Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
Clin Immunol ; 246: 109180, 2023 01.
Article in English | MEDLINE | ID: mdl-36396013

ABSTRACT

Borrelia burgdorferi (Bb) infection causes Lyme disease, for which there is need for more effective therapies. Here, we sequenced the antibody repertoire of plasmablasts in Bb-infected humans. We expressed recombinant monoclonal antibodies (mAbs) representing the identified plasmablast clonal families, and identified their binding specificities. Our recombinant anti-Bb mAbs exhibit a range of activity in mediating macrophage phagocytosis of Bb. To determine if we could increase the macrophage phagocytosis-promoting activity of our anti-Bb mAbs, we generated a TLR9-agonist CpG-oligo-conjugated anti-BmpA mAb. We demonstrated that our CpG-conjugated anti-BmpA mAb exhibited increased peak Bb phagocytosis at 12-24 h, and sustained macrophage phagocytosis over 60+ hrs. Further, our CpG-conjugated anti-BmpA mAb induced macrophages to exhibit a sustained activation morphology. Our findings demonstrate the potential for TLR9-agonist CpG-oligo conjugates to enhance mAb-mediated clearance of Bb, and this approach might also enhance the activity of other anti-microbial mAbs.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Humans , Borrelia burgdorferi/metabolism , Toll-Like Receptor 9/metabolism , Macrophages , Lyme Disease/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/metabolism
2.
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
3.
Front Cell Infect Microbiol ; 12: 892220, 2022.
Article in English | MEDLINE | ID: mdl-35586252

ABSTRACT

The bacterial agent of Lyme disease, Borrelia burgdorferi, relies on an intricate gene regulatory network to transit between the disparate Ixodes tick vector and mammalian host environments. We recently reported that a B. burgdorferi mutant lacking a transcriptionally active intergenic region of lp17 displayed attenuated murine tissue colonization and pathogenesis due to altered expression of multiple antigens. In this study, a more detailed characterization of the putative regulatory factor encoded by the intergenic region was pursued. In cis complemented strains featuring mutations aimed at eliminating potential protein translation were capable of full tissue colonization, suggesting that the functional product encoded by the intergenic region is not a protein as previously predicted. In trans complementation of the intergenic region resulted in elevated transcription of the sequence compared to wild type and was found to completely abolish infectivity in both immunocompetent "and immunodeficient mice. Quantitative analysis of transcription of the intergenic region by wild-type B. burgdorferi showed it to be highly induced during murine infection relative to in vitro culture. Lastly, targeted deletion of this intergenic region resulted in significant changes to the transcriptome, including genes with potential roles in transmission and host adaptation. The findings reported herein strongly suggest that this segment of lp17 serves a potentially critical role in the regulation of genes required for adaptation and persistence of the pathogen in a mammalian host.


Subject(s)
Borrelia burgdorferi , Ixodes , Lyme Disease , Animals , Arthropod Proteins/genetics , Arthropod Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Borrelia burgdorferi/genetics , Borrelia burgdorferi/metabolism , DNA, Intergenic/genetics , Host Adaptation , Ixodes/microbiology , Lyme Disease/genetics , Lyme Disease/metabolism , Lyme Disease/microbiology , Mice , Oligopeptides/genetics , Oligopeptides/metabolism
4.
Infect Immun ; 90(5): e0005922, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35416705

ABSTRACT

The Borrelia burgdorferi BB0323 protein undergoes a complex yet poorly defined proteolytic maturation event that generates N-terminal and C-terminal proteins with essential functions in cell growth and infection. Here, we report that a borrelial protease, B. burgdorferi high temperature requirement A protease (BbHtrA), cleaves BB0323 between asparagine (N) and leucine (L) at positions 236 and 237, while the replacement of these residues with alanine in the mutant protein prevents its cleavage, despite preserving its normal secondary structure. The N-terminal BB0323 protein binds BbHtrA, but its cleavage site mutant displays deficiency in such interaction. An isogenic borrelial mutant with NL-to-AA substitution in BB0323 (referred to as Bbbb0323NL) maintains normal growth yet is impaired for infection of mice or transmission from infected ticks. Notably, the BB0323 protein is still processed in Bbbb0323NL, albeit with lower levels of mature N-terminal BB0323 protein and multiple aberrantly processed polypeptides, which could result from nonspecific cleavages at other asparagine and leucine residues in the protein. The lack of infectivity of Bbbb0323NL is likely due to the impaired abundance or stoichiometry of a protein complex involving BB0238, another spirochete protein. Together, these studies highlight that a precise proteolytic event and a particular protein-protein interaction, involving multiple borrelial virulence determinants, are mutually inclusive and interconnected, playing essential roles in the infectivity of Lyme disease pathogens.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Animals , Asparagine/metabolism , Bacterial Proteins/metabolism , Leucine/metabolism , Lyme Disease/metabolism , Mice , Peptide Hydrolases/metabolism , Proteolysis , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
5.
Sci Rep ; 12(1): 688, 2022 01 13.
Article in English | MEDLINE | ID: mdl-35027599

