Next Article in Journal
Whole-Genome Profile of Greek Patients with Teratozοοspermia: Identification of Candidate Variants and Genes
Next Article in Special Issue
Unravelling Metagenomics Approach for Microbial Biofuel Production
Previous Article in Journal
A Novel 3-Gene Signature for Identifying COVID-19 Patients Based on Bioinformatics and Machine Learning
Previous Article in Special Issue
Vacuolal and Peroxisomal Calcium Ion Transporters in Yeasts and Fungi: Key Role in the Translocation of Intermediates in the Biosynthesis of Fungal Metabolites
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Large-Scale Sequencing of Borreliaceae for the Construction of Pan-Genomic-Based Diagnostics

by
Kayla M. Socarras
1,2,3,
Benjamin S. Haslund-Gourley
3,
Nicholas A. Cramer
4,5,
Mary Ann Comunale
3,
Richard T. Marconi
4,5 and
Garth D. Ehrlich
1,2,3,4,6,*
1
Center for Advanced Microbial Processing, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19102, USA
2
Center for Genomic Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19102, USA
3
Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
4
Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, 1112 East Clay Street, Room 101 Health Sciences Research Building, Richmond, VA 23298, USA
5
Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA
6
Center for Surgical Infections and Biofilms, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19102, USA
*
Author to whom correspondence should be addressed.
Genes 2022, 13(9), 1604; https://doi.org/10.3390/genes13091604
Submission received: 4 August 2022 / Revised: 3 September 2022 / Accepted: 4 September 2022 / Published: 8 September 2022
(This article belongs to the Special Issue Feature Papers in Microbial Genetics)

Abstract

:
The acceleration of climate change has been associated with an alarming increase in the prevalence and geographic range of tick-borne diseases (TBD), many of which have severe and long-lasting effects—particularly when treatment is delayed principally due to inadequate diagnostics and lack of physician suspicion. Moreover, there is a paucity of treatment options for many TBDs that are complicated by diagnostic limitations for correctly identifying the offending pathogens. This review will focus on the biology, disease pathology, and detection methodologies used for the Borreliaceae family which includes the Lyme disease agent Borreliella burgdorferi. Previous work revealed that Borreliaceae genomes differ from most bacteria in that they are composed of large numbers of replicons, both linear and circular, with the main chromosome being the linear with telomeric-like termini. While these findings are novel, additional gene-specific analyses of each class of these multiple replicons are needed to better understand their respective roles in metabolism and pathogenesis of these enigmatic spirochetes. Historically, such studies were challenging due to a dearth of both analytic tools and a sufficient number of high-fidelity genomes among the various taxa within this family as a whole to provide for discriminative and functional genomic studies. Recent advances in long-read whole-genome sequencing, comparative genomics, and machine-learning have provided the tools to better understand the fundamental biology and phylogeny of these genomically-complex pathogens while also providing the data for the development of improved diagnostics and therapeutics.

1. Introduction

Even in this era of modern medicine, including mass vaccination achievements and antibiotic treatment regimens, vector-borne diseases such as Lyme borreliosis, Malaria, Dengue fever, yellow fever, and bubonic plague still prevail. This group of ancient and persistent diseases is transmitted to humans through the bite of infected arthropod vectors including mosquitoes, lice, flies, and ticks. These vectors acquire their associated pathogens through blood feedings from multiple hosts throughout complex life cycles. This life-cycle complexity leads to difficulties in diagnoses and treatments, creating a persistent healthcare burden that has resulted in a push to identify biomarkers for the development of improved diagnostics, vaccines, and therapeutics [1].
Research on these fronts has been divided unevenly across different vector species. While a great deal of research has been performed on diseases transmitted by mosquitoes, tick-borne diseases (TBD) are less studied. As their name implies, TBDs are primarily transmitted through the bite of infected ticks. While these small ectoparasitic arachnids have long been documented to cause disease, some as early as 1550 B.C., their true clinical significance was not realized until 1893, when a publication by Smith and Killborne linked Rhipicephalus annaltus to the transmission of the protist parasite Babesia bigemina in cattle [2,3,4,5,6]. Despite this finding, research on ticks and their corresponding TBDs progressed slowly until the latter half of the 20th century.
Moreover, research in the field progressed on TBDs unevenly, often centering on the most prevalent pathogen (or what was thought to be the most prevalent pathogen) or vector in a given region rather than the wide array of TBD pathogens and their vectors. This is most clearly seen with Lyme borreliosis (Lyme disease), caused by the Borreliella burgdorferi sensu lato spirochetes and their known vectors, Ixodes scapularis and I. pacificus, within the Northeastern and Pacific regions of the United States (US), respectively. Research efforts initially centered on the mammalian immune response towards these spirochetes, leading to developments of first-generation diagnostics that have some efficacy but are fraught with poor sensitivity [7,8]. While this work is important, more research is needed to improve the efficacy of Lyme borreliosis diagnostics, including biomarker discovery, nucleic-acid-based methods, and improved serological methods. This review will cover the TBD field as it stands, with emphasis on borreliosis, specifically Lyme borreliosis, and outline plausible paths for the development of better diagnostics and therapeutics.

2. The Macro and Micro Ecology of Ticks: Their Role in the Environment and Their Microbiomes

The Agrasidae have 186 recognized species and are commonly referred to as ‘soft’ ticks due to the absence of a scutum, a hard protective plate, and the exposure of their mouths [9]. The second, larger Ixodidae tick family has 720 recognized species. These arachnids are commonly known as ‘hard’ ticks due to the presence of their scutum (Figure 1). While both families have distinct morphologies, all can thrive in various geographic niches ranging from the tropics to the subarctic [10]. Though many tick species are found in a range of climates, most tick species are concentrated in tropical/subtropical regions [10].
Recently, due mainly to climate change, the geographic range of many tick species has expanded poleward beyond their traditional habitats [11,12]. This restructuring has resulted from a variety of reasons including prey migration, local environmental degradation, and increased temperatures associated with global warming [11,12,13,14]. The latter facilitates increased tick survival through winters within previously uninhabitable regions [15,16,17,18,19,20]. Evidence of this change is seen in numerous tick species. A tick species that is able to live in both hemispheres and extreme climates is Ixodes uriae, a seabird tick [21,22] that is now prevalent in Nordic Countries and can transmit Borrelia burgdorferi, Borrelellia garinii, and Borrelia bavariensis [23,24,25]. Similarly, Haemaphysalis longicornis, the Asian Longhorn tick, has recently arrived in the US [26,27]. This invasive species has rapidly integrated within its new ecosystem and has joined the list of clinically relevant tick species found within the US (Table 1) [26,28]. Additionally, several native North American tick species are also migrating further north. Some have been documented to parasitize wildlife in the harsh frontiers of Alaska [29,30].
As the habitat zones of ticks change, so does the epidemiology of the pathogens they transmit [2]. This review will focus on the most significant tick species that carry human pathogens (Table 1) including I. scapularis (black-legged tick) and its west-coast sister species I. pacifius (western black-legged tick); Dermacentor variabilis (American dog tick); and Amblyomma americanum (Lone star tick).
The most well-known of these is I. scapularis. This species is fascinating for its biology and its role in medical history. The clinical significance of the deer tick was realized when it was linked with outbreaks of a rheumatoid arthritis-like disease in children in Lyme, Connecticut [31]. This mysterious outbreak lasted for years and went well into the onset of the AIDS epidemic. It was originally postulated as a viral disease [32,33]. In the years that followed, this was disproven when a ‘treponema-like’ spirochete was isolated from the blood of local fauna and afflicted individuals [34,35,36,37]. This spirochete was named Borrelia burgdorferi in honor of the lead scientist Dr. Wilhelm Burgdorfer [38]. In the years that followed, the maladies caused by B. burgdorferi were called Lyme disease, now referred to as Lyme borreliosis (Lb) to specify the clinical infection [36,37]. Throughout the remainder of this review, Lb shall be the predominant term used.
Despite identifying the Lb etiological agent, its true vector remained unknown. Early investigations for the Lyme borreliosis vector focused on the prevalent New England tick species known to transmit pathogens including D. variabilis and Ixodes dammini. The latter Ixodes species was first hypothesized as the host of B. burgdorferi—a concept later proven true after successful isolation of the spirochete [34]. At the time, I. dammini was the sole known vector. Soon, this finding was contested based on growing evidence of Lb beyond the boundaries of New England. Subsequently, it was demonstrated that I. dammini was not a separate species from I. scapularis and that the use of I. dammini as junior subjective of I. scapularis should be avoided to minimize confusion in the research and medical communities [39,40]. Later studies on other Ixodes species then highlighted I. pacificus as a western vector [41,42,43].
Once I. scapularis was recognized as a vector of interest, it was critical to garner more information about its underlying biology and how it impacted pathogen transmission. From these studies, it was determined that I. scapularis required the successful completion of a blood meal to progress through each of its life stages (larva, nymph, adult) [44,45]. During feeding, the ticks obtain the necessary nutrients for growth and survival and acquire microbes that assist in these functions [2,46,47]. This need for feeding for growth and survival is most evident during the initial stages of life. At the nymphal stage, I. scapularis ticks will primarily feed on small, woodland creatures like Peromyscus leucopus, the white-footed mouse, during which it may acquire B. burgdorferi (Figure 2) [16,48,49,50,51,52]. Additionally, other tick-borne pathogens may be acquired from blood meals such as Anaplasma, Bartonella, Ehrlichia, and Babesia spp. [48,53,54,55]. As I. scapularis nymphs molt into adults they may become more species restrictive and feed on larger prey like Odocoileus virginianus, the white-tailed deer, or Ursus americanus, the American black bear [56,57,58]. Additionally, they may also prey upon non-competent hosts such as reptiles, or on incidental hosts such as domestic animals, wildlife, and humans [59,60,61]. Historically, I. scapularis was most prevalent along the eastern seaboard of North America (Table 1), but with climate change its range has expanded both northward and westward.
Another hard tick species, D. variabilis, thrives on many hosts during all of its life stages. Like Ixodes ticks, this hard tick preys on small mammals early in its life cycle before parasitizing larger prey as it matures. While this vector was known as a North American anthropophilic ectoparasite, scientific interest rose with the sudden emergence of Rocky Mountain Spotted Fever outbreaks earlier in the 20th century [33,51,62]. Compounded with the precipitous upsurge of Lb, researchers initially considered this arachnid, and its close cousin D. andersonii, as potential vectors of the disease. In later transmission studies, however, both ectoparasites were disqualified as being capable of transmitting the etiological agents of Lb, and to date, no Borrelia spirochetes have been detected within Dermacentor spp. ticks. Due to the nature of this tick and other TBD pathogens it has been known to carry, it remains clinically relevant and is found ubiquitously throughout North America (Table 2).
Lastly, the most significant tick on the western North American seaboard is I. pacificus [79,80]. Similar to its east coast cousin, I. scapularis, I. pacificus is a generalist feeder that thrives on a variety of hosts (Table 1). In doing so, it can acquire and transmit a broad range of microbes, including the Borrelial spirochetes (Table 2) [81,82,83]. I. pacificus has been found to quest greater distances than other tick species during all life stages [84]. This facilitates nymph and adult I. pacificus to better track prey for blood meals.
As mentioned, each of the discussed tick species are vectors for multiple TBD pathogens, and their acquisition/transmission of each microbe is heavily influenced by their food supply, life stage, and environment. The community of these microbes within a metazoan is called the microbiome. The term refers to commensal, symbiotic, and pathogenic microbes that coexist in a defined space [85]. The tick microbiome includes viruses, eubacteria, archaea, and eukaryotes, and together with metazoan hosts they form a holobiome [86,87,88,89]. Until very recently, microbiome studies focused solely on the bacterial components of the microbiome due to the ease of targeting the bacterial 16S rRNA gene, which is conserved throughout the entire domain. This high degree of sequence conservation permits the identification of all species within the domain through a single PCR amplification followed by DNA sequencing. However, earlier approaches lacked specificity due to technical limitations.
Previously, many bacteria within the tick microbiome have been found critical for development, survival, the balancing of complex metabolic pathways, and the execution of numerous functions of the host they preside in [51,90]. Thus, many of the most significant microbiome exist in mutualistic relationships with their hosts. In ticks, these bacteria include members of the Rickettsia, Rickettsia-like, and Wolbachia genera [51,91]. Their presence/absence impacts their overall morphology, feeding habits, and retention of other microbes [51,91,92]. This is most clearly observed among tick-borne organisms like Coxiella-like genera that are essential for the survival and reproduction of A. americanum [93,94,95]. Similarly, Rickettsia-like spp. are necessary for folic acid biosynthesis within I. scapularis and I. pacificus, and the Wolbachia spp. for reproduction [93,94,95].
In early studies, researchers found that the tick microbiome was subject to change based on its feeding status and environmental stressors. With the advent of next-generation sequencing (NGS), the small nuances behind such changes and the impact of a given host on the microbial consortium became clearer. In these studies, I. scapularis was found to host a wide range of bacterial species within their microbiomes [51,90]. This complex bacterial microbiome was noted to be quite variable in terms of both breadth and diversity based on the life stage [91,96,97]. Additionally, it was noted to change based upon feeding patterns and sex [98,99]. In previous studies of I. scapularis adults, both male and female ticks were noted to have the Rickettsia and Rickettssia-like spp. as predominant members of their microbiome [91,96,99]. Other tick species, including D. variabilis and A. americanum, were not found to be able to sustain such a wide net of bacterial organisms [100,101,102]. Interestingly, while the breadth of the microbiome was different, a similar decrease in diversity occurred among all tick species as they progressed throughout their lifecycles [102,103,104,105].
Recently, the I. pacificus microbiome has also been a subject of research. Like, I. scapularis, it has a diverse microbiome [83,102]. However, it has yet to be determined what role its microbiome members play in the etiology of human infections. Previous efforts in characterizing the I. pacificus microbiome have identified common commensal bacteria including Spiroplasma ixodetis as well as various Rickettsia spp., Rickettsia-like spp., Ehrlichia-like, and Anaplasma-like spp. [106]. Additionally, the major human pathogen as Borreliella burgdorferi and Borrelia miyamotoi have also been found at reduced levels within this tick population [106].
While research on tick microbiomes has improved our understanding of the various TBD pathogens, there are several limitations in most studies. The first is that standard short-read sequencing in NGS systems lack the resolution necessary to identify the bacteria at the species-level. Next is the dearth of applying these technologies to elucidate the full nature of the microbial consortia within ticks. Finally, these platforms do not target microorganisms such as fungi, nematodes, and apicomplexan parasites. Thus, much remains to be elucidated regarding to the various tick microbiomes.

