Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
PLoS Pathog ; 20(4): e1012122, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38558079

ABSTRACT

Lyme disease is a tick-borne infection caused by the spirochete Borrelia (Borreliella) burgdorferi. Borrelia species have highly fragmented genomes composed of a linear chromosome and a constellation of linear and circular plasmids some of which are required throughout the enzootic cycle. Included in this plasmid repertoire by almost all Lyme disease spirochetes are the 32-kb circular plasmid cp32 prophages that are capable of lytic replication to produce infectious virions called ϕBB-1. While the B. burgdorferi genome contains evidence of horizontal transfer, the mechanisms of gene transfer between strains remain unclear. While we know that ϕBB-1 transduces cp32 and shuttle vector DNA during in vitro cultivation, the extent of ϕBB-1 DNA transfer is not clear. Herein, we use proteomics and long-read sequencing to further characterize ϕBB-1 virions. Our studies identified the cp32 pac region and revealed that ϕBB-1 packages linear cp32s via a headful mechanism with preferential packaging of plasmids containing the cp32 pac region. Additionally, we find ϕBB-1 packages fragments of the linear chromosome and full-length plasmids including lp54, cp26, and others. Furthermore, sequencing of ϕBB-1 packaged DNA allowed us to resolve the covalently closed hairpin telomeres for the linear B. burgdorferi chromosome and most linear plasmids in strain CA-11.2A. Collectively, our results shed light on the biology of the ubiquitous ϕBB-1 phage and further implicates ϕBB-1 in the generalized transduction of diverse genes and the maintenance of genetic diversity in Lyme disease spirochetes.


Subject(s)
Bacteriophages , Borrelia burgdorferi , Lyme Disease , Humans , Borrelia burgdorferi/genetics , Bacteriophages/genetics , Plasmids/genetics , Lyme Disease/genetics , Genomics , DNA
2.
J Immunol ; 211(6): 994-1005, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37556156

ABSTRACT

Long-lived T-dependent B cell responses fail to develop during persistent infection of mice with Borrelia burgdorferi, the causative agent of Lyme disease, raising questions about the induction and/or functionality of anti-B. burgdorferi adaptive immune responses. Yet, a lack of reagents has limited investigations into B. burgdorferi-specific T and B cells. We attempted two approaches to track B. burgdorferi-induced CD4 T cells. First, a B. burgdorferi mutant was generated with an influenza hemagglutinin (HA) peptide, HA111-119, inserted into the B. burgdorferi arthritis-related protein (Arp) locus. Although this B. burgdorferi arp::HA strain remained infectious, peptide-specific TCR transgenic CD4 T cells in vitro, or adoptively transferred into B. burgdorferi arp::HA-infected BALB/c mice, did not clonally expand above those of recipients infected with the parental B. burgdorferi strain or a B. burgdorferi mutant containing an irrelevant peptide. Some expansion, however, occurred in B. burgdorferi arp::HA-infected BALB/c SCID mice. Second, a (to our knowledge) newly identified I-Ab-restricted CD4 T cell epitope, Arp152-166, was used to generate Arp MHC class II tetramers. Flow cytometry showed small numbers of Arp-specific CD4 T cells emerging in mice infected with B. burgdorferi but not with Arp-deficient Borrelia afzelii. Although up to 30% of Arp-specific CD4 T cells were ICOS+PD-1+CXCR5+BCL6+ T follicular helper cells, their numbers declined after day 12, before germinal centers (GCs) are prominent. Although some Arp-specific B cells, identified using fluorochrome-labeled rArp proteins, had the phenotype of GC B cells, their frequencies did not correlate with anti-Arp serum IgG. The data suggest a failure not in the induction, but in the maintenance of GC T follicular helper and/or B cells to B. burgdorferi.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Mice , Animals , CD4-Positive T-Lymphocytes , Mice, SCID , B-Lymphocytes
3.
Mol Microbiol ; 119(6): 711-727, 2023 06.
Article in English | MEDLINE | ID: mdl-37086029