ABSTRACT

Even after appropriate treatment, a proportion of Lyme disease patients suffer from a constellation of symptoms, collectively called Post-Treatment Lyme Disease Syndrome (PTLDS). Brain PET scan of patients with PTLDS have demonstrated likely glial activation indicating persistent neuroinflammatory processes. It is possible that unresolved bacterial remnants can continue to cause neuroinflammation. In previous studies, we have shown that non-viable Borrelia burgdorferi can induce neuroinflammation and apoptosis in an oligodendrocyte cell line. In this follow-up study, we analyze the effect of sonicated remnants of B. burgdorferi on primary rhesus frontal cortex (FC) and dorsal root ganglion (DRG) explants. Five FC and three DRG tissue fragments from rhesus macaques were exposed to sonicated B. burgdorferi and analyzed for 26 inflammatory mediators. Live bacteria and medium alone served as positive and negative control, respectively. Tissues were also analyzed for cell types mediating inflammation and overall apoptotic changes. Non-viable B. burgdorferi induced significant levels of several inflammatory mediators in both FC and DRG, similar to live bacteria. However, the levels induced by non-viable B. burgdorferi was often (several fold) higher than those induced by live ones, especially for IL-6, CXCL8 and CCL2. This effect was also more profound in the FC than in the DRG. Although the levels often differed, both live and dead fragments induced the same mediators, with significant overlap between FC and DRG. In the FC, immunohistochemical staining for several inflammatory mediators showed the presence of multiple mediators in astrocytes, followed by microglia and oligodendrocytes, in response to bacterial remnants. Staining was also seen in endothelial cells. In the DRG, chemokine/cytokine staining was predominantly seen in S100 positive (glial) cells. B. burgdorferi remnants also induced significant levels of apoptosis in both the FC and DRG. Apoptosis was confined to S100 + cells in the DRG while distinct neuronal apoptosis was also detected in most FC tissues in response to sonicated bacteria. Non-viable B. burgdorferi can continue to be neuropathogenic to both CNS and PNS tissues with effects likely more profound in the former. Persistence of remnant-induced neuroinflammatory processes can lead to long term health consequences.


Subject(s)
Borrelia burgdorferi/pathogenicity , Frontal Lobe/metabolism , Frontal Lobe/pathology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/microbiology , Animals , Apoptosis , Chemokine CCL2/metabolism , Female , In Vitro Techniques , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Lyme Disease/metabolism , Lyme Disease/pathology , Macaca mulatta , Male
6.
Sci Transl Med ; 13(620): eabj9827, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34788080

ABSTRACT

Ixodes scapularis ticks transmit many pathogens that cause human disease, including Borrelia burgdorferi. Acquired resistance to I. scapularis due to repeated tick exposure has the potential to prevent tick-borne infectious diseases, and salivary proteins have been postulated to contribute to this process. We examined the ability of lipid nanoparticle­containing nucleoside-modified mRNAs encoding 19 I. scapularis salivary proteins (19ISP) to enhance the recognition of a tick bite and diminish I. scapularis engorgement on a host and thereby prevent B. burgdorferi infection. Guinea pigs were immunized with a 19ISP mRNA vaccine and subsequently challenged with I. scapularis. Animals administered 19ISP developed erythema at the bite site shortly after ticks began to attach, and these ticks fed poorly, marked by early detachment and decreased engorgement weights. 19ISP immunization also impeded B. burgdorferi transmission in the guinea pigs. The effective induction of local redness early after I. scapularis attachment and the inability of the ticks to take a normal blood meal suggest that 19ISP may be used either alone or in conjunction with traditional pathogen-based vaccines for the prevention of Lyme disease, and potentially other tick-borne infections.