3. Borreliosis, the Most Common Type of Tick-Borne Disease

The emergence of chronic cutaneous, neurologic, arthritic, and cardiac maladies have been documented worldwide for hundreds of years [31,107,108,109,110,111,112]. In the US, these outbreaks have occurred in waves for centuries. Some of the earliest mentions were documented in Long Island during the early 1600s [113]. These cases were called ‘Montauk’s knees’, ‘Southhampton knee’, or water on the knee due to the arthritic-like symptoms [114]. In the latter half of the twentieth century, another wave occurred at Lyme, Connecticut in the beginning of the 1970s. In this small community, several children presented with numerous non-specific and arthritic-like symptoms. These ailments were later formally recognized as Lyme borreliosis and is commonly referred to as Lyme disease [107,108,109,110,111,112]. Over the years, Lb has continually increased in prevalence within the US and Canada and is now the most common tick-borne disease, accounting for ~500,000 new cases each year in the US alone—due in large part to climate change, deforestation, habitat loss, and loss of predators of the primary mammalian species upon which the ticks feed [115].
In the years that followed the discovery of Lb, its etiological agent, Borreliella burgdorferi, and other Borreliella spirochetes were found to be pathogenic to humans [38,116,117,118,119,120]. Some of these spirochetes do not, however, have the same vectors or pathogenesis. In this instance, other Borreliaceae members can cause Relapsing fever (Rf) and may be transmitted through a tick or louse vector. While these distinctions were made primarily on pathology and geo-locale of origin, later comparative genomic research suggested splitting the genus into two distinct disease-causing genera [121]. The Lb causing spirochetes were then given the new designation of Borreliella, while all other Borreliaceae spirochetes which cause Rf retained its original name of Borrelia.
Lb is a multi-systemic infectious disease with a wide and seemingly unconnected variety of conditions (e.g., polyarthralgias; neurological diseases, including polydysthesias/parathesias, cardiomyopathy, multiple sclerosis, other demylenating diseases, and ataxia; and psychiatric conditions, such as pediatric bipolar disorder and PANS and PANDAS) [122,123,124,125,126,127,128,129,130]. The sole pathognomonic presentation of Lb is erythema migrans, commonly known as the bulls-eye rash. Unfortunately, this presentation may not occur or is not visible to all individuals, occurring in approximately 50% of Lb cases [131,132,133,134]. Lb is divided into distinct stages: localized and disseminated. The disease presentations vary wildly among individuals, as well as by the species of Borreliella. This is illustrated most clearly with common Borreliella spirochetes, B. burgdorferi, B.azfelii, and B. garinii, each of which is endemic to the US or Europe. B. burgdorferi, the most common cause of Lb in the US, is primarily associated with arthritis, while B. afzelii is associated with cutaneous infections and B. garinii with neurological disease in Europe [133].
Due to the spectrum of non-specific symptoms for Lb, diagnoses are often difficult. Currently, clinicians rely on imprecise serological diagnostics and proof of tick-bite before accepting a Lb differential. While the above approaches may be useful in some instances, these current diagnostics have severe limitations, including a highly unreliable negative predictive value. To understand why these diagnostics may fail, it is critical to understand the basic biology of these spirochetes.
Previously, researchers have noted that Borreliellal spirochetes share many features ranging from their obligate parasitic nature within a large network of reservoir hosts, rather organisms that sustain spirochetes and facilitate their reproduction, to dynamic morphology that facilitates their near-constant host invasion [38,135]. Their unique morphology is thought to be created by 11 anti-parallel inter-membrane flagella and a chitobiose peptidoglycan [136,137]. Interestingly, this morphology has been documented to change in response to varying external stimuli [138,139,140,141,142]. It is, however, unclear what the mechanisms underlying the Borreliaceae morphological shifts are.
In addition to altering their morphology as a stress response, Borreliaceace spirochetes can manipulate their host’s immune and inflammatory response to their advantage. This is most clearly seen within I. scapularis ticks where Borreliella spp. reside within the tick midgut. These spirochetes are bound to the tick receptor for OspA (TROSPA) until the initiation of a blood meal [143]. Through feeding, the Borreliella dissociate from TROSPA, then switch their outer membrane surface protein (Osp) composition. The act of feeding induces tick salivary proteins to cover presenting Borreliella OspA and translocate to the tick salivary gland before peritrophic membrane formation [144,145]. Once in the salivary gland, the spirochete can then be transmitted into the new host dermis. During transmission, the Borreliella OspA in the outer membrane decreases and OspC rises [144]. For humans, Borreliella/Borrelia can be transmitted at varying rates depending on the tick species, tick feeding status, microbial strain, and microbial load, e.g., an I. scapularis tick can transmit B. burgdorferi within 24–48 h of initiating blood feeding [146,147,148].
Once Borrelieceae spirochetes have successfully entered the human body, they can persist within the dermis before disseminating. There are two proposed dissemination methods for Borreliella spirochetes: the hematogenous and non-hematogenous routes including the lymphatics or tissue [149]. In both dissemination routes, the spirochetes mitigate the host immune response to prevent recognition by the innate immune system and ultimately delay and distort the development of a T-cell-dependent B-cell response [49,150,151,152,153]. In addition, Borreliaceae spirochetes, can also evade the host immune system through various other means [154]. Both Borrelieceae can utilize the antigenic variation system, vls, present within the genome to evade the complement cascade. Additionally, Borreliella can achieve complement evasion by binding to Factor H, a negative regulator of host complement, to outer membrane proteins CspA, CspZ, and OspE [155,156,157]. Borreliella can also inhibit the classical complement pathway by binding C1r to outer membrane protein BBK32 [158,159]. Through antigenic switching of Borreliella outer membrane proteins, including hypervariable OspC and BBA70, the overall outer membrane composition and pathogenesis of the spirochete can be altered in situ [160,161]. Through these virulence mechanisms, it is believed that if Borrelieceae spirochetes are not successfully cleared by the immune system, they may colonize host tissues to form a persistent infection.
While Lyme borreliosis has become highly prevalent, the impact of elapsing fever (Rf) still remains a significant health concern. The RF-causing Borrelia genus can be transmitted either by ticks or lice around the world [154,162,163,164]. Within the United States, this infectious disease has remained endemic solely within western mountainous regions [164]. Regardless of geo-locale of origin, all variants of Rf have the same symptomology. The illness does have nonspecific symptoms like fatigue, headache, nausea, and muscle/joint aches [165,166]. Important diagnostically, however, is that uniquely induces periodic fever spikes associated with Borrelial septicemias. The fever dissipates during periods of time when there are decreased levels of Borrelia present within the blood but return on a cyclical basis over the course of weeks. Due to the elevated numbers of Borrelia within the blood, Rf is commonly diagnosed through microscopic examination of blood smears (Figure 3).

4. Borreliaceae Diagnostics

In the US, the cost of preventing and treating Lb has been estimated to range from $712 million to $1.3 billion per year, but this is likely a gross underestimate as many Lb patients go undiagnosed for years while seeking care for their ‘nonspecific’ symptoms [167]. Often, patients may pay out of pocket for additional diagnostic tests and treatments. In contrast, acute diagnostics for other common bacteria such as Streptococcus, Treponema pallidum, and Staphylococcus infections are accurate and lead to effective treatment before the bacteria can progress to later stages of infection [168,169,170,171,172]. Meanwhile, the economic impact of Lb infections continues to rise in large part due to an inadequate diagnosis. Thus, it is critical to develop and implement better diagnostics, prognostics, and therapeutics for borrelioses [7,173].
Many factors contribute to acute Lb diagnoses being missed. For example, persons with darker skin pigmentation will often not display a visible EM rash, others lack access to medical care or only have non-specific symptoms of acute Lb, while still others have a non-traditional EM rash that is not recognized during the acute phase of Lb [132,174,175]. If a patient suspected of acute Lb presents to healthcare providers, clinicians will assess risk factors for contracting Lyme borreliosis, including symptom presentation timing, geographic location, recent travel history, pet ownership, and history of other TBDs or rashes [176]. Laboratory-based tests are then utilized by clinicians to confirm a suspected acute case of Lyme borreliosis. Currently, there are several indirect and direct approaches to assist in diagnosing an individual with Lb. The Center for Disease Control and Prevention (CDC) recommends a two-tiered serological (ELISA and Western blot) system to confirm a suspected Lyme borreliosis case [177]. The two-tiered approach relies on a patient’s adaptive immune response towards transiently expressed surface proteins of Borreliella. Producing an IgG antibody response with strong avidity towards specific antigen targets takes between 2–3 weeks following infection [177]. Furthermore, most Lb western blots utilize B. burgdorferi sensu stricto strain B31 (Bb B31) as the source of the proteins utilized in their assay [178]. The B31 subtype was isolated over 30 years ago and the Bb B31 antigens do not represent other Borreliella antigens produced from other closely related Lb causing spirochetes [7,179]. Thus, patients who seroconvert during acute Lb infection could produce antibodies targeting antigens that are not included on the standard western blot. In addition, these serology-based diagnostic approaches cannot serve as prognostics to track treatment outcomes. This forces physicians to primarily rely on a patient’s symptoms to guide clinical outcomes or antibiotic treatment efficacy studies [180].
Serologic Lb diagnostics are further complicated by variation in the human adaptive immune response. If patients are diagnosed with Lb based on an EM rash and antibiotic treatment is promptly initiated, they might not seroconvert [181,182]. This fact further complicates the surveillance and confirmation of Lb. The lack of seroconversion could be due to Bb’s profound immunomodulatory and immunosuppressive effects which depend on the combination of host and pathogen genetics [152,183]. Accurate diagnosis of Lb is further complicated when a patient is co-infected with other Borreliellal spp. or additional tick-borne disease pathogens that are also commonly transferred from the tick’s mid-gut [7,184,185,186,187,188]. Taken together, the average sensitivity of the Lb two-tiered test for the acute Lb is less than 50% [178]. This poor sensitivity produces high rates of false-negatives and delays treatment which contributes to the development of chronic/late-stage Lb. During late-stage Lb, such as Lyme carditis or Lyme arthritis, a two-tiered test can confirm the diagnosis of the patient with high sensitivity. Unfortunately, patients in the later stages of Lb face permanent tissue damage and require longer antibiotic treatments [189,190].
Newer serologically-based diagnostics present recombinantly expressed surface proteins from multiple pathogens and strains of Borreliella [185,191]. These approaches increase the chance of detecting antibodies produced towards B. burgdorferi strains other than B31 or identifying co-infections. These methods, however, still have their limitations, as Bb is immunosuppressive and, depending on the infecting strain and host genetics, a significant percentage of infected persons will fail to appropriately produce antibodies.
Clinicians seeking diagnosis for suspected Lb patients may venture beyond CDC guidelines. Traditional pathologic assays such as dark-field microscopy and primary culture from blood or skin biopsies have poor sensitivity and are not employed as a reliable diagnostic for Lb, however they have a very high positive predictive value [192]. Attempting to culture or detect Borreliella spirochetes from human tissue biopsies using PCR methods (standard or quantitative) is invasive and insensitive [7,193,194,195,196,197]. These direct methods are limited by the low spirochete load in tissues and the bloodstream, unlike many other human bacterial pathologies [7,185,186,198,199,200]. Thus, patients and clinicians require alternative methods of acute Lb diagnosis.
In contrast to the limitations of above-mentioned PCR-based methods for detecting Borreliella DNA within humans, Next-Generation Sequencing (NGS) approaches can be highly specific. As Borreliella spirochetes are rarely present in the blood after initial disease onset, the challenge for NGS is to obtain enough of a sample to confidently detect genes associated with acute Lb bacteria [8,201]. Previously, many Borreliella NGS approaches targeted highly conserved genes throughout the genera such as the ribosomal 16S in ticks or human samples [202]. However, this approach was limited to identifying Lb within ticks rather than humans due to low titers of Borreliella in the bloodstream.
In a new NGS approach, the limitations of the sample sources may be circumvented by using patient urine [203]. While most NGS-based assays are limited by the low counts of Borreliella genomic material present in human samples such as blood, this approach aims to ensure a higher Borreliella DNA yield with claims that Lb bacteria infect the kidneys. While accuracy was stated to be ‘superior’ to the standard two-tiered testing approach, the sensitivity of the test has yet to be reported in the literature. This is slightly different than previous diagnostic iterations which used the same biosample but targeted solely OspA, a protein which would not be expressed in high quantities on the outersurface of Borreliaceae within a mammalian host [204].
Other efforts have been made to increase the sensitivity of Lb bloodborne detection. Traditional PCR-based diagnostics for Lyme borreliosis have been improved by isothermal amplification of DNA, followed by PCR amplification of Borreliellal DNA, which is then detected by electrospray ionization mass spectrometry (PCR/ESI-MSI). In PCR/ESI-MSI, it was possible to detect the presence of B. burgdorferi in 13 of 21 blood samples from patients with an acute Lb cases confirmed with positive serology and a history of at least one EM rash [8,200]. The assay required 1.25 mL of EDTA-treated whole blood and could detect 0.6 or greater copies of Borreliella genomes in whole blood.
In a follow-up study, the PCR/ESI-MSI method attempted to survey the presence of B. burgdorferi sensu stricto within four patients during their antibiotic treatment for Lyme borreliosis [205]. In this study, the investigators increased the blood volume from 1.25 mL to 20 mL, with the aim of increasing the diagnostic sensitivity. B. burgdorferi genes were detected in 2 of the 4 patients acutely infected with the aforementioned spirochete. In addition, they did not determine if the increased blood volume increased sensitivity. The genomic amplification approach relied on detecting and targeting conserved genes present within the Borreliella genome such as rpoC, FlaB, and OspC [206]. While such targets can indicate the presence of this spirochetal genus and may provide species-level resolution, there are some complications [205]. One such complication is that use of consistently expressed proteins like OspC may be insufficient due to the protein’s high diversity.
Another NGS diagnostic approach utilizes unbiased metagenomic cell-free DNA sequencing of human plasma. This cell-free DNA (cfDNA) approach was used to detect B. burgdorferi DNA from 64% (18 of 28) human plasma samples during acute Lb [207]. The cfDNA sequencing method’s sensitivity was further improved by combining the results of the modified two-tiered serology testing to identify 86% of acute Lyme borreliosis cases. Additionally, a recent NGS detection study identified core genes within the Borreliella pangenome to increase the sensitivity of DNA-based Borreliaceae diagnostics [208]. Taken together, genomic Borreliella detection methods have significantly improved over the last decade. However, more work is required to deliver a robust and sensitive diagnosis for patients and clinicians. One possibility is to use innate immune proteins that recognize specific PAMPs, such as Apolipoprotein H linked to paramagnetic beads, to ‘sweep’ a much larger volume of blood [209,210,211,212,213].
Next-generation sequencing has been used to detect host responses to acute Lb rather than attempting to directly detect the Borreliellal genome. Sequencing human T-cell receptors (TCRs) is a novel approach to Lyme disease diagnostics and began clinical trials in 2021 [214]. T-cells respond to Lb infection earlier than B-cells can produce antibodies, and thus the expansion of Borreliellal-specific T-cell receptor sequences in a patient’s circulating lymphocytes has the potential to confirm acute cases of Lyme disease earlier than traditional serologic methods [151]. This TCR immuno-sequencing assay differentiated acute Lb patients from healthy controls with a sensitivity of 54%, while the standard two-tiered serological testing approach had a sensitivity of only 30%. Clearly, human T-cell responses significantly vary between patients and this approach will not detect all acute Lyme disease cases. However, TCR immuno-sequencing assay’s increased sensitivity is a move in the right direction and has the potential to be combined with other diagnostic approaches to further increase sensitivity.
In addition to genomic detection, researchers have explored xenodiagnosis, metabolomics, and biomarker profiling [215,216,217]. Xenodiagnostics use an uninfected, natural vector for the isolation of the targeted pathogen from the infected host [218]. In the case of Borreliellal spirochetes, ticks facilitate the reacquisition of Borreliella from a variety of hosts during a 24-h feeding cycle [219,220]. This feature was noted in the past with Lyme-infected monkeys and mice but, was not substantiated in humans until 2014 [221,222,223]. Over the last 8 years, a clinical trial of tick-based recapture of Borreliellal pathogens from infected human hosts has been underway [218]. No results from this study have been released at this time. This approach could be further bolstered by applying NGS to characterize the pathogens recaptured after the tick feeds on the patient suspected of contracting Lyme borreliosis to increase diagnostic sensitivity. It is important to note, that while it could prove useful, much more work would be necessary to make it feasible as a diagnostic.
Metabolomic analyses of Lyme borreliosis patients has also made great strides in recent years. These studies have identified altered abundances of circulating metabolites produced by host tissues during Lyme borreliosis. A recent assay was able to discriminate between acute Lyme borreliosis and uninfected controls using their metabolic profiles [224,225]. Diagnostics relying on specific metabolic profiles are limited by the time and cost associated to prepare samples for analysis but offer yet another promising avenue for future Lyme borreliosis diagnostics.
Proteomic studies of serum collected from humans afflicted with Lb by Zhou et al. identified host acute phase protein abundance alterations during acute Lyme borreliosis [217]. The abundance of proteins—APOA4, C9, CRP, CST6, PGLYRP2, and S100A9—were validated using a second sample set of acute Lyme borreliosis samples and discriminated between healthy controls and acute Lb patients with a 78% sensitivity. Developing a multiplexed ELISA to identify acute-phase proteins associated with Lyme borreliosis could yield a high-throughput diagnostic, yet the issue of cross-reactivity with other infection markers must first be addressed. “Mimic diseases” such as rheumatoid arthritis or fibromyalgia often have similar acute phase protein alterations. Thus, the Lb proteomic study should be validated for Lb-specificity by testing against a panel of sera from patients with other mimic diseases [224].
Lastly, a glycoproteomic approach using MALDI-FT-ICR mass spectroscopy has been demonstrated to detect changes in the IgG N-glycan profile during acute Lyme disease with a sensitivity of 75% and specificity of 100%. Moreover, this assay can differentiate between acute Lb cases and patients who have received successful doxycycline treatment [226].
In summary, Borreliella diagnostics are improving, but have a long way to go as each has strengths, as well as limitations [178]. In this era of increasing TBDs, the best path forward may be to combine multiple diagnostics to complement the strengths of each method to construct a testing protocol that is highly sensitive and specific. In doing so, better measures can be taken to initiate early treatments and prevent chronic disease progression.