ABSTRACT

PlzA is a c-di-GMP-binding protein crucial for adaptation of the Lyme disease spirochete Borrelia (Borreliella) burgdorferi during its enzootic life cycle. Unliganded apo-PlzA is important for vertebrate infection, while liganded holo-PlzA is important for survival in the tick; however, the biological function of PlzA has remained enigmatic. Here, we report that PlzA has RNA chaperone activity that is inhibited by c-di-GMP binding. Holo- and apo-PlzA bind RNA and accelerate RNA annealing, while only apo-PlzA can strand displace and unwind double-stranded RNA. Guided by the crystal structure of PlzA, we identified several key aromatic amino acids protruding from the N- and C-terminal domains that are required for RNA-binding and unwinding activity. Our findings illuminate c-di-GMP as a switch controlling the RNA chaperone activity of PlzA, and we propose that complex RNA-mediated modulatory mechanisms allow PlzA to regulate gene expression during both the vector and host phases of the B. burgdorferi life cycle.


Subject(s)
Borrelia burgdorferi Group , Borrelia burgdorferi , Ixodes , Lyme Disease , Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Borrelia burgdorferi Group/genetics , Lyme Disease/genetics , RNA/metabolism
4.
PLoS Pathog ; 18(3): e1010385, 2022 03.
Article in English | MEDLINE | ID: mdl-35255112

ABSTRACT

We have identified GpsA, a predicted glycerol-3-phosphate dehydrogenase, as a virulence factor in the Lyme disease spirochete Borrelia (Borreliella) burgdorferi: GpsA is essential for murine infection and crucial for persistence of the spirochete in the tick. B. burgdorferi has a limited biosynthetic and metabolic capacity; the linchpin connecting central carbohydrate and lipid metabolism is at the interconversion of glycerol-3-phosphate and dihydroxyacetone phosphate, catalyzed by GpsA and another glycerol-3-phosphate dehydrogenase, GlpD. Using a broad metabolomics approach, we found that GpsA serves as a dominant regulator of NADH and glycerol-3-phosphate levels in vitro, metabolic intermediates that reflect the cellular redox potential and serve as a precursor for lipid and lipoprotein biosynthesis, respectively. Additionally, GpsA was required for survival under nutrient stress, regulated overall reductase activity and controlled B. burgdorferi morphology in vitro. Furthermore, during in vitro nutrient stress, both glycerol and N-acetylglucosamine were bactericidal to B. burgdorferi in a GlpD-dependent manner. This study is also the first to identify a suppressor mutation in B. burgdorferi: a glpD deletion restored the wild-type phenotype to the pleiotropic gpsA mutant, including murine infectivity by needle inoculation at high doses, survival under nutrient stress, morphological changes and the metabolic imbalance of NADH and glycerol-3-phosphate. These results illustrate how basic metabolic functions that are dispensable for in vitro growth can be essential for in vivo infectivity of B. burgdorferi and may serve as attractive therapeutic targets.


Subject(s)
Borrelia burgdorferi Group , Borrelia burgdorferi , Lyme Disease , Ticks , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Glycerol/metabolism , Glycerolphosphate Dehydrogenase/genetics , Glycerolphosphate Dehydrogenase/metabolism , Mice , NAD/metabolism , Oxidation-Reduction , Phosphates/metabolism
5.
Curr Issues Mol Biol ; 42: 223-266, 2021.
Article in English | MEDLINE | ID: mdl-33300497

ABSTRACT

Borrelia (Borreliella) burgdorferi, along with closely related species, is the etiologic agent of Lyme disease. The spirochete subsists in an enzootic cycle that encompasses acquisition from a vertebrate host to a tick vector and transmission from a tick vector to a vertebrate host. To adapt to its environment and persist in each phase of its enzootic cycle, B. burgdorferi wields three systems to regulate the expression of genes: the RpoN-RpoS alternative sigma factor cascade, the Hk1/Rrp1 two-component system and its product c-di-GMP, and the stringent response mediated by RelBbu and DksA. These regulatory systems respond to enzootic phase-specific signals and are controlled or fine- tuned by transcription factors, including BosR and BadR, as well as small RNAs, including DsrABb and Bb6S RNA. In addition, several other DNA-binding and RNA-binding proteins have been identified, although their functions have not all been defined. Global changes in gene expression revealed by high-throughput transcriptomic studies have elucidated various regulons, albeit technical obstacles have mostly limited this experimental approach to cultivated spirochetes. Regardless, we know that the spirochete, which carries a relatively small genome, regulates the expression of a considerable number of genes required for the transitions between the tick vector and the vertebrate host as well as the adaptation to each.


