You are on page 1of 31

VIRULENCE

2023, VOL. 14, NO. 1, 2265015


https://doi.org/10.1080/21505594.2023.2265015

REVIEW ARTICLE

Pathogenicity and virulence of Borrelia burgdorferi


a,b a a,b
Martin Strnad , Natalie Rudenko , and Ryan O.M. Rego
a
Biology Centre CAS, Institute of Parasitology, České Budějovice, Czech Republic; bFaculty of Science, University of South Bohemia,
Branišovská, Czech Republic

ABSTRACT ARTICLE HISTORY


Infection with Borrelia burgdorferi often triggers pathophysiologic perturbations that are further Received 9 November 2022
augmented by the inflammatory responses of the host, resulting in the severe clinical conditions of Revised 23 August 2023
Lyme disease. While our apprehension of the spatial and temporal integration of the virulence Accepted 25 September
determinants during the enzootic cycle of B. burgdorferi is constantly being improved, there is still 2023
much to be discovered. Many of the novel virulence strategies discussed in this review are undeter­ KEYWORDS
mined. Lyme disease spirochaetes must surmount numerous molecular and mechanical obstacles in Borrelia burgdorferi; Lyme
order to establish a disseminated infection in a vertebrate host. These barriers include borrelial disease; virulence
relocation from the midgut of the feeding tick to its body cavity and further to the salivary glands, determinants; pathogenicity;
deposition to the skin, haematogenous dissemination, extravasation from blood circulation system, clinical manifestations; tick-
evasion of the host immune responses, localization to protective niches, and establishment of local as borne disease
well as distal infection in multiple tissues and organs. Here, the various well-defined but also possible
novel strategies and virulence mechanisms used by B. burgdorferi to evade obstacles laid out by the
tick vector and usually the mammalian host during colonization and infection are reviewed.

Introduction
[1] and 650 000–850 000 cases in Europe annually [2].
The bacterial pathogen Borrelia burgdorferi, also known LD occurs in a variety of clinical manifestations, ran­
as Borreliella burgdorferi, is the causative agent of Lyme ging from skin lesions, through musculoskeletal, neu­
disease (LD) and is transmitted to humans through rological, to cardiovascular symptoms [3]. The
Ixodes ticks. LD is the most common tick infection, organotrophy associated with B. burgdorferi species
with approximately 476 000 cases in the United States has an impact on the development of the disease.

CONTACT Martin Strnad martin.strnad.cze@gmail.com


© 2023 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted
use, distribution, and reproduction in any medium, provided the original work is properly cited. The terms on which this article has been published allow the posting of the
Accepted Manuscript in a repository by the author(s) or with their consent.
2 M. STRNAD ET AL.

Although there is already an abundance of knowledge tick salivary glands, from which they are transmitted to
on the causative agent of the disease, an efficacious the host dermis via saliva. The salivary route of trans­
strategy to reliably overcome LD has not been found. mission is highly favourable to the pathogen as it delays
There is currently no vaccine for human use available, the contact with borreliacidal factors present in the host
although new ways how to cope with the malicious blood. Borrelia then migrate through the extracellular
pathogen are being explored [4]. Multiple genes com­ matrix (ECM) constituents for haematogenous dissemi­
mon for B. burgdorferi sensu lato species (referred to as nation into their colonization niches where they cause
B. burgdorferi in this Review) display high sequence the damage that can result in development of disease
variability between the species of LD spirochaetes. and complete the enzootic cycle [13,14].
Genes required for optimal infectivity often differ To be able to establish an infection and stay inside
markedly at both the nucleotide and amino acid level the host, B. burgdorferi must evade the immune pres­
[5,6]. Such variability significantly complicates the sure exerted by the host’s defence system. The genome
design of efficacious immunization strategies including sequences of B. burgdorferi species revealed that spir­
vaccines since the immune responses do not protect ochaetes lack the classically defined virulence determi­
against all infectious species to the same extent. This nants common to a large number of microbial and,
highlights the current strategy which involves clinical specifically, bacterial pathogens, such as lipopolysac­
development of a multivalent protein vaccine that tar­ charides, specialized secretion systems, and toxins
gets all common serotypes of B. burgdorferi [7]. [15,16]. Borrelia has developed several well-defined
Virulence determinants of B. burgdorferi are found mechanisms such as utilization of tick and host pro­
both on the linear chromosome and on linear and teins, complement evasion, localization into specific
circular plasmids. Chromosomal virulence genes niches, dynamic genetic regulation and antigenic varia­
include p66, bgp, plzA, rpoN, rpoS, or bosR whereas tion, in order to survive in the host [8]. There are also
virulence-associated genes that are present on plasmids several alleged virulence mechanisms not yet uncondi­
encode proteins such as DbpA/B, BBK32, OspC, PncA, tionally accepted by the expert community. These
VlsE [8]. B. burgdorferi possesses a unique, segmented include transformations of spiral-shaped replicating
genome comprised of the largest plasmid complement Borrelia into alternative structural forms such as
of any characterized bacterium. The plasmids are sub­ round bodies or biofilm-like structures [17]. These
stantially variable between species and strains, both in pleomorphic variants are hypothesized to be able to
number and in sequence [9]. In contrast to a majority survive the host immune response or prolonged courses
of other bacteria, circular plasmids represent a minority of antibiotic treatment in cases when the subpopulation
of the genome in B. burgdorferi. With the exception of of Borrelia cells switch the phenotype from susceptible
the cp-26 plasmid, all circular plasmids are related to to persistent under hostile pressure [18]. Intracellular
each other and are derived from the cp-32 plasmid localization of the spirochaetes, shedding of borrelial
family. A unique characteristic of linear replicons is outer membrane vesicles, nutritional virulence, modu­
that they are terminated by covalently closed hairpin lation of tick microbiota and tick behaviour, and
ends [10]. Borrelia-induced structural and mechanical transfor­
B. burgdorferi exists in an enzootic cycle alternating mations of host cells are other potential virulence
between a tick vector and a vertebrate host (Figure 1). mechanisms of LD Borrelia [19–25].
Acquisition of B. burgdorferi by a tick occurs through There are a number of outstanding reviews focus­
haematophagous bites of the tick. In order to feed, tick ing either on a narrow range of topics in borrelial
chelicerae cut the host epidermis and the hypostome virulence mechanisms [26–29] or which provide
penetrates through the layers of the skin to secret saliva a detailed overview of multiple widely accepted viru­
into the tick bite site on the host and draw blood, lence mechanisms [8]. Herein, we guide the reader
possibly, together with Borrelia [11]. The spirochaete not only through the complexities of classical but
enters the tick when the larval/nymphal tick takes importantly also through the trending virulence
a bloodmeal on an infected host and colonizes the mechanisms employed by LD Borrelia. In the context
tick midgut. During subsequent feeding, a handful of of this review, similarly as in the other review of
spirochaetes colonizing the tick exit the midgut by borrelial pathogenic mechanisms [8], the term “viru­
traversing a peritrophic membrane, a layer of epithelial lence determinants” encompasses all genes that
cells and a basal membrane [12]. The spirochaetes then impact the infection process, eventhough these fac­
migrate into the haemocoel as they navigate toward the tors were often studied and defined in the host
VIRULENCE 3

Figure 1. Enzootic cycle responsible for maintaining B. burgdorferi in tick populations. Ixodes ticks undergo a three-stage life
cycle (larva, nymph and adult), with one bloodmeal per stage. Uninfected larval ticks hatch from eggs and feed on a variety of small
mammals that can host B. burgdorferi. The larva takes an infected bloodmeal and moults into a next tick stage, which is a nymph.
Nymphs are responsible for transmitting the majority of infections to humans, leading to variety of clinical manifestations. Deer are
important hosts for adult ticks but they are not effective reservoirs for the spirochaetes. Engorged females lay thousands of eggs,
however, the spirochaetes are not transmitted through the eggs to the larval progeny.

animals which are not very susceptible to the disease Diagnosis of Lyme disease
or show few signs of LD, e.g. mice. This definition
includes also genes involved in fundamental meta­ LD has a broad spectrum of symptoms and clinical
bolic processes necessary for borrelial survival in the manifestations. The initial step in distinguishing LD
host. When the genes associated with a suspected from other illnesses involves the review of all signs
virulence trait are specifically deactivated, it results and symptoms and gathering subjective and objective
in a noticeable reduction in infection potential. To data from the patient. If there is potential tick exposure
sum up, in addition to a better understanding of the in an endemic area and the individual exhibits an
bacterial pathogenesis process, identification of viru­ expanding skin lesion called erythema migrans (EM),
lence factors in the spirochaetal genome may allow immediate suspicion for LD should arise. Recognizing
the design of new, more efficient and, importantly, EM can be challenging, since EM can be present in
more specific intervention strategies than those that various morphological forms, different from the classic
are currently employed in combating of LD. bull’s eye [30].
4 M. STRNAD ET AL.

The current reference method utilized for confirma­ longer treatment periods [41]. When LD is diagnosed
tion of physician-diagnosed LD is serological testing. later, after the spirochaetes have moved from the tick
Standard two-tiered testing (STTT) involves an initial bite site to the secondary sites of infection, the stage is
enzyme-linked immunoassay (ELISA) as the primary called early disseminated infection. This stage can occur
test, followed by supplemental IgM and/or IgG immu­ days, weeks, or even months after an infected tick
noblots. The STTT is recognized for its limited sensi­ transmits the spirochaetes. Either oral or intravenous
tivity in detecting early localized infection, as opposed antibiotic therapy is recommended for treatment.
to very high sensitivity in detecting late infection [31]. Clinical manifestations of late LD include a variety of
Modified two-tier testing (MTTT) is an updated two- symptoms that are often arthritic and neurologic, and
tiered testing algorithm, in which confirmation of the occur months after the transmission of the pathogen
infection is done with an ELISA instead of an immu­ [42,43]. Even though the majority of LD cases are cured
noblot. Multiple studies, both in North America and by antibiotic therapy, around 10–20% of patients
Europe, have shown that MTTT has improved sensitiv­ experience persistent disease that does not resolve
ity compared with STTT, without losing specificity within an extended period of time even after repeated
[32,33]. Some MTTT algorithms have already received courses of antibiotic therapy [44]. Symptoms such as
FDA clearance for use in serological diagnostics of LD. headache, fatigue, and joint pain are commonly asso­
The interpretation of serology results can be intri­ ciated with this stage of the infection, which has been
cate. It usually takes several days to a few weeks for the defined as post-treatment Lyme disease syndrome
immune system to generate a Borrelia-specific IgM (PTLDS) [44]. PTLDS is considered a subset of the
response, while the development of a Borrelia-specific term chronic LD, which is an umbrella term with
IgG response can take up to two months [34]. Methods a wide and imprecise definition, often applied to indi­
to detect IgM antibodies are mainly useful during early viduals experiencing non-specific symptoms that are
disease, since later during infection they often generate believed to stem from an assumed ongoing
false positive signal due to cross-reactions with other B. burgdorferi infection. These patients may or may
infections [35,36]. Another regular weakness of current not exhibit evidence of past or present LD [45].
commercial diagnostic tests is the low sensitivity for Considerable confusion and controversy still exist and
less prevalent or rare Borrelia species, especially those questions about the possible existence of this fourth
which exhibit high polymorphism in the commonly stage of LD, its frequency or the cause of PTLDS is
detected immunogenic proteins [37,38]. In addition, actively discussed by the scientific and medical com­
establishing a clear connection between antibody status munity. The exact mechanisms and molecular triggers
and actual infection can be challenging. Non-infected leading to the long-term sequelae of the infection are
individuals may exhibit immunity long after active LD still undetermined. It is hypothesized that the reduced
and test positive, while infected individuals might efficacy of antibiotic therapy may be caused by: a) host-
experience a delay in their antibody response and test adapted spirochaetes that persist in the tissues or pos­
negative [39]. Furthermore, specifically in Lyme neuro­ sibly intracellularly inside certain cell types such as
borreliosis, the antibody production does not always fibroblasts and thus are inaccessible to antibiotics
follow the typical immune response of initial IgM [19]; b) excessive inflammatory responses to residual
secretion followed by secretion of IgG [40]. antigenic molecules such as peptidoglycan from killed
microbes or autoimmune responses [46]; c) tissue
damage after clearance of Borrelia [42].
Treatment & clinical manifestations of Lyme
The genetic heterogeneity of B. burgdorferi species
disease
seems to be the main factor of the diverse clinical
The course of LD can be divided into three distinct manifestations with serious multi-system disorders of
stages – early localized LD, early disseminated LD and human LD. It is not surprising, as out of 22 currently
late LD. Early localized infection, typically presented as recognized LD spirochaete species, 12 were already
a painless skin rash (EM), is treated with antibiotics. detected or isolated from human patients, diagnosed
The therapeutic success of LD is dependent on how with LD or expected to have a spirochaete infection
early is the treatment with antibiotics initiated. A short [47,48]. Within the species that are detected in, or
course of oral antibiotics in most cases efficiently elim­ isolated from humans, the frequency of recovery varies
inates the pathogen and cures the majority of LD cases. greatly. While B. afzelii, B. bavariensis, B. garinii,
It is recommended that patients with EM are treated B. burgdorferi sensu stricto (ss) or B. mayonii are
with a 10-day course of doxycycline or a 14-day course responsible for the majority of LD cases worldwide,
of amoxycillin or, possibly, cefuroxime rather than the number of patients in which B. bissettii,
VIRULENCE 5

B. kurtenbachii, B. spielmanii or B. valaisiana have been was pulled from the US market in 2002 due to numer­
detected is low, and detection of B. americana, ous reports of severe side effects. There was
B. lusitaniae or B. andersonii in humans is rare [49, a hypothesis suggesting that the vaccine antigen, outer
50]. Diverse clinical manifestations of the disease are surface protein A (OspA), acted as an autoantigen,
connected to different organ tropism that individual potentially causing arthritis. However, the adverse
LD spirochaete species possess. For example, infection effects were never definitively substantiated [75]. The
with B. burgdorferi ss, the major case of LD in the Valneva-Pfizer “VLA15” is currently the only LD vac­
United States, results in the most common for North cine candidate in advanced clinical development
America musculoskeletal disorder, Lyme arthritis, that (phase 3). This multivalent protein subunit vaccine
was diagnosed in at least 60% of untreated human targets OspA and provides protection against the six
patients with EM [51]. In Europe, where B. afzelii and prevalent OspA serotypes expressed by the
B. garinii represent the most pathogenic spirochaete B. burgdorferi species commonly found in North
species and are recovered from environmental and America and Europe [7,76].
samples of human origin much more often than In order to address the considerable variability
B. burgdorferi ss, the number of diagnosed cases of observed in proteins across different strains and species
arthritis did not exceed 15% [52]. Widely distributed of B. burgdorferi, effort has been put on creating
B. garinii was confirmed to be related to increased a multivalent chimeric vaccines that offer protective
number of neuroborreliosis cases in Europe [53,54]. effects against various species of LD, aiming to tackle
Occasionally, infections with B. burgdorferi ss and the heterogeneity inherent in the pathogen [70,77,78].
B. afzelii result in neuroborelliosis as well, however, Multivalent OspA-based formulations are currently
the number of diagnosed cases is comparably lower. common targets in vaccine development. To create self-
B. afzelii predominantly causes skin infections such as assembling nanoparticles, OspA was combined with
EM, acrodermatitis chronica atrophicans (ACA) or bacterial ferritin. These OspA-ferritin nanoparticles
borrelial lymphocytoma [55]. Rare cases of ACA were induced robust and long-lasting antibody responses
described in patients infected with B. garinii as well against the major serotypes in both mice and non-
[56–58]. In addition to the major recognized LD caus­ human primates [79]. To achieve broad protection
ing species, some other species are occasionally with a single recombinant antigen, a strategy called
detected in humans as well. While connection of grafting, or epitope reshaping, has been successfully
B. bissettii [59,60] with cardiovascular disorders was tested for production of neutralizing antibodies against
described in patients from Europe [61,62], the status the six clinically most relevant OspA serotypes [80].
of B. lusitaniae [63], and B. valaisiana [64] as a human Besides vaccines based on recombinant protein tech­
pathogen still needs a valid confirmation [65]. Tick co- nology, DNA tattoo and lipid nanoparticle-
infection with other pathogens such as Rickettsia and A. encapsulated DNA vaccination represents a promising
phagocytophilum does not significantly increase in clin­ strategy for prevention of LD [81–83]. DNA vaccines
ical symptoms or disease duration [66]. present several potential benefits compared to conven­
tional strategies, such as the activation of both B- and
T-cell responses, enhanced vaccine stability, the
Vaccine development
absence of any infectious agents, and the straightfor­
Although there are currently no approved vaccines ward large-scale production process [84].
against LD for human use, multiple studies have built
a solid foundation for further vaccine development.
B. burgdorferi produces multiple antigenic and immu­ Methods used to identify virulence mechanisms
nologically accessible molecules which were shown to and determinants
be promising vaccine candidates but which have never
Genetic modifications
reached the market. These molecules include mostly the
surface-exposed borrelial proteins such as BBA52, Virulence is defined by the ability of a microorganism
BB0405, BBI39, DbpA, BBK32, OspC, with data show­ to cause disease in the host. Modern strategies that are
ing that combination vaccines composed of DbpA, being used to uncover the fine details of bacterial viru­
BBK32, and OspC are more powerful than single for­ lence mechanisms encompass a combination of bio­
mulation or double component cocktails [67–73]. chemical, genetic, imaging, and computational
The only licenced vaccine to prevent LD was techniques. The currently used genetic tools for manip­
LYMERix, with efficacy of nearly 80% [74]. After ulating B. burgdorferi are sufficiently mastered for pre­
being available on the market few years, the vaccine cise and effective genetic investigations and were
6 M. STRNAD ET AL.