Subject(s)
Ixodes , Lyme Disease , Animals , Guinea Pigs , Liposomes , Lyme Disease/metabolism , Lyme Disease/prevention & control , Nanoparticles , RNA, Messenger , Vaccination , Vaccines, Synthetic , mRNA Vaccines
7.
Elife ; 102021 11 16.
Article in English | MEDLINE | ID: mdl-34783654

ABSTRACT

Adiponectin-mediated pathways contribute to mammalian homeostasis; however, little is known about adiponectin and adiponectin receptor signaling in arthropods. In this study, we demonstrate that Ixodes scapularis ticks have an adiponectin receptor-like protein (ISARL) but lack adiponectin, suggesting activation by alternative pathways. ISARL expression is significantly upregulated in the tick gut after Borrelia burgdorferi infection, suggesting that ISARL signaling may be co-opted by the Lyme disease agent. Consistent with this, RNA interference (RNAi)-mediated silencing of ISARL significantly reduced the B. burgdorferi burden in the tick. RNA-seq-based transcriptomics and RNAi assays demonstrate that ISARL-mediated phospholipid metabolism by phosphatidylserine synthase I is associated with B. burgdorferi survival. Furthermore, the tick complement C1q-like protein 3 interacts with ISARL, and B. burgdorferi facilitates this process. This study identifies a new tick metabolic pathway that is connected to the life cycle of the Lyme disease spirochete.


Many countries around the world are seeing an increase in the number of patients diagnosed with Lyme disease, with often serious joint, heart, and neurologic complications. This illness is caused by species of 'spirochete' bacteria that live and multiply inside black-legged ticks, and get injected into mammals upon a bite. Ticks are not simply 'syringes' however, and a complex relationship is established between spirochetes and their host. This is particularly true since Lyme disease-causing bacteria such as Borrelia burgdorferi rely on ticks to obtain energy and nutrients. Tang, Cao et al. delved into these complex interactions by focusing on the molecular cascades (or pathways) involving adiponectin, a hormone essential for regulating sugar levels and processing fats. Analyses of gene and protein databases highlighted that ticks carry a receptor-like protein for adiponectin but not the hormone itself, suggesting that an alternative pathway is at play. This may involve B. burgdorferi, which gets its fats and sugars from its host. And indeed, experiments showed that ticks produced more of the adiponectin receptor-like protein when they carried B. burgdorferi; conversely, silencing the receptor reduced the number of surviving spirochetes inside the tick. Further exploration showed that the receptor mediates molecular cascades that help to process fat molecules; these are associated with spirochete survival. In addition, the receptor-like protein was activated by C1QL3, a 'complement 1q domain-contained' molecule which might be part of the tick energy-making or immune systems. Larger quantities of C1QL3 were found in ticks upon B. burgdorferi infection, suggesting that the spirochete facilitates an interaction that boosts activity of the adiponectin receptor-like protein. Overall, the work by Tang and Cao et al. revealed a new pathway which B. burgdorferi takes advantage of to infect their host and multiply. Targeting this molecular cascade could help to interfere with the life cycle of the spirochete, as well as fight Lyme disease and other insect-borne conditions.


Subject(s)
Borrelia burgdorferi/metabolism , Ixodes/metabolism , Ixodes/microbiology , Receptors, Adiponectin/metabolism , Animals , Arthropod Proteins/metabolism , Arthropod Vectors/metabolism , Arthropod Vectors/microbiology , Lyme Disease/metabolism , Lyme Disease/microbiology , Phospholipids/metabolism , RNA Interference , Receptors, Adiponectin/genetics , Transcriptome
8.
PLoS Pathog ; 17(8): e1009869, 2021 08.
Article in English | MEDLINE | ID: mdl-34415955

