (mTOR) inhibitors. In addition, for LB patients, anti-Borrelia antibody responses were measured. Finally, in a subset of LB patients, gene expression was analysed using RNA-sequencing data from the ex vivo stimulation experiments. RESULTS We identified a previously unknown genetic variant, rs1061632, that was associated with enhanced LB susceptibility. This polymorphism was an eQTL for KCTD20 and ETV7 genes, and its major risk allele was associated with upregulation of the mTOR pathway and cytokine responses, and lower anti-Borrelia antibody production. In addition, we replicated the recently reported SCGB1D2 locus that was suggested to have a protective effect on B. burgdorferi infection, and associated this locus with higher Borrelia burgdorferi antibody indexes and lower IL-10 responses. CONCLUSIONS Susceptibility for LB was associated with higher anti-inflammatory responses and reduced anti-Borrelia antibody production, which in turn may negatively impact bacterial clearance. These findings provide important insights into the immunogenetic susceptibility for LB and may guide future studies on development of preventive or therapeutic measures. TRIAL REGISTRATION The LymeProspect study was registered with the International Clinical Trials Registry Platform (NTR4998, registration date 2015-02-13).
Collapse
Affiliation(s)
- Hedwig D Vrijmoeth
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Jeanine Ursinus
- Department of Internal Medicine, Division of Infectious Diseases, Amsterdam UMC, Location AMC, University of Amsterdam, P.O. Box 22660, Amsterdam, 1100 DD, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Javier Botey-Bataller
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
- Department of Computational Biology for Individualised Infection Medicine, Centre for Individualised Infection Medicine, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
| | - Yunus Kuijpers
- Department of Computational Biology for Individualised Infection Medicine, Centre for Individualised Infection Medicine, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
| | - Xiaojing Chu
- Department of Computational Biology for Individualised Infection Medicine, Centre for Individualised Infection Medicine, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
| | - Freek R van de Schoor
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, MSD 2080, Msida, Malta
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences, University of Malta, MSD 2080, Msida, Malta
| | - Cheng-Jian Xu
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
- Department of Computational Biology for Individualised Infection Medicine, Centre for Individualised Infection Medicine, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
| | - Mihai G Netea
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, 53113, Bonn, Germany
| | - Bart Jan Kullberg
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - Cees C van den Wijngaard
- National Institute for Public Health and Environment (RIVM), Center for Infectious Disease Control, Bilthoven, 3720 BA, the Netherlands
| | - Yang Li
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
- Department of Computational Biology for Individualised Infection Medicine, Centre for Individualised Infection Medicine, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, 30625, Hannover, Germany
| | - Joppe W Hovius
- Department of Internal Medicine, Division of Infectious Diseases, Amsterdam UMC, Location AMC, University of Amsterdam, P.O. Box 22660, Amsterdam, 1100 DD, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands.
| |
Collapse
|
38
|
Giardini HAM, Neves FS, Pereira IA, Cordeiro RA. Lyme disease and Whipple's disease: a comprehensive review for the rheumatologist. Adv Rheumatol 2024; 64:16. [PMID: 38438928 DOI: 10.1186/s42358-024-00359-x] [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: 01/16/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024] Open
Abstract
Despite their rarity, Lyme disease and Whipple's disease are of significant importance in rheumatology, as both can manifest as chronic arthritis, presenting challenges in the differential diagnosis of inflammatory arthropathies. In Lyme disease, arthritis typically emerges as a late manifestation, usually occurring six months after the onset of erythema migrans. The predominant presentation involves mono- or oligoarthritis of large joints, with a chronic or remitting-recurrent course. Even with appropriate antimicrobial treatment, arthritis may persist due to inadequate immunological control triggered by the disease. In contrast, Whipple's disease may present with a migratory and intermittent seronegative poly- or oligoarthritis of large joints, preceding classic gastrointestinal symptoms by several years. Both disorders, particularly Whipple's disease, can be misdiagnosed as more common autoimmune rheumatic conditions such as rheumatoid arthritis and spondyloarthritis. Epidemiology is crucial in suspecting and diagnosing Lyme disease, as the condition is transmitted by ticks prevalent in specific areas of the United States, Europe, and Asia. On the contrary, the causative agent of Whipple's disease is widespread in the environment, yet invasive disease is rare and likely dependent on host genetic factors. In addition to erythema migrans in Lyme disease and gastrointestinal manifestations in Whipple's disease, neurological and cardiac involvement can further complicate the course of both. This article offers a comprehensive review of the epidemiological, pathophysiological, clinical, and therapeutic aspects of both diseases.