recently reviewed in great detail by Rosa and Jewett grown under fed-tick, unfed-tick, and mammalian
[85]. Targeted genes can be selectively rendered inac­ growth conditions, it was shown that mostly the extra­
tive and the resulting mutant phenotype can be exam­ cellular lipoproteins are dynamically regulated during
ined for infectivity in the experimental mouse-tick borrelial transitions between the tick vector and the
transmission cycle, thus elucidating the actual functions host, hinting to their important role in borrelial
of borrelial virulence factors during infection of the pathogenesis [93]. The common drawback of the
host. Genetic manipulation of B. burgdorferi is typically DNA microarray technology is that only the annotated
achieved via allelic exchange. The transformation pro­ coding regions of the genome can be profiled. To meet
tocol often makes use of electroporation for transfer of the limitations of probe performance in DNA micro­
foreign DNA [86]. The selection of a mutant with arrays, RNA sequencing (RNA-Seq) has been used to
either gene disruption or gene complementation is explore for instance the antisense transcripts and non­
achieved by using a selectable antibiotic marker. coding/small RNAs [97,98].
Suicide vectors are commonly utilized for targeted The in vivo expression technology (IVET) is
gene inactivation, as the mutant is generated by allelic a genetic tool that allows selection of genes specifically
replacement of the wild-type gene with a disrupted expressed in vivo in complex environments. In contrast
copy [85,87]. The functional complementation of the to DNA microarrays technology and RNA-Seq, IVET
mutant is typically achieved through shuttle and suicide does not require borrelial RNA isolation for the tran­
vectors for trans and cis complementation, respec­ scriptomic investigation and provides purely qualitative
tively [85]. data with no information about the quantity of studied
Transposon mutagenesis and signature tagged muta­ gene product. This promoter-trapping technique pro­
genesis (STM) are more recent genetic approaches that vides means for genome-wide identification of the bor­
were developed to identify novel bacterial virulence relial genes that are expressed for instance during
factors and were already exploited in borrelial research murine infection. Using IVET for the first time in
[88–90]. Both methods represent a technical advance B. burgdorferi, an RpoS-independent gene bbk46 was
over other random mutagenesis approaches such as identified and shown to be important for evasion of the
radiation and chemical mutangenesis [91] as they uti­ host adaptive immunity [99]. Using an IVET reporter
lize transposable genetic elements to randomly inacti­ cassette containing antibiotic resistance aacC1 gene and
vate multiple genes, with each specific mutation borrelial pncA, Casselli and Bankhead [100] identified
occurring in an individual clone. A Himar1-based a number of constitutively expressed promoters,
transposon suicide vector is often used for the delivery in vitro-induced promoters, and in vivo-induced pro­
of the gene insertions [88,92]. The principle of STM is moters in B. burgdorferi.
based on molecular barcoding with unique DNA
sequences (signature tags), which allow mutants to be
Imaging techniques
differentiated from each other. This allows high-
throughput screens of large number of different The rapid development of imaging technologies has
mutants within a single animal. By comparing the revolutionized our ability to visualize the delicate intri­
signature tags of the input pool of mutants, i.e. those cacies of Borrelia-host interactions at very different
which were inoculated into an animal, and the recov­ scales. Imaging methods and applications encompass
ered pool of mutants, i.e. those which survived in the a palette of options from which one might select, span­
animal, the important virulence factors can be ning from whole animal imaging techniques to mod­
identified. alities that are able to reach subatomic resolution
[101,102]. Bioluminescent imaging has enabled one to
follow the infection of Borrelia in an entire model
Gene expression analysis
animal over time by non-invasive means in natural
DNA microarrays have been used to study changes in environment [101]. Another sophisticated technique
the global expression profile of B. burgdorferi grown that has allowed the investigation of Borrelia pathogen­
under various in vitro and in vivo conditions [93,94]. esis, including dissemination and invasion of the host,
For mimicking the growth conditions in the mamma­ is intravital microscopy [14]. Intravital imaging allows
lian host, a dialysis membrane chamber implanted real-time characterization of a pathogen in a living host
into the rat peritonea is considered a top-notch at specific locations. For intravital imaging, spinning
model as it allows to obtain high number of host- disk confocal microscopy (SDCM) and two-photon
adapted spirochaetes for subsequent analysis [95,96]. microscopy (2PM) are often employed [14,103–105].
By comparison of expression profiles of spirochaetes The spinning disk in the SDCM setup allows for
VIRULENCE 7

imaging of very fast dynamic processes in live speci­ [109]. CyaB ortholog can be found in other patho­
mens and reduces phototoxicity. With improved laser genic bacteria such as Pseudomonas aeruginosa or
penetration by low energy photons and significantly Vibrio vulnificus [110,111]. Cyclic dimeric guanosine
reduced phototoxicity, 2 PM is very convenient for monophosphate (c-di-GMP) is an ubiquitous second
real-time intravital imaging of Borrelia in the mouse messenger found in many prokaryotes such as
dermis [104,105]. Using SDCM in an intravital setting, Mycobacterium tuberculosis, Vibrio cholerae, or
the details of vascular extravasation of B. burgdorferi Salmonella enterica that regulates a wide span of
were visualized, showing that the bacterium engages in physiological processes including bacterial protein
multi-stage interactions with the endothelial cells, secretion, multicellular behaviour, virulence, motility,
including tethering and dragging interactions, and sta­ and cell development and differentiation [112,113].
tionary adhesion to the vasculature [14]. Intravital ima­ Although c-di-GMP has various downstream effec­
ging is typically accomplished by using fluorescent tors in bacteria [114], PlzA protein is the only uni­
labelling of the pathogen, preferably by expression of versal c-di-GMP binding protein in B. burgdorferi.
a fluorescent fusion protein such as GFP. To avoid the The regulatory processes of c-di-GMP are mediated
potential issues with bulkiness of the GFP, new small through binding with the PilZ domain, the
fluorescent probes for investigation of spirochaete mor­ C-terminal part of PlzA [115]. In B. burgdorferi, c-di-
phology and motility are being explored [106]. GMP regulates the adaptation processes to the tick
Light and electron microscopy are usually used to and mammalian environment via the interaction with
provide a cellular-level understanding of mechanisms PlzA [116]. PilZ domains are found in multiple bac­
of Borrelia infection (spirochaete-host cell interac­ terial taxa such as actinobacteria, proteobacteria, or
tions) [22,107]. To address the structure-functional spirochaetes [112].
contributions of borrelial proteins to bacterial viru­ LuxS is another virulence factor found in Borrelia
lence and disease pathogenesis, modern high- [117] as well as many other pathogens such as
resolution imaging modalities can be utilized. Atomic Bacillus anthracis [118], Staphylococcus epidermidis
force microscopy-based techniques enable the study of [119], or Haemophilus parasuis [120]. In addition to
protein–protein interactions at the single-molecule the mostly referred genes, such as ospA, ospB, ospC,
level, for instance, in uncovering the important inter­ crasp(s), erp(s), vls, dbpA/B, flaB, p66, rpo(s), the
actions that facilitate the spirochaete to efficiently group of genes involved in sensing and transduction
translocate in the extracellular matrix of a host [13]. of environmental signals deserves more attention. To
Nuclear magnetic resonance-based techniques can go differentially synthesize proteins during the infectious
even deeper with resolution power and determine the cycle, Borrelia is equipped with regulatory networks
location of the binding site and the dynamic structural to sense its environmental signals. Quorum sensing is
rearrangements that occur upon binding, with amino a mechanism by which many prokaryotes monitor
acid precision [102,108]. their surroundings by producing and responding to
signalling molecules known as autoinducers [121].
The genome of B. burgdorferi encodes for a number
Borrelia-specific versus common virulence
of enzymes involved in quorum sensing, such as
factors
MetK, LuxS, and Pfs [15] that enables the spirochaete
Virulence factors can be classified as pathogen- to synthesize an autoinducer type 2 (AI-2) mediating
specific (no orthologous protein in other bacteria) in this way quorum sensing that can function in both
and common (one or more orthologous proteins in the mammalian host and the tick vector. Therefore, it
different pathogenic bacteria) based on whether the is possible to regulate the differential expression of
virulence determinant is present in a single species or Borrelia genes in different environments [117,122].
it is found in multiple different organisms. The bacterium has to interact with different tissues
B. burgdorferi contains a high number of borrelia- during its life cycle, derive nutrition mostly from
specific virulence factors such as OspC, OspA, VlsE warm-blooded mammalian hosts as well as from the
but also contains several virulence determinants that tick vector, and avoid clearance by host and vector
can be found in other microorganisms. For instance, immune responses. By exploiting quorum sensing,
B. burgdorferi utilizes non-toxin adenylate cyclase a whole borrelial population can synchronize produc­
cyaB, an enzyme that produces secondary messenger tion of molecules required for infection and survival.
cyclic adenosine monophosphate (cAMP), to modify There are additional advantages in having the whole
borrelial gene expression and protein production in population of Borrelia to coordinate particular func­
order to enhance its virulence in the vertebrate host tions. For example, simultaneous transmission of
8 M. STRNAD ET AL.

large numbers of spirochaetes from a tick to a warm- Spirochaetes reside in the midgut during tick starvation
blooded host might improve the odds that at least phases as well as tick moults. Upon blood feeding,
some spirochaetes survive the host’s immune Borrelia escape from the gut to navigate toward the
responses and establish disseminated infection [122]. salivary glands and onwards into a new host [134].
The quorum sensing mediated by LuxS is considered An alternative transmission pathway, in which the
as the main system used for cell-to-cell communica­ tick salivary glands are considered unessential for suc­
tion. Although an earlier study suggested that luxS is cessful transmission, might be exploited by some of the
redundant for B. burgdorferi tick colonization, trans­ major European species. In the study by Pospisilova
mission to a mammal, or establishment of infection et al. [135], the authors show that B. afzelii did not
[123], a more recent study demonstrated that the reach the tick salivary glands at any stage of feeding and
luxS mutant was markedly less infectious than the that tick saliva is redundant for infectivity of this parti­
wild type and that luxS gives a fundamental advan­ cular species. The possible differences in the transmis­
tage to the spirochaete during vertebrate infec­ sion mechanisms in B. burgdorferi genospecies were
tion [124]. further strengthened by a study demonstrating no pre­
sence of B. afzelii in the salivary glands of unfed ticks
[136]. On the contrary, in the same study, other
Tick transmission factors of B. burgdorferi
European species B. lusitaniae, B. spielmanii and
B. burgdorferi naturally persists in an enzootic cycle B. garinii were detected in both midgut and salivary
that primarily involves Ixodes ticks and mammals. glands suggesting that the migration of these spiro­
Adult and nymphal ticks are the most important stages chaetes between midgut and salivary glands might not
in the transmission of the bacterium. B. burgdorferi is be activated by the bloodmeal [136].
not propagated from the adult female tick to their off­
spring, or it happens only rarely [125,126]. The transo­
varial transfer of B. burgdorferi has been occasionally
Protein interactions between LD Borrelia and ticks
documented in the literature, but these records may
probably stem from confusion between LD species Borrelia adapts to the transition between the tick vector
and relapsing fever (RF) Borrelia [44]. Nymphs are and vertebrate host by preferential gene expression. To
the most harmful tick stage for humans, because the optimize colonization and persistence in the tick mid­
risk of an infection in the majority of cases comes from gut, Borrelia has evolved complex strategies, how to
the bite of this developmental stage of the tick vector. exploit the tick midgut milieu (Figure 2). The successful
Tick larvae are very rarely infected and the adult ticks tick colonization depends on and is governed by com­
are large enough to be noticed early and removed plex molecular interplay of spirochaetes with the gut
appropriately. Ixodes ticks take only one bloodmeal epithelium receptors. The temporal pattern of ospA
per life stage (larva, nymph, adult) after hatching (outer surface protein A) gene expression together
from eggs. At least seven tick-borne zoonoses are trans­ with a number of functional studies [137,138] indicate
mitted by members of the Ixodes complex in the north­ that the abundantly expressed OspA plays a vital role in
ern hemisphere, and include diseases such as LD, spirochaete persistence in the vector. OspA-deficient
ehrlichiosis, babesiosis, anaplasmosis, tick-borne ence­ spirochaetes are unable to colonize the tick since they
phalitis, Rocky Mountain spotted fever, Powassan ence­ fail to bind to the tick gut [137]. The impaired attach­
phalitis disease, etc. ment of B. burgdorferi to the tick gut effectively
B. burgdorferi species are host-propagated pathogens obstructs all subsequent downstream transmission
that shuttle between tick vectors and vertebrate hosts. events. Tick receptor for OspA (TROSPA), located on
B. burgdorferi in the unfed ticks is predominantly the luminal surface of gut epithelium, interacts with the
located in the midgut lumen, where the spirochaetes spirochaete OspA lipoprotein and facilitates the tick
have to avoid the tick innate immune responses, man­ colonization. TROSPA interacts specifically with OspA
ifested especially in the form of antimicrobial peptides and was shown to be upregulated when spirochaetes
(AMPs) [127,128]. Further down the road primarily in are ingested [139]. RNA interference of TROSPA pre­
the tick haemocoel, the bacteria have to avoid the vents efficient colonization of the tick and reduces
haemocyte-mediated processes (phagocytosis, encapsu­ borrelia transmission to the host [139]. Similarly, bor­
lation and nodulation) as well as damage mediated by relial outer surface lipoprotein BBE31 binds to the
AMPs, lysozyme, complement-like factors, and reactive vector molecule TRE31 located in the tick gut [140].
oxygen and nitrogen species [129–132]. Borrelia multi­ Borrelia infection induces TRE31 expression and silen­
ply in the midgut to many thousands of cells [133]. cing of TRE31 results in a significant decrease of the
VIRULENCE 9

Figure 2. Molecular interactions at the tick midgut luminal interface involved in transmission and acquisition/persistence
of B. burgdorferi in ticks. When an infected tick starts feeding on the host, spirochaetes present in the tick midgut multiply,
traverse the gut barrier to reach the haemocoel (1), navigate toward the tick salivary glands (2), and ultimately infect the host (3). (a)
upon blood intake, the spirochete changes its protein coat to produce molecules such as OspC and BBE31, which enable the
pathogen to escape from gut and invade the tick salivary glands. Tick epithelial cells express molecules Ixofin3D and TRE31, which
facilitate the process of gut penetration. (b) Borrelia cells ingested in the bloodmeal bind to the tick gut and stay there until a next
tick feeding. B. burgdorferi expresses several-exposed lipoproteins such as OspA, OspB and BptA, which protect spirochaetes from
bactericidal components present in the host blood and enable them to colonize the tick gut. OspA interacts specifically with the tick
receptor TROSPA, thereby enabling efficient colonization of the vector.

borrelial loads in the tick haemolymph and salivary was shown to be reduced [145]. Using a yeast surface
glands [140]. display library, Ixofin3D and I. scapularis dystroglycan-
Outer surface protein B (OspB) is another surface- like protein (ISDLP) were identified as other candidate
exposed lipoprotein essential for adherence, survival, and tick gut binding partners of B. burgdorferi [146,147]. The
persistent infection of the tick midgut [137]. OspB- expression of both Ixofin3D and ISDLP is increased in
deficient microbes are capable of migrating to the feeding Borrelia-infected tick gut during bloodmeal ingestion.
ticks together with the bloodmeal but their ability to Ixofin3D was suggested to facilitate borrelial congregation
adhere to the tick gut and survive within the arthropod to the tick gut that ultimately helps Borrelia to navigate
vector is significantly diminished [141]. While the binding and escape from the gut [146].
partners for BBE31 and OspA are known, the mechanism The migration of the spirochaetes toward the tick
how OspB exerts its function is still unclear. Studies have salivary glands is accompanied by downregulation of
also identified borrelial proteins required for persistence tick gut specific borrelial proteins such as OspA and
inside the tick, which are critical for survival during long concomitant upregulation of proteins such as OspC
off-host periods between bloodmeals. BptA (bbe16), that helps Borrelia to reach the tick salivary glands
a lp25-encoded surface-exposed lipoprotein, was found [148] and infect and colonize the host [149]. The
to be essential for this persistence of Borrelia in ticks but I. scapularis salivary protein Salp15 binds specifically
the molecular mechanisms by which BptA promotes the to OspC, thereby enabling the bacterium to avoid clear­
persistence remains to be elucidated [142]. DksA is ance mediated by host anitbodies [150]. The same
involved in the transcriptional response to nutrient lim­ function was demonstrated for the homologue of
itation and supports the transcription and translation of Salp15 from I. ricinus Iric1. In addition, Iric2 and
genes contributing to B. burgdorferi infectivity such as of Iric3 from I. ricinus are capable of OspC binding with
virulence-associated lipoprotein OspC [143,144]. Another high affinity [151]. Although Borrelia evolved its own
potentially important factor in the tick vector stage of mechanisms to combat immune defence of the host,
B. burgdorferi is an outer membrane protein BBA52. the transmission via tick saliva gives spirochaetes cer­
Tick acquisition of mutants lacking functional bba52 tain adaptive benefits. Salivary gland products also
10 M. STRNAD ET AL.

provide the first line of defence for Borrelia against the antioxidant protein Salp25D also play a fundamental
host innate immune system. Tick salivary proteins play role in the acquisition of B. burgdorferi [154] (Figure 3).
a role in the inhibition of the host immune and homo­
eostatic responses that are raised against them. This
includes the suppression of activation of phagocytic Pathogenic mechanisms at the host–pathogen
dendritic cells and macrophages, including the inhibi­ interface
tion of natural killer cells that are responsible for che­
mokine and cytokine production, and disabling of Genome sequencing revealed that B. burgdorferi lacks
granulocyte recruitment to the tick bite site [152]. virulence factors common to many microbial and bac­
When an uninfected tick feeds on an infected host, terial pathogens, such as specialized secretion systems,
the spirochaetes are directed toward the tick bite site by toxins, or extracellular proteases with the exception of
specific environmental and chemotactic stimuli. Tick HtrA [15,155]. As a notorious extracellular pathogen,
protein Salp12 is released into the skin tissue as the spirochaete most likely does not damage the host
a component of the tick saliva, where it is sensed by cells by penetrating them or multiplying in them.
resident Borrelia [153]. This recognition contributes to Neither does Borrelia produce toxins that would
attraction and relocation of the spirochaetes into the impose direct damage to the host cells membranes,
tick feeding pit. Salp12 is expressed and present in the kill the host immune cells, or inhibit protein synthesis.
salivary glands as well as midgut. It was shown that The outer membrane of B. burgdorferi is structurally
during tick feeding, the expression of Salp12 in midgut distinctive among bacterial cell envelopes and it is
of the tick was significantly higher than Salp12 expres­ a common target of host immunity [156]. Unlike typi­
sion in the salivary glands at various investigated time cal diderms, B. burgdorferi does not produce the endo­
points, possibly creating a concentration gradient toxin lipopolysaccharides but secrete extensive surface
which directs the bacteria toward the tick midgut. lipoproteins at the host–pathogen interface [15]. These
Passive immunization and disruption of Salp12 in the outer membrane lipoproteins play an essential in the
tick salivary glands were shown to decrease acquisition borrelial virulence by suppressing immune responses of
of the spirochaetes by the vector [153]. Using similar the host and by mediating interactions with the targets
experimental procedure, it was shown that the tick tissues [157]. Despite the activation of both the innate

Figure 3. Molecular factors involved in transmission and acquisition of B. burgdorferi by ticks. numerous molecules produced
by both a tick vector and Borrelia are required for successful early-stage infection of the host and tick acquisition of B. burgdorferi.
Many tick proteins (Salp15, evasins, TSLPI, tHRF, etc.) and borrelial proteins (e.g. OspC, DbpA, DbpB, BBK32) are known to be
necessary for transmission. Chemotactic tick protein Salp12 and antioxidant Salp25D, together with borrelial BBA52, were shown to
be critical for host-to-tick acquisition of Borrelia.
VIRULENCE 11

and adaptive immune systems, Borrelia is often able to (FHL-1) appears to depend on the Borrelia genotype
survive in the hostile environment to cause infection, [170, 171]. B. burgdorferi species vary in their suscept­
which then can become persistent. LD has not been ibility to complement. B. afzelii and B. burgdorferi ss
associated with severe inflammatory over-reaction such are strongly serum (complement) resistant while
as cytokine storms known from cytomegalovirus, B. garinii is more sensitive to most mammalian
Epstein-Barr virus, streptococcus, and many other serum [172,173]. The spirochaete has evolved mechan­
microbes. Borrelia is better known for its immune- isms to evade destruction from the complement sys­
evasion mechanisms, expression of immunomodulatory tem by producing a set of outer surface lipoproteins
surface proteins, downregulation of immunogenic pro­ that interact with host complement molecules and
teins, and antigenic variation, which help to elude manipulate the native activities of the defence system
immune responses of the host [158]. [174]. Borrelia can either bind directly to complement
components or the spirochaete can interact with the
host-derived regulators of the complement system in
Evasion of host innate immunity order to attenuate complement activation.
Direct interference is mediated by a spectrum of
The mammalian host immune response to infection with outer surface lipoproteins such as BBK32, OspC, and
LD spirochaetes begins with effector components of the BBA70. BBK32 specifically inhibits the classical path­
innate immune system. Nitric oxide (NO), generated in way through high-affinity binding of the C1r subcom­
multiple biologic tissues and cells by specific nitric oxide ponent of C1, entrapping C1 in its zymogen form
synthases, is a known player in innate immunity [175]. OspC was shown to bind C4b in both
responses against bacterial pathogens [159]. Although B. garinii and B. burgdorferi ss, blocking formation
NO is a powerful antimicrobial agent and its production of C3 convertase and thereby restricting killing via
is elevated during borrelial infection [160], B. burgdorferi the classical and lectin pathways [176]. OspC, together
is not significantly affected by NO during mouse infection with other surface molecules such as OspA, CspA,
[161]. In contrary to elevated levels of NO during infec­ Erp-family proteins, and BBA70, are capable of bind­
tion, the spirochaete is able to suppress the production of ing plasminogen, which is a known complement inhi­
additional common toxic molecules such as reactive oxy­ bitor [177–182]. Indirect strategies include the
gen species (ROS) via the mTOR pathway in order to production of complement-regulator acquiring surface
avoid killing [162,163]. B. burgdorferi produces multiple proteins (CRASPs) which encompass CspA, CspZ, and
proteins which were implicated to ameliorate the effects OspE paralogs to inhibit the complement responses.
of ROS exposure, including BmtA, SodA [164,165]. These molecules attenuate complement activation on
Recently, a gene cluster bb_0554-bb_0556 (xdhACB) the borrelial surface by binding to complement regu­
encoding a xanthine dehygrogenase and transmembrane lator FH or FH-related protein 1 (FHR-1) [183,184].
proteins BB0017, BB0164, and BB0202 were shown to CspA (CRASP-1) attenuates complement on two cen­
play a significant role in ROS resistance [166,167]. In tral activation levels, C3b generation and assembly of
addition, the capacity of lysozyme, an antimicrobial the terminal complement membrane attack complex
enzyme and a major constituent of innate immunity, to [178,185]. European species B. bavariensis produces
kill B. burgdorferi is quite limited [168]. surface proteins BGA66 and BGA71 similar to CspA.
Both molecules bind complement components C7, C8
and C9, and prevent assembly of the terminal comple­
Complement evasion by LD Borrelia ment complex [186]. CspZ (or CRASP-2), a second
The complement system is considered a functional FH/FHL-1-binding protein, also downregulates the
bridge between innate and adaptive immune responses formation of C3 and C5 convertases on the spiro­
and is known to be activated via three pathways – the chaete surface. A third type of FH-binding proteins
classical pathway, lectin pathway, and alternative path­ are OspE paralogs ErpP/ErpC/ErpA (also known as
way. Upon activation by the pathogen, the comple­ CRASP-3, CRASP-4 and CRASP-5). The outer surface
ment system opsonizes bacterial surfaces and induce protein OspE binds to Factor H as well as various
a series of proinflammatory responses that ultimately members of complement factor H-related (CFHR)
lead to bacteriolysis [169]. Vertebrate host range for proteins [187]. ErpA and ErpP, the OspE-related pro­
Borrelia is determined by spirochaetes sensitivity to teins, bind the complement factor H-related proteins
complement of particular animal species; the ability CFHR1, CFHR2, and CFHR5, while ErpC binds only
to bind factor H (FH) and factor-H-like protein 1 CFHR1 and CFHR2 [174,188].
12 M. STRNAD ET AL.