Subject(s)
Borrelia burgdorferi/genetics , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Lyme Disease/microbiology , Transcriptome , Adaptation, Physiological , Animals , Arthropod Vectors/microbiology , Genes, Bacterial , Host-Pathogen Interactions , Humans , Lyme Disease/transmission , Ticks/microbiology
6.
Mol Microbiol ; 113(2): 399-417, 2020 02.
Article in English | MEDLINE | ID: mdl-31742773

ABSTRACT

6S RNA binds to RNA polymerase and regulates gene expression, contributing to bacterial adaptation to environmental stresses. In this study, we examined the role of 6S RNA in murine infectivity and tick persistence of the Lyme disease spirochete Borrelia (Borreliella) burgdorferi. B. burgdorferi 6S RNA (Bb6S RNA) binds to RNA polymerase, is expressed independent of growth phase or nutrient stress in culture, and is processed by RNase Y. We found that rny (bb0504), the gene encoding RNase Y, is essential for B. burgdorferi growth, while ssrS, the gene encoding 6S RNA, is not essential, indicating a broader role for RNase Y activity in the spirochete. Bb6S RNA regulates expression of the ospC and dbpA genes encoding outer surface protein C and decorin binding protein A, respectively, which are lipoproteins important for host infection. The highest levels of Bb6S RNA are found when the spirochete resides in unfed nymphs. ssrS mutants lacking Bb6S RNA were compromised for infectivity by needle inoculation, but injected mice seroconverted, indicating an ability to activate the adaptive immune response. ssrS mutants were successfully acquired by larval ticks and persisted through fed nymphs. Bb6S RNA is one of the first regulatory RNAs identified in B. burgdorferi that controls the expression of lipoproteins involved in host infectivity.


Subject(s)
Adhesins, Bacterial/metabolism , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Borrelia burgdorferi , RNA, Bacterial , RNA, Untranslated , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Borrelia burgdorferi/genetics , Borrelia burgdorferi/metabolism , Gene Expression Regulation, Bacterial , Ixodes/microbiology , Lipoproteins/metabolism , Lyme Disease/microbiology , Mice , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Ribonucleases/genetics , Ribonucleases/metabolism
7.
Annu Rev Genet ; 46: 515-36, 2012.
Article in English | MEDLINE | ID: mdl-22974303

ABSTRACT

The spirochetes in the Borrelia burgdorferi sensu lato genospecies group cycle in nature between tick vectors and vertebrate hosts. The current assemblage of B. burgdorferi sensu lato, of which three species cause Lyme disease in humans, originated from a rapid species radiation that occurred near the origin of the clade. All of these species share a unique genome structure that is highly segmented and predominantly composed of linear replicons. One of the circular plasmids is a prophage that exists as several isoforms in each cell and can be transduced to other cells, likely contributing to an otherwise relatively anemic level of horizontal gene transfer, which nevertheless appears to be adequate to permit strong natural selection and adaptation in populations of B. burgdorferi. Although the molecular genetic toolbox is meager, several antibiotic-resistant mutants have been isolated, and the resistance alleles, as well as some exogenous genes, have been fashioned into markers to dissect gene function. Genetic studies have probed the role of the outer membrane lipoprotein OspC, which is maintained in nature by multiple niche polymorphisms and negative frequency-dependent selection. One of the most intriguing genetic systems in B. burgdorferi is vls recombination, which generates antigenic variation during infection of mammalian hosts.


Subject(s)
Borrelia burgdorferi/genetics , DNA, Bacterial/genetics , Genes, Bacterial , Prophages/metabolism , Alleles , Animals , Antigenic Variation , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacteriophages/genetics , Bacteriophages/metabolism , Bacteriophages/pathogenicity , Borrelia burgdorferi/immunology , Borrelia burgdorferi/pathogenicity , Borrelia burgdorferi/virology , DNA, Bacterial/metabolism , Electroporation , Evolution, Molecular , Genetic Variation , Humans , Ixodes/microbiology , Linkage Disequilibrium , Lipoproteins/genetics , Lipoproteins/immunology , Lipoproteins/metabolism , Lyme Disease/microbiology , Plasmids/genetics , Plasmids/metabolism , Prophages/genetics , Recombination, Genetic , Selection, Genetic , Species Specificity , Transduction, Genetic , Transformation, Genetic
8.
J Bacteriol ; 201(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30478087