Collapse
Affiliation(s)
- Henrique Ayres Mayrink Giardini
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, 455- 3º andar- sala 3192 Cerqueira Cesar, CEP:01246-903, Sao Paulo, SP, Brazil.
| | - Fabricio Souza Neves
- Rheumatology Division, Internal Medicine Department, Health Sciences Center, Universidade Federal de Santa Catarina (UFSC), Florianopolis, SC, Brazil
| | | | - Rafael Alves Cordeiro
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, 455- 3º andar- sala 3192 Cerqueira Cesar, CEP:01246-903, Sao Paulo, SP, Brazil
| |
Collapse
|
39
|
Bowman KA, Wiggins CD, DeRiso E, Paul S, Strle K, Branda JA, Steere AC, Lauffenburger DA, Alter G. Borrelia-specific antibody profiles and complement deposition in joint fluid distinguish antibiotic-refractory from -responsive Lyme arthritis. iScience 2024; 27:108804. [PMID: 38303696 PMCID: PMC10830897 DOI: 10.1016/j.isci.2024.108804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/24/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024] Open
Abstract
Lyme arthritis, caused by the spirochete Borrelia burgdorferi, is the most common feature of late disseminated Lyme disease in the United States. While most Lyme arthritis resolves with antibiotics, termed "antibiotic-responsive", some individuals develop progressive synovitis despite antibiotic therapy, called "antibiotic-refractory" Lyme arthritis (LA). The primary drivers behind antibiotic-refractory arthritis remain incompletely understood. We performed a matched, cross-compartmental comparison of antibody profiles from blood and joint fluid of individuals with antibiotic-responsive (n = 11) or antibiotic-refractory LA (n = 31). While serum antibody profiles poorly discriminated responsive from refractory patients, a discrete profile of B.burgdorferi-specific antibodies in joint fluid discriminated antibiotic-responsive from refractory LA. Cross-compartmental comparison of antibody glycosylation, IgA1, and antibody-dependent complement deposition (ADCD) revealed more poorly coordinated humoral responses and increased ADCD in refractory disease. These data reveal B.burgdorferi-specific serological markers that may support early stratification and clinical management, and point to antibody-dependent complement activation as a key mechanism underlying persistent disease.
Collapse
Affiliation(s)
- Kathryn A. Bowman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Brigham and Women’s Hospital, Division of Infectious Diseases, Boston, MA 02115, USA
| | - Christine D. Wiggins
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Elizabeth DeRiso
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Steffan Paul
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Klemen Strle
- Tufts University School of Medicine Boston, Boston, MA, USA
| | - John A. Branda
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Allen C. Steere
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Moderna Therapeutics Inc., Cambridge, MA 02139, USA
| |
Collapse
|
40
|
Coelho AA, Carvalho RR, Muniz AL, Crispim AA, Meneses AM, Silva CWD, Paula DSD, Alves APNN, Sousa FB, Silva PGDB. CD20 + cells blockage by rituximab delays wound healing in oral traumatic ulcers in rats. Arch Oral Biol 2024; 157:105844. [PMID: 37950958 DOI: 10.1016/j.archoralbio.2023.105844] [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: 08/19/2023] [Revised: 10/18/2023] [Accepted: 11/01/2023] [Indexed: 11/13/2023]
Abstract
OBJECTIVE Wound healing of oral traumatic ulcers (OTU) is strongly associated with cytokines and inflammatory cells, and the reduction of anti-inflammatory cells, such as lymphocyte B, may interfere with OTU repair. We aimed to evaluate the role of CD20 + cells in the healing process of OTU in rats. DESIGN Wistar male rats were divided into four groups: a control group (treated with 0.1 mL/kg of saline) and three groups treated with anti-CD20 rituximab (RTX) at 2.5, 10, or 40 mg/kg 24 h before OTU production. The animals were weighed (day 0) and euthanized on days 3, 7, 14, and 21 after ulceration. With Blood cells (hematological analysis) and the traumatically induced ulcers were clinically measured. The mucosal samples were histologically (scores 0-4), histochemically (collagen assay (picrosirius)), histomorphometrically (cell counting), and immunohistochemically (CD20+, Tumor Necrosis Factor alpha(TNF-α), Interleukin(IL)- 1β, IL-6 and α-smooth-muscle-actin (α-SMA)) analyzed. ANOVA-1-2-way/Bonferroni, Kruskal-Wallis/Dunn, and correlation analyses were performed (GraphPad Prism 5.0, p < 0.05). RESULTS RTX leads to leukopenia, lymphocytopenia, and neutropenia (p < 0.001), and high doses reduced the OTU area (p = 0.001), impaired histologic scores (p < 0.05), and delayed polymorphonuclear (p < 0.001) and mononuclear (p < 0.001) cells, and total (p = 0.011), type-I (p = 0.008), and type-III (p = 0.021) collagen. CONCLUSION RTX treatment reduced CD20+ cells in OTU (p = 0.001), TNF-α (p = 0.006), and α-SMA (p = 0.022) immunostaining and delayed IL-6 reduction (p = 0.006), with no influence in IL-1β immunostaining. CD20 + cell blockage by RTX reduced cell migration, acute inflammation, and wound healing in OTU.