Antigenic diversity and variation in LD Borrelia suggested to play a role in host selectivity [192], plas­
minogen binding in hosts [200], defining Borrelia inva­
After activation of adaptive immunity, antigen present­
siveness in rodents [200], dissemination during
ing cells utilize bacterial peptides and present them to
mammalian infection [201], salivary gland migration
B and T cells. T cells release cytokines and stimulate
in the tick [148], evasion of innate immunity [202],
macrophage activation while B cells produce specific
binding a tick salivary protein that inhibits complement
antibodies that bind borrelial outer membrane epitopes.
[150,203], conferring bloodstream survival [204], and
The escape from adaptive immune responses is
contributing to Borrelia strain-specific differences in
mediated primarly by the VlsE antigenic variation sys­ tissue tropism [205]. OspC suppresses the classical
tem [189]. Antigenic diversity can fundamentally and lectin complement pathways and competes with
extend the time a pathogen maintains an infection complement protein C2 for C4b binding [176]. OspC
within a host and avoids eradication by the host is important for Borrelia in macrophage phagocytosis,
immune system. The initial set of surface-presented reducing the uptake of the bacterium by human and
antigenic molecules stimulate an immune response murine macrophages [206].
against the dominant antigens. If the pathogen changes Decorin binding proteins A and B (DbpA/B), that
the composition of its antigenic coat to new variants, are important for motility, bind primarily to the con­
the microbe escapes the immune response and con­ nective tissue proteoglycan decorin and facilitate host
tinues infection until the host generates a new response colonization by the spirochaete [13]. Although not
against the latest variants [190]. LD Borrelia species are absolutely essential for infection, the important role of
well known for differential gene expression and altera­ these adhesins for the overall virulence of B. burgdorferi
tions in antigenic structure during their life cycle. was demonstrated in a number of studies [67,207,208].
A prototypical example is downregulation of OspA DbpA and DbpB deletion mutants display marked
lipoprotein during transmission while the expression attenuation in mammals, but particularly early during
of OspC is upregulated. Subsequently, OspC, which is the course of infection [209,210]. Recovery of mutant
critical for early stage of mammalian infection is down­ bacteria from tissues distant to the inoculation site is
regulated and Borrelia produces genetic variants at the diminished as well [208,210]. Besides binding to speci­
vls (variable membrane protein-like sequences) locus to fic glycosaminoglycan (GAG) ligands in order to
enable long-term infection of the host [149,191]. anchor the spirochaete in the destination niches,
OspC is a dominant highly polymorphic antigen of DbpA and DbpB help in host colonization by enhan­
LD bacteria, that is heavily targeted by the host cing borrelial translational motility in the low shear
immune system. Borrelia ospC is more diverse than stress environments [13]. DbpA sequence variability
any other studied gene to date; it is known to be able among different species of B. burgdorferi is high, result­
to establish the secondary immune response or ing in pronounced differences in their GAG affinities
immune memory in hosts [192], association between [6,211,212]. It was shown that allelic variations of
ospC genotypes and invasiveness in human patients and B. burgdorferi DbpA affect tissue tropism and disease
infected animals have been reported in multiple studies manifestation of different LD genospecies [213].
[193–195]. The gene encoding for OspC is mapped to Besides temporal changes in the antigenic coat as
cp26, a 26-kb circular plasmid that is a stable compo­ a result of tick vector-host circulation and high
nent of the segmented B. burgdorferi genome [196]. sequence variability between borrelial genospecies,
OspC is transiently but absolutely required during the Borrelia are capable of in situ antigenic variation to
early stage of infection and neither vlsE nor ospA can avoid detection from host adaptive immune response.
compensate for the absence of OspC and restore infec­ Recombinational switching of a gene locus, or the pro­
tivity to an ospC mutant [191]. OspC is a protective cess of alteration of the pathogen’s surface antigens in
antigen [197], due to its high sequence variability, pro­ order to avoid detection from host adaptive immune
tection is generally strain specific [198]. Multiple alleles response, is one of the most effective attributes of
are circulating among reservoir hosts and tick vectors immune evasion by human pathogens. Concomitant
[192]. OspC alleles A, B, and L were detected in Europe with the development of the host acquired immune
and North America in vectors and hosts including response and OspC downregulation, robust synthesis
humans. Six ospC alleles are prevalent in Europe and of VlsE is initiated. In B. burgdorferi strain B31, the vlsE
four of them were detected in human samples. Ten gene is encoded on the 28-kb linear plasmid, lp28–1,
ospC alleles were identified in the western United less than 100 bp from the right telomere. VlsE is dis­
States. Four ospC alleles were abundant in the south­ similar to OspC, OspA, or Dbps, as it is abundantly
eastern United States [199]. OspC has also been present on the spirochaetes surface during persistent
VIRULENCE 13

infection [214, 215]. The silent cassettes at the vls locus mechanical stress that B. burgdorferi needs to over­
vary during the course of infection as well as between come in order to infect the target tissues. During the
species and within species [216–218]. The VlsE anti­ vascular extravasation and after reaching the extra­
genic variation system contains the vlsE expression site cellular space, the bacterium faces a milieu of much
and 15 silent cassettes, thus providing multiple possible lower shear stress than is experienced in the vascu­
recombination events for producing a variety of VlsE lature. Here, however, the pathogen needs to able to
epitopes through unidirectional, segmental gene con­ penetrate through dense and highly viscous struc­
version [189,219]. The vlsE cassette region can have tures with pore sizes much smaller than the diameter
over 90% nucleotide sequence identity with the vls of borrelial cell. This is allowed by forming transi­
cassettes. European species B. garinii and B. afzelii ently stable interactions that enable pushing against
were shown to have less vls silent cassettes than the surrounding structures [104]. It was shown that
B. burgdorferi B31. B. garinii strain Ip90 carries 11 vls DbpA and DbpB are critical borrelial surface mole­
silent cassettes and B. afzelii strain ACAI encodes 14 vls cules that allow for efficient dissemination of Borrelia
silent cassettes [220]. The cassettes can recombine in in low shear stress and dense environments such as
seemingly random manner (although there is some the extracellular matrix [13].
preference for certain silent cassettes), ranging from In contrast to pathogens that circulate only pas­
single nucleotide substitutions to almost full replace­ sively in the vasculature of a host, B. burgdorferi can
ment of the vlsE site, with switch rate of approximately actively influence its dissemination in the host in
0.033 per cell generation [221]. The RuvAB helicase order to infect various organs and tissues. Multiple
complex apparently facilitates the vlsE recombination studies have shown that motility of B. burgdorferi is
[222]. There are two types of sequence changes; tem­ a critical virulence determinant and absolutely vital
plated and non-templated. The templated changes are for pathogenesis throughout the enzootic cycle and
those which correspond to sequences in the vls silent the loss of motility leads to a non-infectious or
cassettes, whereas non-templated changes are often attenuated phenotype [228–230]. The bacterium is
represented by point mutations. The degree of recom­ able to adopt distinct motility states and transition
bination events and switch length of nucleotides are between them as the pathogen colonizes the host
influenced by the immune pressure. The spirochaetes [104]. The unique flat wave rotational movement of
in wild-type immunocompetent mice display longer Borrelia is generated by 7–11 flagella located beneath
stretches of recombination and average switching rate the outer membrane in the periplasmic space. Since
than in immunodeficient mice [221]. The antigenic borrelial flagella do not extend outwards from the
variability of VlsE facilitates another novel escape bacterial surface, B. burgdorferi is able to migrate
mechanism from innate immunity described as epitope with ease in highly viscous media, in which most
shielding [223]. The VlsE protein can effectively block other bacteria are significantly decelerated [231].
the binding of antibodies which target immunogenic Therefore, the spirochaetes like to hide in the extra­
borrelial proteins such as arthritis-related protein Arp. cellular structures, making them less subject to cir­
culating leukocytes. Borrelia with a mutation in the
major flagellin gene flaB that did not synthesize
Motility-driven pathogenesis
flagella, was shown to be non-motile and rod-
B. burgdorferi possesses surface adhesins that are well shaped [232]. As such, the flagella are arguably one
adapted to aid in its dissemination and colonization of, if not the most critical virulence factors in the LD
strategies (Figure 4). During its infectious cycle, infection. Additionally, B. burgdorferi possesses an
B. burgdorferi needs to cope with high blood flow- unusual peptidoglycan cell wall which significantly
mediated shear stress in the host vasculature. BBK32 contributes to structural and morphological integrity
and P66 borrelial proteins are key players mediating of the bacterium and allows the spirochaete to with­
the stabilizing interactions and the adhesion to the stand the high torque generated by the periplasmic
cells lining the vascular lumen [103,224,225]. flagella during borrelial translocation [233]. BpiP
Recently, it was suggested that VlsE also promotes protein aids in sustaining the cellular integrity cell
transient binding to the vasculature under flow via by binding the peptidoglycan wall and it influences
binding to dermatan sulphate and that a complex the borrelial virulence in the host [234]. Notably,
temporal choreography of P66, DbpA/B and OspC peptidoglycan was shown to be a persistent immuno­
is required for the escape process from postcapillary gen contributing to inflammation during acute infec­
venules [226,227]. Blood circulation is not the only tion and post-Lyme arthritis [46].
14 M. STRNAD ET AL.

Figure 4. Molecular factors directly involved in active migration and dissemination of Borrelia in humans. (a) B. burgdorferi
has to overcome high shear forces generated by blood flow in order to adhere to vascular surfaces for extravasation. The interactions
of borrelial protein BBK32 with vascular fibronectin allow active migration in high shear stress environment and borrelial proteins
P66 and VlsE aid in vascular transmigration. (b) in low shear stress niches such as extracellular matrix of target skin tissue, borrelial
surface-exposed proteins DbpA and DbpB facilitate the translational motion by interactions with matrix molecules such as decorin.

Localization into specific niches in extracellular extravasation, the spirochaetes colonize the ECM of
matrix multiple tissues and organs, which contain molecules
that can be bound by Borrelia. These are commonly
In contrast to RF Borrelia, LD Borrelia are rapidly
proteoglycans, biomolecules composed of a core pro­
cleared from the blood circulation and cannot multiply
tein together with long and negatively charged polysac­
there [235]. When the adaptive immune responses of
charide chains called glycosaminoglycans (GAGs). For
the host are fully activated, B. burgdorferi disappears
example in joints, ECM of the articular cartilage con­
from the bloodstream and disseminates to the organs,
tains a variety of GAGs such as chondroitin sulphates,
collagenous tissues, joints, and synovial fluid of the host
[236]. The spirochaetes need to survive until they are keratan sulphate, dermatan sulphate as well as multiple
transmitted back to competent ticks. At this time the collagen types [239,240]. LD Borrelia bind to different
successful persistence of the spirochaete within the host proteoglycans, which promotes tissue colonization and
depends on evading the host’s immune system, rather bacterial attachment to the cells [211,213,241,242]. The
than exploiting the host tissues for reproduction or binding interactions with the host ECM components
growth [237]. The extracellular matrix provides an are mediated by outer surface molecules of Borrelia
immune-privileged milieu for the pathogen [238]. The (Table 1). B. burgdorferi displays a dizzying diversity
generally accepted theory is, after vascular of abundant surface lipoproteins. At least 120 proteins
VIRULENCE 15

Table 1. Surface proteins of B. burgdorferi which are known to mediate attachment to the
extracellular matrix (ECM) components of the vertebrate host.
ECM component Protein Reference
Collagen CspA, BBA33 [243, 244]
Decorin DbpA, DbpB [245, 246]
Fibronectin BBK32, RevA, RevB, BB0347, OspC [205, 242, 247–249]
Integrins BBK32, P66, BB0172, BBB07 [250–253]
Laminin BmpA, ErpX, BB0406, CspA [,256]
Glycosaminoglycans DbpA, DbpB, BBK32, Bgp, OspC, Lmp1, VlsE [205, 226, 257–260]

that possess a lipid moiety are encoded in the borrelial persistence. Interestingly, the discussed novel virulence
genome [261], which represent nearly 8% of all borre­ mechanisms bear almost exclusively on the persistent
lial open reading frames [262]. Out of 125 lipoproteins, stage of the disease.
86 of these were shown to be secreted to the outer
surface [261]. Structural variants of B. burgdorferi
DbpA and DbpB exhibit binding activity toward
various components of the ECM, including the proteins Whereas no significant debate surrounds the issue of
decorin, biglycan and various GAG molecules such as B. burgdorferi dissemination pathways within the host,
dermatan sulphate or heparin [108,211,241,245,263]. the strategy for persistence inside the host is a different
Fibronectin-binding activity was demonstrated for at story. Under optimal conditions, B. burgdorferi has the
least four outer membrane proteins, BBK32, RevA, typical flat wave morphology. The bacterium adopts the
RevB, and BB0347 [242,247–249]. BBK32 mutants dis­ spiral form during multiple phases of its enzootic
play a significant defect in infectivity in the mouse cycle – migration from the feeding tick midgut to the
infection studies and highlight the fundamental role haemocoel and salivary glands, deposition in the skin,
of the protein in B. burgdorferi pathogenesis haematogenous dissemination, adhesion to and trans­
[248,264,265]. Similarly to BBK32, the lack of RevA migration through the endothelium, and establishment
significantly affects the pathogenesis in the mouse of infection in distal niches, as it was evidenced by live
model of LD, although revA is not absolutely essential and intravital imaging [12,14,105,267].
for infection [266]. The role of RevB and BB0347 in In the face of the hostile microenvironment conditions,
pathogenesis remains to be elucidated. BmpA, ErpX the situation around the morphology of Borrelia becomes
and BB0406 were shown to interact with laminin less clear. It was shown in vitro that B. burgdorferi can alter
[254–256]. B. burgdorferi produces also collagen- its cell shape into a different pleomorphic form under
binding proteins such as CspA and BBA33 [243,244] physiological stress, immune pressure, or antibiotic treat­
and it is able to bind several types of host integrins such ment [17,19,268–271], even inside tick midgut [272]. The
as α3β1 integrin using P66, BB0172, and BBB07 pro­ morphologic variants, usually spherical and non-motile
teins [250–253]. forms, of the B. burgdorferi are known under various
terms in the literature. The terms such as L-forms, cystic
forms, spheroplasts, or round bodies are commonly used. It
was shown that the round bodies retain their flagella in the
Alternative pathogenic mechanisms
periplasmic space, have an intact and flexible cell envelope,
With the rise of modern research technologies and and are able to dynamically transition back into the motile
inflow of new studies, novel virulence mechanisms spirochaete under favourable conditions in vitro [17,271].
that might be exploited by the LD pathogen have In addition, it was determined that the round bodies display
come to light. These novel virulence activities are still lower metabolic activity and present different protein pro­
either not fully accepted by the expert borrelial com­ files and antigenicity than the spiral form [17,268].
munity or have not been thoroughly investigated yet. The morphologic variants are often suggested to be
Confirmation of any of the purported virulence associated with chronic or persistent LD and PTLDS, as
mechanisms (Figure 5) would significantly expedite they may enhance survival in hostile environmental
our comprehension of B. burgdorferi infection, provid­ conditions [18]. Chronic LD describes the cluster of
ing valuable insights to leverage in the creation of novel symptoms that start after getting LD, persist despite
vaccines and other countermeasures. The pattern of the antibiotic therapy and that result into long-term seque­
disease can be resolved into several discrete stages – lae of infection. Although a systematic review of
host invasion, dissemination, colonization, and B. burgdorferi pleomorphs does not reveal a clear role
16 M. STRNAD ET AL.