ABSTRACT

The pathogenic spirochete Borrelia burgdorferi senses and responds to changes in the environment, including changes in nutrient availability, throughout its enzootic cycle in Ixodes ticks and vertebrate hosts. This study examined the role of DnaK suppressor protein (DksA) in the transcriptional response of B. burgdorferi to starvation. Wild-type and dksA mutant B. burgdorferi strains were subjected to starvation by shifting cultures grown in rich complete medium, Barbour-Stoenner-Kelly II (BSK II) medium, to a defined mammalian tissue culture medium, RPMI 1640, for 6 h under microaerobic conditions (5% CO2, 3% O2). Microarray analyses of wild-type B. burgdorferi revealed that genes encoding flagellar components, ribosomal proteins, and DNA replication machinery were downregulated in response to starvation. DksA mediated transcriptomic responses to starvation in B. burgdorferi, as the dksA-deficient strain differentially expressed only 47 genes in response to starvation compared to the 500 genes differentially expressed in wild-type strains. Consistent with a role for DksA in the starvation response of B. burgdorferi, fewer CFU of dksA mutants were observed after prolonged starvation in RPMI 1640 medium than CFU of wild-type B. burgdorferi spirochetes. Transcriptomic analyses revealed a partial overlap between the DksA regulon and the regulon of RelBbu, the guanosine tetraphosphate and guanosine pentaphosphate [(p)ppGpp] synthetase that controls the stringent response; the DksA regulon also included many plasmid-borne genes. Additionally, the dksA mutant exhibited constitutively elevated (p)ppGpp levels compared to those of the wild-type strain, implying a regulatory relationship between DksA and (p)ppGpp. Together, these data indicate that DksA, along with (p)ppGpp, directs the stringent response to effect B. burgdorferi adaptation to its environment.IMPORTANCE The Lyme disease bacterium Borrelia burgdorferi survives diverse environmental challenges as it cycles between its tick vectors and various vertebrate hosts. B. burgdorferi must withstand prolonged periods of starvation while it resides in unfed Ixodes ticks. In this study, the regulatory protein DksA is shown to play a pivotal role controlling the transcriptional responses of B. burgdorferi to starvation. The results suggest that DksA gene regulatory activity impacts B. burgdorferi metabolism, virulence gene expression, and the ability of this bacterium to complete its natural life cycle.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Gene Expression Regulation, Bacterial , Stress, Physiological , Transcription Factors/metabolism , Adaptation, Physiological , Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Borrelia burgdorferi/growth & development , Colony Count, Microbial , Culture Media/chemistry , Gene Deletion , Gene Expression Profiling , Guanosine Pentaphosphate/metabolism , Guanosine Tetraphosphate/metabolism , Microarray Analysis , Microbial Viability , Regulon , Transcription Factors/genetics
9.
Curr Top Microbiol Immunol ; 415: 113-140, 2018.
Article in English | MEDLINE | ID: mdl-28918538

ABSTRACT

The spirochetes Borrelia (Borreliella) burgdorferi and Borrelia hermsii, the etiologic agents of Lyme disease and relapsing fever, respectively, cycle in nature between an arthropod vector and a vertebrate host. They have extraordinarily unusual genomes that are highly segmented and predominantly linear. The genetic analyses of Lyme disease spirochetes have become increasingly more sophisticated, while the age of genetic investigation in the relapsing fever spirochetes is just dawning. Molecular tools available for B. burgdorferi and related species range from simple selectable markers and gene reporters to state-of-the-art inducible gene expression systems that function in the animal model and high-throughput mutagenesis methodologies, despite nearly overwhelming experimental obstacles. This armamentarium has empowered borreliologists to build a formidable genetic understanding of the cellular physiology of the spirochete and the molecular pathogenesis of Lyme disease.