5. The Borreliella and Borrelia Genomes

This near-constant cycle of host transmission, acquisition, and host immune evasion suggests that all Borreliaceae spirochetes would contain a large, complex pan-genome. This hypothesis was challenged when the first B. burgdorferi sensu stricto genome was sequenced in the late 1990s [227]. It showed that B. burgdorferi held a small singular linear chromosome and several linear and circular episomes and prophages. These findings were echoed in later sequencing projects in other Borreliella spp. Additionally, early B. burgdorferi genomic studies, revealed many housekeeping genes were contained on the chromosome, progressing from the center [227,228]. Within them were genes for ribosomal machinery, 32 transfer (t)RNAs, and tandem repeats. Surprisingly, genes necessary for de novo biosynthesis of carbohydrates, amino acids, nucleosides, and lipids were not among them. Borreliella also lacked genes for iron-requiring proteins. In their place were genes for several kinds of scavenger proteins or proteins that relied on manganese, magnesium, or zinc for cofactors [229,230,231,232,233,234,235]. This absence of genes for key enzymes was not unique among obligate pathogens. In later years, these stark absences were mirrored within the chromosomes of many Lb spirochetes, to the point where each species varied by less than 8% in terms of gene content [66,117,119,236,237,238,239,240].
While similar studies for Rf spirochetes have been conducted, less is known about their genome and gene content. The first Borrelia genome was sequenced in 2008 by Lescot et al. [241]. Since then, researchers have found that Borrelia spirochetes, like Borreliella spp., have a singular linear chromosome and many plasmids [209,242,243,244]. The Borrelia chromosome is also similar in its GC skew and contains many housekeeping genes [241,245]. It does vary in that it has fewer copies for the 5S and 23S rRNA genes in comparison to Borreliella chromosomes [241]. Additionally, the Borrelia chromosome still retains many of the genes critical for purine salvaging and glycerol biosynthesis which is suspected of facilitating survival and rapid growth [244]. Others have found a high degree of synteny within the chromosome amongst Borrelia species with less than 2% difference, or the absence of one gene [244]. Currently, a quarter of the chromosomal genes remain uncharacterized.
As mentioned, both the Borreliella and Borrelia spirochetes have a large plasmid repertoire. Borreliella spp. more so than Borrelia spp. The gene content within the Borreliellal plasmids is sparse. The few identifiable genes contained within are unique to an individual species that could assist in host adaptation [246]. The distribution of these virulence genes can be on either the small linear or circular replicons. To date, the Borrelial linear plasmids (lp) have been suggested to contain fewer functional genes and are laced with pseudogenes [247,248,249]. In contrast, the circular plasmids hold more coding genes [250]. It is possible that the lp-associated pseudogene set serves as part of an evolutionary mechanism to provide for the rapid emergence of new functional genes through recombinational processes, or that they play roles in gene regulation [250,251]. This can be seen in different studies such as those conducted by Dulebohn et al. [252]. In one instance the Borreliella lp28-3 investigated, and it was found that several genes, though considered as non-essential to Borreliella survival or as pseudogenes, facilitated spirochete survival throughout the infectious cycle [252].
Previous Borreliella genomic studies have found plasmids to contain a significant fraction of the genes necessary for spirochetal adaptation, survival, or pathogenesis. Most of these plasmids are under the stringent control and are present as single copies within the genome—making the Borreliellal genome, in this sense, more similar to multi-replicon genomes of eukaryotes. The classification of these episomal elements has been fraught with multiple difficulties as the genes are largely unannotated and can only be identified via paralogous family (Pfam) groups, leading to classifications based on size in kilobases and topology. Current plasmid naming conventions do not have any relation to actual biological functions. Within the Borreliella genome, three plasmids are thought to be the most relevant physiologically: linear plasmid 54, linear plasmid 17, and circular plasmid (cp) 26.
Lp54 is uniform in gene density and without repeat elements. The genes present on this plasmid create surface localized lipoproteins that interact directly with host immunomodulators necessary for tick transmission and mammalian infection. The smaller episome, lp17, differs from lp54 by containing genes for homologous proteins with unknown functions and an end with high homology to other linear Borrleiella replicons. Lp17 also varies greatly among different Borreliaceae species, amounting to 16 variants identified to date [247]. Lastly, cp26 is highly syntenic and contains genes that encode key metabolic and nutrient import proteins. Interestingly, cp26 also contains the OspC genes critical for mammalian infection and the resolvase T enzyme which has been documented as necessary for genome replication [253,254]. As stated, most of the Lb plasmids are present as single copies except for cp32 [249]. This replicon is highly syntenic among various species and strains and carries numerous osp genes necessary for survival in murine, and tick hosts. Genes needed for successful host adaptation and immune evasion are also found in the lp28 replicon including the has the VLS antigenic variation system [247,255].
Borrelia spirochetes contain fewer, but slightly larger, plasmids. Within these replicons are genes for metabolism, replicon maintenance, and pathogenesis [244]. Borrelia, similar to their Lyme borreliosis cousins, have both linear and circular episomes. The naming of Rf episomes also follows similar conventions. RF linear plasmids are divided into two categories based on size. The first group of linear plasmids is composed of replicons that are approximately 200 Kb in size. The second group is much smaller, averaging at 30 Kb, much like those seen in Lb spirochetes. These smaller plasmids encode genes utilized for antigenic variation which provide RF spirochetes the capacity to switch between antigenic variants, quickly lending to immune evasion, and explaining the ‘relapsing fever’ phenotype [244]. It is important to note that there is one known RF cp that is syntenic with Lb cp32 [256]. The contents of the replicons are also primarily for the generation of surface proteins.

6. Pangenomic Applications

Considering the genomic differences among the Borrelieace spirochetes and their pathogenic heterogeneity, there are understandably profound therapeutic differences. As the incidence and prevalence of borrelioses rise, it is imperative to develop new approaches to diagnostics. Recently, a new field called pangenomics has provided researchers with a large toolset of comparative genomic technologies to characterize compositional, functional, and structural differences among large numbers of genomes at any taxonomic level. This has been exemplified by recent pangenomic analyses of the pathogenic bacterial species Clostridioides difficile, Escherichia coli, Haemophilus influenzae, and Moraxella catarrhalis [257,258,259,260,261,262,263,264,265,266,267]. These studies have helped identify new targets for the development of vaccines, diagnostics, and therapeutics [257,258,259,260,261]. Genus-level pangenomes have also been described [257,261,268,269,270,271,272].
In the case of Borreliella/Borrelia, discerning key features of the genome has been difficult due to biological features and technical limitations. Borrelia’s complex genome includes repetitive sequences, covalently-bound hairpin ends (akin to telomeres), and a multitude of linear and circular plasmids. These challenges were difficult to overcome until the advent of next-generation sequencing. Recently, various Illumina platforms have been used due to their technological improvements, allowing for the sequencing of Borrelia’s many plasmids. Unfortunately, while this is a step forward in Borreliellal/Borrelial genomics, such platforms are still ill-suited for properly capturing the large stretches of repetitive sequences in some of these episomes [273]. Thus, establishing near-complete genomes was exceedingly challenging without the use of additional platforms.
This drove Borrelial geneticists to search for ways to capture the entire genome in all of its complexity. The arrival of the latest models of Pacific Bioscience’s long-read sequencing platforms that utilize the high-fidelity circular consensus sequencing protocols has allowed researchers to attain much longer and more accurate sequence reads. This method has been applied to both Lb and Rf spirochetes [66,242]. There were concerns, however, about capturing smaller episomes within the spirochete’s genome using this technology.
Concurrently with the growth of Borreliellal/Borrelial genomics, a tentative preliminary pangenome, rather defined as the complete collection of all genes available within a species (or higher taxonomic unit), from existing sequenced and annotated Borrelia genomes was created. This research focused on B. burgforferi sensu stricto and the B. burgdorferi sensu lato complexes [208,274]. In this first pangenome study, several Lb spirochetes were chosen based on their host of origin, geo-locale origin, vector, and if they had been isolated from symptomatic individuals. These 22 Borreliella genomes were then used to construct genome-wide, single-nucleotide-polymorphism phylogenies [274]. The researchers checked the pangenome size for B. burgdorferi alone and at the genus-species level for the entire Borreliella burgdorferi sensu lato complex. It was suggested that B. burgdorferi by itself had a ‘closed’ pangenome. This finding suggests that there were a limited number of novel additions of new genes added to the genome for each B. burgdorferi genome used in the analysis. Further expansion of the pangenome project to encompass other Borreliella members revealed a different pangenome composition. It was shown that as a genus, Borreliella had an open pangenome that would continue to expand with each new genome addition. While this study advanced the field of Borreliellal comparative genomics and pangenomics, there were limitations brought primarily from the technical capabilities of sequencing platforms, which were prevalent at the time of research.
Subsequent studies on the B. burgdorferi pangenome were smaller in scale. One study centered on the design and application of a potential xenodiagnostic through the use of the pangenome by targeting a fragment of the B. burgdorferi 16S rRNA gene [208]. This was then applied with limited success on I. ricinius ticks [208]. This could stem from the limited sequences of the Borreliella 16S gene. Additionally, this gene has been documented to exist as a single copy within the Borreliellal genome, which is commensurate with its slow growth and long doubling times.
In the age of long read, next-generation sequencing, it is possible to overcome many of the afore-mentioned limitations. With the recent expansion of PacBio sequencing platforms, a B. burgdorferi, Borreliella burgdorferi sensu lato complex being constructed as well as establishing the first pangenome for the Borreliaceae family. Thus, novel biomarkers can be identified as diagnostic targets at the species and genus levels. It is anticipated that this new and expanded pangenome for the Borreliaceae family will yield more accurate diagnostics, new antibiotic targets specific to Borreliella, and improve outcomes for patients diagnosed with Borreliella-based infections.