Figure 5. Potential virulence mechanisms of B. burgdorferi. (a) B. burgdorferi can change its morphology as a response to
environmental stress. Pleomorphic forms of B. burgdorferi such as biofilm-like structures (or aggregates) and round bodies could
possibly help the spirochaetes to overcome prolonged stress conditions exerted for instance by antibiotics. (b) Although
B. burgdorferi is considered extracellular parasite, the spirochete is occasionally found inside the nonphagocytic cells. The
intracellular niche might help them to hide from immune responses. (c) outer membrane vesicles produced by the bacterium
may modulate host immune responses. (d) structural transformation in cell shape and actin cytoskeleton of human cells were shown
upon contact with Borrelia. (e) B. burgdorferi relies on uptake of essential nutrients such as amino acids, fatty acids and nucleosides
from its host environments for survival and infection. Nutritional virulence might constitute important virulence factors for B.
burgdorferi.

in chronic LD [273], it was demonstrated that LD Taken together, it is evident that the spiral, highly
patients might have more intense responses to borrelial motile form is absolutely required for borrelial dis­
circular forms in comparison to spiral forms of semination and colonization of the tick vector and
Borrelia. These results suggest that round bodies the vertebrate host. We know that the motility states
might play a certain role in LD pathogenesis [268]. are dynamic as the spirochaetes were observed to
Using an ex-vivo murine skin model, it was observed transition between them in the dermis of the host
that Borrelia can also form biofilm-like colonies made [104] and the midgut of the tick [12]. It is also
by spiral-shaped bacteria [274]. Biofilm represents an apparent that B. burgdorferi is exposed to many hos­
alternative lifestyle in which the microbes grow as tile environments and antimicrobial molecules dur­
structrured aggregates and adopt a multicellular beha­ ing the pathogenic cycle and that the importance of
viour that facilitates their survival under unfavourable morphologic variants for survival in hostile condi­
conditions. The bacteria are typically held together by tions should be considered in future research.
self-produced polymer matrixes. The presence of algi­ Further studies need to be done to assess the poten­
nate-like polysaccharide was revealed in a study that tial contribution of morphologic and motility var­
explored the Borrelia aggregates in vitro [275]. iants to virulence of B. burgdorferi.
VIRULENCE 17

Intracellular niche metabolic pathways needed to produce its own nutri­


ents. Recently, it has been suggested that Borrelia is
The ability of B. burgdorferi to maintain themselves for
able to hijack the molecular machinery of host cells
extended periods of time in the vertebrate hosts is
to gain survival benefits [162,282], in a similar man­
critical for continuation of their enzootic cycle. The
ner as was evidenced in many other human patho­
spirochaete can persist in multiple tissue sites despite
gens [283–285]. Metabolism generates energy as well
strong immune pressure. Since B. burgdorferi is known
as fundamental building blocks for every vital aspect
to be an extracellular pathogen, the ECM is considered
of cell biology, including the formation of the cytos­
to be the ultimate protective niche for the spirochaete,
keleton and extracellular matrix. Since many pro­
where the organism is sequestered from the immune
ducts of the cellular machinery are structural
responses [238]. Nonetheless, one of the questions that
components of the cell and at the same time LD is
is still a matter of debate, is whether B. burgdorferi is
characterized by tissue transformations of the host, it
truly an obligate extracellular pathogen, or if the spir­
might be tempting to hypothesize that Borrelia
ochaete is capable of living, and possibly reproducing,
directly affects the structural and mechanical proper­
inside the host cells, when the conditions outside the
ties of the host cells. Significant upregulation of
cells are not favourable (for instance due to presence of
genes coding for decorin or collagen type I was
strong immunity or antibiotics). A number of studies
demonstrated in human fibroblasts upon cultivation
reported the intracellular localization of the bacteria
with B. burdorferi species [278]. Induced alterations
inside non-phagocytic cells, especially fibroblasts
in cell shape and actin cytoskeleton of human cells
[276–278], but also in synovial cells [279], endothelial
were also confirmed upon contact with Borrelia
cells [280], or glial and neuronal cells [281]. On the
[286]. The structural rearrangements of the host
other hand, a recent study was not able to confirm the
cells will likely be highly beneficial for the pathogen.
intracellular niche in the non-phagocytic mammalian
For instance, the skeletal changes can provide a way
cells using advanced correlative imaging approach, but
for Borrelia to escape phagocytosis. Specifically, it
they proved the propensity of the spirochaete for extra­
was shown that uptake of Borrelia by macrophages
cellular surface structures [107]. The stimuli that drives
is not a simple, one-directional process but rather
the pathogen to internalize into the non-phagocytic
a lengthy battle, where the back-and-forth movement
cells has yet to be determined.
of the spirochaete might create “tunnels” inside the
Studies on B. burgdorferi infected murine fibroblasts
macrophage [22]. To what extent is the pathogenesis
has also suggested cyst-like Borrelia located inside these
caused by structural changes in the host organism is
cells to possibly aid in host immune evasion by har­
now a question worth pursuing as mechanical prop­
bouring the bacterium in an inactive state. When the
erties of the host cells might also be the critical
friendly environment is restored, the spirochaetes can
determinant of the pathogen virulence [287].
revert back to a motile spirochaetal form [270,271,276].
Based on the intimate interactions that Borrelia
Notably, coiling phagocytosis, the preferential phago­
form with the host cells, one can assume that the
cytic mechanism for B. burgdoferi, has been demon­
physical contact of Borrelia with the cell (probably
strated to occur less often with non-motile forms than
with some of the outer surface virulent molecules of
motile spirochaetes [268], suggesting that different
Borrelia) triggers the modulatory effects on cellular
receptors are present in the bacterial membrane of
metabolism, which ultimately results in changes of
spiral and non-spiral Borrelia. Moreover, intracellular
cellular architecture and nanomechanical properties
spirochaetes were observed in a variety of shapes, while
of the cell. In the literature, there are numerous studies
simultaneously avoiding lysosomal colocalization dur­
showing that cells affected by a disease exhibit differ­
ing the coculture [19]. The differences in antigenic
ent nanomechanical properties [288] and that cell
expression between borrelial pleomorphs plausibly
elasticity is a critical determinant of proper cell func­
explain the ability of the bacterium to adapt to different
tion [289]. Specifically in LD Borrelia, exposure of
milieus and survive in highly adverse environ­
endothelial cells to spirochaetes led to decreased moti­
ments [19].
lity and physical forces in host cell monolayers and
affected the mechanotransduction of the endothelial
cells [25]. An understanding of the structural rearran­
Borrelia-induced structural transformations of host
gements and mechanical responses of human cells to
cells
pathogen infection may shed light on the origin of
Borrelia is a pathogen that depends distinctly on its structure-related clinical manifestations such as skin
host to survive, since the spirochaete lacks many fibrosis, morphea, or hyperkeratosis.
18 M. STRNAD ET AL.

Secretion to extracellular space can also transfer lipids and glycolipids to host epithelial
cells. This lipid exchange could be an important process
The capacity of bacteria to secrete proteins, polysac­
leading to antigen presentation and immune recogni­
charides, and various macromolecular membrane-
tion that contributes to the pathogenesis of LD [302].
derived complexes beyond the bacterial cell surface
Nonetheless, the precise roles of OMVs in the patho­
is often essential in the understanding of microbial
genesis of Borrelia infections remain to be defined.
pathogenesis. Frequently, bacterial pathogenicity
depends fundamentally on the ability to secrete viru­
lence-associated molecules which are located on the
Nutritional virulence of LD Borrelia
bacterial outer surface, released into the extracellular
space or introduced directly into the host cell. Although LD spirochaetes are not known to significantly
Secretion systems have been thoroughly studied in affect the host by directly competing for scarce nutrients
a broad spectrum of Gram-negative species [290]. with the host cells, the term “nutritional virulence” has
Contrary to the other bacterial systems, secretion of been recently mentioned in connection with borrelial
soluble proteins into the surrounding environment virulence [21,303]. Like many other obligate parasites,
has been demonstrated rather sporadically in LD Borrelia species have significantly reduced their
Borrelia due to limited secretory capabilities of spir­ metabolic and biosynthetic capabilities. As
ochaetes [291]. a consequence, B. burgdorferi lacks genes encoding
Bacterial outer membrane vesicles (OMVs) have enzymes necessary for nucleic acid, amino acid, and
multifaceted roles as they can function as signal deliv­ fatty acid biosynthesis. The survival of the spirochaete
ery vehicles for proteins, toxins, and other virulence in nature is thus fully dependent on salvage of essential
factors, as well as genetic material and have the ability nutrients from their vertebrate hosts and tick vectors. B.
to modulate host immune responses [20,292]. OMVs burgdorferi requires uptake of numerous components,
are usually formed by two different mechanisms: by such as cholesterol and long chain saturated and unsa­
membrane blebbing or explosive cell lysis, with size turated fatty acids [302,304], purines [305], transition
between 10 and 300 nm and with a single membrane metals [164], and various carbon sources [306,307].
bilayer, which consist of almost the same outer mem­ Several lines of evidence indicate that factors involved
brane proteins as the membranes of the bacteria that in nutrient acquisition might constitute key virulence
the OMVs originated from [293,294]. Although the determinants for B. burgdorferi [8]. Borrelia utilizes var­
secretion of proteins to extracellular space is rare and ious scavenging strategies, obtaining macromolecules
largely uncharacterized in Borrelia, several studies have from the microenvironment and breaking them down
shown that LD spirochaetes are able to produce OMVs. to produce substrates for their own anabolic processes.
Borrelia OMVs, also known as blebs in the literature, Typically, the bacterium utilizes highly selective/unspe­
have been revealed to be released near the sites of cell cific porins (P66, P13, Oms28, BB0406) that open and
division [295], and shedding can be observed both in close in response to specific stimuli and allow passive
culture [20] or in vivo inside tick midguts mostly at the diffusion, or, active transport, which exploit the free
outset of tick feeding [12]. The blebs were shown to be energy from ATP hydrolysis to pump the solutes against
likely a transitional stage between the spiral and the concentration gradients [256,308,309].
round bodies form, with an expanded outer envelope Owing to its parasitic lifestyle, B. burgdorferi is
and with a folded protoplasmic cylinder inside [17]. restricted to utilizing only the nutrients that are avail­
OMVs from B. burgdorferi were demonstrated to be able in its microenvironment. It is probable that the
on average 33 nm in diameter and contained antigenic ability to exploit a variety of available carbohydrate
proteins OspA, OspC, p39, peptidoglycan, and neutro­ sources during the enzootic cycle is essential for the
phil attracting protein NapA [296,297]. Another survival of B. burgdorferi [307]. It has been reported
B. burgdorferi protein termed Oms28 was reported to that B. burgdorferi can utilize at least seven carbohy­
be secreted from borrelial cells into extracellular envir­ drates as the carbon source – glucose, mannose,
onment [298] and associated with borrelial OMVs N-acetylglucosamine, chitobiose, maltose, trehalose,
[299]. GAG-binding outer surface membrane protein and glycerol [310,311]. When multiple nutrient sources
Bgp has been shown to be secreted from the borrelial are available, the spirochaete likely takes advantage of
cell as well [298,300]. The spirochaetes produce OMVs the rapidly absorbable and high energy carbon sources,
that contain double stranded DNA [296,301], which typically glucose. During the off-host starvation periods
could presumably be a mechanism of horizontal gene in the tick, the spirochaetes were shown to use glycerol
transfer. Through the release of OMVs, B. burgdorferi as a carbohydrate source for glycolysis to maintain
VIRULENCE 19

maximal Borrelia fitness [306]. A common regulatory new ways to detect and combat the pathogen. In this
mechanism in bacteria that represses genes associated review, we have summarized the critical findings
with the utilization of secondary carbon sources has regarding the well-defined virulence factors of
been coined carbon catabolite repression (CCR) [312]. B. burgdorferi and potential novel virulence mechan­
There is no evidence of simple CCR in B. burgdorferi isms that are being investigated under the scope of
[15], rather the switch in carbon usage is orchestrated current research. Although much has been explored
by mechanisms including various regulatory ele­ about the genes and proteins that play roles in disease
ments [313]. development [8], the need for further extensive
B. burgdorferi is unable to synthesize fatty acids de research is obvious to completely characterize the dif­
novo, elongate fatty acid side chains or process exogenous ferent factors underlying the infection caused by
fatty acids via beta-oxidation [13]. Another noteworthy B. burgdorferi. The recent improvements in high-end
aspect is that B. burgdorferi differs from other bacteria and technologies, which allow for instance whole-genome
spirochaetes in that it mostly contains only two types of sequencing and enable much better resolution in
phospholipids in the outer and cytoplasmic membranes, microscopy and other structural biology techniques
phosphatidylglycerol and phosphatidylcholine [14]. In such as NMR, has directly benefitted the understanding
order to obtain exogenous choline and fatty acids, the of various aspects of LD, including borrelial phylogeny,
spirochaete utilizes lipases to break down the lipids of virulence mechanisms, subversion of immune system,
the host [314]. Using the phosphatidylcholine synthase or host association of different Borrelia species. In this
proteins, the spirochaete then synthesizes new lipids using regard, especially targeted mutagenesis and comple­
the building blocks acquired from the host [315]. The mentation are key methods for studying genes of
lipolytic activity of multiple borrelial enzymes was unknown function among bacterial pathogens includ­
shown to contribute to optimal infectivity of the bacter­ ing the LD spirochaete. In addition, genome sequen­
ium [21,314], supporting fatty acid salvage as a virulence cing of multiple B. burdorferi genospecies should
mechanism of B. burgdorferi. In addition, fatty acids are significantly accelerate the knowledge on this bacterial
not only fundamental components of borrelial membrane complex. Until now, most of the data about borrelial
bilayers, but these moieties are part of many virulence- virulence determinants come from genetic experiments
associated, surface-exposed lipoproteins, needed for opti­ performed on North American strains of a single spe­
mal infectivity during tick and host colonization. Osp-, cies, B. burgdorferi ss [9]. Recent whole-genome
Csp-, and Erp- lipoprotein families, DbpA/B, RevA/B, sequences of several European and US originated spe­
and VlsE belong among the well-known, surface lipopro­ cies/strains makes adaptation and use of genetic tech­
teins tightly associated with borrelial pathogenic potential niques feasible in studying inherent differences between
[261]. Palmitic acid is the most common fatty acid found them [16,319–321].
in the lipoproteins of B. burgdorferi [316]. The intercon­ The complexity of the clinical picture of LD requires
nection of glycerol metabolism and glycolysis with lipo­ a reliable diagnosis and a wide variety of pathologies. In
protein biosynthesis have been found to depend on case of untreated infection or absence of antibiotic ther­
a function of two dehydrogenases, glycerol-3-phosphate apy, disseminated spirochaetes can persist in the patient
dehydrogenase (GlpD) and glycerol-3-phosphate dehy­ for many months while evading immune responses.
drogenase (GpsA) [317]. GpsA is required for survival Antibiotic treatment at the early stage of the disease is
during nutrient stress and for mammalian infection [317]. the only effective measure to clear the infection as no
The spirochaete is unable of de novo synthesis of NAD+ vaccine for human use is currently available. There are
and therefore the molecule has to be recycled through the a few novel promising vaccine candidates in the develop­
salvage pathways of nicotinamide. The enzyme that cata­ ment or exploratory stage [4,7]. In addition, the direct
lyzes the reaction (PncA) is a key element for mammalian anti-Borrelia measures can be coupled with anti-tick stra­
infection [318]. tegies to deliver a whole new level of human protection
[322]. Full characterization of the drivers involved in the
pathogenic processes, understanding of the regulatory
Conclusion
mechanisms, and further molecular and genetic analysis,
An understanding of the key aspects in the pathogen­ will enable us to design better control and diagnostic
esis of LD is decisive to the effective care of patients approaches.
affected with the disease. The virulence strategies of the We aimed at highlighting a lot of the advances
infectious microbes are constantly evolving to maintain made in understanding the various virulence determi­
their ability to infect, disseminate, and proliferate in the nants of B. burgdorferi. It has also brought into shar­
host, which, in turn, forces the researchers to evolve per focus how much data we still need, to obtain
20 M. STRNAD ET AL.

a more in depth understanding of the virulence [4] Strnad M, Grubhoffer L, Rego ROM. Novel targets and
mechanisms that have been identified and those yet strategies to combat borreliosis. Appl Microbiol
to be identified. The STM analysis was a major step Biotechnol. 2020;104(5):1915–1925. doi: 10.1007/
s00253-020-10375-8
toward this goal but needs to be followed through at [5] Jauris-Heipke S, Fuchs R, Motz M, et al. Genetic het­
the individual level for observing the virulence pheno­ erogenity of the genes coding for the outer surface
type associated with each of the STM mutants. It is protein C (OspC) and the flagellin of Borrelia
abundantly clear that to be able to obtain better drug- burgdorferi. Med Microbiol Immunol. 1993;182
related interventions for patients, who suffer because (1):37–50. doi: 10.1007/BF00195949
[6] Roberts WC, Mullikin BA, Lathigra R, et al. Molecular
of misdiagnosis or belated treatment, use of faster
analysis of sequence heterogeneity among genes encod­
genetic methods/’omics’ studies is important. The ing decorin binding proteins a and B of Borrelia burg­
uniqueness/plasticity of the genomes of the various dorferi Sensu Lato. Infect Immun. 1998;66
Borrelia genospecies is the biggest challenge we face (11):5275–5285. doi: 10.1128/IAI.66.11.5275-5285.1998
in obtaining a broader spectrum of medical/diagnostic [7] Comstedt P, Schüler W, Meinke A, et al. The novel
options to counter this disease-causing bacterial Lyme borreliosis vaccine VLA15 shows broad protec­
tion against Borrelia species expressing six different
pathogen.
OspA serotypes. PLoS One. 2017;12(9):e0184357. doi:
10.1371/journal.pone.0184357
[8] Coburn J, Garcia B, Hu LT, et al. Lyme disease
Acknowledgements pathogenesis. Curr Issues Mol Biol. 2021;42:473–518.
This work was supported by the Czech Science Foundation doi: 10.21775/cimb.042.473
grants No. 22-18647K and 23-06525J. MS is supported by the [9] Strnad M, Rego ROM. The need to unravel the twisted
Program for Prospective Human Resources by Czech nature of the Borrelia burgdorferi sensu lato complex
Academy of Sciences. NR is supported by grant from the across Europe. Microbiol (Reading). 2020;166
Ministry of Health of the Czech Republic (NV19-05-00191). (5):428–435. doi: 10.1099/mic.0.000899
Figures were created with BioRender.com. [10] Stewart PE, Byram R, Grimm D, et al. The plasmids of
Borrelia burgdorferi: essential genetic elements of a
pathogen. Plasmid. 2005;53(1):1–13. doi: 10.1016/j.plas
Disclosure statement mid.2004.10.006
[11] Vancová M, Bílý T, Šimo L, et al. Three-dimensional
No potential conflict of interest was reported by the reconstruction of the feeding apparatus of the tick
authors. Ixodes ricinus (Acari: ixodidae): a new insight into
the mechanism of blood-feeding. Sci Rep. 2020;10
(1):165. doi: 10.1038/s41598-019-56811-2
Data Availability statement [12] Dunham-Ems SM, Caimano MJ, Pal U, et al. Live
imaging reveals a biphasic mode of dissemination of
Data sharing is not applicable to this article as no datasets Borrelia burgdorferi within ticks. J Clin Invest.
were generated or analysed in the current study. 2009;119(12):3652–3665. doi: 10.1172/JCI39401
[13] Strnad M, Oh YJ, Vancová M, et al. Nanomechanical
mechanisms of Lyme disease spirochete motility
ORCID enhancement in extracellular matrix. Commun Biol.
Martin Strnad http://orcid.org/0000-0002-1671-6252 2021;4(1):1–9. doi: 10.1038/s42003-021-01783-1
Natalie Rudenko http://orcid.org/0000-0002-0506-2249 [14] Moriarty TJ, Norman MU, Colarusso P, et al. Real-
Ryan O.M. Rego http://orcid.org/0000-0001-6932-0940 time high resolution 3D imaging of the lyme disease
spirochete adhering to and escaping from the vascula­
ture of a living host. PLOS Pathog. 2008;4(6):e1000090.
References doi: 10.1371/journal.ppat.1000090
[15] Fraser CM, Casjens S, Huang WM, et al. Genomic
[1] Kugeler KJ, Schwartz AM, Delorey MJ, et al. sequence of a Lyme disease spirochaete, Borrelia
Estimating the frequency of Lyme disease Diagnoses, burgdorferi. Nature. 1997;390(6660):580–586. doi: 10.
United States, 2010–2018. Emerg Infect Dis. 2021;27 1038/37551
(2):616–619. doi: 10.3201/eid2702.202731 [16] Casjens SR, Mongodin EF, Qiu W-G, et al. Whole-
[2] Sylvie Goddyn WF, Peterle A, Octavia Sârbu D, et al. genome sequences of two Borrelia afzelii and two
MOTION for a RESOLUTION on Lyme disease (bor­ Borrelia garinii Lyme disease agent isolates.
reliosis) | B8-0514/2018 | European Parliament J Bacteriol. 2011;193(24):6995–6996. doi: 10.1128/JB.
[Internet]. [cited 2022 Oct 27]. Available from: 05951-11
https://www.europarl.europa.eu/doceo/document/ [17] Meriläinen L, Herranen A, Schwarzbach A, et al.
B-8-2018-0514_EN.html Morphological and biochemical features of Borrelia
[3] Stanek G, Strle F. Lyme borreliosis–from tick bite to burgdorferi pleomorphic forms. Microbiology
diagnosis and treatment. FEMS Microbiol Rev. 2018;42 (Reading). 2015;161(3):516–527. doi: 10.1099/mic.0.
(3):233–258. doi: 10.1093/femsre/fux047 000027
VIRULENCE 21