Subject(s)
Borrelia/genetics , Genetic Engineering , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Borrelia/classification , Borrelia burgdorferi/genetics , Lyme Disease/microbiology , Molecular Biology , Relapsing Fever/microbiology
10.
J Bacteriol ; 200(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29632096

ABSTRACT

The rRNA genes of Borrelia (Borreliella) burgdorferi are unusually organized; the spirochete has a single 16S rRNA gene that is more than 3 kb from a tandem pair of 23S-5S rRNA operons. We generated an rnc null mutant in B. burgdorferi that exhibits a pleiotropic phenotype, including decreased growth rate and increased cell length. Here, we demonstrate that endoribonuclease III (RNase III) is, as expected, involved in processing the 23S rRNA in B. burgdorferi The 5' and 3' ends of the three rRNAs were determined in the wild type and rncBb mutants; the results suggest that RNase III in B. burgdorferi is required for the full maturation of the 23S rRNA but not for the 5S rRNA nor, curiously, for the 16S rRNA.IMPORTANCE Lyme disease, the most common tick-borne zoonosis in the Northern Hemisphere, is caused by the bacterium Borrelia (Borreliella) burgdorferi, a member of the deeply branching spirochete phylum. B. burgdorferi carries a limited suite of ribonucleases, enzymes that cleave RNA during processing and degradation. Several ribonucleases, including RNase III, are involved in the production of ribosomes, which catalyze translation and are a major target of antibiotics. This is the first study to dissect the role of an RNase in any spirochete. We demonstrate that an RNase III mutant is viable but has altered processing of rRNA.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/enzymology , RNA, Bacterial/metabolism , RNA, Ribosomal, 23S/metabolism , Ribonuclease III/metabolism , Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Borrelia burgdorferi/metabolism , Humans , Lyme Disease/microbiology , Operon , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , RNA, Ribosomal, 23S/genetics , RNA, Ribosomal, 5S/genetics , RNA, Ribosomal, 5S/metabolism , Ribonuclease III/genetics
11.
PLoS Pathog ; 11(9): e1005160, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26371761

ABSTRACT

As the Lyme disease bacterium Borrelia burgdorferi traverses its enzootic cycle, alternating between a tick vector and a vertebrate host, the spirochete must adapt and persist in the tick midgut under prolonged nutrient stress between blood meals. In this study, we examined the role of the stringent response in tick persistence and in regulation of gene expression during nutrient limitation. Nutritionally starving B. burgdorferi in vitro increased the levels of guanosine tetraphosphate (ppGpp) and guanosine pentaphosphate (pppGpp), collectively referred to as (p)ppGpp, products of the bifunctional synthetase/hydrolase RelBbu (RelA/SpoT homolog). Conversely, returning B. burgdorferi to a nutrient-rich medium decreased (p)ppGpp levels. B. burgdorferi survival in ticks between the larval and nymph blood meals, and during starvation in vitro, was dependent on RelBbu. Furthermore, normal morphological conversion from a flat-wave shape to a condensed round body (RB) form during starvation was dependent on RelBbu; relBbu mutants more frequently formed RBs, but their membranes were compromised. By differential RNA sequencing analyses, we found that RelBbu regulates an extensive transcriptome, both dependent and independent of nutrient stress. The RelBbu regulon includes the glp operon, which is important for glycerol utilization and persistence in the tick, virulence factors and the late phage operon of the 32-kb circular plasmid (cp32) family. In summary, our data suggest that RelBbu globally modulates transcription in response to nutrient stress by increasing (p)ppGpp levels to facilitate B. burgdorferi persistence in the tick.


Subject(s)
Arachnid Vectors/microbiology , Bacterial Proteins/metabolism , Borrelia burgdorferi/physiology , Gene Expression Regulation, Bacterial , Ixodes/microbiology , Pyrophosphatases/metabolism , Stress, Physiological , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Borrelia burgdorferi/growth & development , Borrelia burgdorferi/ultrastructure , Gastrointestinal Tract/microbiology , Guanosine Pentaphosphate/metabolism , Guanosine Tetraphosphate/metabolism , Ixodes/physiology , Larva/microbiology , Larva/physiology , Microbial Viability , Microscopy, Electron, Scanning , Mutation , Nymph/microbiology , Nymph/physiology , Operon , Pyrophosphatases/genetics , Regulon , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transcriptome
12.
Cell Microbiol ; 18(7): 919-27, 2016 07.
Article in English | MEDLINE | ID: mdl-27147446