ABSTRACT

The Lyme disease spirochete Borrelia burgdorferi relies on uptake of essential nutrients from its host environments for survival and infection. Therefore, nutrient acquisition mechanisms constitute key virulence properties of the pathogen, yet these mechanisms remain largely unknown. In vivo expression technology applied to B. burgdorferi (BbIVET) during mammalian infection identified gene bb0562, which encodes a hypothetical protein comprised of a conserved domain of unknown function, DUF3996. DUF3996 is also found across adjacent encoded hypothetical proteins BB0563 and BB0564, suggesting the possibility that the three proteins could be functionally related. Deletion of bb0562, bb0563 and bb0564 individually and together demonstrated that bb0562 alone was important for optimal disseminated infection in immunocompetent and immunocompromised mice by needle inoculation and tick bite transmission. Moreover, bb0562 promoted spirochete survival during the blood dissemination phase of infection. Gene bb0562 was also found to be important for spirochete growth in low serum media and the growth defect of Δbb0562 B. burgdorferi was rescued with the addition of various long chain fatty acids, particularly oleic acid. In mammals, fatty acids are primarily stored in fat droplets in the form of triglycerides. Strikingly, addition of glyceryl trioleate, the triglyceride form of oleic acid, to the low serum media did not rescue the growth defect of the mutant, suggesting bb0562 may be important for the release of fatty acids from triglycerides. Therefore, we searched for and identified two canonical GXSXG lipase motifs within BB0562, despite the lack of homology to known bacterial lipases. Purified BB0562 demonstrated lipolytic activity dependent on the catalytic serine residues within the two motifs. In sum, we have established that bb0562 is a novel nutritional virulence determinant, encoding a lipase that contributes to fatty acid scavenge for spirochete survival in environments deficient in free fatty acids including the mammalian host.


Subject(s)
Bacterial Proteins/metabolism , Fatty Acids/deficiency , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions , Lipase/metabolism , Lyme Disease/microbiology , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi/physiology , Female , Ixodes/microbiology , Lyme Disease/immunology , Lyme Disease/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred NOD , Virulence Factors/genetics
9.
PLoS Pathog ; 17(7): e1009725, 2021 07.
Article in English | MEDLINE | ID: mdl-34265024

ABSTRACT

In this study, we examined the relationship between c-di-GMP and its only known effector protein, PlzA, in Borrelia burgdorferi during the arthropod and mammalian phases of the enzootic cycle. Using a B. burgdorferi strain expressing a plzA point mutant (plzA-R145D) unable to bind c-di-GMP, we confirmed that the protective function of PlzA in ticks is c-di-GMP-dependent. Unlike ΔplzA spirochetes, which are severely attenuated in mice, the plzA-R145D strain was fully infectious, firmly establishing that PlzA serves a c-di-GMP-independent function in mammals. Contrary to prior reports, loss of PlzA did not affect expression of RpoS or RpoS-dependent genes, which are essential for transmission, mammalian host-adaptation and murine infection. To ascertain the nature of PlzA's c-di-GMP-independent function(s), we employed infection models using (i) host-adapted mutant spirochetes for needle inoculation of immunocompetent mice and (ii) infection of scid mice with in vitro-grown organisms. Both approaches substantially restored ΔplzA infectivity, suggesting that PlzA enables B. burgdorferi to overcome an early bottleneck to infection. Furthermore, using a Borrelia strain expressing a heterologous, constitutively active diguanylate cyclase, we demonstrate that 'ectopic' production of c-di-GMP in mammals abrogates spirochete virulence and interferes with RpoS function at the post-translational level in a PlzA-dependent manner. Structural modeling and SAXS analysis of liganded- and unliganded-PlzA revealed marked conformational changes that underlie its biphasic functionality. This structural plasticity likely enables PlzA to serve as a c-di-GMP biosensor that in its respective liganded and unliganded states promote vector- and host-adaptation by the Lyme disease spirochete.


Subject(s)
Adaptation, Physiological/physiology , Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Borrelia burgdorferi/pathogenicity , Virulence/physiology , Animals , Cyclic GMP/analogs & derivatives , Female , Host-Pathogen Interactions/physiology , Immune Evasion/physiology , Ixodes/parasitology , Lyme Disease/metabolism , Mice
10.
PLoS Pathog ; 17(5): e1009546, 2021 05.
Article in English | MEDLINE | ID: mdl-33984073