Collapse
Affiliation(s)
| | | | - Ana Luiza Muniz
- Department of Dentistry, Unichristus, Fortaleza, Ceara, Brazil
| | | | | | | | | | - Ana Paula Negreiros Nunes Alves
- Department of Dental Clinic, Division of Oral Pathology, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Fabrício Bitu Sousa
- Department of Dentistry, Unichristus, Fortaleza, Ceara, Brazil; Department of Dental Clinic, Division of Oral Pathology, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | | |
Collapse
|
41
|
Ilchovska D. Lyme Disease and Autoimmune Diseases. INFECTION AND AUTOIMMUNITY 2024:473-488. [DOI: 10.1016/b978-0-323-99130-8.00041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
42
|
Rouse JR, Danner R, Wahhab A, Pereckas M, McClune ME, Steere AC, Strle K, Jutras BL, Lochhead RB. Human leukocyte antigen HLA-DR-expressing fibroblast-like synoviocytes are inducible antigen presenting cells that present autoantigens in Lyme arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.568066. [PMID: 38045407 PMCID: PMC10690166 DOI: 10.1101/2023.11.21.568066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Background HLA-DR-expressing fibroblast-like synoviocytes (FLS) are a prominent cell type in synovial tissue in chronic inflammatory forms of arthritis. We recently showed that peptides from several extracellular matrix (ECM) proteins, including fibronectin-1 (FN1), contained immunogenic CD4+ T cell epitopes in patients with postinfectious Lyme arthritis (LA). However, the role of FLS in presentation of these T cell epitopes remains uncertain. Methods Primary LA FLS and primary murine FLS stimulated with interferon gamma (IFNγ), Borrelia burgdorferi, and/or B. burgdorferi peptidoglycan (PG) were assessed for properties associated with antigen presentation. HLA-DR-presented peptides from stimulated LA FLS were identified by immunopeptidomics analysis. OT-II T cells were cocultured with stimulated murine FLS in the presence of cognate ovalbumin antigen to determine the potential of FLS to act as inducible antigen presenting cells (APC). Results FLS expressed HLA-DR molecules within inflamed synovial tissue and tendons from patients with post-infectious LA patients in situ. MHC class II and costimulatory molecules were expressed by FLS following in vitro stimulation with IFNγ and B. burgdorferi and presented both foreign and self MHC-II peptides, including T cell epitopes derived from two Lyme autoantigens fibronectin-1 (FN1) and endothelial cell growth factor (ECGF). Stimulated murine FLS induced proliferation of naïve OT-II CD4+ T cells, particularly when FLS were stimulated with both IFNγ and PG. Conclusions MHC-II+ FLS are inducible APCs that can induce CD4+ T cell activation and can present Lyme autoantigens derived from ECM proteins, thereby amplifying tissue-localized autoimmune CD4+ T cell responses in LA.