ABSTRACT

Borrelia burgdorferi, the causative agent of Lyme disease (along with closely related genospecies), is in the deeply branching spirochete phylum. The bacterium is maintained in nature in an enzootic cycle that involves transmission from a tick vector to a vertebrate host and acquisition from a vertebrate host to a tick vector. During its arthropod sojourn, B. burgdorferi faces a variety of stresses, including nutrient deprivation. Here, we review some of the spirochetal factors that promote persistence, maintenance and dissemination of B. burgdorferi in the tick, and then focus on the utilization of available carbohydrates as well as the exquisite regulatory systems invoked to adapt to the austere environment between blood meals and to signal species transitions as the bacteria traverse their enzootic cycle. The spirochetes shift their source of carbon and energy from glucose in the vertebrate to glycerol in the tick. Regulation of survival under limiting nutrients requires the classic stringent response in which RelBbu controls the levels of the alarmones guanosine tetraphosphate and guanosine pentaphosphate (collectively termed (p)ppGpp), while regulation at the tick-vertebrate interface as well as regulation of protective responses to the blood meal require the two-component system Hk1/Rrp1 to activate production of the second messenger cyclic-dimeric-GMP (c-di-GMP).


Subject(s)
Arachnid Vectors/microbiology , Borrelia burgdorferi/physiology , Lyme Disease/microbiology , Ticks/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Borrelia burgdorferi/pathogenicity , Carbon/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Gene Expression Regulation, Bacterial , Guanosine Pentaphosphate/metabolism , Host-Pathogen Interactions
13.
Yale J Biol Med ; 90(2): 317-323, 2017 06.
Article in English | MEDLINE | ID: mdl-28656017

ABSTRACT

Borrelia (Borreliella) burgdorferi and closely related genospecies are the causative agents of Lyme disease, the most common tick-borne disease north of the equator. The bacterium, a member of the spirochete phylum, is acquired by a tick vector that feeds on an infected vertebrate host and is transmitted to another vertebrate during subsequent feeding by the next tick stage. The precise navigation of this enzootic cycle entails the regulation of genes required for these two host-specific phases as well as the transitions between them. Recently, an expansive swath of small RNAs has been identified in B. burgdorferi and likely many, if not most, are involved in regulating gene expression. Regardless, with only a few exceptions, the functions of these RNAs are completely unknown. However, several state-of-the-art approaches are available to identify the targets of these RNAs and provide insight into their role in the enzootic cycle and infection.


Subject(s)
Borrelia burgdorferi/physiology , RNA, Small Untranslated/physiology , Borrelia burgdorferi/genetics , Borrelia burgdorferi/pathogenicity , Humans , Lyme Disease/microbiology , RNA, Antisense/genetics , RNA, Antisense/physiology , RNA, Small Untranslated/genetics
14.
Annu Rev Microbiol ; 65: 479-99, 2011.
Article in English | MEDLINE | ID: mdl-21801026

ABSTRACT

Borrelia burgdorferi, the spirochete that causes Lyme disease, is maintained in nature via an enzootic cycle that comprises a tick vector and a vertebrate host. Transmission from the tick to the mammal, acquisition from the mammal back to the tick, and adaptation to the two disparate environments require sensing signals and responding by regulating programs of gene expression. The molecular mechanisms utilized to effect these lifestyle changes have begun to be elucidated and feature an alternative sigma factor cascade in which RpoN (σ(54)) and RpoS (σ(S)) globally control the genes required for the different phases of the enzootic cycle. The RpoN-RpoS pathway is surprisingly complex, entailing Rrp2, an unusual enhancer-binding protein and two-component regulatory system response regulator activated by acetyl phosphate; BosR, an unorthodox DNA-binding protein; DsrA(Bb), a small noncoding RNA; and Hfq and CsrA, two RNA-binding proteins. B. burgdorferi also has a c-di-GMP signaling system that regulates the tick side of the enzootic cycle and whose function is only beginning to be appreciated.