Author Contributions

Conceptualization: G.D.E., K.M.S. and B.S.H.-G.; Writing: K.M.S., B.S.H.-G., M.A.C., N.A.C. and R.T.M.; Figure construction: K.M.S., N.A.C. and R.T.M. All authors have read and agreed to the published version of the manuscript.

Funding

KMS & GDE were supported in part by the Cook Foundation and the Oskar Fischer Project as well as DUCOM. MAC and BSGH were supported by DUCOM. RTM is supported from the NIH: 5R01AI141801-04, 1R01AI65876-01, and Zoetis grant: FP15713. Support for NAC and RTM were in part from the Christian Foundation and the Reynolds Family Foundation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gubler, D.J. Arthropods in Disease Transmission. In Hunter’s Tropical Medicine and Emerging Infectious Disease; Saunders: Philadelphia, PA, USA, 2013. [Google Scholar]
  2. Anderson, J.F.; Magnarelli, L.A. Biology of Ticks. Infect. Dis. Clin. North Am. 2008, 22, 195–215. [Google Scholar] [CrossRef] [PubMed]
  3. Anderson, J.F. The Natural History of Ticks. Med. Clin. North Am. 2002, 86, 205–218. [Google Scholar] [CrossRef]
  4. Obenchain, F.D.; Galun, R. Physiology of Ticks: Current Themes in Tropical Science; Elsevier: Amsterdam, The Netherlands, 2013; ISBN 9781483162348. [Google Scholar]
  5. Smith, T.; Kilborne, F.L. Investigations into the Nature, Causation, and Prevention of Texas Or Southern Cattle Fever; U.S. Government Printing Office: Washington, DC, USA, 1893.
  6. Beard, C.B.; Eisen, L.; Eisen, R.J. The Rise of Ticks and Tickborne Diseases in the United States-Introduction. J. Med. Entomol. 2021, 58, 1487–1489. [Google Scholar] [CrossRef]
  7. Aguero-Rosenfeld, M.E.; Wang, G.; Schwartz, I.; Wormser, G.P. Diagnosis of Lyme Borreliosis. Clin. Microbiol. Rev. 2005, 18, 484–509. [Google Scholar] [CrossRef] [PubMed]
  8. Mosel, M.R.; Aucott, J.; Schutzer, S.E.; Marques, A.; Arnaboldi, P.M.; Dattwyler, R.; Eshoo, M.W. Lyme Disease Diagnostics. In Lyme Disease and Relapsing Fever Spirochetes: Genomics, Molecular Biology, Host Interactions and Disease Pathogenesis 2021; Caister Academic Press: Wymondham, UK, 2021. [Google Scholar]
  9. Bowman, A.S.; Nuttall, P.A. Ticks: Biology, Disease and Control; Cambridge University Press: Cambridge, UK, 2008; ISBN 9781107321076. [Google Scholar]
  10. Entomological Society of America Bloodsucking Ticks (Ixodoidea)—Vectors of Diseases of Man and Animals—Translated From Russia; Entomological Society of America: Annapolis, MD, USA, 1908.
  11. Khatchikian, C.E.; Prusinski, M.A.; Stone, M.; Backenson, P.B.; Wang, I.-N.; Foley, E.; Seifert, S.N.; Levy, M.Z.; Brisson, D. Recent and Rapid Population Growth and Range Expansion of the Lyme Disease Tick Vector, Ixodes Scapularis, in North America. Evolution 2015, 69, 1678–1689. [Google Scholar] [CrossRef]
  12. Leighton, P.A.; Koffi, J.K.; Pelcat, Y.; Lindsay, L.R.; Ogden, N.H. Predicting the Speed of Tick Invasion: An Empirical Model of Range Expansion for the Lyme Disease vectorIxodes Scapularisin Canada. J. Appl. Ecol. 2012, 49, 457–464. [Google Scholar] [CrossRef]
  13. Ogden, N.H.; Maarouf, A.; Barker, I.K.; Bigras-Poulin, M.; Lindsay, L.R.; Morshed, M.G.; O’callaghan, C.J.; Ramay, F.; Waltner-Toews, D.; Charron, D.F. Climate Change and the Potential for Range Expansion of the Lyme Disease Vector Ixodes Scapularis in Canada. Int. J. Parasitol. 2006, 36, 63–70. [Google Scholar] [CrossRef]
  14. Ogden, N.H.; St-Onge, L.; Barker, I.K.; Brazeau, S.; Bigras-Poulin, M.; Charron, D.F.; Francis, C.M.; Heagy, A.; Lindsay, R.; Maarouf, A.; et al. Risk Maps for Range Expansion of the Lyme Disease Vector, Ixodes Scapularis, in Canada Now and with Climate Change. Int. J. Health Geogr. 2008, 7, 24. [Google Scholar] [CrossRef]
  15. Gardner, A.M.; Pawlikowski, N.C.; Hamer, S.A.; Hickling, G.J.; Miller, J.R.; Schotthoefer, A.M.; Tsao, J.I.; Allan, B.F. Landscape Features Predict the Current and Forecast the Future Geographic Spread of Lyme Disease. Proc. Biol. Sci. 2020, 287, 20202278. [Google Scholar] [CrossRef]
  16. Eisen, R.J.; Eisen, L.; Beard, C.B. County-Scale Distribution of Ixodes Scapularis and Ixodes Pacificus (Acari: Ixodidae) in the Continental United States. J. Med. Entomol. 2016, 53, 349–386. [Google Scholar] [CrossRef] [Green Version]
  17. Clover, J.R.; Lane, R.S. Evidence Implicating Nymphal Ixodes Pacificus (Acari: Ixodidae) in the Epidemiology of Lyme Disease in California. Am. J. Trop. Med. Hyg. 1995, 53, 237–240. [Google Scholar] [CrossRef] [PubMed]
  18. Sonenshine, D.E. Range Expansion of Tick Disease Vectors in North America: Implications for Spread of Tick-Borne Disease. Int. J. Environ. Res. Public Health 2018, 15, 478. [Google Scholar] [CrossRef] [PubMed]
  19. Hahn, M.B.; Jarnevich, C.S.; Monaghan, A.J.; Eisen, R.J. Modeling the Geographic Distribution of Ixodes Scapularis and Ixodes Pacificus (Acari: Ixodidae) in the Contiguous United States. J. Med. Entomol. 2016, 53, 1176–1191. [Google Scholar] [CrossRef] [PubMed]
  20. Ogden, N.H.; Trudel, L.; Artsob, H.; Barker, I.K.; Beauchamp, G.; Charron, D.F.; Drebot, M.A.; Galloway, T.D.; O’Handley, R.; Thompson, R.A.; et al. Ixodes Scapularis Ticks Collected by Passive Surveillance in Canada: Analysis of Geographic Distribution and Infection with Lyme Borreliosis Agent Borrelia Burgdorferi. J. Med. Entomol. 2006, 43, 600–609. [Google Scholar] [CrossRef]
  21. Descamps, S. Winter Temperature Affects the Prevalence of Ticks in an Arctic Seabird. PLoS ONE 2013, 8, e65374. [Google Scholar]
  22. Dietrich, M.; Gómez-Díaz, E.; McCoy, K.D. Worldwide Distribution and Diversity of Seabird Ticks: Implications for the Ecology and Epidemiology of Tick-Borne Pathogens. Vector Borne Zoonotic Dis. 2011, 11, 453–470. [Google Scholar] [CrossRef]
  23. Smith, R.P., Jr.; Muzaffar, S.B.; Lavers, J.; Lacombe, E.H.; Cahill, B.K.; Lubelczyk, C.B.; Kinsler, A.; Mathers, A.J.; Rand, P.W. Borrelia Garinii in Seabird Ticks (Ixodes Uriae), Atlantic Coast, North America. Emerg. Infect. Dis. 2006, 12, 1909–1912. [Google Scholar] [CrossRef]
  24. Munro, H.J.; Ogden, N.H.; Lindsay, L.R.; Robertson, G.J.; Whitney, H.; Lang, A.S. Evidence for Borrelia Bavariensis Infections of Ixodes Uriae within Seabird Colonies of the North Atlantic Ocean. Appl. Environ. Microbiol. 2017, 83, e01087-17. [Google Scholar] [CrossRef]
  25. Olsén, B.; Jaenson, T.G.; Noppa, L.; Bunikis, J.; Bergström, S. A Lyme Borreliosis Cycle in Seabirds and Ixodes Uriae Ticks. Nature 1993, 362, 340–342. [Google Scholar] [CrossRef]
  26. U.S. Centers for Disease Control and Prevention. Others What You Need To Know about Asian Longhorned Ticks-A New Tick in the United States; CDC: Atlanta, GA, USA, 2019. [Google Scholar]
  27. Nuttall, P. Climate, Ticks and Disease; CABI: Wallingford, UK, 2021; ISBN 9781789249637. [Google Scholar]
  28. Beard, C.B.; Ben Beard, C.; Occi, J.; Bonilla, D.L.; Egizi, A.M.; Fonseca, D.M.; Mertins, J.W.; Backenson, B.P.; Bajwa, W.I.; Barbarin, A.M.; et al. Multistate Infestation with the Exotic Disease–Vector Tick Haemaphysalis Longicornis—United States, August 2017–September 2018. MMWR. Morbid. Mortal. Wkly Rep. 2018, 67, 1310–1313. [Google Scholar] [CrossRef]
  29. CDC Regions Where Ticks Live. Available online: https://www.cdc.gov/ticks/geographic_distribution.html (accessed on 9 February 2022).
  30. Hahn, M.B.; Disler, G.; Durden, L.A.; Coburn, S.; Witmer, F.; George, W.; Beckmen, K.; Gerlach, R. Establishing a Baseline for Tick Surveillance in Alaska: Tick Collection Records from 1909-2019. Ticks Tick Borne Dis. 2020, 11, 101495. [Google Scholar] [CrossRef] [PubMed]
  31. Steere, A.C.; Malawista, S.E.; Snydman, D.R.; Shope, R.E.; Andiman, W.A.; Ross, M.R.; Steele, F.M. Lyme arthritis: An epidemic of oligoarticular arthritis in children and adults in three Connecticut communities. Arthritis Rheum. 1977, 20, 7–17. [Google Scholar] [CrossRef] [PubMed]
  32. Steere, A.C.; Hardin, J.A.; Malawista, S.E. Lyme Arthritis: A New Clinical Entity. Hosp. Pract. 1978, 13, 143–158. [Google Scholar] [CrossRef] [PubMed]
  33. Barbour, A.G.; Benach, J.L. Discovery of the Lyme Disease Agent. MBio 2019, 10, e02166-19. [Google Scholar] [CrossRef]
  34. Anderson, J.F.; Magnarelli, L.A.; Burgdorfer, W.; Barbour, A.G. Spirochetes in Ixodes Dammini and Mammals from Connecticut. Am. J. Trop. Med. Hyg. 1983, 32, 818–824. [Google Scholar] [CrossRef] [PubMed]
  35. Bosler, E.M.; Coleman, J.L.; Benach, J.L.; Massey, D.A.; Hanrahan, J.P.; Burgdorfer, W.; Barbour, A.G. Natural Distribution of the Ixodes Dammini Spirochete. Science 1983, 220, 321–322. [Google Scholar] [CrossRef]
  36. Benach, J.L.; Bosler, E.M.; Hanrahan, J.P.; Coleman, J.L.; Habicht, G.S.; Bast, T.F.; Cameron, D.J.; Ziegler, J.L.; Barbour, A.G.; Burgdorfer, W.; et al. Spirochetes Isolated from the Blood of Two Patients with Lyme Disease. N. Engl. J. Med. 1983, 308, 740–742. [Google Scholar] [CrossRef]
  37. Steere, A.C.; Grodzicki, R.L.; Kornblatt, A.N.; Craft, J.E.; Barbour, A.G.; Burgdorfer, W.; Schmid, G.P.; Johnson, E.; Malawista, S.E. The Spirochetal Etiology of Lyme Disease. N. Engl. J. Med. 1983, 308, 733–740. [Google Scholar] [CrossRef]
  38. Burgdorfer, W.; Barbour, A.G.; Hayes, S.F.; Benach, J.L.; Grunwaldt, E.; Davis, J.P. Lyme Disease—A Tick-Borne Spirochetosis? Science 1982, 216, 1317–1319. [Google Scholar] [CrossRef]
  39. Oliver, J.H., Jr.; Owsley, M.R.; Hutcheson, H.J.; James, A.M.; Chen, C.; Irby, W.S.; Dotson, E.M.; McLain, D.K. Conspecificity of the Ticks Ixodes Scapularis and I. Dammini (Acari: Ixodidae). J. Med. Entomol. 1993, 30, 54–63. [Google Scholar] [CrossRef]
  40. Piesman, J.; Mather, T.N.; Sinsky, R.J.; Spielman, A. Duration of Tick Attachment and Borrelia Burgdorferi Transmission. J. Clin. Microbiol. 1987, 25, 557–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Peavey, C.A.; Lane, R.S. Transmission of Borrelia Burgdorferi by Ixodes Pacificus Nymphs and Reservoir Competence of Deer Mice (Peromyscus Maniculatus) Infected by Tick-Bite. J. Parasitol. 1995, 81, 175–178. [Google Scholar] [CrossRef] [PubMed]