Collapse
Affiliation(s)
- Joseph R Rouse
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Rebecca Danner
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amanda Wahhab
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michaela Pereckas
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mecaila E McClune
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, USA
- Center for Emerging, Zoonotic and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Allen C Steere
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Klemen Strle
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Brandon L Jutras
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, USA
- Center for Emerging, Zoonotic and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Robert B Lochhead
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Division of Rheumatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
43
|
Helble JD, Walsh MJ, McCarthy JE, Smith NP, Tirard AJ, Arnold BY, Villani AC, Hu LT. Single-cell RNA sequencing of murine ankle joints over time reveals distinct transcriptional changes following Borrelia burgdorferi infection. iScience 2023; 26:108217. [PMID: 37953958 PMCID: PMC10632114 DOI: 10.1016/j.isci.2023.108217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/06/2023] [Accepted: 10/11/2023] [Indexed: 11/14/2023] Open
Abstract
Lyme disease is caused by the bacterial pathogen Borrelia burgdorferi, which can be readily modeled in laboratory mice. In order to understand the cellular and transcriptional changes that occur during B. burgdorferi infection, we conducted single-cell RNA sequencing (scRNA-seq) of ankle joints of infected C57BL/6 mice over time. We found that macrophages/monocytes, T cells, synoviocytes and fibroblasts all showed significant differences in gene expression of both inflammatory and non-inflammatory genes that peaked early and returned to baseline before the typical resolution of arthritis. Predictions of cellular interactions showed that macrophages appear to communicate extensively between different clusters of macrophages as well as with fibroblasts and synoviocytes. Our data give unique insights into the interactions between B. burgdorferi and the murine immune system over time and allow for a better understanding of mechanisms by which the dysregulation of the immune response may lead to prolonged symptoms in some patients.
Collapse
Affiliation(s)
- Jennifer D. Helble
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Michael J. Walsh
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Julie E. McCarthy
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Neal P. Smith
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alice J. Tirard
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Benjamin Y. Arnold
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Linden T. Hu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
44
|
Clark IA, Vissel B. Autocrine positive feedback of tumor necrosis factor from activated microglia proposed to be of widespread relevance in chronic neurological disease. Pharmacol Res Perspect 2023; 11:e01136. [PMID: 37750203 PMCID: PMC10520644 DOI: 10.1002/prp2.1136] [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: 06/29/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/27/2023] Open
Abstract
Over a decade's experience of post-stroke rehabilitation by administering the specific anti-TNF biological, etanercept, by the novel perispinal route, is consistent with a wide range of chronically diminished neurological function having been caused by persistent excessive cerebral levels of TNF. We propose that this TNF persistence, and cerebral disease chronicity, largely arises from a positive autocrine feedback loop of this cytokine, allowing the persistence of microglial activation caused by the excess TNF that these cells produce. It appears that many of these observations have never been exploited to construct a broad understanding and treatment of certain chronic, yet reversible, neurological illnesses. We propose that this treatment allows these chronically activated microglia to revert to their normal quiescent state, rather than simply neutralizing the direct harmful effects of this cytokine after its release from microglia. Logically, this also applies to the chronic cerebral aspects of various other neurological conditions characterized by activated microglia. These include long COVID, Lyme disease, post-stroke syndromes, traumatic brain injury, chronic traumatic encephalopathy, post-chemotherapy, post-irradiation cerebral dysfunction, cerebral palsy, fetal alcohol syndrome, hepatic encephalopathy, the antinociceptive state of morphine tolerance, and neurogenic pain. In addition, certain psychiatric states, in isolation or as sequelae of infectious diseases such as Lyme disease and long COVID, are candidates for being understood through this approach and treated accordingly. Perispinal etanercept provides the prospect of being able to treat various chronic central nervous system illnesses, whether they are of infectious or non-infectious origin, through reversing excess TNF generation by microglia.
Collapse
Affiliation(s)
- Ian A. Clark
- Research School of Biology, Australian National UniversityCanberraAustralian Capital TerritoryAustralia
| | - Bryce Vissel
- St Vincent's Hospital Centre for Applied Medical ResearchSt Vincent's HospitalDarlinghurstAustralia
- UNSW Medicine & Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and HealthSchool of Clinical Medicine, UNSW SydneySydneyNew South WalesAustralia
| |
Collapse
|
45
|
Horowitz RI, Fallon J, Freeman PR. Comparison of the Efficacy of Longer versus Shorter Pulsed High Dose Dapsone Combination Therapy in the Treatment of Chronic Lyme Disease/Post Treatment Lyme Disease Syndrome with Bartonellosis and Associated Coinfections. Microorganisms 2023; 11:2301. [PMID: 37764145 PMCID: PMC10537894 DOI: 10.3390/microorganisms11092301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/27/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Twenty-five patients with relapsing and remitting Borreliosis, Babesiosis, and bartonellosis despite extended anti-infective therapy were prescribed double-dose dapsone combination therapy (DDDCT), followed by one or several courses of High Dose Dapsone Combination Therapy (HDDCT). A retrospective chart review of these 25 patients undergoing DDDCT therapy and HDDCT demonstrated that 100% improved their tick-borne symptoms, and patients completing 6-7 day pulses of HDDCT had superior levels of improvement versus 4-day pulses if Bartonella was present. At the completion of treatment, 7/23 (30.5%) who completed 8 weeks of DDDCT followed by a 5-7 day pulse of HDDCT remained in remission for 3-9 months, and 3/23 patients (13%) who recently finished treatment were 1 ½ months in full remission. In conclusion, DDDCT followed by 6-7 day pulses of HDDCT could represent a novel, effective anti-infective strategy in chronic Lyme disease/Post Treatment Lyme Disease Syndrome (PTLDS) and associated co-infections, including Bartonella, especially in individuals who have failed standard antibiotic protocols.
Collapse
Affiliation(s)
- Richard I. Horowitz
- Lyme and Tick-Borne Diseases Working Group, New York State Department of Health, Albany, NY 12224, USA
- Hudson Valley Healing Arts Center, Hyde Park, NY 12538, USA; (J.F.); (P.R.F.)
| | - John Fallon
- Hudson Valley Healing Arts Center, Hyde Park, NY 12538, USA; (J.F.); (P.R.F.)
| | - Phyllis R. Freeman
- Hudson Valley Healing Arts Center, Hyde Park, NY 12538, USA; (J.F.); (P.R.F.)
| |
Collapse
|
46
|
Barbour AG, Duong JV, Long AD. Lyme Disease Agent Reservoirs Peromyscus leucopus and P. maniculatus Have Natively Inactivated Genes for the High-Affinity Immunoglobulin Gamma Fc Receptor I (CD64). Pathogens 2023; 12:1056. [PMID: 37624016 PMCID: PMC10458454 DOI: 10.3390/pathogens12081056] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
The abundant and widely distributed deermice Peromyscus leucopus and P. maniculatus are important reservoirs for several different zoonotic agents in North America. For the pathogens they persistently harbor, these species are also examples of the phenomenon of infection tolerance. In the present study a prior observation of absent expression of the high-affinity Fc immunoglobulin gamma receptor I (FcγRI), or CD64, in P. leucopus was confirmed in an experimental infection with Borreliella burgdorferi, a Lyme disease agent. We demonstrate that the null phenotype is attributable to a long-standing inactivation of the Fcgr1 gene in both species by a deletion of the promoter and coding sequence for the signal peptide for FcγRI. The Fcgr1 pseudogene was also documented in the related species P. polionotus. Six other Peromyscus species, including P. californicus, have coding sequences for a full-length FcγRI, including a consensus signal peptide. An inference from reported phenotypes for null Fcgr1 mutations engineered in Mus musculus is that one consequence of pseudogenization of Fcgr1 is comparatively less inflammation during infection than in animals, including humans, with undisrupted, fully active genes.