Subject(s)
Borrelia burgdorferi/genetics , Gene Expression Regulation, Bacterial , Lyme Disease/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Humans , Sigma Factor/genetics , Sigma Factor/metabolism , Ticks/microbiology , Ticks/physiology
15.
bioRxiv ; 2024 Jan 13.
Article in English | MEDLINE | ID: mdl-38260690

ABSTRACT

Lyme disease is a tick-borne infection caused by the spirochete Borrelia (Borreliella) burgdorferi. Borrelia species have highly fragmented genomes composed of a linear chromosome and a constellation of linear and circular plasmids some of which are required throughout the enzootic cycle. Included in this plasmid repertoire by almost all Lyme disease spirochetes are the 32-kb circular plasmid cp32 prophages that are capable of lytic replication to produce infectious virions called ϕBB-1. While the B. burgdorferi genome contains evidence of horizontal transfer, the mechanisms of gene transfer between strains remain unclear. While we know that ϕBB-1 transduces cp32 and shuttle vector DNA during in vitro cultivation, the extent of ϕBB-1 DNA transfer is not clear. Herein, we use proteomics and long-read sequencing to further characterize ϕBB-1 virions. Our studies identified the cp32 pac region and revealed that ϕBB-1 packages linear cp32s via a headful mechanism with preferentially packaging of plasmids containing the cp32 pac region. Additionally, we find ϕBB-1 packages fragments of the linear chromosome and full-length plasmids including lp54, cp26, and others. Furthermore, sequencing of ϕBB-1 packaged DNA allowed us to resolve the covalently closed hairpin telomeres for the linear B. burgdorferi chromosome and most linear plasmids in strain CA-11.2A. Collectively, our results shed light on the biology of the ubiquitous ϕBB-1 phage and further implicates ϕBB-1 in the generalized transduction of diverse genes and the maintenance of genetic diversity in Lyme disease spirochetes.

16.
Infect Immun ; 81(5): 1663-73, 2013 May.
Article in English | MEDLINE | ID: mdl-23460518

ABSTRACT

The laboratory mouse model of Lyme disease has revealed that Borrelia burgdorferi differentially expresses numerous outer surface proteins that influence different stages of infection (tick-borne transmission, tissue colonization, dissemination, persistence, and tick acquisition). Deletion of two such outer surface proteins, decorin-binding proteins A and B (DbpA/B), has been documented to decrease infectivity, impede early dissemination, and, possibly, prevent persistence. In this study, DbpA/B-deficient spirochetes were confirmed to exhibit an early dissemination defect in immunocompetent, but not immunodeficient, mice, and the defect was found to resolve with chronicity. Development of disease (arthritis and carditis) was attenuated only in the early stage of infection with DbpA/B-deficient spirochetes in both types of mice. Persistence of the DbpA/B-deficient spirochetes occurred in both immunocompetent and immunodeficient mice in a manner indistinguishable from that of wild-type spirochetes. Dissemination through the lymphatic system was evaluated as an underlying mechanism for the early dissemination defect. At 12 h, 3 days, 7 days, and 14 days postinoculation, DbpA/B-deficient spirochetes were significantly less prevalent and in lower numbers in lymph nodes than wild-type spirochetes. However, in immunodeficient mice, deficiency of DbpA/B did not significantly decrease the prevalence or spirochete numbers in lymph nodes. Complementation of DbpA/B restored a wild-type phenotype. Thus, the results indicated that deficiency of DbpA/B allows the acquired immune response to restrict early dissemination of spirochetes, which appears to be at least partially mediated through the lymphatic system.


Subject(s)
Adhesins, Bacterial/physiology , Borrelia burgdorferi/pathogenicity , Lyme Disease/microbiology , Adaptive Immunity/physiology , Animals , Bacterial Adhesion/physiology , Chronic Disease , Female , Immunocompromised Host , Lyme Disease/immunology , Lymph Nodes/microbiology , Mice , Mice, SCID
18.
Mol Microbiol ; 78(3): 622-35, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20815822