  42. Lane, R.S.; Brown, R.N.; Piesman, J.; Peavey, C.A. Vector Competence of Ixodes Pacificus and Dermacentor Occidentalis (Acari: Ixodidae) for Various Isolates of Lyme Disease Spirochetes. J. Med. Entomol. 1994, 31, 417–424. [Google Scholar] [CrossRef] [PubMed]
  43. Piesman, J.; Sinsky, R.J. Ability of Ixodes Scapularis, Dermacentor Variabilis, and Amblyomma Americanum (Acari: Ixodidae) to Acquire, Maintain, and Transmit Lyme Disease Spirochetes (Borrelia Burgdorferi). J. Med. Entomol. 1988, 25, 336–339. [Google Scholar] [CrossRef] [PubMed]
  44. Eisen, R.J.; Eisen, L. The Blacklegged Tick, Ixodes Scapularis: An Increasing Public Health Concern. Trends Parasitol. 2018, 34, 295–309. [Google Scholar] [CrossRef]
  45. Hamer, S.A.; Hickling, G.J.; Sidge, J.L.; Walker, E.D.; Tsao, J.I. Synchronous Phenology of Juvenile Ixodes Scapularis, Vertebrate Host Relationships, and Associated Patterns of Borrelia Burgdorferi Ribotypes in the Midwestern United States. Ticks Tick Borne Dis. 2012, 3, 65–74. [Google Scholar] [CrossRef]
  46. Hunter, D.J.; Torkelson, J.L.; Bodnar, J.; Mortazavi, B.; Laurent, T.; Deason, J.; Thephavongsa, K.; Zhong, J. The Rickettsia Endosymbiont of Ixodes Pacificus Contains All the Genes of De Novo Folate Biosynthesis. PLoS ONE 2015, 10, e0144552. [Google Scholar] [CrossRef]
  47. Sojka, D.; Franta, Z.; Horn, M.; Caffrey, C.R.; Mareš, M.; Kopáček, P. New Insights into the Machinery of Blood Digestion by Ticks. Trends Parasitol. 2013, 29, 276–285. [Google Scholar] [CrossRef]
  48. Brown, R.N.; Lane, R.S. Reservoir Competence of Four Chaparral-Dwelling Rodents for Borrelia Burgdorferi in California. Am. J. Trop. Med. Hyg. 1996, 54, 84–91. [Google Scholar] [CrossRef]
  49. Tracy, K.E.; Baumgarth, N. Borrelia Burgdorferi Manipulates Innate and Adaptive Immunity to Establish Persistence in Rodent Reservoir Hosts. Front. Immunol. 2017, 8, 116. [Google Scholar] [CrossRef]
  50. Barbour, A.G.; Bunikis, J.; Travinsky, B.; Hoen, A.G.; Diuk-Wasser, M.A.; Fish, D.; Tsao, J.I. Niche Partitioning of Borrelia Burgdorferi and Borrelia Miyamotoi in the Same Tick Vector and Mammalian Reservoir Species. Am. J. Trop. Med. Hyg. 2009, 81, 1120–1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Narasimhan, S.; Fikrig, E. Tick Microbiome: The Force within. Trends Parasitol. 2015, 31, 315–323. [Google Scholar] [CrossRef] [PubMed]
  52. Institute of Medicine (US) Committee on Lyme Disease; Other Tick-Borne Diseases: The State of the Science. An Overview of Tick-Borne Diseases; National Academies Press (US): Washington, DC, USA, 2011. [Google Scholar]
  53. Salkeld, D.J.; Leonhard, S.; Girard, Y.A.; Hahn, N.; Mun, J.; Padgett, K.A.; Lane, R.S. Identifying the Reservoir Hosts of the Lyme Disease Spirochete Borrelia Burgdorferi in California: The Role of the Western Gray Squirrel (Sciurus Griseus). Am. J. Trop. Med. Hyg. 2008, 79, 535–540. [Google Scholar] [CrossRef] [PubMed]
  54. McKee, C.D.; Bai, Y.; Webb, C.T.; Kosoy, M.Y. Bats Are Key Hosts in the Radiation of Mammal-Associated Bartonella Bacteria. Infect. Genet. Evol. 2021, 89, 104719. [Google Scholar] [CrossRef] [PubMed]
  55. Hersh, M.H.; Tibbetts, M.; Strauss, M.; Ostfeld, R.S.; Keesing, F. Reservoir Competence of Wildlife Host Species for Babesia Microti. Emerg. Infect. Dis. 2012, 18, 1951–1957. [Google Scholar] [CrossRef]
  56. Zolnik, C.P.; Makkay, A.M.; Falco, R.C.; Daniels, T.J. American Black Bears as Hosts of Blacklegged Ticks (Acari: Ixodidae) in the Northeastern United States. J. Med. Entomol. 2015, 52, 1103–1110. [Google Scholar] [CrossRef]
  57. Halsey, S.J.; Allan, B.F.; Miller, J.R. The Role of Ixodes Scapularis, Borrelia Burgdorferi and Wildlife Hosts in Lyme Disease Prevalence: A Quantitative Review. Ticks Tick Borne Dis. 2018, 9, 1103–1114. [Google Scholar] [CrossRef]
  58. Sidge, J.L.; Foster, E.S.; Buttke, D.E.; Hojgaard, A.; Graham, C.B.; Tsao, J.I. Lake Michigan Insights from Island Studies: The Roles of Chipmunks and Coyotes in Maintaining Ixodes Scapularis and Borrelia Burgdorferi in the Absence of White-Tailed Deer. Ticks Tick Borne Dis. 2021, 12, 101761. [Google Scholar] [CrossRef]
  59. Lane, R.S.; Loye, J.E. Lyme Disease in California: Interrelationship of Ixodes Pacificus (Acari: Ixodidae), the Western Fence Lizard (Sceloporus Occidentalis), and Borrelia Burgdorferi. J. Med. Entomol. 1989, 26, 272–278. [Google Scholar] [CrossRef]
  60. Eisen, R.J.; Eisen, L.; Lane, R.S. Habitat-Related Variation in Infestation of Lizards and Rodents with Ixodes Ticks in Dense Woodlands in Mendocino County, California. Exp. Appl. Acarol. 2004, 33, 215–233. [Google Scholar] [CrossRef]
  61. Casher, L.; Lane, R.; Barrett, R.; Eisen, L. Relative Importance of Lizards and Mammals as Hosts for Ixodid Ticks in Northern California. Exp. Appl. Acarol. 2002, 26, 127. [Google Scholar] [CrossRef] [PubMed]
  62. Parola, P.; Raoult, D. Ticks and Tickborne Bacterial Diseases in Humans: An Emerging Infectious Threat. Clin. Infect. Dis. 2001, 32, 897–928. [Google Scholar] [CrossRef]
  63. Yabsley, M.J.; Shock, B.C. Natural History of Zoonotic Babesia: Role of Wildlife Reservoirs. Int. J. Parasitol. Parasites Wildl. 2013, 2, 18–31. [Google Scholar] [CrossRef]
  64. Woldehiwet, Z. The Natural History of Anaplasma Phagocytophilum. Vet. Parasitol. 2010, 167, 108–122. [Google Scholar] [CrossRef] [PubMed]
  65. Fukunaga, M.; Takahashi, Y.; Tsuruta, Y.; Matsushita, O.; Ralph, D.; McClelland, M.; Nakao, M. Genetic and Phenotypic Analysis of Borrelia Miyamotoi Sp. Nov., Isolated from the Ixodid Tick Ixodes Persulcatus, the Vector for Lyme Disease in Japan. Int. J. Syst. Bacteriol. 1995, 45, 804–810. [Google Scholar] [CrossRef] [PubMed]
  66. Kingry, L.C.; Batra, D.; Replogle, A.; Rowe, L.A.; Pritt, B.S.; Petersen, J.M. Whole Genome Sequence and Comparative Genomics of the Novel Lyme Borreliosis Causing Pathogen, Borrelia Mayonii. PLoS ONE 2016, 11, e0168994. [Google Scholar] [CrossRef] [PubMed]
  67. Paddock, C.D.; Childs, J.E. Ehrlichia Chaffeensis: A Prototypical Emerging Pathogen. Clin. Microbiol. Rev. 2003, 16, 37–64. [Google Scholar] [CrossRef]
  68. Pritt, B.S.; Sloan, L.M.; Johnson, D.K.H.; Munderloh, U.G.; Paskewitz, S.M.; McElroy, K.M.; McFadden, J.D.; Binnicker, M.J.; Neitzel, D.F.; Liu, G.; et al. Emergence of a New Pathogenic Ehrlichia Species, Wisconsin and Minnesota, 2009. N. Engl. J. Med. 2011, 365, 422–429. [Google Scholar] [CrossRef]
  69. McCoy, G.W. Chapin Bacterium Tularense, the Cause of a Plague-like Disease of Rodents. J. Infect. Dis. (Public Health Bull.) 1912, 10, 61–72. [Google Scholar]
  70. Paddock, C.D.; Sumner, J.W.; Comer, J.A.; Zaki, S.R.; Goldsmith, C.S.; Goddard, J.; McLellan, S.L.F.; Tamminga, C.L.; Ohl, C.A. Rickettsia Parkeri: A Newly Recognized Cause of Spotted Fever Rickettsiosis in the United States. Clin. Infect. Dis. 2004, 38, 805–811. [Google Scholar] [CrossRef]
  71. Kosoy, O.I.; Lambert, A.J.; Hawkinson, D.J.; Pastula, D.M.; Goldsmith, C.S.; Charles Hunt, D.; Erin Staples, J. Novel Thogotovirus Associated with Febrile Illness and Death, United States, 2014. Emerg. Infect. Dis. 2015, 21, 760–764. [Google Scholar] [CrossRef] [PubMed]
  72. Florio, F.; Miller, M.S.; Mugrage, E.R. Colorado Tick Fever: Isolation of the Virus from Dermacentor Andersoni in Nature and a Laboratory Study of the Transmission of the Virus in the Tick. J. Immunol. 1950, 64, 257–263. [Google Scholar] [PubMed]
  73. McMullan, L.K.; Folk, S.M.; Kelly, A.J.; MacNeil, A.; Goldsmith, C.S.; Metcalfe, M.G.; Batten, B.C.; Albariño, C.G.; Zaki, S.R.; Rollin, P.E.; et al. A New Phlebovirus Associated with Severe Febrile Illness in Missouri. N. Engl. J. Med. 2012, 367, 834–841. [Google Scholar] [CrossRef] [PubMed]
  74. McLEAN, D.M.; Donohue, W.L. Powassan Virus: Isolation of Virus from a Fatal Case of Encephalitis. Can. Med. Assoc. J. 1959, 80, 708–711. [Google Scholar] [PubMed]
  75. Cutler, S.; Vayssier-Taussat, M.; Estrada-Peña, A.; Potkonjak, A.; Mihalca, A.D.; Zeller, H. A New Borrelia on the Block: Borrelia Miyamotoi—A Human Health Risk? Euro. Surveill. 2019, 24, 180017. [Google Scholar] [CrossRef]
  76. Johnson, T.L.; Graham, C.B.; Hojgaard, A.; Breuner, N.E.; Maes, S.E.; Boegler, K.A.; Replogle, A.J.; Kingry, L.C.; Petersen, J.M.; Eisen, L.; et al. Isolation of the Lyme Disease Spirochete Borrelia Mayonii From Naturally Infected Rodents in Minnesota. J. Med. Entomol. 2017, 54, 1088–1092. [Google Scholar] [CrossRef]
  77. Rochlin, I.; Egizi, A.; Lindström, A. The Original Scientific Description of the Lone Star Tick (Amblyomma Americanum, Acari: Ixodidae) and Implications for the Species’ Past and Future Geographic Distributions. J. Med. Entomol. 2022, 59, 412–420. [Google Scholar] [CrossRef]
  78. Stromdahl, E.Y.; Nadolny, R.M.; Hickling, G.J.; Hamer, S.A.; Ogden, N.H.; Casal, C.; Heck, G.A.; Gibbons, J.A.; Cremeans, T.F.; Pilgard, M.A. Amblyomma Americanum (Acari: Ixodidae) Ticks Are Not Vectors of the Lyme Disease Agent, Borrelia Burgdorferi (Spirocheatales: Spirochaetaceae): A Review of the Evidence. J. Med. Entomol. 2018, 55, 501–514. [Google Scholar] [CrossRef]
  79. Burgdorfer, W.; Lane, R.S.; Barbour, A.G.; Gresbrink, R.A.; Anderson, J.R. The Western Black-Legged Tick, Ixodes Pacificus: A Vector of Borrelia Burgdorferi. Am. J. Trop. Med. Hyg. 1985, 34, 925–930. [Google Scholar] [CrossRef]
  80. Padgett, K.A.; Lane, R.S. Life Cycle of Ixodes Pacificus (Acari: Ixodidae): Timing of Developmental Processes under Field and Laboratory Conditions. J. Med. Entomol. 2001, 38, 684–693. [Google Scholar] [CrossRef]
  81. Chang, C.C.; Chomel, B.B.; Kasten, R.W.; Romano, V.; Tietze, N. Molecular Evidence of Bartonella Spp. in Questing Adult Ixodes Pacificus Ticks in California. J. Clin. Microbiol. 2001, 39, 1221–1226. [Google Scholar] [CrossRef]
  82. Dingler, R.J.; Wright, S.A.; Donohue, A.M.; Macedo, P.A.; Foley, J.E. Surveillance for Ixodes Pacificus and the Tick-Borne Pathogens Anaplasma Phagocytophilum and Borrelia Burgdorferi in Birds from California’s Inner Coast Range. Ticks Tick Borne Dis. 2014, 5, 436–445. [Google Scholar] [CrossRef] [PubMed]