ABSTRACT

The bacterial pathogen responsible for causing Lyme disease, Borrelia burgdorferi, is an atypical Gram-negative spirochete that is transmitted to humans via the bite of an infected Ixodes tick. In diderms, peptidoglycan (PG) is sandwiched between the inner and outer membrane of the cell envelope. In many other Gram-negative bacteria, PG is bound by protein(s), which provide both structural integrity and continuity between envelope layers. Here, we present evidence of a peptidoglycan-associated protein (PAP) in B. burgdorferi. Using an unbiased proteomics approach, we identified Neutrophil Attracting Protein A (NapA) as a PAP. Interestingly, NapA is a Dps homologue, which typically functions to bind and protect cellular DNA from damage during times of stress. While B. burgdorferi NapA is known to be involved in the oxidative stress response, it lacks the critical residues necessary for DNA binding. Biochemical and cellular studies demonstrate that NapA is localized to the B. burgdorferi periplasm and is indeed a PAP. Cryo-electron microscopy indicates that mutant bacteria, unable to produce NapA, have structural abnormalities. Defects in cell-wall integrity impact growth rate and cause the napA mutant to be more susceptible to osmotic and PG-specific stresses. NapA-linked PG is secreted in outer membrane vesicles and augments IL-17 production, relative to PG alone. Using microfluidics, we demonstrate that NapA acts as a molecular beacon-exacerbating the pathogenic properties of B. burgdorferi PG. These studies further our understanding of the B. burgdorferi cell envelope, provide critical information that underlies its pathogenesis, and highlight how a highly conserved bacterial protein can evolve mechanistically, while maintaining biological function.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/physiology , Cell Wall/chemistry , Chemokines, CXC/metabolism , Lyme Disease/pathology , Peptidoglycan/metabolism , Bacterial Proteins/genetics , Cell Wall/microbiology , Chemokines, CXC/genetics , Humans , Lyme Disease/metabolism , Lyme Disease/microbiology
11.
Commun Biol ; 4(1): 268, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33649506

ABSTRACT

As opposed to pathogens passively circulating in the body fluids of their host, pathogenic species within the Spirochetes phylum are able to actively coordinate their movement in the host to cause systemic infections. Based on the unique morphology and high motility of spirochetes, we hypothesized that their surface adhesive molecules might be suitably adapted to aid in their dissemination strategies. Designing a system that mimics natural environmental signals, which many spirochetes face during their infectious cycle, we observed that a subset of their surface proteins, particularly Decorin binding protein (Dbp) A/B, can strongly enhance the motility of spirochetes in the extracellular matrix of the host. Using single-molecule force spectroscopy, we disentangled the mechanistic details of DbpA/B and decorin/laminin interactions. Our results show that spirochetes are able to leverage a wide variety of adhesion strategies through force-tuning transient molecular binding to extracellular matrix components, which concertedly enhance spirochetal dissemination through the host.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion , Borrelia burgdorferi/metabolism , Extracellular Matrix/microbiology , Ixodes/microbiology , Lyme Disease/microbiology , Adhesins, Bacterial/genetics , Animals , Borrelia burgdorferi/genetics , Borrelia burgdorferi/pathogenicity , Decorin/metabolism , Extracellular Matrix/metabolism , Female , Host-Pathogen Interactions , Kinetics , Laminin/metabolism , Lyme Disease/metabolism , Movement , Protein Binding , Rabbits , Single Molecule Imaging
12.
PLoS Pathog ; 17(1): e1009180, 2021 01.
Article in English | MEDLINE | ID: mdl-33428666

ABSTRACT

Borrelia burgdorferi must acquire all of its amino acids (AAs) from its arthropod vector and vertebrate host. Previously, we determined that peptide uptake via the oligopeptide (Opp) ABC transporter is essential for spirochete viability in vitro and during infection. Our prior study also suggested that B. burgdorferi employs temporal regulation in concert with structural variation of oligopeptide-binding proteins (OppAs) to meet its AA requirements in each biological niche. Herein, we evaluated the contributions to the B. burgdorferi enzootic cycle of three of the spirochete's five OppAs (OppA1, OppA2, and OppA5). An oppA1 transposon (tn) mutant lysed in the hyperosmolar environment of the feeding tick, suggesting that OppA1 imports amino acids required for osmoprotection. The oppA2tn mutant displayed a profound defect in hematogenous dissemination in mice, yet persisted within skin while inducing only a minimal antibody response. These results, along with slightly decreased growth of the oppA2tn mutant within DMCs, suggest that OppA2 serves a minor nutritive role, while its dissemination defect points to an as yet uncharacterized signaling function. Previously, we identified a role for OppA5 in spirochete persistence within the mammalian host. We now show that the oppA5tn mutant displayed no defect during the tick phase of the cycle and could be tick-transmitted to naïve mice. Instead of working in tandem, however, OppA2 and OppA5 appear to function in a hierarchical manner; the ability of OppA5 to promote persistence relies upon the ability of OppA2 to facilitate dissemination. Structural homology models demonstrated variations within the binding pockets of OppA1, 2, and 5 indicative of different peptide repertoires. Rather than being redundant, B. burgdorferi's multiplicity of Opp binding proteins enables host-specific functional compartmentalization during the spirochete lifecycle.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/physiology , Host-Pathogen Interactions , Ixodes/microbiology , Lyme Disease/microbiology , Membrane Transport Proteins/metabolism , Oligopeptides/metabolism , Animals , Biological Transport , Female , Gene Expression Regulation, Bacterial , Lyme Disease/genetics , Lyme Disease/metabolism , Mice , Mice, Inbred C3H , Rats , Rats, Sprague-Dawley , Virulence
13.
Biochem Biophys Res Commun ; 537: 1-6, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33373861

ABSTRACT

Borrelia burgdorferi is the causative agent of Lyme disease, the leading tick-borne illness in the United States. However, due to, in part, to the significant number of proteins of unknown function encoded across the complex fragmented genome, the molecular mechanisms of B. burgdorferi infection remain largely undefined. Previous work identified the virulence determinant gene, bbk13, which is critical for B. burgdorferi's ability to establish a productive disseminated infection. BBK13 is an immunogenic, non-surface exposed protein of unknown function predicted to harbor an N-terminal transmembrane domain and annotated as a member of the SIMPL domain protein superfamily (PF04402). In eukaryotes, SIMPL domain proteins have been shown to contribute to NF-kappa-B signaling but have no known functions in prokaryotes. Herein we investigated the biochemical and biophysical properties of BBK13 toward elucidation of its function. Bioinformatics analysis revealed secondary and tertiary structural homology between BBK13 and two other prokaryotic SIMPL domain proteins for which the crystal structures have been solved, Brucella abortus BP26 and Campylobacter jejuni cjSLP. Furthermore, comparable to BP26, recombinant BBK13 self-assembled into multimeric complexes in vitro and endogenous BBK13 was found in large oligomeric complexes in the spirochete membrane. Together these data suggest that the oligomeric structure of BBK13 may be important for the molecular function of this critical infection protein.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Cell Membrane/metabolism , Lyme Disease/metabolism , Lyme Disease/microbiology , Protein Multimerization , Amino Acid Sequence , Protein Domains , Protein Interaction Maps , Recombinant Proteins/chemistry , Structural Homology, Protein
14.
PLoS Pathog ; 16(10): e1008953, 2020 10.
Article in English | MEDLINE | ID: mdl-33048986

ABSTRACT

Borrelia burgdorferi, the Lyme disease pathogen causes persistent infection by evading the host immune response. Differential expression of the surface-exposed lipoprotein VlsE that undergoes antigenic variation is a key immune evasion strategy employed by B. burgdorferi. Most studies focused on the mechanism of VlsE antigen variation, but little is known about VlsE regulation and factor(s) that regulates differential vlsE expression. In this study, we investigated BB0025, a putative YebC family transcriptional regulator (and hence designated BB0025 as YebC of B. burgdorferi herein). We constructed yebC mutant and complemented strain in an infectious strain of B. burgdorferi. The yebC mutant could infect immunocompromised SCID mice but not immunocompetent mice, suggesting that YebC plays an important role in evading host adaptive immunity. RNA-seq analyses identified vlsE as one of the genes whose expression was most affected by YebC. Quantitative RT-PCR and Western blot analyses confirmed that vlsE expression was dependent on YebC. In vitro, YebC and VlsE were co-regulated in response to growth temperature. In mice, both yebC and vlsE were inversely expressed with ospC in response to the host adaptive immune response. Furthermore, EMSA proved that YebC directly binds to the vlsE promoter, suggesting a direct transcriptional control. These data demonstrate that YebC is a new regulator that modulates expression of vlsE and other genes important for spirochetal infection and immune evasion in the mammalian host.


Subject(s)
Antigenic Variation/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/metabolism , Borrelia burgdorferi/immunology , Immune Evasion/immunology , Lipoproteins/metabolism , Lyme Disease/immunology , Amino Acid Sequence , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Lipoproteins/genetics , Lipoproteins/immunology , Lyme Disease/metabolism , Lyme Disease/microbiology , Mice , Mice, Inbred C3H , Mice, SCID , Mutation , Protein Conformation , Sequence Homology
15.
PLoS Pathog ; 16(5): e1008516, 2020 05.
Article in English | MEDLINE | ID: mdl-32413091

ABSTRACT

Lyme disease, caused by Borrelia burgdorferi, B. afzelii and B. garinii, is a chronic, multi-systemic infection and the spectrum of tissues affected can vary with the Lyme disease strain. For example, whereas B. garinii infection is associated with neurologic manifestations, B. burgdorferi infection is associated with arthritis. The basis for tissue tropism is poorly understood, but has been long hypothesized to involve strain-specific interactions with host components in the target tissue. OspC (outer surface protein C) is a highly variable outer surface protein required for infectivity, and sequence differences in OspC are associated with variation in tissue invasiveness, but whether OspC directly influences tropism is unknown. We found that OspC binds to the extracellular matrix (ECM) components fibronectin and/or dermatan sulfate in an OspC variant-dependent manner. Murine infection by isogenic B. burgdorferi strains differing only in their ospC coding region revealed that two OspC variants capable of binding dermatan sulfate promoted colonization of all tissues tested, including joints. However, an isogenic strain producing OspC from B. garinii strain PBr, which binds fibronectin but not dermatan sulfate, colonized the skin, heart and bladder, but not joints. Moreover, a strain producing an OspC altered to recognize neither fibronectin nor dermatan sulfate displayed dramatically reduced levels of tissue colonization that were indistinguishable from a strain entirely deficient in OspC. Finally, intravital microscopy revealed that this OspC mutant, in contrast to a strain producing wild type OspC, was defective in promoting joint invasion by B. burgdorferi in living mice. We conclude that OspC functions as an ECM-binding adhesin that is required for joint invasion, and that variation in OspC sequence contributes to strain-specific differences in tissue tropism displayed among Lyme disease spirochetes.


Subject(s)
Borrelia burgdorferi/metabolism , Dermatan Sulfate/metabolism , Extracellular Matrix/metabolism , Joint Diseases/metabolism , Joints/metabolism , Lyme Disease/metabolism , Animals , Antigens, Bacterial , Bacterial Adhesion , Bacterial Outer Membrane Proteins , Borrelia burgdorferi/genetics , Borrelia burgdorferi/pathogenicity , Dermatan Sulfate/genetics , Extracellular Matrix/genetics , Extracellular Matrix/microbiology , Extracellular Matrix/pathology , Female , Fibronectins/genetics , Fibronectins/metabolism , Joint Diseases/genetics , Joint Diseases/microbiology , Joint Diseases/pathology , Joints/microbiology , Joints/pathology , Lyme Disease/genetics , Lyme Disease/microbiology , Lyme Disease/pathology , Mice , Mice, SCID , Mutation , Organ Specificity
17.
J Neuropathol Exp Neurol ; 79(5): 518-529, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32196082

ABSTRACT

Borrelia burgdorferi (Bb), which is neurotropic, can attack the central nervous system (CNS), leading to the development of various neurologic symptoms. The pathogenesis of Lyme neuroborreliosis (LNB) remains poorly understood. Presently, there is a lack of knowledge of the changes in mRNA and proteins in the CNS following early disseminated Lyme disease. Explants from the frontal cortex of 3 rhesus brains were incubated with medium alone or with medium containing live Bb for 6, 12, or 24 hours. Then, we analyzed identified mRNA and proteins in the frontal cortex tissues, allowing for an in-depth view of the transcriptome and proteome for a macroscopic and unbiased understanding of early disseminated Lyme disease in the brain. Through bioinformatics analysis, a complex network of enriched pathways that were mobilized during the progression of Lyme spirochete infection was described. Furthermore, based on the analysis of omics data, translational regulation, glycosaminoglycan/proteoglycan-binding activity in colonization and dissemination to tissues, disease-associated genes, and synaptic function were enriched, which potentially play a role in pathogenesis during the interaction between frontal cortex tissues and spirochetes. These integrated omics results provide unbiased and comprehensive information for the further understanding of the molecular mechanisms of LNB.


Subject(s)
Frontal Lobe/metabolism , Frontal Lobe/microbiology , Gene Expression Profiling , Lyme Disease/metabolism , Proteomics , Animals , Female , Gene Expression , Macaca mulatta , Male , RNA, Messenger/metabolism
18.
Ticks Tick Borne Dis ; 11(3): 101386, 2020 05.
Article in English | MEDLINE | ID: mdl-32035898

ABSTRACT

Changes in cellular metabolism have proven to be important factors in driving cell behavior. It has been shown that cellular metabolism of immune cells changes when exposed to or infected by several pathogens: while this is often an adaptation of the host cells to the infection, sometimes it represents a mechanism through which the pathogens evade immune activation. Borrelia burgdorferi sensu lato, the causative agent of Lyme borreliosis, is a pathogen that highly depends on the host to survive, as the bacterium lacks many central metabolic pathways to generate its own nutrients. It is therefore quite likely that the bacterium interacts with host cells to obtain these metabolites and thereby affects metabolism in the host. Previously, several studies have assessed metabolic pathways in B. burgdorferi s.l. and how it adapts to its different host species. However, few studies have looked into how the interaction with the bacterium might affect the host cell metabolism. In this review we present the major metabolic pathways activated during Lyme borreliosis, viewed from both bacterium and host metabolism, and we discuss how these pathways interact with each other, and how they influence pathogenesis of Lyme borreliosis.


Subject(s)
Borrelia burgdorferi/physiology , Host-Pathogen Interactions , Lyme Disease/metabolism , Metabolic Networks and Pathways , Lyme Disease/immunology
19.
FASEB J ; 34(2): 2840-2852, 2020 02.
Article in English | MEDLINE | ID: mdl-31908031

ABSTRACT

Eicosanoids are powerful mediators of inflammation and are known to drive both the progression and regression of arthritis. We previously reported the infection of C3H 5-lipoxygenase (LO)-deficient mice with Borrelia burgdorferi results in prolonged nonresolving Lyme arthritis. Here we define the role of the 5-LO metabolite leukotriene (LT)B4 and its high-affinity receptor, BLT1, in this response. C3H and C3H BLT1-/- mice were infected with B. burgdorferi and arthritis progression was monitored by ankle swelling over time. Similar to 5-LO-/- mice, BLT1-/- mice developed nonresolving Lyme arthritis characterized by increased neutrophils in the joint at later time points than WT mice, but with fewer apoptotic (caspase-3+ ) neutrophils. In vitro, BLT1-/- neutrophils were defective in their ability to undergo apoptosis due to the lack of LTB4 -mediated down-regulation of cAMP, subsequent failure to induce Death-Inducing Signaling Complex (DISC) components, and decreased FasL and CD36 expression. Inhibition of adenylyl cyclase with SQ 22,536 restored BLT1-/- BMN apoptosis, FasL and CD36 expression, and clearance by macrophages. We conclude that LTB4/BLT1 signaling has an unexpected critical role in mediating neutrophil apoptosis via the down-regulation of cAMP. Loss of BLT1 signaling led to defective clearance of neutrophils from the inflamed joint and failed arthritis resolution.


Subject(s)
Apoptosis , Borrelia burgdorferi/metabolism , Lyme Disease/metabolism , Neutrophils/metabolism , Receptors, Leukotriene B4/metabolism , Signal Transduction , Animals , Disease Models, Animal , Lyme Disease/genetics , Lyme Disease/pathology , Mice , Mice, Knockout , Neutrophils/pathology , Receptors, Leukotriene B4/genetics
20.
Biochim Biophys Acta Gen Subj ; 1864(3): 129499, 2020 03.
Article in English | MEDLINE | ID: mdl-31785327

ABSTRACT

Lyme disease is a tick-borne infection caused by Borrelia burgdorferi sensu lato complex spirochetes. The spirochete is located in the gut of the tick; as the infected tick starts the blood meal, the spirochete must travel through the hemolymph to the salivary glands, where it can spread to and infect the new host organism. In this study, we determined the crystal structures of the key outer surface protein BBE31 from B. burgdorferi and its orthologous protein BSE31 (BSPA14S_RS05060 gene product) from B. spielmanii. BBE31 is known to be important for the transfer of B. burgdorferi from the gut to the hemolymph in the tick after a tick bite. While BBE31 exerts its function by interacting with the Ixodes scapularis tick gut protein TRE31, structural and mass spectrometry data revealed that BBE31 has a glutathione (GSH) covalently attached to Cys142 suggesting that the protein may have acquired some additional functions in contrast to its orthologous protein BSE31, which lacks any interactions with GSH. In the current study, in addition to analyzing the potential reasons for GSH binding, the three-dimensional structure of BBE31 provides new insights into the molecular details of the transmission process as the protein plays an important role in the initial phase before the spirochete is physically transferred to the new host. This knowledge will be potentially used for the development of new strategies to fight against Lyme disease.


Subject(s)
Antigens, Bacterial/ultrastructure , Bacterial Outer Membrane Proteins/ultrastructure , Borrelia burgdorferi/metabolism , Lyme Disease/metabolism , Animals , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Borrelia burgdorferi/genetics , Borrelia burgdorferi/pathogenicity , Glutathione/metabolism , Humans , Ixodes/metabolism , Lyme Disease/transmission , Spirochaetales , Spirochaetales Infections/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...