[18] Rudenko N, Golovchenko M, Kybicova K, et al. Infect Dis. 2020;39(11):2143–2152. doi: 10.1007/
Metamorphoses of Lyme disease spirochetes: phenom­ s10096-020-03946-0
enon of Borrelia persisters. Parasites Vectors. 2019;12 [33] Davis IRC, McNeil SA, Allen W, et al. Performance of
(1):237. doi: 10.1186/s13071-019-3495-7 a modified two-tiered testing enzyme immunoassay
[19] Karvonen K, Nykky J, Marjomäki V, et al. Distinctive algorithm for serologic diagnosis of Lyme disease in
evasion mechanisms to allow persistence of Borrelia burg­ Nova Scotia. J Clin Microbiol. 2020;58(7):e01841–19.
dorferi in different human cell lines. Front Microbiol. doi: 10.1128/JCM.01841-19
2021;12:711291. doi: 10.3389/fmicb.2021.711291 [34] Branda JA, Steere AC. Laboratory Diagnosis of Lyme
[20] Karvonen K, Tammisto H, Nykky J, et al. Borrelia Borreliosis. Clin Microbiol Rev. 2021;34(2):e00018–19.
burgdorferi Outer Membrane Vesicles Contain doi: 10.1128/CMR.00018-19
Antigenic Proteins, but Do Not Induce Cell Death in [35] Ang CW, Brandenburg AH, van Burgel ND, et al.
Human Cells. Microorganisms. 2022;10(2):212. doi: 10. A Dutch nationwide evaluation of serological assays for
3390/microorganisms10020212 detection of Borrelia antibodies in clinically well-defined
[21] Kuhn HW, Lasseter AG, Adams PP, et al. BB0562 is patients. Diagn Microbiol Infect Dis. 2015;83
a nutritional virulence determinant with lipase activity (3):222–228. doi: 10.1016/j.diagmicrobio.2015.07.007
important for Borrelia burgdorferi infection and survi­ [36] Joyner G, Mavin S, Milner R, et al. Introduction of IgM
val in fatty acid deficient environments. PLOS testing for the diagnosis of acute Lyme borreliosis:
Pathogens. 2021;17(8):e1009869. doi: 10.1371/journal. a study of the benefits, limitations and costs. Eur
ppat.1009869 J Clin Microbiol Infect Dis. 2022;41(4):671–675. doi:
[22] Klose M, Scheungrab M, Luckner M, et al. FIB-SEM- 10.1007/s10096-021-04366-4
based analysis of Borrelia intracellular processing by [37] Sabin AP, Scholze BP, Lovrich SD, et al. Clinical eva­
human macrophages. J Cell Sci. 2021;134:jcs252320. luation of a Borrelia modified two-tiered testing
doi: 10.1242/jcs.252320 (MTTT) shows increased early sensitivity for Borrelia
[23] Narasimhan S, Rajeevan N, Liu L, et al. Gut microbiota burgdorferi but not other endemic Borrelia species in
of the tick vector Ixodes scapularis modulate coloniza­ a high incidence region for Lyme disease in Wisconsin.
tion of the Lyme disease spirochete. Cell Host Microbe. Diagn Microbiol Infect Dis. 2023;105(1):115837. doi:
2014;15(1):58–71. doi: 10.1016/j.chom.2013.12.001 10.1016/j.diagmicrobio.2022.115837
[24] Herrmann C, Gern L. Search for blood or water is [38] Wilske B, Fingerle V, Schulte-Spechtel U.
influenced by Borrelia burgdorferi in Ixodes ricinus. Microbiological and serological diagnosis of Lyme
Parasites Vectors. 2015;8(1):6. doi: 10.1186/s13071- borreliosis. FEMS Immunol Med Microbiol.
014-0526-2 2007;49(1):13–21. doi: 10.1111/j.1574-695X.2006.
[25] Yuste RA, Muenkel M, Axarlis K, et al. Borrelia burg­ 00139.x
dorferi modulates the physical forces and immunity [39] Kalish RA, McHugh G, Granquist J, et al. Persistence
signaling in endothelial cells. iScience. 2022;25 of Immunoglobulin M or Immunoglobulin G Antibody
(8):104793. doi: 10.1016/j.isci.2022.104793 Responses to Borrelia burgdorferi 10–20 Years after
[26] Lin Y-P, Diuk-Wasser MA, Stevenson B, et al. Active Lyme Disease. Clin Infect Dis. 2001;33
Complement evasion contributes to Lyme borreliae– (6):780–785. doi: 10.1086/322669
host associations. Trends Parasitol. 2020;36 [40] Krogen I, Skarphédinsson S, Jensen TG, et al. No
(7):634–645. doi: 10.1016/j.pt.2020.04.011 correlation between symptom duration and intrathecal
[27] Coburn J, Leong J, Chaconas G. Illuminating the roles production of IgM and/or IgG antibodies in Lyme
of the Borrelia burgdorferi adhesins. Trends Microbiol. neuroborreliosis – a retrospective cohort study in
2013;21(8):372–379. doi: 10.1016/j.tim.2013.06.005 Denmark. J Infect. 2022;85(5):507–512. doi: 10.1016/j.
[28] Hyde JA. Borrelia burgdorferi keeps moving and car­ jinf.2022.08.045
ries on: a review of borrelial dissemination and [41] Lantos PM, Rumbaugh J, Bockenstedt LK, et al. Clinical
invasion. Front Immunol. 2017;8:114. doi: 10.3389/ practice guidelines by the infectious Diseases Society of
fimmu.2017.00114 America (IDSA), American Academy of Neurology
[29] Kurokawa C, Lynn GE, Pedra JHF, et al. Interactions (AAN), and American College of Rheumatology (ACR):
between Borrelia burgdorferi and ticks. Nat Rev 2020 guidelines for the Prevention, diagnosis, and treat­
Microbiol. 2020;18(10):587–600. doi: 10.1038/s41579- ment of Lyme disease. Arthritis Care Res (Hoboken).
020-0400-5 2021;73(1):1–9. doi: 10.1002/acr.24495
[30] Schutzer SE, Berger BW, Krueger JG, et al. Atypical [42] Crossland NA, Alvarez X, Embers ME. Late dissemi­
erythema migrans in patients with PCR-Positive Lyme nated Lyme disease. Am J Pathol. 2018;188(3):672–682.
disease. Emerg Infect Dis. 2013;19(5):815–817. doi: 10. doi: 10.1016/j.ajpath.2017.11.005
3201/eid1905.120796 [43] Stanek G, Fingerle V, Hunfeld K-P, et al. Lyme borre­
[31] Waddell LA, Greig J, Mascarenhas M, et al. The accu­ liosis: Clinical case definitions for diagnosis and man­
racy of diagnostic tests for Lyme disease in humans, agement in Europe. Clin Microbiol Infect. 2011;17
a Systematic review and meta-analysis of North (1):69–79. doi: 10.1111/j.1469-0691.2010.03175.x
American research. PLoS One. 2016;11(12):e0168613. [44] Bobe JR, Jutras BL, Horn EJ, et al. Recent Progress in
doi: 10.1371/journal.pone.0168613 Lyme Disease and Remaining Challenges. Front Med.
[32] Baarsma ME, Schellekens J, Meijer BC, et al. Diagnostic 2021;8:666554. doi: 10.3389/fmed.2021.666554
parameters of modified two-tier testing in European [45] Wong KH, Shapiro ED, Soffer GK. A review of
patients with early Lyme disease. Eur J Clin Microbiol post-treatment Lyme disease syndrome and chronic
22 M. STRNAD ET AL.

Lyme disease for the practicing immunologist. Clin Rev [60] Margos G, Lane RS, Fedorova N, et al. Borrelia bisset­
Allergy Immunol. 2022;62(1):264–271. doi: 10.1007/ tiae sp. nov. and Borrelia californiensis sp. nov. prevail
s12016-021-08906-w in diverse enzootic transmission cycles. Int J Syst Evol
[46] Jutras BL, Lochhead RB, Kloos ZA, et al. Borrelia Microbiol. 2016;66(3):1447–1452. doi: 10.1099/ijsem.0.
burgdorferi peptidoglycan is a persistent antigen in 000897
patients with Lyme arthritis. Proc Natl Acad Sci [61] Rudenko N, Golovchenko M, Růzek D, et al. Molecular
U S A. 2019;116(27):13498–13507. doi: 10.1073/pnas. detection of Borrelia bissettii DNA in serum samples
1904170116 from patients in the Czech Republic with suspected
[47] Rudenko N, Golovchenko M. Sexual transmission of borreliosis. FEMS Microbiol Lett. 2009;292
Lyme Borreliosis? The question that calls for an (2):274–281. doi: 10.1111/j.1574-6968.2009.01498.x
answer. Trop Med Infect Dis. 2021;6(2):87. doi: 10. [62] Rudenko N, Golovchenko M, Mokrácek A, et al.
3390/tropicalmed6020087 Detection of Borrelia bissettii in cardiac valve tissue
[48] Majerová K, Hönig V, Houda M, et al. Hedgehogs, of a patient with endocarditis and aortic valve stenosis
squirrels, and blackbirds as sentinel hosts for active in the Czech Republic. J Clin Microbiol. 2008;46
surveillance of Borrelia miyamotoi and Borrelia burg­ (10):3540–3543. doi: 10.1128/JCM.01032-08
dorferi complex in urban and rural environments. [63] Collares-Pereira M, Couceiro S, Franca I, et al. First
Microorganisms. 2020;8(12):E1908. doi: 10.3390/ isolation of Borrelia lusitaniae from a human patient.
microorganisms8121908 J Clin Microbiol. 2004;42(3):1316–1318. doi: 10.1128/
[49] Rudenko N, Golovchenko M, Grubhoffer L, et al. JCM.42.3.1316-1318.2004
Updates on Borrelia burgdorferi sensu lato complex [64] Diza E, Papa A, Vezyri E, et al. Borrelia valaisiana in
with respect to public health. Ticks Tick Borne Dis. cerebrospinal fluid. Emerging Infect Dis. 2004;10
2011;2(3):123–128. doi: 10.1016/j.ttbdis.2011.04.002 (9):1692–1693. doi: 10.3201/eid1009.030439
[50] Clark KL, Leydet B, Hartman S. Lyme borreliosis in [65] Margos G, Sing A, Fingerle V. Published data do not
human patients in Florida and Georgia, USA. support the notion that Borrelia valaisiana is human
Int J Med Sci. 2013;10(7):915–931. doi: 10.7150/ijms. pathogenic. Infection. 2017;45(4):567–569. doi: 10.
6273 1007/s15010-017-1032-1
[51] Steere AC. Lyme disease. N Engl J Med. 1989;321 [66] Eliassen KE, Ocias LF, Krogfelt KA, et al. Tick-
(9):586–596. doi: 10.1056/NEJM198908313210906 transmitted co-infections among erythema migrans
[52] Oschmann P, Dorndorf W, Hornig C, et al. Stages and patients in a general practice setting in Norway:
syndromes of neuroborreliosis. J Neurol. 1998;245 a clinical and laboratory follow-up study. BMC Infect
(5):262–272. doi: 10.1007/s004150050216 Dis. 2021;21(1):1044. doi: 10.1186/s12879-021-06755-8
[53] Ornstein K, Berglund J, Nilsson I, et al. [67] Cassatt DR, Patel NK, Ulbrandt ND, et al. DbpA, but
Characterization of Lyme borreliosis isolates from not OspA, is expressed by Borrelia burgdorferi during
patients with erythema migrans and neuroborreliosis spirochetemia and is a target for protective antibodies.
in southern Sweden. J Clin Microbiol. 2001;39 Infect Immun. 1998;66(11):5379–5387. doi: 10.1128/
(4):1294–1298. doi: 10.1128/JCM.39.4.1294-1298.2001 IAI.66.11.5379-5387.1998
[54] Ruzić-Sabljić E, Maraspin V, Lotric-Furlan S, et al. [68] Hanson MS, Cassatt DR, Guo BP, et al. Active and
Characterization of Borrelia burgdorferi sensu lato passive immunity against Borrelia burgdorferi decorin
strains isolated from human material in Slovenia. binding protein a (DbpA) protects against infection.
Wien Klin Wochenschr. 2002;114:544–550. Infect Immun. 1998;66(5):2143–2153. doi: 10.1128/IAI.
[55] Steere AC, Strle F, Wormser GP, et al. Lyme 66.5.2143-2153.1998
borreliosis. Nat Rev Dis Primers. 2016;2(1):16090. doi: [69] Brown EL, Kim JH, Reisenbichler ES, et al.
10.1038/nrdp.2016.90 Multicomponent Lyme vaccine: three is not a crowd.
[56] Grange F, Wechsler J, Guillaume J-C, et al. Borrelia Vaccine. 2005;23(28):3687–3696. doi: 10.1016/j.vac
burgdorferi-associated lymphocytoma cutis simulating cine.2005.02.006
a primary cutaneous large B-cell lymphoma. J Am [70] Earnhart CG, Marconi RT. An octavalent lyme disease
Acad Dermatol. 2002;47(4):530–534. doi: 10.1067/ vaccine induces antibodies that recognize all incorpo­
mjd.2002.120475 rated OspC type-specific sequences. Hum Vaccin.
[57] Smetanick MT, Zellis SL, Ermolovich T. 2007;3(6):281–289. doi: 10.4161/hv.4661
Acrodermatitis chronica atrophicans: a case report [71] Kumar M, Kaur S, Kariu T, et al. Borrelia burgdorferi
and review of the literature. Cutis. 2010;85:247–252. BBA52 is a potential target for transmission blocking
[58] Picken RN, Strle F, Picken MM, et al. Identification of Lyme disease vaccine. Vaccine. 2011;29(48):9012–9019.
three species of Borrelia burgdorferi sensu lato doi: 10.1016/j.vaccine.2011.09.035
(B. burgdorferi sensu stricto, B. garinii, and B. afzelii) [72] Klouwens MJ, Trentelman JJ, Ersoz JI, et al.
among isolates from acrodermatitis chronica atrophi­ Investigating BB0405 as a novel Borrelia afzelii vacci­
cans lesions. J Invest Dermatol. 1998;110(3):211–214. nation candidate in Lyme borreliosis. Sci Rep. 2021;11
doi: 10.1046/j.1523-1747.1998.00130.x (1):4775. doi: 10.1038/s41598-021-84130-y
[59] Strle F, Picken RN, Cheng Y, et al. Clinical findings for [73] Singh P, Verma D, Backstedt BT, et al. Borrelia burg­
patients with Lyme borreliosis caused by Borrelia burg­ dorferi BBI39 paralogs, targets of protective immunity,
dorferi sensu lato with genotypic and phenotypic simi­ reduce pathogen persistence either in hosts or in the
larities to strain 25015. Clin Infect Dis. 1997;25 vector. J Infect Dis. 2017;215(6):1000–1009. doi: 10.
(2):273–280. doi: 10.1086/514551 1093/infdis/jix036
VIRULENCE 23

[74] Nigrovic LE, Thompson KM. The Lyme vaccine: Microbiol. 2004;70(10):5973–5979. doi: 10.1128/AEM.
a cautionary tale. Epidemiol Infect. 2007;135(1):1–8. 70.10.5973-5979.2004
doi: 10.1017/S0950268806007096 [89] Botkin DJ, Abbott AN, Stewart PE, et al. Identification
[75] Shaffer L. Inner workings: lyme disease vaccines face of potential virulence determinants by Himar1 trans­
familiar challenges, both societal and scientific. Proc position of infectious Borrelia burgdorferi B31. Infect
Natl Acad Sci U S A. 2019;116(39):19214–19217. doi: Immun. 2006;74(12):6690–6699. doi: 10.1128/IAI.
10.1073/pnas.1913923116 00993-06
[76] Bézay N, Hochreiter R, Kadlecek V, et al. Safety and [90] Lin T, Gao L, Zhang C, et al. Analysis of an ordered,
immunogenicity of a novel multivalent OspA-based Comprehensive STM mutant Library in infectious
vaccine candidate against Lyme borreliosis: Borrelia burgdorferi: insights into the genes required
a randomised, phase 1 study in healthy adults. Lancet for mouse infectivity. PLoS One. 2012;7(10):e47532.
Infect Dis. 2023;S1473-3099(23):210–214. doi: 10.1016/ doi: 10.1371/journal.pone.0047532
S1473-3099(23)00210-4 [91] Bose JL. Chemical and UV mutagenesis. Methods Mol
[77] Earnhart CG, Buckles EL, Marconi RT. Development Biol. 2016;1373:111–115.
of an OspC-based tetravalent, recombinant, chimeric [92] Lin T, Gao L. Genome-Wide Mutagenesis in Borrelia
vaccinogen that elicits bactericidal antibody against burgdorferi. Methods Mol Biol. 2018;1690:201–223.
diverse Lyme disease spirochete strains. Vaccine. [93] Revel AT, Talaat AM, Norgard MV. DNA microarray
2007;25(3):466–480. doi: 10.1016/j.vaccine.2006.07.052 analysis of differential gene expression in Borrelia
[78] Wressnigg N, Pöllabauer E-M, Aichinger G, et al. burgdorferi, the Lyme disease spirochete. Proc Natl
Safety and immunogenicity of a novel multivalent Acad Sci U S A. 2002;99(3):1562–1567. doi: 10.1073/
OspA vaccine against Lyme borreliosis in healthy pnas.032667699
adults: a double-blind, randomised, dose-escalation [94] Liang FT, Nelson FK, Fikrig E. DNA microarray
phase 1/2 trial. Lancet Infect Dis. 2013;13(8):680–689. assessment of putative Borrelia burgdorferi lipoprotein
doi: 10.1016/S1473-3099(13)70110-5 genes. Infect Immun. 2002;70(6):3300–3303. doi: 10.
[79] Kamp HD, Swanson KA, Wei RR, et al. Design of 1128/IAI.70.6.3300-3303.2002
a broadly reactive Lyme disease vaccine. NPJ Vaccines. [95] Akins DR, Bourell KW, Caimano MJ, et al. A new
2020;5(1):33. doi: 10.1038/s41541-020-0183-8 animal model for studying Lyme disease spirochetes
[80] Nayak A, Schüler W, Seidel S, et al. Broadly protective in a mammalian host-adapted state. J Clin Invest.
multivalent OspA vaccine against Lyme borreliosis, 1998;101(10):2240–2250. doi: 10.1172/JCI2325
developed based on surface shaping of the [96] Caimano MJ. Cultivation of Borrelia burgdorferi in
C-Terminal fragment. Infect Immun. 2020;88(4): dialysis membrane chambers in rat peritonea. Curr
e00917–19. doi: 10.1128/IAI.00917-19 Protoc Microbiol. 2005;(1). doi: 10.1002/
[81] Klouwens MJ, Trentelman JJA, Wagemakers A, et al. 9780471729259.mc12c03s00
Tick-Tattoo: DNA vaccination against B. burgdorferi [97] Arnold WK, Savage CR, Brissette CA, et al. RNA-Seq
or Ixodes scapularis Tick proteins. Front Immunol. of Borrelia burgdorferi in multiple phases of growth
2021;12:615011. doi: 10.3389/fimmu.2021.615011 reveals insights into the dynamics of gene expression,
[82] Wagemakers A, Mason LMK, Oei A, et al. Rapid transcriptome architecture, and noncoding RNAs.
outer-surface protein C DNA tattoo vaccination pro­ PLoS One. 2016;11(10):e0164165. doi: 10.1371/jour
tects against Borrelia afzelii infection. Gene Ther. nal.pone.0164165
2014;21(12):1051–1057. doi: 10.1038/gt.2014.87 [98] Lybecker MC, Samuels DS. Small RNAs of Borrelia
[83] Pfeifle A, Thulasi Raman SN, Lansdell C, et al. DNA burgdorferi: characterizing functional regulators in
lipid nanoparticle vaccine targeting outer surface pro­ a sea of sRnas. Yale J Biol Med. 2017;90(2):317–323.
tein C affords protection against homologous Borrelia [99] Ellis TC, Jain S, Linowski AK, et al. In vivo expression
burgdorferi needle challenge in mice. Front Immunol. technology identifies a novel virulence factor critical for
2023;14:1020134. doi: 10.3389/fimmu.2023.1020134 Borrelia burgdorferi persistence in mice. PLOS Pathog.
[84] Saade F, Petrovsky N. Technologies for enhanced effi­ 2013;9(8):e1003567. doi: 10.1371/journal.ppat.1003567
cacy of DNA vaccines. Expert Rev Vaccines. 2012;11 [100] Casselli T, Bankhead T. Use of in vivo expression
(2):189–209. doi: 10.1586/erv.11.188 technology for the identification of putative host adap­
[85] Rosa PA, Jewett MW. Genetic manipulation of tation factors of the Lyme disease spirochete. J Mol
Borrelia. Curr Issues Mol Biol. 2021;42:307–332. doi: Microbiol Biotechnol. 2015;25(5):349–361. doi: 10.
10.21775/cimb.042.307 1159/000439305
[86] Samuels DS. Electrotransformation of the spirochete [101] Hyde JA, Weening EH, Chang M, et al. Bioluminescent
Borrelia burgdorferi. Methods Mol Biol. imaging of Borrelia burgdorferi in vivo demonstrates
1995;47:253–259. that the fibronectin-binding protein BBK32 is required
[87] Rosa PA, Tilly K, Stewart PE. The burgeoning mole­ for optimal infectivity. Mol Microbiol. 2011;82
cular genetics of the Lyme disease spirochaete. Nat Rev (1):99–113. doi: 10.1111/j.1365-2958.2011.07801.x
Microbiol. 2005;3(2):129–143. doi: 10.1038/ [102] Hejduk L, Rathner P, Strnad M, et al. Resonance
nrmicro1086 assignment and secondary structure of DbpA protein
[88] Stewart PE, Hoff J, Fischer E, et al. Genome-wide from the European species, Borrelia afzelii. Biomol
transposon mutagenesis of Borrelia burgdorferi for NMR Assign. 2021;15(2):415–420. doi: 10.1007/
identification of phenotypic mutants. Appl Environ s12104-021-10039-2
24 M. STRNAD ET AL.

[103] Niddam AF, Ebady R, Bansal A, et al. Plasma fibro­ Infect Immun. 2002;70(8):4099–4105. doi: 10.1128/IAI.
nectin stabilizes Borrelia burgdorferi–endothelial inter­ 70.8.4099-4105.2002
actions under vascular shear stress by a catch-bond [118] Jones MB, Peterson SN, Benn R, et al. Role of luxS in
mechanism. PNAS. 2017;114(17):E3490–8. doi: 10. bacillus anthracis growth and virulence factor
1073/pnas.1615007114 expression. Virulence. 2010;1(2):72–83. doi: 10.4161/
[104] Harman MW, Dunham-Ems SM, Caimano MJ, et al. viru.1.2.10752
The heterogeneous motility of the Lyme disease spir­ [119] Xu L, Li H, Vuong C, et al. Role of the luxS
ochete in gelatin mimics dissemination through tissue. Quorum-Sensing System in Biofilm Formation and
Proc Natl Acad Sci, USA. 2012;109(8):3059–3064. doi: Virulence of Staphylococcus epidermidis. Infect
10.1073/pnas.1114362109 Immun. 2006;74(1):488–496. doi: 10.1128/IAI.74.1.
[105] Bockenstedt LK, Gonzalez D, Mao J, et al. What ticks 488-496.2006
do under your skin: Two-Photon intravital imaging of [120] Zhang B, Ku X, Zhang X, et al. The AI-2/luxS quorum
Ixodes scapularis feeding in the presence of the Lyme sensing system affects the growth characteristics, bio­
disease spirochete. Yale J Biol Med. 2014;87:3–13. film formation, and virulence of haemophilus parasuis.
[106] Hillman C, Stewart PE, Strnad M, et al. Visualization of Front Cell Infect Microbiol. 2019;9:62. doi: 10.3389/
spirochetes by labeling membrane proteins with fluor­ fcimb.2019.00062
escent biarsenical dyes. Front Cell Infect Microbiol. [121] Taga ME, Bassler BL. Chemical communication among
2019;9:287. doi: 10.3389/fcimb.2019.00287 bacteria. Proc Natl Acad Sci U S A. 2003;100
[107] Strnad M, Elsterová J, Schrenková J, et al. Correlative (2):14549–14554. doi: 10.1073/pnas.1934514100
cryo-fluorescence and cryo-scanning electron micro­ [122] Stevenson B, von Lackum K, Wattier RL, et al. Quorum
scopy as a straightforward tool to study sensing by the Lyme disease spirochete. Microbes
host-pathogen interactions. Sci Rep. 2015;5(1):18029. Infect. 2003;5(11):991–997. doi: 10.1016/S1286-
doi: 10.1038/srep18029 4579(03)00184-9
[108] Wang X. Solution structure of decorin-binding protein [123] Blevins JS, Revel AT, Caimano MJ, et al. The luxS gene
a from Borrelia burgdorferi. Biochemistry. 2012;51 is not required for Borrelia burgdorferi tick coloniza­
(42):8353–8362. doi: 10.1021/bi3007093 tion, transmission to a mammalian host, or induction
[109] Ante VM, Farris LC, Saputra EP, et al. The Borrelia of disease. Infect Immun. 2004;72(8):4864–4867. doi:
burgdorferi adenylate cyclase, CyaB, is important for 10.1128/IAI.72.8.4864-4867.2004
virulence factor production and mammalian infection. [124] Arnold WK, Savage CR, Antonicello AD, et al.
Front Microbiol. 2021;12:676192. doi: 10.3389/fmicb. Apparent Role for Borrelia burgdorferi LuxS during
2021.676192 Mammalian Infection. Infect Immun. 2015;83
[110] Topal H, Fulcher NB, Bitterman J, et al. Crystal struc­ (4):1347–1353. doi: 10.1128/IAI.00032-15
ture and Regulation mechanisms of the CyaB Adenylyl [125] Strnad M, Hönig V, Růžek D, et al. Europe-wide
cyclase from the human pathogen Pseudomonas meta-analysis of Borrelia burgdorferi Sensu Lato pre­
aeruginosa. J Mol Biol. 2012;416(2):271–286. doi: 10. valence in questing Ixodes ricinus ticks. Appl
1016/j.jmb.2011.12.045 Environ Microbiol. 2017;83(15). doi: 10.1128/AEM.
[111] Kim YR, Kim SY, Kim CM, et al. Essential role of an 00609-17
adenylate cyclase in regulating Vibrio vulnificus [126] van Duijvendijk G, Coipan C, Wagemakers A, et al.
virulence. FEMS Microbiol Lett. 2005;243(2):497–503. Larvae of Ixodes ricinus transmit Borrelia afzelii and
doi: 10.1016/j.femsle.2005.01.016 B. miyamotoi to vertebrate hosts. Parasites Vectors.
[112] Galperin MY, Chou S-H, Stock AM. Structural 2016;9(1):97. doi: 10.1186/s13071-016-1389-5
Conservation and diversity of PilZ-Related Domains. [127] Tonk M, Cabezas-Cruz A, Valdés JJ, et al. Defensins
J Bacteriol. 2020;202(4):10.1128/jb.00664–19. doi: 10. from the tick Ixodes scapularis are effective against
1128/JB.00664-19 phytopathogenic fungi and the human bacterial patho­
[113] Hong Y, Zhou X, Fang H, et al. Cyclic di-GMP med­ gen Listeria grayi. Parasites Vectors. 2014;7(1):554. doi:
iates Mycobacterium tuberculosis dormancy and 10.1186/s13071-014-0554-y
pathogenecity. Tuberculosis (Edinb). 2013;93 [128] Cruz CE, Fogaça AC, Nakayasu ES, et al.
(6):625–634. doi: 10.1016/j.tube.2013.09.002 Characterization of proteinases from the midgut of
[114] Amikam D, Galperin MY. PilZ domain is part of the rhipicephalus (boophilus) microplus involved in the
bacterial c-di-GMP binding protein. Bioinformatics. generation of antimicrobial peptides. Parasites
2006;22(1):3–6. doi: 10.1093/bioinformatics/bti739 Vectors. 2010;3(1):63. doi: 10.1186/1756-3305-3-63
[115] Jusufovic N, Savage CR, Saylor TC, et al. Borrelia [129] Pereira LS, Oliveira PL, Barja-Fidalgo C, et al. Production
burgdorferi PlzA is a c-di-GMP dependent DNA and of reactive oxygen species by hemocytes from the cattle
RNA binding protein. bioRxiv. 2023. tick boophilus microplus. Exp Parasitol. 2001;99
[116] Groshong AM, Grassmann AA, Luthra A, et al. PlzA is (2):66–72. doi: 10.1006/expr.2001.4657
a bifunctional c-di-GMP biosensor that promotes tick [130] Eggenberger LR, Lamoreaux WJ, Coons LB. Hemocytic
and mammalian host-adaptation of Borrelia encapsulation of implants in the tick dermacentor
burgdorferi. PLOS Pathog. 2021;17(7):e1009725. doi: variabilis. Exp Appl Acarol. 1990;9(3–4):279–287. doi:
10.1371/journal.ppat.1009725 10.1007/BF01193434
[117] Stevenson B, Babb K. LuxS-Mediated quorum sensing [131] Fogaça AC, Sousa G, Pavanelo DB, et al. Tick immune
in Borrelia burgdorferi, the Lyme disease spirochete. system: what is known, the interconnections, the gaps,
VIRULENCE 25

and the challenges. Front Immunol. 2021;12:628054. transmission. PLOS Pathogens. 2014;10(8):e1004278.
doi: 10.3389/fimmu.2021.628054 doi: 10.1371/journal.ppat.1004278
[132] Tanaka T, Kawano S, Nakao S, et al. The identification [147] Coumou J, Narasimhan S, Trentelman JJ, et al.
and characterization of lysozyme from the hard tick Ixodes scapularis dystroglycan-like protein pro­
haemaphysalis longicornis. Ticks Tick Borne Dis. motes Borrelia burgdorferi migration from the
2010;1(4):178–185. doi: 10.1016/j.ttbdis.2010.09.001 gut. J Mol Med (Berl). 2016;94(3):361–370. doi:
[133] De Silva AM, Fikrig E. Growth and migration of 10.1007/s00109-015-1365-0
Borrelia burgdorferi in Ixodes ticks during blood [148] Pal U, Yang X, Chen M, et al. OspC facilitates Borrelia
feeding. Am J Trop Med Hyg. 1995;53(4):397–404. burgdorferi invasion of Ixodes scapularis salivary
doi: 10.4269/ajtmh.1995.53.397 glands. J Clin Invest. 2004;113(2):220–230. doi: 10.
[134] Ribeiro JM, Mather TN, Piesman J, et al. 1172/JCI200419894
Dissemination and salivary delivery of Lyme disease [149] Tilly K, Krum JG, Bestor A, et al. Borrelia burgdorferi
spirochetes in vector ticks (Acari: ixodidae). J Med OspC protein required exclusively in a crucial early
Entomol. 1987;24(2):201–205. doi: 10.1093/jmedent/ stage of mammalian infection. Infect Immun. 2006;74
24.2.201 (6):3554–3564. doi: 10.1128/IAI.01950-05
[135] Pospisilova T, Urbanova V, Hes O, et al. Tracking of [150] Ramamoorthi N, Narasimhan S, Pal U, et al. The Lyme
Borrelia afzelii transmission from infected Ixodes rici­ disease agent exploits a tick protein to infect the mam­
nus Nymphs to mice. Infect Immun. 2019;87(6): malian host. Nature. 2005;436(7050):573–577. doi: 10.
e00896–18. doi: 10.1128/IAI.00896-18 1038/nature03812
[136] Lejal E, Moutailler S, Šimo L, et al. Tick-borne patho­ [151] Bierwagen P, Sliwiak J, Jaskolski M, et al. Strong inter­
gen detection in midgut and salivary glands of adult actions between Salp15 homologues from the tick
Ixodes ricinus. Parasites Vectors. 2019;12(1):152. doi: I. ricinus and distinct types of the outer surface OspC
10.1186/s13071-019-3418-7 protein from Borrelia. Ticks Tick Borne Dis. 2021;12
[137] Yang XF, Pal U, Alani SM, et al. Essential role for (2):101630. doi: 10.1016/j.ttbdis.2020.101630
OspA/B in the life cycle of the Lyme disease [152] Kotál J, Langhansová H, Lieskovská J, et al. Modulation
spirochete. J Exp Med. 2004;199(5):641–648. doi: 10. of host immunity by tick saliva. J Proteomics.
1084/jem.20031960 2015;128:58–68. doi: 10.1016/j.jprot.2015.07.005
[138] Pal U, de Silva AM, Montgomery RR, et al. Attachment [153] Murfin KE, Kleinbard R, Aydin M, et al. Borrelia
of Borrelia burgdorferi within Ixodes scapularis burgdorferi chemotaxis toward tick protein Salp12
mediated by outer surface protein a. J Clin Invest. contributes to acquisition. Ticks Tick Borne Dis.
2000;106(4):561–569. doi: 10.1172/JCI9427 2019;10(5):1124–1134. doi: 10.1016/j.ttbdis.2019.06.002
[139] Pal U, Li X, Wang T, et al. TROSPA, an Ixodes scapu­ [154] Narasimhan S, Sukumaran B, Bozdogan U, et al. A tick
laris receptor for Borrelia burgdorferi. Cell. 2004;119 antioxidant facilitates the Lyme disease agent’s success­
(4):457–468. doi: 10.1016/j.cell.2004.10.027 ful migration from the mammalian host to the arthro­
[140] Zhang L, Zhang Y, Adusumilli S, et al. Molecular inter­ pod vector. Cell Host Microbe. 2007;2(1):7–18. doi: 10.
actions that enable movement of the Lyme disease agent 1016/j.chom.2007.06.001
from the tick gut into the hemolymph. PLOS Pathog. [155] Coleman JL, Crowley JT, Toledo AM, et al. The HtrA
2011;7(6):e1002079. doi: 10.1371/journal.ppat.1002079 protease of Borrelia burgdorferi degrades outer mem­
[141] Neelakanta G, Li X, Pal U, et al. Outer surface protein brane protein BmpD and chemotaxis phosphatase
B is critical for Borrelia burgdorferi adherence and CheX. Mol Microbiol. 2013;88(3):619–633. doi: 10.
survival within Ixodes ticks. PLOS Pathogens. 2007;3 1111/mmi.12213
(3):e33. doi: 10.1371/journal.ppat.0030033 [156] Boylan JA, Lawrence KA, Downey JS, et al. Borrelia
[142] Revel AT, Blevins JS, Almazán C, et al. bptA (bbe16) is burgdorferi membranes are the primary targets of reac­
essential for the persistence of the Lyme disease spir­ tive oxygen species. Mol Microbiol. 2008;68
ochete, Borrelia burgdorferi, in its natural tick vector. (3):786–799. doi: 10.1111/j.1365-2958.2008.06204.x
Proc Natl Acad Sci U S A. 2005;102(19):6972–6977. [157] Radolf JD, Caimano MJ, Stevenson B, et al. Of ticks,
doi: 10.1073/pnas.0502565102 mice and men: understanding the dual-host lifestyle of
[143] Mason C, Thompson C, Ouyang Z. DksA plays an Lyme disease spirochaetes. Nat Rev Microbiol. 2012;10
essential role in regulating the virulence of Borrelia (2):87–99. doi: 10.1038/nrmicro2714
burgdorferi. Mol Microbiol. 2020;114(1):172–183. doi: [158] Tracy KE, Baumgarth N. Borrelia burgdorferi manip­
10.1111/mmi.14504 ulates innate and adaptive immunity to establish per­
[144] Boyle WK, Groshong AM, Drecktrah D, et al. DksA sistence in rodent reservoir hosts. Front Immunol.
controls the response of the Lyme disease spirochete 2017;8:116. doi: 10.3389/fimmu.2017.00116
Borrelia burgdorferi to starvation. J Bacteriol. 2019;201 [159] Bruch-Gerharz D, Ruzicka T, Kolb-Bachofen V. Nitric
(4):e00582–18. doi: 10.1128/JB.00582-18 oxide in human skin: current status and future
[145] Kumar M, Yang X, Coleman AS, et al. BBA52 facil­ prospects. J Invest Dermatol. 1998;110(1):1–7. doi: 10.
itates Borrelia burgdorferi transmission from feeding 1046/j.1523-1747.1998.00084.x
ticks to murine hosts. J Infect Dis. 2010;201 [160] Ma Y, Seiler KP, Tai KF, et al. Outer surface lipopro­
(7):1084–1095. doi: 10.1086/651172 teins of Borrelia burgdorferi stimulate nitric oxide pro­
[146] Narasimhan S, Coumou J, Schuijt TJ, et al. A tick gut duction by the cytokine-inducible pathway. Infect
protein with fibronectin III domains aids Borrelia Immun. 1994;62(9):3663–3671. doi: 10.1128/iai.62.9.
burgdorferi congregation to the gut during 3663-3671.1994
26 M. STRNAD ET AL.

[161] Seiler KP, Vavrin Z, Eichwald E, et al. Nitric oxide [174] Skare JT, Garcia BL. Complement evasion by Lyme
production during murine Lyme disease: lack of invol­ disease spirochetes. Trends Microbiol. 2020;28
vement in host resistance or pathology. Infect Immun. (11):889–899. doi: 10.1016/j.tim.2020.05.004
1995;63(10):3886–3895. doi: 10.1128/iai.63.10.3886- [175] Garcia BL, Zhi H, Wager B, et al. Borrelia burgdorferi
3895.1995 BBK32 inhibits the classical pathway by blocking acti­
[162] Kerstholt M, Vrijmoeth H, Lachmandas E, et al. Role vation of the C1 complement complex. PLOS Pathog.
of glutathione metabolism in host defense against 2016;12(1):e1005404. doi: 10.1371/journal.ppat.
Borrelia burgdorferi infection. Proc Natl Acad Sci 1005404
U S A. 2018;115(10):E2320–8. doi: 10.1073/pnas. [176] Caine JA, Lin Y-P, Kessler JR, et al. Borrelia burgdor­
1720833115 feri outer surface protein C (OspC) binds complement
[163] Kerstholt M, Brouwer M, Te Vrugt M, et al. Borrelia component C4b and confers bloodstream survival. Cell
burgdorferi inhibits NADPH-mediated reactive oxygen Microbiol. 2017;19(12):e12786. doi: 10.1111/cmi.12786
species production through the mTOR pathway. Ticks [177] Barthel D, Schindler S, Zipfel PF. Plasminogen Is
Tick Borne Dis. 2022;13(4):101943. doi: 10.1016/j. a Complement Inhibitor. J Biol Chem. 2012;287
ttbdis.2022.101943 (22):18831–18842. doi: 10.1074/jbc.M111.323287
[164] Ouyang Z, He M, Oman T, et al. A manganese trans­ [178] Hammerschmidt C, Koenigs A, Siegel C, et al. Versatile
porter, BB0219 (BmtA), is required for virulence by the roles of CspA orthologs in complement Inactivation of
Lyme disease spirochete, Borrelia burgdorferi. Proc serum-resistant Lyme disease spirochetes. Infect
Natl Acad Sci U S A. 2009;106(9):3449–3454. doi: 10. Immun. 2014;82(1):380–392. doi: 10.1128/IAI.01094-13
1073/pnas.0812999106 [179] Koenigs A, Hammerschmidt C, Jutras BL, et al. BBA70
[165] Esteve-Gassent MD, Elliott NL, Seshu J. sodA is essen­ of Borrelia burgdorferi is a novel plasminogen-binding
tial for virulence of Borrelia burgdorferi in the murine protein. J Biol Chem. 2013;288(35):25229–25243. doi:
model of Lyme disease. Mol Microbiol. 2009;71 10.1074/jbc.M112.413872
(3):594–612. doi: 10.1111/j.1365-2958.2008.06549.x [180] Önder Ö, Humphrey PT, McOmber B, et al. OspC Is
[166] Phelan JP, Bourgeois JS, McCarthy JE, et al. A putative Potent Plasminogen Receptor on Surface of Borrelia
xanthine dehydrogenase is critical for Borrelia burg­ burgdorferi. J Biol Chem. 2012;287(20):16860–16868.
dorferi survival in ticks and mice. Microbiology doi: 10.1074/jbc.M111.290775
(Reading). 2023;169(1):001286. doi: 10.1099/mic.0. [181] Fuchs H, Wallich R, Simon MM, et al. The outer sur­
001286 face protein a of the spirochete Borrelia burgdorferi is
[167] Ramsey ME, Hyde JA, Medina-Perez DN, et al. A a plasmin(ogen) receptor. Proc Natil Acad Sci.
high-throughput genetic screen identifies previously 1994;91:12594–12598.
uncharacterized Borrelia burgdorferi genes important [182] Brissette CA, Haupt K, Barthel D, et al. Borrelia burg­
for resistance against reactive oxygen and nitrogen dorferi Infection-Associated Surface Proteins ErpP,
species. PLOS Pathog. 2017;13(2):e1006225. doi: 10. ErpA, and ErpC Bind Human Plasminogen. Infect
1371/journal.ppat.1006225 Immun. 2009;77(1):300–306. doi: 10.1128/IAI.01133-08
[168] Lusitani D, Malawista SE, Montgomery RR. Borrelia [183] Haupt K, Kraiczy P, Wallich R, et al. FHR-1, an addi­
burgdorferi are susceptible to killing by a variety of tional human plasma protein, binds to complement
human polymorphonuclear leukocyte components. regulator-acquiring surface proteins of Borrelia
J Infect Dis. 2002;185(6):797–804. doi: 10.1086/339341 burgdorferi. Int J Med Microbiol. 2008;298:287–291.
[169] Dunkelberger JR, Song W-C. Complement and its role doi: 10.1016/j.ijmm.2007.11.010
in innate and adaptive immune responses. Cell Res. [184] Kraiczy P, Rossmann E, Brade V, et al. Binding of
2010;20(1):34–50. doi: 10.1038/cr.2009.139 human complement regulators FHL-1 and factor H to
[170] Kurtenbach K, De Michelis S, Etti S, et al. Host asso­ CRASP-1 orthologs of Borrelia burgdorferi. Wien Klin
ciation of Borrelia burgdorferi sensu lato–the key role Wochenschr. 2006;118(21–22):669–676. doi: 10.1007/
of host complement. Trends Microbiol. 2002;10 s00508-006-0691-1
(2):74–79. doi: 10.1016/S0966-842X(01)02298-3 [185] Hallström T, Siegel C, Mörgelin M, et al. CspA from
[171] Stevenson B, El-Hage N, Hines MA, et al. Differential Borrelia burgdorferi inhibits the terminal complement
binding of host complement inhibitor factor H by pathway. MBio. 2013;4(4):4. doi: 10.1128/mBio.00481-13
Borrelia burgdorferi Erp surface proteins: a possible [186] Hammerschmidt C, Klevenhaus Y, Koenigs A, et al.
mechanism underlying the expansive host range of BGA66 and BGA71 facilitate complement resistance of
Lyme disease spirochetes. Infect Immun. 2002;70 Borrelia bavariensis by inhibiting assembly of the
(2):491–497. doi: 10.1128/IAI.70.2.491-497.2002 membrane attack complex. Mol Microbiol. 2016;99
[172] van Dam AP, Oei A, Jaspars R, et al. Complement- (2):407–424. doi: 10.1111/mmi.13239
mediated serum sensitivity among spirochetes that [187] Hellwage J, Meri T, Heikkilä T, et al. The complement
cause Lyme disease. Infect Immun. 1997;65:1228–1236. regulator factor H binds to the surface protein OspE of
doi: 10.1128/iai.65.4.1228-1236.1997 Borrelia burgdorferi. J Biol Chem. 2001;276
[173] Kurtenbach K, Peacey M, Rijpkema SG, et al. (11):8427–8435. doi: 10.1074/jbc.M007994200
Differential transmission of the genospecies of [188] Kraiczy P, Hellwage J, Skerka C, et al. Immune evasion of
Borrelia burgdorferi sensu lato by game birds and Borrelia burgdorferi: mapping of a complement-inhibitor
small rodents in England. Appl Environ Microbiol. factor H-binding site of BbCRASP-3, a novel member of
1998;64(4):1169–1174. doi: 10.1128/AEM.64.4.1169- the Erp protein family. Eur J Immunol. 2003;33
1174.1998 (3):697–707. doi: 10.1002/eji.200323571
VIRULENCE 27

[189] Zhang J-R, Norris SJ. Genetic variation of the Borrelia [202] Xu Q, McShan K, Liang FT. Essential protective role
burgdorferi gene vlsE involves cassette-specific, seg­ attributed to the surface lipoproteins of Borrelia burg­
mental gene conversion. Infect Immun. 1998;66 dorferi against innate defences. Mol Microbiol. 2008;69
(8):3698–3704. doi: 10.1128/IAI.66.8.3698-3704.1998 (1):15–29. doi: 10.1111/j.1365-2958.2008.06264.x
[190] Frank SA Benefits of antigenic variation [Internet]. [203] Hovius JW, Schuijt TJ, de Groot KA, et al. Preferential
Immunology and Evolution of infectious disease. protection of Borrelia burgdorferi Sensu Stricto by
Princeton University Press; 2002 [cited 2022 Oct 12]. a Salp15 homologue in Ixodes ricinus Saliva. J Infect
Available from: https://www.ncbi.nlm.nih.gov/books/ Dis. 2008;198(8):1189–1197. doi: 10.1086/591917
NBK2405/ [204] Caine JA, Coburn J, Morrison RP. A short-term
[191] Tilly K, Bestor A, Rosa PA. Lipoprotein succession in Borrelia burgdorferi infection model identifies tissue
Borrelia burgdorferi: similar but distinct roles for OspC tropisms and bloodstream survival conferred by adhe­
and VlsE at different stages of mammalian infection. sion proteins. Infect Immun. 2015;83(8):3184–3194.
Mol Microbiol. 2013;89(2):216–227. doi: 10.1111/mmi. doi: 10.1128/IAI.00349-15
12271 [205] Lin Y-P, Tan X, Caine JA, et al. Strain-specific joint
[192] Brisson D, DE D. ospC diversity in Borrelia burgdor­ invasion and colonization by Lyme disease spirochetes
feri: different hosts are different niches. Genetics. is promoted by outer surface protein C. PLOS
2004;168(2):713–722. doi: 10.1534/genetics.104.028738 Pathogens. 2020;16(5):e1008516. doi: 10.1371/journal.
[193] Brisson D, Baxamusa N, Schwartz I, et al. Biodiversity ppat.1008516
of Borrelia burgdorferi strains in tissues of Lyme dis­ [206] Carrasco SE, Troxell B, Yang Y, et al. Outer surface
ease patients. PLoS One. 2011;6(8):e22926. doi: 10. protein OspC is an antiphagocytic factor that protects
1371/journal.pone.0022926 Borrelia burgdorferi from phagocytosis by
[194] Ivanova L, Christova I, Neves V, et al. Comprehensive macrophages. Infect Immun. 2015;83(12):4848–4860.
seroprofiling of sixteen B. burgdorferi OspC: implica­ doi: 10.1128/IAI.01215-15
tions for Lyme disease diagnostics design. Clin [207] Salo J, Jaatinen A, Söderström M, et al. Decorin bind­
Immunol. 2009;132(3):393–400. doi: 10.1016/j.clim. ing proteins of Borrelia burgdorferi promote arthritis
2009.05.017 development and joint specific post-treatment DNA
[195] Dykhuizen DE, Brisson D, Sandigursky S, et al. The persistence in mice. PLoS One. 2015;10(3):e0121512.
propensity of different Borrelia burgdorferi sensu doi: 10.1371/journal.pone.0121512
stricto genotypes to cause disseminated infections in [208] Shi Y, Xu Q, McShan K, et al. Both decorin-binding
humans. Am J Trop Med Hyg. 2008;78(5):806–810. proteins a and B are critical for the overall virulence of
doi: 10.4269/ajtmh.2008.78.806 Borrelia burgdorferi. Infect Immun. 2008;76
[196] Sadziene A, Wilske B, Ferdows MS, et al. The cryptic (3):1239–1246. doi: 10.1128/IAI.00897-07
ospC gene of Borrelia burgdorferi B31 is located on [209] Imai DM, Samuels DS, Feng S, et al. The early disse­
a circular plasmid. Infect Immun. 1993;61 mination defect attributed to disruption of
(5):2192–2195. doi: 10.1128/iai.61.5.2192-2195.1993 decorin-binding proteins is abolished in chronic mur­
[197] Gilmore RD, Kappel KJ, Dolan MC, et al. Outer surface ine Lyme borreliosis. Infect Immun. 2013;81
protein C (OspC), but not P39, is a protective immu­ (5):1663–1673. doi: 10.1128/IAI.01359-12
nogen against a tick-transmitted Borrelia burgdorferi [210] Weening EH, Parveen N, Trzeciakowski JP, et al. Borrelia
challenge: evidence for a conformational protective burgdorferi lacking DbpBA exhibits an early survival
epitope in OspC. Infect Immun. 1996;64 defect during experimental infection. Infect Immun.
(6):2234–2239. doi: 10.1128/iai.64.6.2234-2239.1996 2008;76(12):5694–5705. doi: 10.1128/IAI.00690-08
[198] Bockenstedt LK, Hodzic E, Feng S, et al. Borrelia burg­ [211] Benoit VM, Fischer JR, Lin Y-P, et al. Allelic variation
dorferi strain-specific Osp C-mediated immunity in of the Lyme disease spirochete adhesin DbpA influ­
mice. Infect Immun. 1997;65(11):4661–4667. doi: 10. ences spirochetal binding to Decorin, dermatan sulfate,
1128/iai.65.11.4661-4667.1997 and mammalian cells. Infect Immun. 2011;79
[199] Rudenko N, Golovchenko M, Hönig V, et al. Detection (9):3501–3509. doi: 10.1128/IAI.00163-11
of Borrelia burgdorferi sensu stricto ospC alleles asso­ [212] Salo J, Loimaranta V, Lahdenne P, et al. Decorin bind­
ciated with human lyme borreliosis worldwide in ing by DbpA and B of Borrelia garinii, Borrelia afzelii,
non-human-biting tick Ixodes affinis and rodent and Borrelia burgdorferi sensu stricto. J Infect Dis.
hosts in Southeastern United States. Appl Environ 2011;204(1):65–73. doi: 10.1093/infdis/jir207
Microbiol. 2013;79(5):1444–1453. doi: 10.1128/AEM. [213] Lin Y-P, Benoit V, Yang X, et al. Strain-specific varia­
02749-12 tion of the decorin-binding adhesin DbpA influences
[200] Lagal V, Portnoï D, Faure G, et al. Borrelia burgdorferi the tissue tropism of the lyme disease spirochete. PLOS
sensu stricto invasiveness is correlated with OspC– Pathog. 2014;10(7):e1004238. doi: 10.1371/journal.
plasminogen affinity. Microbes Infect. 2006;8 ppat.1004238
(3):645–652. doi: 10.1016/j.micinf.2005.08.017 [214] Crother TR, Champion CI, Wu X-Y, et al. Antigenic
[201] Seemanapalli SV, Xu Q, McShan K, et al. Outer surface composition of Borrelia burgdorferi during infection of
protein C is a dissemination-facilitating factor of SCID mice. Infect Immun. 2003;71(6):3419–3428. doi:
Borrelia burgdorferi during mammalian infection. 10.1128/IAI.71.6.3419-3428.2003
PLoS One. 2010;5(12):e15830. doi: 10.1371/journal. [215] Crother TR, Champion CI, Whitelegge JP, et al.
pone.0015830 Temporal analysis of the antigenic composition of
28 M. STRNAD ET AL.

Borrelia burgdorferi during infection in rabbit skin. burgdorferi. Infect Immun. 2013;81(6):2012–2021.
Infect Immun. 2004;72(9):5063–5072. doi: 10.1128/ doi: 10.1128/IAI.01228-12
IAI.72.9.5063-5072.2004 [229] Ge Y, Old I, Saint Girons I, et al. FliH and fliI of
[216] Coutte L, Botkin DJ, Gao L, et al. Detailed analysis of Borrelia burgdorferi are similar to flagellar and viru­
sequence changes occurring during vlsE antigenic var­ lence factor export proteins of other bacteria. Gene.
iation in the mouse model of Borrelia burgdorferi 1996;168(1):73–75. doi: 10.1016/0378-1119(95)00743-1
infection. PLOS Pathog. 2009;5(2):e1000293. doi: 10. [230] Li C, Xu H, Zhang K, et al. Inactivation of a putative
1371/journal.ppat.1000293 flagellar motor switch protein FliG1 prevents Borrelia
[217] Bankhead T, Chaconas G. The role of VlsE antigenic burgdorferi from swimming in highly viscous media
variation in the Lyme disease spirochete: persistence and blocks its infectivity. Mol Microbiol. 2010;75
through a mechanism that differs from other (6):1563–1576. doi: 10.1111/j.1365-2958.2010.07078.x
pathogens. Mol Microbiol. 2007;65(6):1547–1558. doi: [231] Charon NW, Goldstein SF. Genetics of motility and
10.1111/j.1365-2958.2007.05895.x chemotaxis of a fascinating group of bacteria: the
[218] Glöckner G, Schulte-Spechtel U, Schilhabel M, et al. spirochetes. Ann Rev Genet. 2002;36(1):47–73. doi:
Comparative genome analysis: selection pressure on 10.1146/annurev.genet.36.041602.134359
the Borrelia vls cassettes is essential for infectivity. [232] Motaleb MA, Corum L, Bono JL, et al. Borrelia burg­
BMC Genomics. 2006;7(1):211. doi: 10.1186/1471- dorferi periplasmic flagella have both skeletal and
2164-7-211 motility functions. Proc Natl Acad Sci, USA. 2000;97
[219] Zhang JR, Hardham JM, Barbour AG, et al. Antigenic (20):10899–10904. doi: 10.1073/pnas.200221797
variation in Lyme disease borreliae by promiscuous [233] DeHart TG, Kushelman MR, Hildreth SB, et al. The
recombination of VMP-like sequence cassettes. Cell. unusual cell wall of the Lyme disease spirochaete
1997;89(2):275–285. doi: 10.1016/S0092-8674(00) Borrelia burgdorferi is shaped by a tick sugar. Nat
80206-8 Microbiol. 2021;6(12):1583–1592. doi: 10.1038/s41564-
[220] Wang D, Botkin DJ, Norris SJ. Characterization of the 021-01003-w
vls antigenic variation loci of the Lyme disease spiro­ [234] Chen Y, Vargas SM, Smith TC, et al. Borrelia peptido­
chaetes Borrelia garinii Ip90 and Borrelia afzelii ACAI. glycan interacting protein (BpiP) contributes to the
Mol Microbiol. 2003;47(5):1407–1417. doi: 10.1046/j. fitness of Borrelia burgdorferi against host-derived fac­
1365-2958.2003.03386.x tors and influences virulence in mouse models of Lyme
[221] Verhey TB, Castellanos M, Chaconas G. Antigenic disease. PLOS Pathog. 2021;17(4):e1009535. doi: 10.
variation in the Lyme spirochete: insights into recom­ 1371/journal.ppat.1009535
binational switching with a suggested role for [235] Liang L, Wang J, Schorter L, et al. Rapid clearance of
error-prone repair. Cell Rep. 2018;23(9):2595–2605. Borrelia burgdorferi from the blood circulation.
doi: 10.1016/j.celrep.2018.04.117 Parasites Vectors. 2020;13(1):191. doi: 10.1186/
[222] Lin T, Gao L, Edmondson DG, et al. Central role of the s13071-020-04060-y
holliday junction helicase RuvAB in vlsE recombina­ [236] Hodzic E, Feng S, Freet KJ, et al. Borrelia burgdorferi
tion and infectivity of Borrelia burgdorferi. PLOS population dynamics and prototype gene expression
Pathog. 2009;5(12):e1000679. doi: 10.1371/journal. during infection of immunocompetent and immuno­
ppat.1000679 deficient mice. Infect Immun. 2003;71(9):5042–5055.
[223] Lone AG, Bankhead T. The Borrelia burgdorferi VlsE doi: 10.1128/IAI.71.9.5042-5055.2003
lipoprotein prevents antibody binding to an [237] Tilly K, Rosa PA, Stewart PE. Biology of infection with
arthritis-related surface Antigen. Cell Rep. 2020;30 Borrelia burgdorferi. Infect Dis Clin North Am.
(11):3663–3670.e5. doi: 10.1016/j.celrep.2020.02.081 2008;22(2):217–234. doi: 10.1016/j.idc.2007.12.013
[224] Kumar D, Ristow LC, Shi M, et al. Intravital imaging of [238] Cabello FC, Godfrey HP, Newman SA. Hidden in plain
vascular transmigration by the Lyme spirochete: sight: borrelia burgdorferi and the extracellular matrix.
requirement for the integrin binding residues of the Trends Microbiol. 2007;15(8):350–354. doi: 10.1016/j.
B. burgdorferi P66 protein. PLOS Pathog. 2015;11(12): tim.2007.06.003
e1005333. doi: 10.1371/journal.ppat.1005333 [239] Paulsen F, Tillmann B. Composition of the extracellu­
[225] Ebady R, Niddam AF, Boczula AE, et al. Biomechanics of lar matrix in human cricoarytenoid joint articular
Borrelia burgdorferi Vascular Interactions. Cell Rep. cartilage. Arch Histol Cytol. 1999;62(2):149–163. doi:
2016;16(10):2593–2604. doi: 10.1016/j.celrep.2016.08.013 10.1679/aohc.62.149
[226] Tan X, Lin Y-P, Pereira MJ, et al. VlsE, the nexus for [240] Vechtova P, Sterbova J, Sterba J, et al. A bite so sweet:
antigenic variation of the Lyme disease spirochete, also the glycobiology interface of tick-host-pathogen
mediates early bacterial attachment to the host micro­ interactions. Parasites Vectors. 2018;11(1):594. doi:
vasculature under shear force. PLOS Pathog. 2022;18 10.1186/s13071-018-3062-7
(5):e1010511. doi: 10.1371/journal.ppat.1010511 [241] Salo J, Pietikäinen A, Söderström M, et al. Flow-
[227] Tan X, Castellanos M, Chaconas G, et al. tolerant adhesion of a bacterial pathogen to human
Choreography of Lyme disease spirochete adhesins to endothelial cells through interaction with biglycan.
promote vascular escape. Microbiol Spectr. 2023;11(4): J Infect Dis. 2016;213(10):1623–1631. doi: 10.1093/
e0125423. doi: 10.1128/spectrum.01254-23 infdis/jiw003
[228] Sultan SZ, Manne A, Stewart PE, et al. Motility is [242] Brissette CA, Bykowski T, Cooley AE, et al. Borrelia
crucial for the infectious life cycle of Borrelia burgdorferi RevA antigen binds host fibronectin. Infect
VIRULENCE 29

Immun. 2009;77(7):2802–2812. doi: 10.1128/IAI. disease pathogens. J Infect Dis. 2020;221(9):1438–1447.


00227-09 doi: 10.1093/infdis/jiz626
[243] Hallström T, Haupt K, Kraiczy P, et al. Complement [257] Lin Y-P, Chen Q, Ritchie JA, et al. Glycosaminoglycan
Regulator–Acquiring Surface Protein 1 of Borrelia binding by Borrelia burgdorferi adhesin BBK32 speci­
burgdorferi Binds to Human Bone Morphogenic fically and uniquely promotes joint colonization. Cell
Protein 2, Several Extracellular Matrix Proteins, and Microbiol. 2015;17(6):860–875. doi: 10.1111/cmi.12407
Plasminogen. J Infect Dis. 2010;202(3):490–498. doi: [258] Parveen N, Leong JM. Identification of a candidate
10.1086/653825 glycosaminoglycan-binding adhesin of the Lyme dis­
[244] Zhi H, Weening EH, Barbu EM, et al. The BBA33 ease spirochete Borrelia burgdorferi. Mol Microbiol.
lipoprotein binds collagen and impacts Borrelia burg­ 2000;35(5):1220–1234. doi: 10.1046/j.1365-2958.2000.
dorferi pathogenesis. Mol Microbiol. 2015;96(1):68–83. 01792.x
doi: 10.1111/mmi.12921 [259] Fischer JR, Parveen N, Magoun L, et al. Decorin-
[245] Guo BP, Norris SJ, Rosenberg LC, et al. Adherence of binding proteins a and B confer distinct mammalian
Borrelia burgdorferi to the proteoglycan decorin. Infect cell type-specific attachment by Borrelia burgdorferi,
Immun. 1995;63(9):3467–3472. doi: 10.1128/iai.63.9. the Lyme disease spirochete. Proc Natl Acad Sci U S A.
3467-3472.1995 2003;100(12):7307–7312. doi: 10.1073/pnas.
[246] Guo BP, Brown EL, Dorward DW, et al. Decorin- 1231043100
binding adhesins from Borrelia burgdorferi. Mol [260] Yang X, Lin Y-P, Heselpoth RD, et al. Middle region of
Microbiol. 1998;30(4):711–723. doi: 10.1046/j.1365- the Borrelia burgdorferi surface-located protein 1
2958.1998.01103.x (Lmp1) interacts with host chondroitin-6-sulfate and
[247] Fischer JR, LeBlanc KT, Leong JM. Fibronectin binding independently facilitates infection. Cell Microbiol.
protein BBK32 of the Lyme disease spirochete pro­ 2016;18(1):97–110. doi: 10.1111/cmi.12487
motes bacterial attachment to glycosaminoglycans. [261] Dowdell AS, Murphy MD, Azodi C, et al.
Infect Immun. 2006;74(1):435–441. doi: 10.1128/IAI. Comprehensive spatial analysis of the Borrelia burg­
74.1.435-441.2006 dorferi lipoproteome reveals a Compartmentalization
[248] Moriarty TJ, Shi M, Lin Y-P, et al. Vascular binding of Bias toward the bacterial surface. J Bacteriol. 2017;199
a pathogen under shear force through mechanistically (6):e00658–16. doi: 10.1128/JB.00658-16
distinct sequential interactions with host [262] Setubal JC, Reis M, Matsunaga J, et al. Lipoprotein
macromolecules. Mol Microbiol. 2012;86 computational prediction in spirochaetal genomes.
(5):1116–1131. doi: 10.1111/mmi.12045 Microbiology (Reading). 2006;152(1):113–121. doi: 10.
[249] Gaultney RA, Gonzalez T, Floden AM, et al. BB0347, 1099/mic.0.28317-0
from the lyme disease spirochete Borrelia burgdorferi, is [263] Leong JM, Robbins D, Rosenfeld L, et al. Structural
surface exposed and interacts with the CS1 requirements for glycosaminoglycan recognition by the
heparin-binding domain of human fibronectin. PLoS Lyme disease spirochete, Borrelia burgdorferi. Infect
One. 2013;8(9):e75643. doi: 10.1371/journal.pone. Immun. 1998;66(12):6045–6048. doi: 10.1128/IAI.66.
0075643 12.6045-6048.1998
[250] Coburn J, Magoun L, Bodary SC, et al. Integrins αvβ3 [264] Seshu J, Esteve-Gassent MD, Labandeira-Rey M, et al.
and α5β1 mediate attachment of Lyme disease spiro­ Inactivation of the fibronectin-binding adhesin gene
chetes to human cells. Infect Immun. 1998;66 bbk32 significantly attenuates the infectivity potential of
(5):1946–1952. doi: 10.1128/IAI.66.5.1946-1952.1998 Borrelia burgdorferi. Mol Microbiol. 2006;59
[251] Behera AK, Durand E, Cugini C, et al. Borrelia burgdor­ (5):1591–1601. doi: 10.1111/j.1365-2958.2005.05042.x
feri BBB07 interaction with integrin alpha3beta1 stimu­ [265] Li X, Liu X, Beck DS, et al. Borrelia burgdorferi lacking
lates production of pro-inflammatory mediators in BBK32, a fibronectin-binding protein, retains full
primary human chondrocytes. Cell Microbiol. pathogenicity. Infect Immun. 2006;74(6):3305–3313.
2008;10:320–331. doi: 10.1111/j.1462-5822.2007.01043.x doi: 10.1128/IAI.02035-05
[252] Ristow LC, Bonde M, Lin Y, et al. Integrin binding by [266] Byram R, Gaultney RA, Floden AM, et al. Borrelia
B orrelia burgdorferi P66 facilitates dissemination but burgdorferi RevA significantly affects pathogenicity
is not required for infectivity. Cell Microbiol. and host response in the mouse model of Lyme
2015;17:1021–1036. doi: 10.1111/cmi.12418 disease. Infect Immun. 2015;83(9):3675–3683. doi: 10.
[253] Wood E, Tamborero S, Mingarro I, et al. BB0172, 1128/IAI.00530-15
a Borrelia burgdorferi Outer Membrane Protein That [267] Norman MU, Moriarty TJ, Dresser AR, et al. Molecular
Binds Integrin α3β1. J Bacteriol. 2013;195 mechanisms involved in vascular interactions of the
(15):3320–3330. doi: 10.1128/JB.00187-13 Lyme disease pathogen in a living host. PLOS Pathog.
[254] Verma A, Brissette CA, Bowman A, et al. Borrelia burg­ 2008;4(10):e1000169. doi: 10.1371/journal.ppat.
dorferi BmpA is a laminin-binding protein. Infect Immun. 1000169
2009;77(11):4940–4946. doi: 10.1128/IAI.01420-08 [268] Meriläinen L, Brander H, Herranen A, et al.
[255] Brissette CA, Verma A, Bowman A, et al. The Borrelia Pleomorphic forms of Borrelia burgdorferi induce dis­
burgdorferi outer-surface protein ErpX binds mamma­ tinct immune responses. Microbes Infect. 2016;18(7–
lian laminin. Microbiol (Reading). 2009;55:863–872. 8):484–495. doi: 10.1016/j.micinf.2016.04.002
[256] Bista S, Singh P, Bernard Q, et al. A novel [269] Vancová M, Rudenko N, Vaněček J, et al.
laminin-binding protein mediates microbial-endothelial Pleomorphism and viability of the Lyme disease patho­
cell interactions and facilitates dissemination of Lyme gen Borrelia burgdorferi exposed to physiological stress
30 M. STRNAD ET AL.

conditions: a correlative cryo-fluorescence and [284] Eisenreich W, Rudel T, Heesemann J, et al. How viral
cryo-scanning electron microscopy study. Front and intracellular bacterial pathogens reprogram the
Microbiol. 2017;8:596. doi: 10.3389/fmicb.2017.00596 metabolism of host cells to allow their intracellular
[270] Brorson O, Brorson SH. Transformation of cystic replication. Front Cell Infect Microbiol. 2019;9:42.
forms of Borrelia burgdorferi to normal, mobile doi: 10.3389/fcimb.2019.00042
spirochetes. Infection. 1997;25(4):240–246. doi: 10. [285] Gehre L, Gorgette O, Perrinet S, et al. Sequestration of
1007/BF01713153 host metabolism by an intracellular pathogen. Elife.
[271] Brorson O, Brorson SH. In vitro conversion of Borrelia 2016;5:e12552. doi: 10.7554/eLife.12552
burgdorferi to cystic forms in spinal fluid, and trans­ [286] Linder S, Heimerl C, Fingerle V, et al. Coiling pha­
formation to mobile spirochetes by incubation in gocytosis of Borrelia burgdorferi by primary human
BSK-H medium. Infection. 1998;26(3):144–150. doi: macrophages is controlled by CDC42Hs and Rac1
10.1007/BF02771839 and involves recruitment of wiskott-aldrich syn­
[272] Dunham-Ems SM, Caimano MJ, Eggers CH, et al. drome protein and Arp2/3 complex. Infect Immun.
Borrelia burgdorferi requires the alternative sigma fac­ 2001;69(3):1739–1746. doi: 10.1128/IAI.69.3.1739-
tor RpoS for dissemination within the vector during 1746.2001
tick-to-Mammal transmission. PLOS Pathog. 2012;8 [287] Siryaporn A, Kuchma SL, O’Toole GA, et al. Surface
(2):e1002532. doi: 10.1371/journal.ppat.1002532 attachment induces Pseudomonas aeruginosa
[273] Lantos PM, Auwaerter PG, Wormser GP. A systematic virulence. Proc Natl Acad Sci U S A. 2014;111
review of Borrelia burgdorferi morphologic variants (47):16860–16865. doi: 10.1073/pnas.1415712111
does not support a role in chronic Lyme disease. Clin [288] Carapeto AP, Vitorino MV, Santos JD, et al.
Infect Dis. 2014;58(5):663. doi: 10.1093/cid/cit810 Mechanical properties of human bronchial epithelial
[274] Torres JP, Senejani AG, Gaur G, et al. Ex Vivo Murine cells expressing wt- and mutant CFTR. Int J Mol Sci.
Skin Model for B. burgdorferi Biofilm. Antibiotics. 2020;21(8):E2916. doi: 10.3390/ijms21082916
2020;9(9):528. doi: 10.3390/antibiotics9090528 [289] Patel NR, Bole M, Chen C, et al. Cell Elasticity
[275] Sapi E, Bastian SL, Mpoy CM, et al. Characterization of Determines Macrophage Function. PLoS One. 2012;7
biofilm formation by Borrelia burgdorferi in vitro. (9):e41024. doi: 10.1371/journal.pone.0041024
PLoS One. 2012;7(10):e48277. doi: 10.1371/journal. [290] Costa TRD, Felisberto-Rodrigues C, Meir A, et al.
pone.0048277 Secretion systems in Gram-negative bacteria: structural
[276] Wu J, Weening EH, Faske JB, et al. Invasion of and mechanistic insights. Nat Rev Microbiol. 2015;13
Eukaryotic cells by Borrelia burgdorferi requires β1 (6):343–359. doi: 10.1038/nrmicro3456
Integrins and src kinase activity. Infect Immun. [291] Schulze RJ, Zückert WR. Borrelia burgdorferi lipopro­
2011;79(3):1338–1348. doi: 10.1128/IAI.01188-10 teins are secreted to the outer surface by default. Mol
[277] Klempner MS, Noring R, Rogers RA. Invasion of Microbiol. 2006;59(5):1473–1484. doi: 10.1111/j.1365-
human skin fibroblasts by the Lyme disease spirochete, 2958.2006.05039.x
Borrelia burgdorferi. J Infect Dis. 1993;167 [292] Jan AT. Outer membrane vesicles (OMVs) of
(5):1074–1081. doi: 10.1093/infdis/167.5.1074 gram-negative bacteria: a perspective update. Front
[278] Aberer E, Surtov-Pudar M, Wilfinger D, et al. Co- Microbiol. 2017;8:1053. doi: 10.3389/fmicb.2017.01053
culture of human fibroblasts and Borrelia burgdorferi [293] Anand D, Chaudhuri A. Bacterial outer membrane vesi­
enhances collagen and growth factor mRNA. Arch cles: new insights and applications. Mol Membr Biol.
Dermatol Res. 2018;310(2):117–126. doi: 10.1007/ 2016;33(6–8):125–137. doi: 10.1080/09687688.2017.
s00403-017-1797-1 1400602
[279] Girschick HJ, Huppertz HI, Rüssmann H, et al. [294] Kuehn MJ, Kesty NC. Bacterial outer membrane vesi­
Intracellular persistence of Borrelia burgdorferi in cles and the host-pathogen interaction. Genes Dev.
human synovial cells. Rheumatol Int. 1996;16 2005;19:2645–2655. doi: 10.1101/gad.1299905
(3):125–132. doi: 10.1007/BF01409985 [295] Kudryashev M, Cyrklaff M, Baumeister W, et al.
[280] Ma Y, Sturrock A, Weis JJ. Intracellular localization of Comparative cryo-electron tomography of pathogenic
Borrelia burgdorferi within human endothelial cells. Lyme disease spirochetes. Mol Microbiol. 2009;71
Infect Immun. 1991;59(2):671–678. doi: 10.1128/iai. (6):1415–1434. doi: 10.1111/j.1365-2958.2009.06613.x
59.2.671-678.1991 [296] Karvonen K Mechanisms and elimination of Borrelia
[281] Livengood JA, Gilmore RD. Invasion of human neuro­ burgdorferi persistence in vitro. JYU Dissertations
nal and glial cells by an infectious strain of Borrelia [Internet]. 2022 [cited 2022 Oct 11]. Available from:
burgdorferi. Microbes Infect. 2006;8(14– https://jyx.jyu.fi/handle/123456789/79467
15):2832–2840. doi: 10.1016/j.micinf.2006.08.014 [297] Davis MM, Brock AM, DeHart TG, et al. The
[282] Kerstholt M, Netea MG, Joosten LAB. Borrelia burg­ peptidoglycan-associated protein NapA plays an
dorferi hijacks cellular metabolism of immune cells: important role in the envelope integrity and in the
Consequences for host defense. Ticks Tick Borne Dis. pathogenesis of the lyme disease spirochete. PLOS
2020;11(3):101386. doi: 10.1016/j.ttbdis.2020.101386 Pathog. 2021;17(5):e1009546. doi: 10.1371/journal.
[283] Mayer KA, Stöckl J, Zlabinger GJ, et al. Hijacking the ppat.1009546
supplies: metabolism as a novel facet of virus-host [298] Cluss RG, Silverman DA, Stafford TR. Extracellular
interaction. Front Immunol. 2019;10:1533. doi: 10. secretion of the Borrelia burgdorferi Oms28 porin
3389/fimmu.2019.01533 and Bgp, a glycosaminoglycan binding protein. Infect
VIRULENCE 31

Immun. 2004;72(11):6279–6286. doi: 10.1128/IAI.72. disaccharides and traverse the enzootic cycle. FEMS
11.6279-6286.2004 Immunol Med Microbiol. 2012;66(2):157–165. doi: 10.
[299] Skare JT, Shang ES, Foley DM, et al. Virulent strain 1111/j.1574-695X.2012.00996.x
associated outer membrane proteins of Borrelia [312] Görke B, Stülke J. Carbon catabolite repression in
burgdorferi. J Clin Invest. 1995;96(5):2380–2392. doi: bacteria: many ways to make the most out of
10.1172/JCI118295 nutrients. Nat Rev Microbiol. 2008;6(8):613–624. doi:
[300] Parveen N, Cornell KA, Bono JL, et al. Bgp, a secreted 10.1038/nrmicro1932
glycosaminoglycan-binding protein of Borrelia burg­ [313] Zhang J-J, Ranghunandanan S, Wang Q, et al.
dorferi strain N40, displays nucleosidase activity and Mechanism of repression of the glycerol utilization
is not essential for infection of immunodeficient mice. operon in Borrelia burgdorferi Internet].
Infect Immun. 2006;74(5):3016–3020. doi: 10.1128/IAI. bioRxivAvailable from. 2022 cited 2023 Jun
74.5.3016-3020.2006 12:2022.11.01.514788. 10.1101/2022.11.01.514788v1.
[301] Garon CF, Dorward DW, Corwin MD. Structural fea­ [314] Shaw DK, Hyde JA, Skare JT. The BB0646 protein
tures of Borrelia burgdorferi–the Lyme disease spiro­ demonstrates lipase and hemolytic activity associated
chete: silver staining for nucleic acids. Scanning with Borrelia burgdorferi, the aetiological agent of
Microsc Suppl. 1989;3:109–115. Lyme disease. Mol Microbiol. 2012;83:319–334. doi:
[302] Crowley JT, Toledo AM, LaRocca TJ, et al. Lipid 10.1111/j.1365-2958.2011.07932.x
Exchange between Borrelia burgdorferi and Host [315] Wang X-G, Scagliotti JP, Hu LT. Phospholipid synth­
Cells. PLOS Pathog. 2013;9(1):e1003109. doi: 10.1371/ esis in Borrelia burgdorferi: BB0249 and BB0721
journal.ppat.1003109 encode functional phosphatidylcholine synthase and
[303] Groshong AM, McLain MA, Radolf JD, et al. Host- phosphatidylglycerolphosphate synthase proteins.
specific functional compartmentalization within the Microbiology (Reading). 2004;150(2):391–397. doi: 10.
oligopeptide transporter during the Borrelia burgdor­ 1099/mic.0.26752-0
feri enzootic cycle. PLOS Pathogens. 2021;17(1): [316] Belisle JT, Brandt ME, Radolf JD, et al. Fatty acids of
e1009180. doi: 10.1371/journal.ppat.1009180 Treponema pallidum and Borrelia burgdorferi
[304] LaRocca TJ, Crowley JT, Cusack BJ, et al. Cholesterol lipoproteins. J Bacteriol. 1994;176(8):2151–2157. doi:
lipids of Borrelia burgdorferi form lipid rafts and are 10.1128/jb.176.8.2151-2157.1994
required for the bactericidal mechanism of a [317] Drecktrah D, Hall LS, Crouse B, et al. The
complement-independent antibody. Cell Host Microbe. glycerol-3-phosphate dehydrogenases GpsA and GlpD
2010;8(4):331–342. doi: 10.1016/j.chom.2010.09.001 constitute the oxidoreductive metabolic linchpin for
[305] Jain S, Showman AC, Jewett MW, et al. Molecular Lyme disease spirochete host infectivity and persistence
Dissection of a Borrelia burgdorferi In Vivo Essential in the tick. PLOS Pathog. 2022;18(3):e1010385. doi: 10.
Purine Transport System. Infect Immun. 2015;83 1371/journal.ppat.1010385
(6):2224–2233. doi: 10.1128/IAI.02859-14 [318] Purser JE, Lawrenz MB, Caimano MJ, et al. A
[306] Pappas CJ, Iyer R, Petzke MM, et al. Borrelia burgdor­ plasmid-encoded nicotinamidase (PncA) is essential
feri requires glycerol for maximum fitness during the for infectivity of Borrelia burgdorferi in a mammalian
tick phase of the enzootic cycle. PLOS Pathog. 2011;7 host. Mol Microbiol. 2003;48(3):753–764. doi: 10.1046/
(7):e1002102. doi: 10.1371/journal.ppat.1002102 j.1365-2958.2003.03452.x
[307] Corona A, Schwartz I, Conway T, et al. Borrelia burg­ [319] Schüler W, Bunikis I, Weber-Lehman J, et al. Complete
dorferi: Carbon metabolism and the tick-Mammal genome sequence of Borrelia afzelii K78 and compara­
enzootic cycle. Microbiol Spectr. 2015;3(3):10.1128/ tive genome analysis. PLoS One. 2015;10(3):e0120548.
microbiolspec.MBP-0011–2014. doi: 10.1128/microbiol doi: 10.1371/journal.pone.0120548
spec.MBP-0011-2014 [320] Schutzer SE, Fraser-Liggett CM, Qiu W-G, et al.
[308] Skare JT, Mirzabekov TA, Shang ES, et al. The Oms66 Whole-Genome Sequences of Borrelia bissettii,
(p66) protein is a Borrelia burgdorferi porin. Infect Borrelia valaisiana, and Borrelia spielmanii.
Immun. 1997;65(9):3654–3661. doi: 10.1128/iai.65.9. J Bacteriol. 2012;194(2):545–546. doi: 10.1128/JB.
3654-3661.1997 06263-11
[309] Skare JT, Champion CI, Mirzabekov TA, et al. Porin activ­ [321] Kingry LC, Batra D, Replogle A, et al. Whole genome
ity of the native and recombinant outer membrane protein sequence and Comparative genomics of the novel
Oms28 of Borrelia burgdorferi. J Bacteriol. 1996;178 Lyme borreliosis causing pathogen, Borrelia mayonii.
(16):4909–4918. doi: 10.1128/jb.178.16.4909-4918.1996 PLoS One. 2016;11(12):e0168994. doi: 10.1371/journal.
[310] von Lackum K, Stevenson B. Carbohydrate utilization pone.0168994
by the Lyme borreliosis spirochete, Borrelia [322] Rego ROM, Trentelman JJA, Anguita J, et al.
burgdorferi. FEMS Microbiol Lett. 2005;243 Counterattacking the tick bite: towards a rational
(1):173–179. doi: 10.1016/j.femsle.2004.12.002 design of anti-tick vaccines targeting pathogen
[311] Hoon-Hanks LL, Morton EA, Lybecker MC, et al. transmission. Parasites Vectors. 2019;12(1):229. doi:
Borrelia burgdorferi malQ mutants utilize 10.1186/s13071-019-3468-x

You might also like