ABSTRACT

Hfq is a global regulatory RNA-binding protein. We have identified and characterized an atypical Hfq required for gene regulation and infectivity in the Lyme disease spirochete Borrelia burgdorferi. Sequence analyses of the putative B. burgdorferi Hfq protein revealed only a modest level of similarity with the Hfq from Escherichia coli, although a few key residues are retained and the predicted tertiary structure is similar. Several lines of evidence suggest that the B. burgdorferi bb0268 gene encodes a functional Hfq homologue. First, the hfq(Bb) gene (bb0268) restores the efficient translation of an rpoS::lacZ fusion in an E. coli hfq null mutant. Second, the Hfq from B. burgdorferi binds to the small RNA DsrA(Bb) and the rpoS mRNA. Third, a B. burgdorferi hfq null mutant was generated and has a pleiotropic phenotype that includes increased cell length and decreased growth rate, as found in hfq mutants in other bacteria. The hfq(Bb) mutant phenotype is complemented in trans with the hfq gene from either B. burgdorferi or, surprisingly, E. coli. This is the first example of a heterologous bacterial gene complementing a B. burgdorferi mutant. The alternative sigma factor RpoS and the outer membrane lipoprotein OspC, which are induced by increased temperature and required for mammalian infection, are not upregulated in the hfq mutant. Consequently, the hfq mutant is not infectious by needle inoculation in the murine model. These data suggest that Hfq plays a key role in the regulation of pathogenicity factors in B. burgdorferi and we hypothesize that the spirochete has a complex Hfq-dependent sRNA network.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Host Factor 1 Protein/metabolism , Lyme Disease/microbiology , Molecular Chaperones/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Borrelia burgdorferi/chemistry , Borrelia burgdorferi/genetics , Borrelia burgdorferi/pathogenicity , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Gene Expression Regulation, Bacterial , Genetic Complementation Test , Host Factor 1 Protein/chemistry , Host Factor 1 Protein/genetics , Humans , Mice , Mice, Inbred C3H , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Sequence Data , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sequence Alignment , Virulence
19.
Mol Microbiol ; 74(6): 1295-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19943896

ABSTRACT

Borrelia burgdorferi encodes a novel DNA-binding protein in the Fur/PerR family of transcriptional regulators termed BosR (BB0647). This issue of Molecular Microbiology contains two molecular genetic studies that help to clarify the function of BB0647 and resolve longstanding controversies. Loss of BB0647 appears to have a pronounced effect on borrelial gene expression and, in one study, caused significant in vitro growth defects. BB0647 was also found to be essential for infection of the mammalian host but not the tick vector. Both Ouyang et al. and Hyde et al. also demonstrate, quite unexpectedly, that BB0647 is required for induction of RpoS, an alternative sigma factor that controls a cadre of B. burgdorferi genes, most notably ospC, which enable the spirochetes to establish mammalian infection following tick inoculation. There are still many unanswered questions regarding the precise physiological role of BB0647, the most important of which relate to its homologues Fur and PerR: to what extent does it regulate either the response to oxidative stress and/or transition metal uptake? The mechanism(s) whereby BB0647 interfaces with the Rrp2-RpoN-RpoS pathway also remains to be discerned. However, these two seminal papers establish BB0647 (BosR) as a central player in the molecular biology and physiology of B. burgdorferi as well as the pathogenesis of Lyme disease.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial , Animals , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Borrelia burgdorferi/growth & development , Borrelia burgdorferi/pathogenicity , DNA-Binding Proteins/genetics , Gene Deletion , Humans , Sigma Factor/biosynthesis
20.
Appl Environ Microbiol ; 76(3): 985-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19966024

ABSTRACT

We have demonstrated that rpsL, encoding the S12 protein of the small ribosomal subunit, can be used as a counterselectable marker in Borrelia burgdorferi, the causative agent of Lyme disease. Mutations in rpsL confer streptomycin resistance. Streptomycin susceptibility is dominant in an rpsL merodiploid, and streptomycin selects for the loss of wild-type rpsL carried in trans. This is the first description of a counterselectable marker in B. burgdorferi.


Subject(s)
Anti-Bacterial Agents/pharmacology , Borrelia burgdorferi/genetics , Ribosomal Proteins/genetics , Streptomycin/pharmacology , Bacterial Proteins/genetics , Biomarkers , Borrelia burgdorferi/drug effects , Cloning, Molecular , DNA, Bacterial/genetics , Drug Resistance, Bacterial , Genes, Bacterial , Genetic Engineering , Microbial Sensitivity Tests , Models, Genetic , Mutagenesis, Site-Directed , Mutation , Protein Synthesis Inhibitors/pharmacology , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Selection, Genetic , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL