Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Appl Environ Microbiol ; : e0082224, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38899883

ABSTRACT

Borrelia burgdorferi, a Lyme disease spirochete, causes a range of acute and chronic maladies in humans. However, a primary vertebrate reservoir in the United States, the white-footed deermouse Peromyscus leucopus, is reported not to have reduced fitness following infection. Although laboratory strains of Mus musculus mice have successfully been leveraged to model acute human Lyme disease, the ability of these rodents to model B. burgdorferi-P. leucopus interactions remains understudied. Here, we compared infection of P. leucopus with B. burgdorferi B31 with infection of the traditional B. burgdorferi murine models-C57BL/6J and C3H/HeN Mus musculus, which develop signs of inflammation akin to human disease. We find that B. burgdorferi was able to reach much higher burdens (10- to 30-times higher) in multiple M. musculus skin sites and that the overall dynamics of infection differed between the two rodent species. We also found that P. leucopus remained transmissive to larval Ixodes scapularis for a far shorter period than either M. musculus strain. In line with these observations, we found that P. leucopus does launch a modest but sustained inflammatory response against B. burgdorferi in the skin, which we hypothesize leads to reduced bacterial viability and rodent-to-tick transmission in these hosts. Similarly, we also observe evidence of inflammation in infected P. leucopus hearts. These observations provide new insight into reservoir species and the B. burgdorferi enzootic cycle.IMPORTANCEA Lyme disease-causing bacteria, Borrelia burgdorferi, must alternate between infecting a vertebrate host-usually rodents or birds-and ticks. In order to be successful in that endeavor, the bacteria must avoid being killed by the vertebrate host before it can infect a new larval tick. In this work, we examine how B. burgdorferi and one of its primary vertebrate reservoirs, Peromyscus leucopus, interact during an experimental infection. We find that B. burgdorferi appears to colonize its natural host less successfully than conventional laboratory mouse models, which aligns with a sustained seemingly anti-bacterial response by P. leucopus against the microbe. These data enhance our understanding of P. leucopus host-pathogen interactions and could potentially serve as a foundation to uncover ways to disrupt the spread of B. burgdorferi in nature.

2.
bioRxiv ; 2024 May 05.
Article in English | MEDLINE | ID: mdl-38746284

ABSTRACT

Ixodes scapularis ticks are an important vector for at least six tick-borne human pathogens, including the predominant North American Lyme disease spirochete Borrelia burgdorferi . The ability for these ticks to survive in nature is credited, in part, to their ability to feed on a variety of hosts without excessive activation of the proinflammatory branch of the vertebrate immune system. While the ability for nymphal ticks to feed on a variety of hosts has been well-documented, the host-parasite interactions between larval I. scapularis and different vertebrate hosts is relatively unexplored. Here we report on the changes in the vertebrate transcriptome present at the larval tick bite site using the natural I. scapularis host Peromyscus leucopus deermouse, a non-natural rodent host Mus musculus (BALB/c), and humans. We note substantially less evidence of activation of canonical proinflammatory pathways in P. leucopus compared to BALB/c mice and pronounced evidence of inflammation in humans. Pathway enrichment analyses revealed a particularly strong signature of interferon gamma, tumor necrosis factor, and interleukin 1 signaling at the BALB/c and human tick bite site. We also note that bite sites on BALB/c mice and humans, but not deermice, show activation of wound-healing pathways. These data provide molecular evidence of the coevolution between larval I. scapularis and P. leucopus as well as expand our overall understanding of I. scapularis feeding. Significance: Ixodes scapularis tick bites expose humans to numerous diseases in North America. While larval tick feeding enables pathogens to enter the tick population and eventually spread to humans, how larval ticks interact with mammals has been understudied compared to other tick stages. Here we examined the transcriptomic response of a natural I. scapularis rodent host ( Peromyscus leucopus ), a non-native I. scapularis rodent host ( Mus musculus ), and an incidental host (humans). We find that there are differences in how all three species respond to larval I. scapularis , with the natural host producing the smallest transcriptomic signature of a canonical proinflammatory immune response and the incidental human host producing the most robust signature of inflammation in response to the larval tick. These data expand our understanding of the pressures on ticks in the wild and inform our ability to model these interactions in laboratory settings.

3.
PLoS Pathog ; 19(12): e1011886, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38157387

ABSTRACT

Borrelia burgdorferi (Bb), the causative agent of Lyme disease, establishes a long-term infection and leads to disease manifestations that are the result of host immune responses to the pathogen. Inflammatory manifestations resolve spontaneously despite continued bacterial presence, suggesting inflammatory cells become less responsive over time. This is mimicked by in vitro repeated stimulations, resulting in tolerance, a phenotypic subset of innate immune memory. We performed comparative transcriptional analysis of macrophages in acute and memory states and identified sets of Tolerized, Hyper-Induced, Secondary-Induced and Hyper-Suppressed genes resulting from memory induction, revealing previously unexplored networks of genes affected by cellular re-programming. Tolerized gene families included inflammatory mediators and interferon related genes as would be predicted by the attenuation of inflammation over time. To better understand how cells mediate inflammatory hypo-responsiveness, we focused on genes that could mediate maintenance of suppression, such as Hyper-Induced genes which are up-regulated in memory states. These genes were notably enriched in stress pathways regulated by anti-inflammatory modulators. We examined one of the most highly expressed negative regulators of immune pathways during primary stimulation, Aconitate decarboxylase 1 (Acod1), and tested its effects during in vivo infection with Bb. As predicted by our in vitro model, we show its inflammation-suppressive downstream effects are sustained during in vivo long-term infection with Bb, with a specific role in Lyme carditis.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Humans , Inflammation , Lyme Disease/microbiology , Macrophages , Anti-Inflammatory Agents
4.
iScience ; 26(11): 108217, 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-37953958

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.

5.
mSystems ; 8(6): e0083523, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37855615

ABSTRACT

IMPORTANCE: Lyme disease is often treated using long courses of antibiotics, which can cause side effects for patients and risks the evolution of antimicrobial resistance. Narrow-spectrum antimicrobials would reduce these risks, but their development has been slow because the Lyme disease bacterium, Borrelia burgdorferi, is difficult to work with in the laboratory. To accelerate the drug discovery pipeline, we developed a computational model of B. burgdorferi's metabolism and used it to predict essential enzymatic reactions whose inhibition prevented growth in silico. These predictions were validated using small-molecule enzyme inhibitors, several of which were shown to have specific activity against B. burgdorferi. Although the specific compounds used are not suitable for clinical use, we aim to use them as lead compounds to develop optimized drugs targeting the pathways discovered here.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Humans , Lyme Disease/drug therapy , Anti-Bacterial Agents/pharmacology , Drug Discovery
6.
Microbiology (Reading) ; 169(1)2023 01.
Article in English | MEDLINE | ID: mdl-36748545

ABSTRACT

Borrelia burgdorferi is a pathogenic bacterium and the causative agent of Lyme disease. It is exposed to reactive oxygen species (ROS) in both the vertebrate and tick hosts. While some mechanisms by which B. burgdorferi ameliorates the effects of ROS exposure have been studied, there are likely other unknown mechanisms of ROS neutralization that contribute to virulence. Here, we follow up on a three gene cluster of unknown function, bb_0554, bb_0555, and bb_0556, that our prior unbiased transposon insertional sequencing studies implicated in both ROS survival and survival in Ixodes scapularis. We confirmed these findings through genetic knockout and provide evidence that these genes are co-transcribed as an operon to produce a xanthine dehydrogenase. In agreement with these results, we found that B. burgdorferi exposure to either uric acid (a product of xanthine dehydrogenase) or allopurinol (an inhibitor of xanthine dehydrogenase) could modulate sensitivity to ROS in a bb_0554-bb_0556 dependent manner. Together, this study identifies a previously uncharacterized three gene operon in B. burgdorferi as encoding a putative xanthine dehydrogenase critical for virulence. We propose renaming this locus xdhACB.


Subject(s)
Borrelia burgdorferi , Ixodes , Lyme Disease , Animals , Mice , Borrelia burgdorferi/genetics , Xanthine Dehydrogenase/genetics , Reactive Oxygen Species , Lyme Disease/microbiology , Ixodes/microbiology
7.
PLoS Pathog ; 18(10): e1010903, 2022 10.
Article in English | MEDLINE | ID: mdl-36265003

ABSTRACT

The Lyme disease bacterial pathogen, Borrelia burgdorferi, establishes a long-term infection inside its mammalian hosts. Despite the continued presence of the bacteria in animal models of disease, inflammation is transitory and resolves spontaneously. T cells with limited effector functions and the inability to become activated by antigen, termed exhausted T cells, are present in many long-term infections. These exhausted T cells mediate a balance between pathogen clearance and preventing tissue damage resulting from excess inflammation. Exhausted T cells express a variety of immunoinhibitory molecules, including the molecule PD-1. Following B. burgdorferi infection, we found that PD-1 and its ligand PD-L1 are significantly upregulated on CD4+ T cells and antigen presenting cell subsets, respectively. Using mice deficient in PD-1, we found that the PD-1/PD-L1 pathway did not impact bacterial clearance but did impact T cell expansion and accumulation in the ankle joint and popliteal lymph nodes without affecting B cell populations or antibody production, suggesting that the PD-1/PD-L1 pathway may play a role in shaping the T cell populations present in affected tissues.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Mice , Animals , Programmed Cell Death 1 Receptor , B7-H1 Antigen , Lyme Disease/microbiology , CD4-Positive T-Lymphocytes , Inflammation , Mammals
8.
Traffic ; 23(12): 558-567, 2022 12.
Article in English | MEDLINE | ID: mdl-36224049

ABSTRACT

Intracellular compartmentalization of ligands, receptors and signaling molecules has been recognized as an important regulator of inflammation. The toll-like receptor (TLR) 2 pathway utilizes the trafficking molecule adaptor protein 3 (AP-3) to activate interleukin (IL)-6 signaling from within phagosomal compartments. To better understand the vesicular pathways that may contribute to intracellular signaling and cooperate with AP-3, we performed a vesicular siRNA screen. We identified Rab8 and Rab11 GTPases as important in IL-6 induction upon stimulation with the TLR2 ligand Pam3 CSK4 or the pathogen, Borrelia burgdorferi (Bb), the causative agent of Lyme disease. These Rabs were recruited to late and lysosomal stage phagosomes and co-transported with TLR2 signaling adaptors and effectors, such as MyD88, TRAM and TAK1, in an AP-3-dependent manner. Our data support a model where AP-3 mediates the recruitment of recycling and secretory vesicles and the assembly of signaling complexes at the phagosome.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Adaptor Proteins, Signal Transducing/metabolism , Borrelia burgdorferi/metabolism , Ligands , Lyme Disease/genetics , Lyme Disease/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phagosomes/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , rab GTP-Binding Proteins , Animals , Mice
9.
J Clin Invest ; 132(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35289310

ABSTRACT

A close association with its vertebrate and tick hosts allows Borrelia burgdorferi, the bacterium responsible for Lyme disease, to eliminate many metabolic pathways and instead scavenge key nutrients from the host. A lipid-defined culture medium was developed to demonstrate that exogenous lipids are an essential nutrient of B. burgdorferi, which can accumulate intact phospholipids from its environment to support growth. Antibody responses to host phospholipids were studied in mice and humans using an antiphospholipid ELISA. Several of these environmentally acquired phospholipids including phosphatidylserine and phosphatidic acid, as well as borrelial phosphatidylcholine, are the targets of antibodies that arose early in infection in the mouse model. Patients with acute infections demonstrated antibody responses to the same lipids. The elevation of antiphospholipid antibodies predicted early infection with better sensitivity than did the standardized 2-tier tests currently used in diagnosis. Sera obtained from patients with Lyme disease before and after antibiotic therapy showed declining antiphospholipid titers after treatment. Further study will be required to determine whether these antibodies have utility in early diagnosis of Lyme disease, tracking of the response to therapy, and diagnosis of reinfection, areas in which current standardized tests are inadequate.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Animals , Antibodies, Antiphospholipid/metabolism , Antibodies, Bacterial , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Phospholipids/metabolism
10.
J Infect Dis ; 225(6): 1113, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34623448
11.
Cell ; 184(21): 5405-5418.e16, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34619078

ABSTRACT

Lyme disease is on the rise. Caused by a spirochete Borreliella burgdorferi, it affects an estimated 500,000 people in the United States alone. The antibiotics currently used to treat Lyme disease are broad spectrum, damage the microbiome, and select for resistance in non-target bacteria. We therefore sought to identify a compound acting selectively against B. burgdorferi. A screen of soil micro-organisms revealed a compound highly selective against spirochetes, including B. burgdorferi. Unexpectedly, this compound was determined to be hygromycin A, a known antimicrobial produced by Streptomyces hygroscopicus. Hygromycin A targets the ribosomes and is taken up by B. burgdorferi, explaining its selectivity. Hygromycin A cleared the B. burgdorferi infection in mice, including animals that ingested the compound in a bait, and was less disruptive to the fecal microbiome than clinically relevant antibiotics. This selective antibiotic holds the promise of providing a better therapeutic for Lyme disease and eradicating it in the environment.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Lyme Disease/drug therapy , Animals , Borrelia burgdorferi/drug effects , Calibration , Cinnamates/chemistry , Cinnamates/pharmacology , Cinnamates/therapeutic use , Drug Evaluation, Preclinical , Feces/microbiology , Female , HEK293 Cells , Hep G2 Cells , Humans , Hygromycin B/analogs & derivatives , Hygromycin B/chemistry , Hygromycin B/pharmacology , Hygromycin B/therapeutic use , Lyme Disease/microbiology , Mice , Microbial Sensitivity Tests , Microbiota/drug effects
12.
Sci Rep ; 11(1): 12384, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34117323

ABSTRACT

Inadequate sensitivity has been the primary limitation for implementing high-throughput sequencing for studies of tick-borne agents. Here we describe the development of TBDCapSeq, a sequencing assay that uses hybridization capture probes that cover the complete genomes of the eleven most common tick-borne agents found in the United States. The probes are used for solution-based capture and enrichment of pathogen nucleic acid followed by high-throughput sequencing. We evaluated the performance of TBDCapSeq to surveil samples that included human whole blood, mouse tissues, and field-collected ticks. For Borrelia burgdorferi and Babesia microti, the sensitivity of TBDCapSeq was comparable and occasionally exceeded the performance of agent-specific quantitative PCR and resulted in 25 to > 10,000-fold increase in pathogen reads when compared to standard unbiased sequencing. TBDCapSeq also enabled genome analyses directly within vertebrate and tick hosts. The implementation of TBDCapSeq could have major impact in studies of tick-borne pathogens by improving detection and facilitating genomic research that was previously unachievable with standard sequencing approaches.


Subject(s)
Babesia microti/genetics , Babesiosis/microbiology , Borrelia burgdorferi/genetics , Genotyping Techniques/methods , Lyme Disease/microbiology , Molecular Diagnostic Techniques/methods , Sequence Analysis, DNA/methods , Animals , Babesia microti/pathogenicity , Babesiosis/diagnosis , Borrelia burgdorferi/pathogenicity , Genome, Bacterial , Genotyping Techniques/standards , Humans , Lyme Disease/diagnosis , Mice , Molecular Diagnostic Techniques/standards , Sensitivity and Specificity , Sequence Analysis, DNA/standards , Ticks/microbiology
13.
J Clin Invest ; 131(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-33914704

ABSTRACT

Disrupting transmission of Borrelia burgdorferi sensu lato complex (B. burgdorferi) from infected ticks to humans is one strategy to prevent the significant morbidity from Lyme disease. We have previously shown that an anti-OspA human mAb, 2217, prevents transmission of B. burgdorferi from infected ticks in animal models. Maintenance of a protective plasma concentration of a human mAb for tick season presents a significant challenge for a preexposure prophylaxis strategy. Here, we describe the optimization of mAb 2217 by amino acid substitutions (2217LS: M428L and N434S) in the Fc domain. The LS mutation led to a 2-fold increase in half-life in cynomolgus monkeys. In a rhesus macaque model, 2217LS protected animals from tick transmission of spirochetes at a dose of 3 mg/kg. Crystallographic analysis of Fab in complex with OspA revealed that 2217 bound an epitope that was highly conserved among the B. burgdorferi, B. garinii, and B. afzelii species. Unlike most vaccines that may require boosters to achieve protection, our work supports the development of 2217LS as an effective preexposure prophylaxis in Lyme-endemic regions, with a single dose at the beginning of tick season offering immediate protection that remains for the duration of exposure risk.


Subject(s)
Antibodies, Bacterial , Antibodies, Monoclonal/pharmacology , Borrelia burgdorferi , Lyme Disease , Amino Acid Substitution , Animals , Antibodies, Bacterial/genetics , Antibodies, Bacterial/immunology , Antibodies, Bacterial/pharmacology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antigens, Surface/genetics , Antigens, Surface/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Borrelia burgdorferi/genetics , Borrelia burgdorferi/immunology , Disease Models, Animal , Humans , Lipoproteins/genetics , Lipoproteins/immunology , Lyme Disease/drug therapy , Lyme Disease/genetics , Lyme Disease/immunology , Lyme Disease/transmission , Macaca fascicularis , Macaca mulatta , Male , Mice , Mice, Transgenic , Mutation, Missense , Ticks/immunology , Ticks/microbiology
14.
Parasite Immunol ; 43(5): e12816, 2021 05.
Article in English | MEDLINE | ID: mdl-33368329

ABSTRACT

The bacterial pathogen Borrelia burgdorferi is the causative agent of Lyme disease and is transmitted to humans through an Ixodes tick vector. B. burgdorferi is able to survive in both mammalian and tick hosts through careful modulation of its gene expression. This allows B. burgdorferi to adapt to the environmental and nutritional changes that occur when it is transmitted between the two hosts. Distinct interactions between the spirochete and its host occur at every step of the enzootic cycle and dictate the ability of the spirochete to survive until the next stage of the cycle. Studying the interface between B. burgdorferi, the Ixodes tick vector and the natural mammalian reservoirs has been made significantly more feasible through the complete genome sequences of the organisms and the advent of high throughput screening technologies. Ultimately, a thorough investigation of the interplay between the two domains (and two phyla within one domain) is necessary in order to completely understand how the pathogen is transmitted.


Subject(s)
Arachnid Vectors/microbiology , Borrelia burgdorferi/physiology , Host Microbial Interactions/physiology , Ixodes/microbiology , Lyme Disease/microbiology , Mammals/microbiology , Animals , Arachnid Vectors/immunology , Borrelia burgdorferi/genetics , Gene Expression , Humans , Ixodes/immunology , Lyme Disease/epidemiology , Lyme Disease/transmission , Mammals/blood , Mammals/parasitology , Microbiota , Nymph/microbiology , Salivary Glands/microbiology
15.
Curr Issues Mol Biol ; 42: 473-518, 2021.
Article in English | MEDLINE | ID: mdl-33353871

ABSTRACT

Lyme disease Borrelia are obligately parasitic, tick- transmitted, invasive, persistent bacterial pathogens that cause disease in humans and non-reservoir vertebrates primarily through the induction of inflammation. During transmission from the infected tick, the bacteria undergo significant changes in gene expression, resulting in adaptation to the mammalian environment. The organisms multiply and spread locally and induce inflammatory responses that, in humans, result in clinical signs and symptoms. Borrelia virulence involves a multiplicity of mechanisms for dissemination and colonization of multiple tissues and evasion of host immune responses. Most of the tissue damage, which is seen in non-reservoir hosts, appears to result from host inflammatory reactions, despite the low numbers of bacteria in affected sites. This host response to the Lyme disease Borrelia can cause neurologic, cardiovascular, arthritic, and dermatologic manifestations during the disseminated and persistent stages of infection. The mechanisms by which a paucity of organisms (in comparison to many other infectious diseases) can cause varied and in some cases profound inflammation and symptoms remains mysterious but are the subjects of diverse ongoing investigations. In this review, we provide an overview of virulence mechanisms and determinants for which roles have been demonstrated in vivo, primarily in mouse models of infection.


Subject(s)
Borrelia , Disease Susceptibility , Lyme Disease/microbiology , Animals , Arthropod Vectors/microbiology , Borrelia/genetics , Disease Models, Animal , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions , Humans , Lyme Disease/transmission , Ticks/microbiology , Virulence , Virulence Factors/genetics
16.
J Infect Dis ; 224(2): 345-350, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33216133

ABSTRACT

Unrecognized immunodeficiency has been proposed as a possible cause of failure of antibiotics to resolve symptoms of Lyme disease. Here, we examined the efficacy of doxycycline in different immunodeficient mice to identify defects that impair antibiotic treatment outcomes. We found that doxycycline had significantly lower efficacy in the absence of adaptive immunity, specifically B cells. This effect was most pronounced in immunodeficient C3H mice compared with C57BL/6 mice, suggesting a role for genetic background beyond immunodeficiency. Addition of a single dose of ceftriaxone to doxycycline treatment effectively cleared infection in C3H mice with severe combined immunodeficiency.


Subject(s)
Anti-Bacterial Agents , Genetic Background , Immunologic Deficiency Syndromes , Lyme Disease , Animals , Anti-Bacterial Agents/therapeutic use , Borrelia burgdorferi , Doxycycline , Immunologic Deficiency Syndromes/genetics , Lyme Disease/drug therapy , Lyme Disease/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
18.
J Immunol ; 205(12): 3383-3389, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33168577

ABSTRACT

Borrelia burgdorferi, the causative agent of Lyme disease, is transmitted by the bite of an infected tick. Once inoculated into the host dermis, it disseminates to various organs including distant skin sites, the heart, the joint and the nervous system. Most humans will develop an early skin manifestation called erythema migrans at the tick bite site. This can be followed by symptoms such as carditis, neuritis, meningitis, or arthritis if not treated. A specific mouse strain, C3H/HeN develops arthritis with B. burgdorferi infection whereas another strain, C57BL/6, develops minimal to no arthritis. Neither strain of mice show any skin signs of rash or inflammation. Factors that determine the presence of skin inflammation and the joint arthritis susceptibility in the host are only partially characterized. We show in this study that murine fibroblast-like synoviocytes display trained immunity, a program in some cells that results in increased inflammatory responses if the cell has previously come in contact with a stimulus, and that trained immunity in fibroblast-like synoviocytes tested ex vivo correlates with Lyme arthritis susceptibility. Conversely, skin fibroblasts do not exhibit trained immunity, which correlates with the absence of skin symptoms in these mice. Moreover, we demonstrate that the trained phenotype in FLS is affected by the cell environment, which depends on the host genetic background. Future studies expanding this initial report of the role of trained immunity on symptoms of B. burgdorferi infection may provide insight into the pathogenesis of disease in murine models.


Subject(s)
Arthritis/immunology , Borrelia burgdorferi/immunology , Immunity, Innate , Immunologic Memory , Lyme Disease/immunology , Synoviocytes/immunology , Animals , Arthritis/genetics , Arthritis/pathology , Female , Inflammation/immunology , Inflammation/pathology , Lyme Disease/genetics , Lyme Disease/pathology , Mice , Mice, Knockout , Synoviocytes/pathology
19.
PLoS Pathog ; 16(5): e1008423, 2020 05.
Article in English | MEDLINE | ID: mdl-32365143

ABSTRACT

Post-transcriptional regulation via small regulatory RNAs (sRNAs) has been implicated in diverse regulatory processes in bacteria, including virulence. One class of sRNAs, termed trans-acting sRNAs, can affect the stability and/or the translational efficiency of regulated transcripts. In this study, we utilized a collaborative approach that employed data from infection with the Borrelia burgdorferi Tn library, coupled with Tn-seq, together with borrelial sRNA and total RNA transcriptomes, to identify an intergenic trans-acting sRNA, which we designate here as ittA for infectivity-associated and tissue-tropic sRNA locus A. The genetic inactivation of ittA resulted in a significant attenuation in infectivity, with decreased spirochetal load in ear, heart, skin and joint tissues. In addition, the ittA mutant did not disseminate to peripheral skin sites or heart tissue, suggesting a role for ittA in regulating a tissue-tropic response. RNA-Seq analysis determined that 19 transcripts were differentially expressed in the ittA mutant relative to its genetic parent, including vraA, bba66, ospD and oms28 (bba74). Subsequent proteomic analyses also showed a significant decrease of OspD and Oms28 (BBA74) proteins. To our knowledge this is the first documented intergenic sRNA that alters the infectivity potential of B. burgdorferi.


Subject(s)
Borrelia burgdorferi/genetics , RNA, Small Untranslated/metabolism , Tropism/genetics , Borrelia burgdorferi/metabolism , Borrelia burgdorferi/pathogenicity , Gene Expression Regulation, Bacterial/genetics , Gene Library , Genome, Bacterial , Lyme Disease/microbiology , Proteomics , RNA, Bacterial/genetics , RNA, Small Untranslated/genetics , Transcriptome/genetics , Virulence
20.
NPJ Vaccines ; 5(1): 33, 2020.
Article in English | MEDLINE | ID: mdl-32377398

ABSTRACT

A growing global health concern, Lyme disease has become the most common tick-borne disease in the United States and Europe. Caused by the bacterial spirochete Borrelia burgdorferi sensu lato (sl), this disease can be debilitating if not treated promptly. Because diagnosis is challenging, prevention remains a priority; however, a previously licensed vaccine is no longer available to the public. Here, we designed a six component vaccine that elicits antibody (Ab) responses against all Borrelia strains that commonly cause Lyme disease in humans. The outer surface protein A (OspA) of Borrelia was fused to a bacterial ferritin to generate self-assembling nanoparticles. OspA-ferritin nanoparticles elicited durable high titer Ab responses to the seven major serotypes in mice and non-human primates at titers higher than a previously licensed vaccine. This response was durable in rhesus macaques for more than 6 months. Vaccination with adjuvanted OspA-ferritin nanoparticles stimulated protective immunity from both B. burgdorferi and B. afzelii infection in a tick-fed murine challenge model. This multivalent Lyme vaccine offers the potential to limit the spread of Lyme disease.

SELECTION OF CITATIONS
SEARCH DETAIL
...