Collapse
Affiliation(s)
- Alan G. Barbour
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California Irvine, Irvine, CA 92697, USA;
- Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Jonathan V. Duong
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California Irvine, Irvine, CA 92697, USA;
| | - Anthony D. Long
- Department of Ecology & Evolutionary Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA;
| |
Collapse
|
47
|
Gutierrez-Hoffmann M, Fan J, O’Meally RN, Cole RN, Florea L, Antonescu C, Talbot CC, Tiniakou E, Darrah E, Soloski MJ. The Interaction of Borrelia burgdorferi with Human Dendritic Cells: Functional Implications. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:612-625. [PMID: 37405694 PMCID: PMC10527078 DOI: 10.4049/jimmunol.2300235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023]
Abstract
Dendritic cells bridge the innate and adaptive immune responses by serving as sensors of infection and as the primary APCs responsible for the initiation of the T cell response against invading pathogens. The naive T cell activation requires the following three key signals to be delivered from dendritic cells: engagement of the TCR by peptide Ags bound to MHC molecules (signal 1), engagement of costimulatory molecules on both cell types (signal 2), and expression of polarizing cytokines (signal 3). Initial interactions between Borrelia burgdorferi, the causative agent of Lyme disease, and dendritic cells remain largely unexplored. To address this gap in knowledge, we cultured live B. burgdorferi with monocyte-derived dendritic cells (mo-DCs) from healthy donors to examine the bacterial immunopeptidome associated with HLA-DR. In parallel, we examined changes in the expression of key costimulatory and regulatory molecules as well as profiled the cytokines released by dendritic cells when exposed to live spirochetes. RNA-sequencing studies on B. burgdorferi-pulsed dendritic cells show a unique gene expression signature associated with B. burgdorferi stimulation that differs from stimulation with lipoteichoic acid, a TLR2 agonist. These studies revealed that exposure of mo-DCs to live B. burgdorferi drives the expression of both pro- and anti-inflammatory cytokines as well as immunoregulatory molecules (e.g., PD-L1, IDO1, Tim3). Collectively, these studies indicate that the interaction of live B. burgdorferi with mo-DCs promotes a unique mature DC phenotype that likely impacts the nature of the adaptive T cell response generated in human Lyme disease.
Collapse
Affiliation(s)
- Maria Gutierrez-Hoffmann
- Lyme Disease Research Center, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
- Division of Rheumatology, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
| | - Jinshui Fan
- Division of Rheumatology, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
| | - Robert N. O’Meally
- Mass Spectrometry and Proteomics Facility,
Department of Biological Chemistry, Johns Hopkins University School of Medicine,
Baltimore, MD 21205, USA
| | - Robert N. Cole
- Mass Spectrometry and Proteomics Facility,
Department of Biological Chemistry, Johns Hopkins University School of Medicine,
Baltimore, MD 21205, USA
| | - Liliana Florea
- Department of Genetic Medicine, Johns Hopkins
University, School of Medicine, Baltimore, MD 21205, USA
| | - Corina Antonescu
- Department of Genetic Medicine, Johns Hopkins
University, School of Medicine, Baltimore, MD 21205, USA
| | - C. Conover Talbot
- Institute for Basic Biomedical Sciences, Johns
Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Eleni Tiniakou
- Division of Rheumatology, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
| | - Erika Darrah
- Lyme Disease Research Center, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
- Division of Rheumatology, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
| | - Mark J. Soloski
- Lyme Disease Research Center, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
- Division of Rheumatology, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
| |
Collapse
|
48
|
Wang D, Zheng X, Chai L, Zhao J, Zhu J, Li Y, Yang P, Mao Q, Xia H. FAM76B regulates NF-κB-mediated inflammatory pathway by influencing the translocation of hnRNPA2B1. eLife 2023; 12:e85659. [PMID: 37643469 PMCID: PMC10446823 DOI: 10.7554/elife.85659] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
FAM76B has been reported to be a nuclear speckle-localized protein with unknown function. In this study, FAM76B was first demonstrated to inhibit the NF-κB-mediated inflammatory pathway by affecting the translocation of hnRNPA2B1 in vitro. We further showed that FAM76B suppressed inflammation in vivo using a traumatic brain injury (TBI) mouse model. Lastly, FAM76B was shown to interact with hnRNPA2B1 in human tissues taken from patients with acute, organizing, and chronic TBI, and with different neurodegenerative diseases. The results suggested that FAM76B mediated neuroinflammation via influencing the translocation of hnRNPA2B1 in vivo during TBI repair and neurodegenerative diseases. In summary, we for the first time demonstrated the role of FAM76B in regulating inflammation and further showed that FAM76B could regulate the NF-κB-mediated inflammatory pathway by affecting hnRNPA2B1 translocation, which provides new information for studying the mechanism of inflammation regulation.
Collapse
Affiliation(s)
- Dongyang Wang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal UniversityXi'anChina
- Translational Medicine Center, Northwest Women’s and Children’s HospitalXi'anChina
| | - Xiaojing Zheng
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal UniversityXi'anChina
| | - Lihong Chai
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal UniversityXi'anChina
| | - Junli Zhao
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal UniversityXi'anChina
| | - Jiuling Zhu
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal UniversityXi'anChina
| | - Yanqing Li
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal UniversityXi'anChina
| | - Peiyan Yang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal UniversityXi'anChina
| | - Qinwen Mao
- Department of Pathology, University of UtahSalt LakeUnited States
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal UniversityXi'anChina
| |
Collapse
|
49
|
Roberts J, Merchant E. Trauma-related Lyme arthritis. BMJ Case Rep 2023; 16:e255532. [PMID: 37407232 PMCID: PMC10335588 DOI: 10.1136/bcr-2023-255532] [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] [Indexed: 07/07/2023] Open
Abstract
A man in his 40s with no prior orthopaedic history presented to an infectious disease clinic with persistent left knee pain and swelling following a traumatic meniscal tear and ensuing prodromal period of fever and chills. Aspiration of the left knee joint revealed a white cell count of 21.0 ×109/L (83% neutrophils) with negative Gram stain and culture. However, Lyme PCR was positive and accompanied by serologies consistent with Lyme arthritis. He was treated with a standard course of antibiotic therapy with subsequent resolution of joint effusion and significant improvement in pain.This is to our knowledge the first report in the literature of Lyme arthritis seemingly provoked by traumatic knee injury. We propose disruption of normal joint anatomy and ensuing inflammation in response to acute injury incited and accelerated migration of previously latent Borrelia burgdorferi spirochetal infection into surrounding synovial tissue, leading to enhanced inflammatory activity and exacerbation of knee pain.
Collapse
Affiliation(s)
- John Roberts
- Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Elisabeth Merchant
- Tufts University School of Medicine, Boston, Massachusetts, USA
- Infectious Diseases, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
50
|
Sanes JT, Costello CE, Steere AC. Heightened Proinflammatory Glycosylation of Borrelia burgdorferi IgG Antibodies in Synovial Fluid in Patients With Antibiotic-Refractory Lyme Arthritis. Arthritis Rheumatol 2023; 75:1263-1274. [PMID: 36716113 PMCID: PMC10313735 DOI: 10.1002/art.42465] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Terminal glycans on the Fc portion of IgG antibodies are critical for antibody-triggered, proinflammatory or antiinflammatory responses. We undertook this study to compare glycan profiles of total IgG1 and Borrelia burgdorferi (Bb)-specific IgG1 antibodies in patients with oral antibiotic-responsive or antibiotic-refractory Lyme arthritis (LA). METHODS Following affinity-column processing, glycan profiles of IgG antibodies were determined in serum and synovial fluid (SF) samples of 21 LA patients using glycoblotting with hydrazide glycan enrichment and determination of glycan structure by matrix-assisted laser desorption ionization-time-of-flight mass spectrometry. Correlations between glycan profiles and treatment outcomes were analyzed. RESULTS Compared with patients with antibiotic-refractory LA, those with antibiotic-responsive LA had total and Bb-specific IgG1 antibody glycans with less intense inflammatory profiles, containing lower percentages of N-acetylglucosamine (GlcNAc) and bisecting GlcNAc and higher percentages of galactose and fucose. In contrast, patients with antibiotic-refractory LA prior to receiving IV antibiotic therapy had total IgG1 and Bb IgG1 antibodies with maximal, minimally opposed, proinflammatory glycan profiles, containing high percentages of GlcNAc and bisecting GlcNAc, intermediate percentages with galactose and fucose, and low percentages with N-acetylneuraminic acid (sialic acid). Patients with refractory LA who were first seen with synovitis after receiving IV antibiotic therapy still had Bb IgG1 antibodies with strongly inflammatory glycan profiles, but their inflammatory potential appeared to be waning. CONCLUSION Patients with oral antibiotic-responsive LA had Bb IgG1 antibodies with more balanced proinflammatory/antiinflammatory glycan profiles, whereas patients with antibiotic-refractory LA had Bb IgG1 antibodies with maximal, minimally opposed, proinflammatory glycan profiles. Among patients with antibiotic-refractory LA, antibodies with this unbalanced inflammatory glycan profile may have a role in sustaining maladaptive joint inflammation.
Collapse
Affiliation(s)
- Jurgen T Sanes
- Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Catherine E Costello
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, Massachusetts
| | - Allen C Steere
- Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|