  83. Kwan, J.Y.; Griggs, R.; Chicana, B.; Miller, C.; Swei, A. Vertical vs. Horizontal Transmission of the Microbiome in a Key Disease Vector, Ixodes Pacificus. Mol. Ecol. 2017, 26, 6578–6589. [Google Scholar] [CrossRef]
  84. Loye, J.E.; Lane, R.S. Questing Behavior of Ixodes Pacificus (Acari: Ixodidae) in Relation to Meteorological and Seasonal Factors. J. Med. Entomol. 1988, 25, 391–398. [Google Scholar] [CrossRef] [PubMed]
  85. Lederberg, J.; McCray, A.T. Ome SweetOmics—A Genealogical Treasury of Words. Scientist 2001, 15, 8. [Google Scholar]
  86. Jansson, J.K.; Hofmockel, K.S. The Soil Microbiome—From Metagenomics to Metaphenomics. Curr. Opin. Microbiol. 2018, 43, 162–168. [Google Scholar] [CrossRef]
  87. Sunagawa, S.; Coelho, L.P.; Chaffron, S.; Kultima, J.R.; Labadie, K.; Salazar, G.; Djahanschiri, B.; Zeller, G.; Mende, D.R.; Alberti, A.; et al. Ocean Plankton. Structure and Function of the Global Ocean Microbiome. Science 2015, 348, 1261359. [Google Scholar] [CrossRef]
  88. Turnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; Knight, R.; Gordon, J.I. The Human Microbiome Project. Nature 2007, 449, 804–810. [Google Scholar] [CrossRef]
  89. Ehrlich, G.D.; Hiller, N.L.; Hu, F.Z. What Makes Pathogens Pathogenic. Genome Biol. 2008, 9, 225. [Google Scholar] [CrossRef]
  90. Bonnet, S.I.; Binetruy, F.; Hernández-Jarguín, A.M.; Duron, O. The Tick Microbiome: Why Non-Pathogenic Microorganisms Matter in Tick Biology and Pathogen Transmission. Front. Cell. Infect. Microbiol. 2017, 7, 236. [Google Scholar] [CrossRef]
  91. Estrada-Peña, A.; Cabezas-Cruz, A.; Obregón, D. Behind Taxonomic Variability: The Functional Redundancy in the Tick Microbiome. Microorganisms 2020, 8, 1829. [Google Scholar] [CrossRef] [PubMed]
  92. Varela-Stokes, A.S.; Park, S.H.; Kim, S.A.; Ricke, S.C. Microbial Communities in North American Ixodid Ticks of Veterinary and Medical Importance. Front. Vet. Sci. 2017, 4, 179. [Google Scholar] [CrossRef]
  93. Zhong, J.; Jasinskas, A.; Barbour, A.G. Antibiotic Treatment of the Tick Vector Amblyomma Americanum Reduced Reproductive Fitness. PLoS ONE 2007, 2, e405. [Google Scholar] [CrossRef] [PubMed]
  94. Hussain, S.; Perveen, N.; Hussain, A.; Song, B.; Aziz, M.U.; Zeb, J.; Li, J.; George, D.; Cabezas-Cruz, A.; Sparagano, O. The Symbiotic Continuum Within Ticks: Opportunities for Disease Control. Front. Microbiol. 2022, 13, 854803. [Google Scholar] [CrossRef] [PubMed]
  95. Zhong, Z.; Zhong, T.; Peng, Y.; Zhou, X.; Wang, Z.; Tang, H.; Wang, J. Symbiont-Regulated Serotonin Biosynthesis Modulates Tick Feeding Activity. Cell Host Microbe 2021, 29, 1545–1557.e4. [Google Scholar] [CrossRef]
  96. Ross, B.D.; Hayes, B.; Radey, M.C.; Lee, X.; Josek, T.; Bjork, J.; Neitzel, D.; Paskewitz, S.; Chou, S.; Mougous, J. Ixodes Scapularis Does Not Harbor a Stable Midgut Microbiome. ISME J. 2018, 12, 2596–2607. [Google Scholar] [CrossRef]
  97. Zolnik, C.P.; Falco, R.C.; Daniels, T.J.; Kolokotronis, S.-O. Transient Influence of Blood Meal and Natural Environment on Blacklegged Tick Bacterial Communities. Ticks Tick Borne Dis. 2018, 9, 563–572. [Google Scholar] [CrossRef]
  98. Thapa, S.; Zhang, Y.; Allen, M.S. Bacterial Microbiomes of Ixodes Scapularis Ticks Collected from Massachusetts and Texas, USA. BMC Microbiol. 2019, 19, 138. [Google Scholar] [CrossRef]
  99. Obregón, D.; Bard, E.; Abrial, D.; Estrada-Peña, A.; Cabezas-Cruz, A. Sex-Specific Linkages Between Taxonomic and Functional Profiles of Tick Gut Microbiomes. Front. Cell. Infect. Microbiol. 2019, 9, 298. [Google Scholar] [CrossRef]
  100. Travanty, N.V.; Ponnusamy, L.; Kakumanu, M.L.; Nicholson, W.L.; Apperson, C.S. Diversity and Structure of the Bacterial Microbiome of the American Dog Tick, Dermacentor Variabilis, Is Dominated by the Endosymbiont Francisella. Symbiosis 2019, 79, 239–250. [Google Scholar] [CrossRef]
  101. Brinkerhoff, R.J.; Clark, C.; Ocasio, K.; Gauthier, D.T.; Hynes, W.L. Factors Affecting the Microbiome of Ixodes Scapularis and Amblyomma Americanum. PLoS ONE 2020, 15, e0232398. [Google Scholar] [CrossRef] [PubMed]
  102. Chicana, B.; Couper, L.I.; Kwan, J.Y.; Tahiraj, E.; Swei, A. Comparative Microbiome Profiles of Sympatric Tick Species from the Far-Western United States. Insects 2019, 10, 353. [Google Scholar] [CrossRef] [PubMed]
  103. Wu-Chuang, A.; Hodžić, A.; Mateos-Hernández, L.; Estrada-Peña, A.; Obregon, D.; Cabezas-Cruz, A. Current Debates and Advances in Tick Microbiome Research. Curr Res Parasitol Vector Borne Dis 2021, 1, 100036. [Google Scholar] [CrossRef] [PubMed]
  104. Williams-Newkirk, A.J.; Rowe, L.A.; Mixson-Hayden, T.R.; Dasch, G.A. Characterization of the Bacterial Communities of Life Stages of Free Living Lone Star Ticks (Amblyomma Americanum). PLoS ONE 2014, 9, e102130. [Google Scholar]
  105. Zhang, R.; Yu, G.; Huang, Z.; Zhang, Z. Microbiota Assessment across Different Developmental Stages of Dermacentor Silvarum (Acari: Ixodidae) Revealed Stage-Specific Signatures. Ticks Tick Borne Dis. 2020, 11, 101321. [Google Scholar] [CrossRef]
  106. Eshoo, M.W.; Carolan, H.E.; Massire, C.; Chou, D.M.; Crowder, C.D.; Rounds, M.A.; Phillipson, C.A.; Schutzer, S.E.; Ecker, D.J. Survey of Ixodes Pacificus Ticks in California Reveals a Diversity of Microorganisms and a Novel and Widespread Anaplasmataceae Species. PLoS ONE 2015, 10, e0135828. [Google Scholar] [CrossRef]
  107. Afzelius, A. Verhandlungen Der Dermatologischen Gesellschaft Zu Stockholm. Arch. Dermatol. Syph. 1910, 101, 404. [Google Scholar]
  108. Hellerstrom, S. Erythema Chronicum Migrans Afzelius with Meningitis. Acta Derm. Venereol. 1951, 31, 227–234. [Google Scholar] [CrossRef]
  109. Garin, C.H.; Bujadoux, C.H. Paralysie Par Les Tiques. J. Med. Lyon 1922, 71, 7. [Google Scholar]
  110. Benach, J.; Garcia Monco, J. The Woldwide Saga of Lyme Borreliosis. Borrelia. Molecular Biology, Host Interaction and Pathogenesis; Caister Academic Press: Norwich, UK, 2010; pp. 7–26. [Google Scholar]
  111. Steere, A.C.; Malawista, S.E.; Hardin, J.A.; Ruddy, S.; Askenase, W.; Andiman, W.A. Erythema Chronicum Migrans and Lyme Arthritis. The Enlarging Clinical Spectrum. Ann. Intern. Med. 1977, 86, 685–698. [Google Scholar] [CrossRef]
  112. Reik, L.; Steere, A.C.; Bartenhagen, N.H.; Shope, R.E.; Malawista, S.E. Neurologic Abnormalities of Lyme Disease. Medicine 1979, 58, 281–294. [Google Scholar] [CrossRef] [PubMed]
  113. Walter, K.S.; Carpi, G.; Caccone, A.; Diuk-Wasser, M.A. Genomic Insights into the Ancient Spread of Lyme Disease across North America. Nat Ecol Evol 2017, 1, 1569–1576. [Google Scholar] [CrossRef] [PubMed]
  114. Earnhart, C.G.; Marconi, R.T. Chapter 52—Lyme Disease. In Vaccines for Biodefense and Emerging and Neglected Diseases; Barrett, A.D.T., Stanberry, L.R., Eds.; Academic Press: London, UK, 2009; pp. 1031–1060. ISBN 9780123694089. [Google Scholar]
  115. Data and Surveillance Lyme Disease|CDC. Available online: https://www.cdc.gov/lyme/datasurveillance/index.html (accessed on 25 April 2019).
  116. Dworkin, M.S.; Schwan, T.G.; Anderson, D.E., Jr.; Borchardt, S.M. Tick-Borne Relapsing Fever. Infect. Dis. Clin. North Am. 2008, 22, 449–468. [Google Scholar] [CrossRef] [PubMed]
  117. Pritt, B.S.; Mead, P.S.; Johnson, D.K.H.; Neitzel, D.F.; Respicio-Kingry, L.B.; Davis, J.P.; Schiffman, E.; Sloan, L.M.; Schriefer, M.E.; Replogle, A.J.; et al. Identification of a Novel Pathogenic Borrelia Species Causing Lyme Borreliosis with Unusually High Spirochaetaemia: A Descriptive Study. Lancet Infect. Dis. 2016, 16, 556–564. [Google Scholar] [CrossRef]
  118. Crowder, C.D.; Matthews, H.E.; Schutzer, S.; Rounds, M.A.; Luft, B.J.; Nolte, O.; Campbell, S.R.; Phillipson, C.A.; Li, F.; Sampath, R.; et al. Genotypic Variation and Mixtures of Lyme Borrelia in Ixodes Ticks from North America and Europe. PLoS ONE 2010, 5, e10650. [Google Scholar] [CrossRef]
  119. Margos, G.; Vollmer, S.A.; Ogden, N.H.; Fish, D. Population Genetics, Taxonomy, Phylogeny and Evolution of Borrelia Burgdorferi Sensu Lato. Infect. Genet. Evol. 2011, 11, 1545–1563. [Google Scholar] [CrossRef] [Green Version]
  120. Ras, N.M.; Lascola, B.; Postic, D.; Cutler, S.J.; Rodhain, F.; Baranton, G.; Raoult, D. Phylogenesis of Relapsing Fever Borrelia spp. Int. J. Syst. Bacteriol. 1996, 46, 859–865. [Google Scholar]
  121. Adeolu, M.; Gupta, R.S. A Phylogenomic and Molecular Marker Based Proposal for the Division of the Genus Borrelia into Two Genera: The Emended Genus Borrelia Containing Only the Members of the Relapsing Fever Borrelia, and the Genus Borreliella Gen. Nov. Containing the Members of the Lyme Disease Borrelia (Borrelia Burgdorferi Sensu Lato Complex). Antonie Van Leeuwenhoek 2014, 105, 1049–1072. [Google Scholar]
  122. Müllegger, R.R.; Glatz, M. Skin Manifestations of Lyme Borreliosis. Am. J. Clin. Dermatol. 2008, 9, 355–368. [Google Scholar] [CrossRef]
  123. Verma, V.; Roman, M.; Shah, D.; Zaretskaya, M.; Yassin, M.H. A Case of Chronic Progressive Lyme Encephalitis as a Manifestation of Late Lyme Neuroborreliosis. Infect. Dis. Rep. 2014, 6, 5496. [Google Scholar] [CrossRef]
  124. Brodziński, S.; Nasierowski, T. Psychosis in Borrelia Burgdorferi Infection—Part I: Epidemiology, Pathogenesis, Diagnosis and Treatment of Neuroborreliosis. Psychiatr. Pol. 2019, 53, 629–640. [Google Scholar] [CrossRef] [PubMed]
  125. Arav-Boger, R.; Crawford, T.; Steere, A.C.; Halsey, N.A. Cerebellar Ataxia as the Presenting Manifestation of Lyme Disease. Pediatr. Infect. Dis. J. 2002, 21, 353–356. [Google Scholar] [CrossRef] [PubMed]
  126. Rauer, S.; Kastenbauer, S.; Fingerle, V.; Hunfeld, K.-P.; Huppertz, H.-I.; Dersch, R. Lyme Neuroborreliosis. Dtsch. Arztebl. Int. 2018, 115, 751–756. [Google Scholar] [CrossRef] [PubMed]
  127. Cardenas-de la Garza, J.A.; De la Cruz-Valadez, E.; Ocampo-Candiani, J.; Welsh, O. Clinical Spectrum of Lyme Disease. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 201–208. [Google Scholar] [CrossRef]
  128. Cassarino, D.S.; Quezado, M.M.; Ghatak, N.R.; Duray, P.H. Lyme-Associated Parkinsonism: A Neuropathologic Case Study and Review of the Literature. Arch. Pathol. Lab. Med. 2003, 127, 1204–1206. [Google Scholar] [CrossRef]
  129. Stanek, G.; Wormser, G.P.; Gray, J.; Strle, F. Lyme Borreliosis. Lancet 2012, 379, 461–473. [Google Scholar] [CrossRef]
  130. Blanc, F.; Philippi, N.; Cretin, B.; Kleitz, C.; Berly, L.; Jung, B.; Kremer, S.; Namer, I.J.; Sellal, F.; Jaulhac, B.; et al. Lyme Neuroborreliosis and Dementia. J. Alzheimers. Dis. 2014, 41, 1087–1093. [Google Scholar] [CrossRef]
  131. Steere, A.C.; Malawista, S.E.; Bartenhagen, N.H.; Spieler, P.N.; Newman, J.H.; Rahn, D.W.; Hutchinson, G.J.; Green, J.; Snydman, D.R.; Taylor, E. The Clinical Spectrum and Treatment of Lyme Disease. Yale J. Biol. Med. 1984, 57, 453–461. [Google Scholar]
  132. Fix, A.D.; Peña, C.A.; Strickland, G.T. Racial Differences in Reported Lyme Disease Incidence. Am. J. Epidemiol. 2000, 152, 756–759. [Google Scholar] [CrossRef]
  133. Coburn, J.; Garcia, B.; Hu, L.T.; Jewett, M.W.; Kraiczy, P.; Norris, S.J.; Skare, J. Lyme Disease Pathogenesis. Curr. Issues Mol. Biol. 2020, 42, 473–518. [Google Scholar]
  134. Torbahn, G.; Hofmann, H.; Rücker, G.; Bischoff, K.; Freitag, M.H.; Dersch, R.; Fingerle, V.; Motschall, E.; Meerpohl, J.J.; Schmucker, C. Efficacy and Safety of Antibiotic Therapy in Early Cutaneous Lyme Borreliosis: A Network Meta-Analysis. JAMA Dermatol. 2018, 154, 1292–1303. [Google Scholar] [CrossRef] [PubMed]
  135. Tilly, K.; Rosa, P.A.; Stewart, P.E. Biology of Infection with Borrelia Burgdorferi. Infect. Dis. Clin. North Am. 2008, 22, 217–234. [Google Scholar] [CrossRef] [PubMed]
  136. DeHart, T.G.; Kushelman, M.R.; Hildreth, S.B.; Helm, R.F.; Jutras, B.L. The Unusual Cell Wall of the Lyme Disease Spirochaete Borrelia Burgdorferi Is Shaped by a Tick Sugar. Nat. Microbiol. 2021, 6, 1583–1592. [Google Scholar] [CrossRef] [PubMed]
  137. Motaleb, M.A.; Corum, L.; Bono, J.L.; Elias, A.F.; Rosa, P.; Samuels, D.S.; Charon, N.W. Borrelia Burgdorferi Periplasmic Flagella Have Both Skeletal and Motility Functions. Proc. Natl. Acad. Sci. USA 2000, 97, 10899–10904. [Google Scholar] [CrossRef] [PubMed]
  138. Sapi, E.; Bastian, S.L.; Mpoy, C.M.; Scott, S.; Rattelle, A.; Pabbati, N.; Poruri, A.; Burugu, D.; Theophilus, P.A.S.; Pham, T.V.; et al. Characterization of Biofilm Formation by Borrelia Burgdorferi in Vitro. PLoS ONE 2012, 7, e48277. [Google Scholar] [CrossRef]
  139. Sapi, E.; Balasubramanian, K.; Poruri, A.; Maghsoudlou, J.S.; Socarras, K.M.; Timmaraju, A.V.; Filush, K.R.; Gupta, K.; Shaikh, S.; Theophilus, P.A.S.; et al. Evidence of In Vivo Existence of Borrelia Biofilm in Borrelial Lymphocytomas. Eur. J. Microbiol. Immunol. 2016, 6, 9–24. [Google Scholar] [CrossRef]
  140. Timmaraju, V.A.; Theophilus, P.A.S.; Balasubramanian, K.; Shakih, S.; Luecke, D.F.; Sapi, E. Biofilm Formation by Borrelia Burgdorferi Sensu Lato. FEMS Microbiol. Lett. 2015, 362, fnv120. [Google Scholar] [CrossRef]
  141. Shaikh, S.; Timmaraju, V.A.; Torres, J.P.; Socarras, K.M.; Theophilus, P.A.S.; Sapi, E. Influence of Tick and Mammalian Physiological Temperatures on Borrelia Burgdorferi Biofilms. Microbiology 2016, 162, 1984–1995. [Google Scholar] [CrossRef]
  142. Rudenko, N.; Golovchenko, M.; Kybicova, K.; Vancova, M. Metamorphoses of Lyme Disease Spirochetes: Phenomenon of Borrelia Persisters. Parasit. Vectors 2019, 12, 237. [Google Scholar] [CrossRef]
  143. Pal, U.; Li, X.; Wang, T.; Montgomery, R.R.; Ramamoorthi, N.; Desilva, A.M.; Bao, F.; Yang, X.; Pypaert, M.; Pradhan, D.; et al. TROSPA, an Ixodes Scapularis Receptor for Borrelia Burgdorferi. Cell 2004, 119, 457–468. [Google Scholar] [CrossRef] [Green Version]
  144. Kurokawa, C.; Lynn, G.E.; Pedra, J.H.F.; Pal, U.; Narasimhan, S.; Fikrig, E. Interactions between Borrelia Burgdorferi and Ticks. Nat. Rev. Microbiol. 2020, 18, 587–600. [Google Scholar] [CrossRef] [PubMed]
  145. Hovius, J.W.R.; van Dam, A.P.; Fikrig, E. Tick–host–pathogen Interactions in Lyme Borreliosis. Trends Parasitol. 2007, 23, 434–438. [Google Scholar] [CrossRef] [PubMed]
  146. CDC Transmission. Available online: https://www.cdc.gov/lyme/transmission/index.html (accessed on 17 March 2022).
  147. Eisen, L. Pathogen Transmission in Relation to Duration of Attachment by Ixodes Scapularis Ticks. Ticks Tick Borne Dis. 2018, 9, 535–542. [Google Scholar] [CrossRef] [PubMed]
  148. Cook, M.J. Lyme Borreliosis: A Review of Data on Transmission Time after Tick Attachment. Int. J. Gen. Med. 2015, 8, 1–8. [Google Scholar] [CrossRef] [PubMed]
  149. Hyde, J.A. Borrelia Burgdorferi Keeps Moving and Carries on: A Review of Borrelial Dissemination and Invasion. Front. Immunol. 2017, 8, 114. [Google Scholar] [CrossRef]
  150. Oosting, M.; Buffen, K.; van der Meer, J.W.M.; Netea, M.G.; Joosten, L.A.B. Innate Immunity Networks during Infection with Borrelia Burgdorferi. Crit. Rev. Microbiol. 2016, 42, 233–244. [Google Scholar]
  151. Bockenstedt, L.K.; Wooten, R.M.; Baumgarth, N. Immune response to Borrelia: Lessons from Lyme disease spirochetes. Curr. Issues Mol. Biol. 2021, 42, 145–190. [Google Scholar]
  152. Elsner, R.A.; Barthold, S.W.; Baumgarth, N. Delays and Diversions Mark the Development of B Cell Responses to Borrelia Burgdorferi Infection. J. Immunol. 2012, 188, 5612–5622. [Google Scholar]
  153. Elsner, R.A.; Hastey, C.J.; Baumgarth, N. CD4+ T Cells Promote Antibody Production but Not Sustained Affinity Maturation during Borrelia Burgdorferi Infection. Infect. Immun. 2015, 83, 48–56. [Google Scholar] [CrossRef]
  154. Röttgerding, F.; Kraiczy, P. Immune Evasion Strategies of Relapsing Fever Spirochetes. Front. Immunol. 2020, 11, 1560. [Google Scholar] [CrossRef]
  155. Hallström, T.; Siegel, C.; Mörgelin, M.; Kraiczy, P.; Skerka, C.; Zipfel, P.F. CspA from Borrelia Burgdorferi Inhibits the Terminal Complement Pathway. MBio 2013, 4, e00481-13. [Google Scholar] [CrossRef] [PubMed]
  156. Hart, T.; Nguyen, N.T.T.; Nowak, N.A.; Zhang, F.; Linhardt, R.J.; Diuk-Wasser, M.; Ram, S.; Kraiczy, P.; Lin, Y.-P. Polymorphic Factor H-Binding Activity of CspA Protects Lyme Borreliae from the Host Complement in Feeding Ticks to Facilitate Tick-to-Host Transmission. PLoS Pathog. 2018, 14, e1007106. [Google Scholar] [CrossRef]
  157. Lin, Y.-P.; Frye, A.M.; Nowak, T.A.; Kraiczy, P. New Insights Into CRASP-Mediated Complement Evasion in the Lyme Disease Enzootic Cycle. Front. Cell. Infect. Microbiol. 2020, 10, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Garcia, B.L.; Zhi, H.; Wager, B.; Höök, M.; Skare, J.T. Borrelia Burgdorferi BBK32 Inhibits the Classical Pathway by Blocking Activation of the C1 Complement Complex. PLoS Pathog. 2016, 12, e1005404. [Google Scholar] [CrossRef] [PubMed]
  159. Kraiczy, P. Hide and Seek: How Lyme Disease Spirochetes Overcome Complement Attack. Front. Immunol. 2016, 7, 385. [Google Scholar] [CrossRef] [PubMed]
  160. Locke, J.W. Complement Evasion in Borrelia Spirochetes: Mechanisms and Opportunities for Intervention. Antibiotics 2019, 8, 80. [Google Scholar] [CrossRef]
  161. Koenigs, A.; Hammerschmidt, C.; Jutras, B.L.; Pogoryelov, D.; Barthel, D.; Skerka, C.; Kugelstadt, D.; Wallich, R.; Stevenson, B.; Zipfel, P.F.; et al. BBA70 of Borrelia Burgdorferi Is a Novel Plasminogen-Binding Protein. J. Biol. Chem. 2013, 288, 25229–25243. [Google Scholar] [CrossRef]
  162. Talagrand-Reboul, E.; Boyer, P.H.; Bergström, S.; Vial, L.; Boulanger, N. Relapsing Fevers: Neglected Tick-Borne Diseases. Front. Cell. Infect. Microbiol. 2018, 8, 98. [Google Scholar] [CrossRef]
  163. Cutler, S.J. Relapsing Fever—A Forgotten Disease Revealed. J. Appl. Microbiol. 2010, 108, 1115–1122. [Google Scholar] [CrossRef]
  164. Forrester, J.D.; Kjemtrup, A.M.; Fritz, C.L.; Marsden-Haug, N.; Nichols, J.B.; Tengelsen, L.A.; Sowadsky, R.; DeBess, E.; Cieslak, P.R.; Weiss, J.; et al. Tickborne Relapsing Fever—United States, 1990–2011. MMWR Morb. Mortal. Wkly. Rep. 2015, 64, 58–60. [Google Scholar]
  165. CDC Symptoms. Available online: https://www.cdc.gov/relapsing-fever/symptoms/index.html (accessed on 10 February 2022).
  166. Lopez, J.; Hovius, J.W.; Bergström, S. Pathogenesis of Relapsing Fever. In Lyme Disease and Relapsing Fever Spirochetes: Genomics, Molecular Biology, Host Interactions and Disease Pathogenesis 2021; Caister Academic Press: Wymondham, UK, 2021. [Google Scholar]
  167. Adrion, E.R.; Aucott, J.; Lemke, K.W.; Weiner, J.P. Health Care Costs, Utilization and Patterns of Care Following Lyme Disease. PLoS ONE 2015, 10, e0116767. [Google Scholar]
  168. Pfoh, E.; Wessels, M.R.; Goldmann, D.; Lee, G.M. Burden and Economic Cost of Group A Streptococcal Pharyngitis. Pediatrics 2008, 121, 229–234. [Google Scholar] [CrossRef] [PubMed]
  169. Luo, Y.; Xie, Y.; Xiao, Y. Laboratory Diagnostic Tools for Syphilis: Current Status and Future Prospects. Front. Cell. Infect. Microbiol. 2020, 10, 574806. [Google Scholar] [CrossRef] [PubMed]
  170. Park, I.U.; Fakile, Y.F.; Chow, J.M.; Gustafson, K.J.; Jost, H.; Schapiro, J.M.; Novak-Weekley, S.; Tran, A.; Nomura, J.H.; Chen, V.; et al. Performance of Treponemal Tests for the Diagnosis of Syphilis. Clin. Infect. Dis. 2019, 68, 913–918. [Google Scholar] [CrossRef] [PubMed]
  171. Worrall, G.; Hutchinson, J.; Sherman, G.; Griffiths, J. Diagnosing Streptococcal Sore Throat in Adults: Randomized Controlled Trial of in-Office Aids. Can. Fam. Phys. 2007, 53, 666–671. [Google Scholar]
  172. Tansarli, G.S.; LeBlanc, L.; Auld, D.B.; Chapin, K.C. Diagnostic Accuracy of Presurgical Staphylococcus Aureus PCR Assay Compared with Culture and Post-PCR Implementation Surgical Site Infection Rates. J. Mol. Diagn. 2020, 22, 1063–1069. [Google Scholar] [CrossRef]
  173. Schmidt, B.L. PCR in Laboratory Diagnosis of Human Borrelia Burgdorferi Infections. Clin. Microbiol. Rev. 1997, 10, 185–201. [Google Scholar] [CrossRef]
  174. Aucott, J.; Morrison, C.; Munoz, B.; Rowe, P.C.; Schwarzwalder, A.; West, S.K. Diagnostic Challenges of Early Lyme Disease: Lessons from a Community Case Series. BMC Infect. Dis. 2009, 9, 79. [Google Scholar] [CrossRef]
  175. Barrett, J. It’s about Lyme: Why Congress Must Enact Medical Insurance Coverage Laws for Lyme Disease Patients Now. Seattle U. L. Rev. SUpra 2022, 45, 50. [Google Scholar]
  176. St Pierre, S.E.; Gould, O.N.; Lloyd, V. Knowledge and Knowledge Needs about Lyme Disease among Occupational and Recreational Users of the Outdoors. Int. J. Environ. Res. Public Health 2020, 17, 355. [Google Scholar] [CrossRef]
  177. Moore, A.; Nelson, C.; Molins, C.; Mead, P.; Schriefer, M. Current Guidelines, Common Clinical Pitfalls, and Future Directions for Laboratory Diagnosis of Lyme Disease, United States. Emerg. Infect. Dis. 2016, 22, 1167–1177. [Google Scholar] [CrossRef] [PubMed]
  178. Waddell, L.A.; Greig, J.; Mascarenhas, M.; Harding, S.; Lindsay, R.; Ogden, N. The Accuracy of Diagnostic Tests for Lyme Disease in Humans, A Systematic Review and Meta-Analysis of North American Research. PLoS ONE 2016, 11, e0168613. [Google Scholar] [CrossRef] [PubMed]
  179. Radolf, J.D.; Scott Samuels, D. Borrelia: Molecular Biology, Host Interaction and Pathogenesis; Horizon Scientific Press: Norwich, UK, 2010; ISBN 9781904455585. [Google Scholar]
  180. Marques, A.R. Laboratory Diagnosis of Lyme Disease: Advances and Challenges. Infect. Dis. Clin. North Am. 2015, 29, 295–307. [Google Scholar] [CrossRef] [PubMed]
  181. Zagorac, G.B.; Kezele, T.G. Ceftriaxone and Doxycycline Induced Seroconversion in Previously Seronegative Patient with Clinically Suspected Disseminated Lyme Disease: Case Report. Infect Chemother. 2021, 53, 582–588. [Google Scholar] [CrossRef]
  182. Coyle, P.K. Advances and Pitfalls in the Diagnosis of Lyme Disease. FEMS Immunol. Med. Microbiol. 1997, 19, 103–109. [Google Scholar] [CrossRef]
  183. Anderson, C.; Brissette, C.A. The Brilliance of Borrelia: Mechanisms of Host Immune Evasion by Lyme Disease-Causing Spirochetes. Pathogens 2021, 10, 281. [Google Scholar] [CrossRef]
  184. Wang, G.; Aguero-Rosenfeld, M.E.; Wormser, G.P. Detection of Borrelia Burgdorferi; Caister Academic Press: Norfolk, UK, 2010. [Google Scholar]
  185. Dumler, J. Molecular Diagnosis of Lyme Disease: Review and Meta-Analysis. Mol. Diagn. 2001, 6, 1–11. [Google Scholar] [CrossRef]
  186. Singh, S.K.; Girschick, H.J. Lyme Borreliosis: From Infection to Autoimmunity. Clin. Microbiol. Infect. 2004, 10, 598–614. [Google Scholar] [CrossRef]
  187. Norris, S.J.; Coburn, J.; Leong, J.M.; Hu, L.T.; Hook, M. Pathobiology of Lyme Disease Borrelia. In Borrelia: Molecular Biology, Host Interaction And Pathogenesis; Horizon Scientific Press: Norwich, UK, 2010; pp. 293–325. [Google Scholar]
  188. Barbour, A.G.; Guo, B.P. Pathogenesis of Relapsing Fever. Borrelia Mol. Biol. Host Interact. Pathogenesis 2010, 333–357. [Google Scholar]
  189. Dattwyler, R.J.; Wormser, G.P.; Rush, T.J.; Finkel, M.F.; Schoen, R.T.; Grunwaldt, E.; Franklin, M.; Hilton, E.; Bryant, G.L.; Agger, W.A.; et al. A Comparison of Two Treatment Regimens of Ceftriaxone in Late Lyme Disease. Wien. Klin. Wochenschr. 2005, 117, 393–397. [Google Scholar] [CrossRef]
  190. Donta, S.T. What We Know and Don’t Know About Lyme Disease. Front Public Health 2021, 9, 819541. [Google Scholar] [CrossRef] [PubMed]
  191. Tokarz, R.; Mishra, N.; Tagliafierro, T.; Sameroff, S.; Caciula, A.; Chauhan, L.; Patel, J.; Sullivan, E.; Gucwa, A.; Fallon, B.; et al. A Multiplex Serologic Platform for Diagnosis of Tick-Borne Diseases. Sci. Rep. 2018, 8, 3158. [Google Scholar] [CrossRef]
  192. Perronne, C. Lyme and Associated Tick-Borne Diseases: Global Challenges in the Context of a Public Health Threat. Front. Cell. Infect. Microbiol. 2014, 4, 74. [Google Scholar] [CrossRef]
  193. Maraspin, V.; Ogrinc, K.; Ružić-Sabljić, E.; Lotrič-Furlan, S.; Strle, F. Isolation of Borrelia Burgdorferi Sensu Lato from Blood of Adult Patients with Borrelial Lymphocytoma, Lyme Neuroborreliosis, Lyme Arthritis and Acrodermatitis Chronica Atrophicans. Infection 2011, 39, 35–40. [Google Scholar] [CrossRef] [PubMed]
  194. Schmidli, J.; Hunziker, T.; Moesli, P.; Schaad, U.B. Cultivation of Borrelia Burgdorferi from Joint Fluid Three Months after Treatment of Facial Palsy due to Lyme Borreliosis. J. Infect. Dis. 1988, 158, 905–906. [Google Scholar] [CrossRef] [PubMed]
  195. Stanek, G.; Klein, J.; Bittner, R.; Glogar, D. Isolation of Borrelia Burgdorferi from the Myocardium of a Patient with Long-Standing Cardiomyopathy. N. Engl. J. Med. 1990, 322, 249–252. [Google Scholar] [CrossRef] [PubMed]
  196. Preac-Mursic, V.; Pfister, H.W.; Spiegel, H.; Burk, R.; Wilske, B.; Reinhardt, S.; Böhmer, R. First Isolation of Borrelia Burgdorferi from an Iris Biopsy. J. Clin. Neuroophthalmol. 1993, 13, 155–161, discussion 162. [Google Scholar] [PubMed]
  197. Chao, L.-L.; Chen, Y.-J.; Shih, C.-M. First Isolation and Molecular Identification of Borrelia Burgdorferi Sensu Stricto and Borrelia Afzelii from Skin Biopsies of Patients in Taiwan. Int. J. Infect. Dis. 2011, 15, e182–e187. [Google Scholar] [CrossRef] [Green Version]
  198. Liveris, D.; Schwartz, I.; McKenna, D.; Nowakowski, J.; Nadelman, R.; DeMarco, J.; Iyer, R.; Bittker, S.; Cooper, D.; Holmgren, D.; et al. Comparison of Five Diagnostic Modalities for Direct Detection of Borrelia Burgdorferi in Patients with Early Lyme Disease. Diagn. Microbiol. Infect. Dis. 2012, 73, 243–245. [Google Scholar] [CrossRef]
  199. Liveris, D.; Wang, G.; Girao, G.; Byrne, D.W.; Nowakowski, J.; McKenna, D.; Nadelman, R.; Wormser, G.P.; Schwartz, I. Quantitative Detection of Borrelia Burgdorferi in 2-Millimeter Skin Samples of Erythema Migrans Lesions: Correlation of Results with Clinical and Laboratory Findings. J. Clin. Microbiol. 2002, 40, 1249–1253. [Google Scholar] [CrossRef]
  200. Eshoo, M.W.; Crowder, C.C.; Rebman, A.W.; Rounds, M.A.; Matthews, H.E.; Picuri, J.M.; Soloski, M.J.; Ecker, D.J.; Schutzer, S.E.; Aucott, J.N. Direct Molecular Detection and Genotyping of Borrelia Burgdorferi from Whole Blood of Patients with Early Lyme Disease. PLoS ONE 2012, 7, e36825. [Google Scholar] [CrossRef] [PubMed]
  201. Branda, J.A.; Steere, A.C. Laboratory Diagnosis of Lyme Borreliosis. Clin. Microbiol. Rev. 2021, 34, e00018-19. [Google Scholar] [CrossRef] [PubMed]
  202. Lambert, J.S.; Cook, M.J.; Healy, J.E.; Murtagh, R.; Avramovic, G.; Lee, S.H. Metagenomic 16S rRNA Gene Sequencing Survey of Borrelia Species in Irish Samples of Ixodes Ricinus Ticks. PLoS ONE 2019, 14, e0209881. [Google Scholar] [CrossRef] [PubMed]
  203. Schettig, R.; Tan-Lim, R.; Warren, D.; Poteet, Z.; Sears, R.; Hummel, M.; Coffin, C.; Quart, K.; Aussems, C.; Matthias, R., Jr.; et al. PathoDNA, an Advanced Diagnostic for Lyme Disease & Co-Infections Utilizing next Generation DNA Sequencing with Greater Sensitivity and Selectivity than ELISA/western Blot. Adv. Infect. Dis. 2021, 11, 405–429. [Google Scholar]
  204. Rauter, C.; Mueller, M.; Diterich, I.; Zeller, S.; Hassler, D.; Meergans, T.; Hartung, T. Critical Evaluation of Urine-Based PCR Assay for Diagnosis of Lyme Borreliosis. Clin. Diagn. Lab. Immunol. 2005, 12, 910–917. [Google Scholar] [CrossRef]
  205. Mosel, M.R.; Carolan, H.E.; Rebman, A.W.; Castro, S.; Massire, C.; Ecker, D.J.; Soloski, M.J.; Aucott, J.N.; Eshoo, M.W. Molecular Testing of Serial Blood Specimens from Patients with Early Lyme Disease during Treatment Reveals Changing Coinfection with Mixtures of Borrelia Burgdorferi Genotypes. Antimicrob. Agents Chemother. 2019, 63, e00237-19. [Google Scholar] [CrossRef]
  206. Eshoo, M.W.; Schutzer, S.E.; Crowder, C.D.; Carolan, H.E.; Ecker, D.J. Achieving Molecular Diagnostics for Lyme Disease. Expert Rev. Mol. Diagn. 2013, 13, 875–883. [Google Scholar] [CrossRef]
  207. Branda, J.A.; Lemieux, J.E.; Blair, L.; Ahmed, A.A.; Hong, D.K.; Bercovici, S.; Blauwkamp, T.A.; Hollemon, D.; Ho, C.; Strle, K.; et al. Detection of Borrelia Burgdorferi Cell-Free DNA in Human Plasma Samples for Improved Diagnosis of Early Lyme Borreliosis. Clin. Infect. Dis. 2021, 73, e2355–e2361. [Google Scholar] [CrossRef]
  208. Lee, S.H.; Healy, J.E.; Lambert, J.S. Single Core Genome Sequencing for Detection of Both Borrelia Burgdorferi Sensu Lato and Relapsing Fever Borrelia Species. Int. J. Environ. Res. Public Health 2019, 16, 1779. [Google Scholar] [CrossRef] [Green Version]
  209. Stefas, I.; Dubois, G.; Tigrett, S.; Lucarz, E.; Veas, F. Apolipoprotein H, an acute phase protein, a performing tool for ultra-sensitive detection and isolation of microorganisms from different origins. In Acute Phase Proteins as Early Non-Specific Biomarkers of Human and Veterinary Diseases; Veas, F., Ed.; InTechOpen: London, UK, 2011; pp. 21–42. [Google Scholar]
  210. Lacout, A.; Mone, Y.; Franck, M.; Marcy, P.-Y.; Mas, M.; Veas, F.; Perronne, C. Blood Cell Disruption to Significantly Improve the Borrelia PCR Detection Sensitivity in Borreliosis in Humans. Med. Hypotheses 2018, 116, 1–3. [Google Scholar] [CrossRef]
  211. Adlhoch, C.; Kaiser, M.; Hoehne, M.; Mas Marques, A.; Stefas, I.; Veas, F.; Ellerbrok, H. Highly Sensitive Detection of the Group A Rotavirus Using Apolipoprotein H-Coated ELISA Plates Compared to Quantitative Real-Time PCR. Virol. J. 2011, 8, 63. [Google Scholar] [CrossRef] [PubMed]
  212. Stefas, I.; Rucheton, M.; D’Angeac, A.D.; Morel-Baccard, C.; Seigneurin, J.M.; Zarski, J.P.; Martin, M.; Cerutti, M.; Bossy, J.P.; Missé, D.; et al. Hepatitis B Virus Dane Particles Bind to Human Plasma Apolipoprotein H. Hepatology 2001, 33, 207–217. [Google Scholar] [CrossRef]
  213. Stefas, E.; Rucheton, M.; Graafland, H.; Moynier, M.; Sompeyrac, C.; Bahraoui, E.M.; Veas, F. Human Plasmatic Apolipoprotein H Binds Human Immunodeficiency Virus Type 1 and Type 2 Proteins. AIDS Res. Hum. Retroviruses 1997, 13, 97–104. [Google Scholar] [CrossRef]
  214. Greissl, J.; Pesesky, M.; Dalai, S.C.; Rebman, A.W.; Soloski, M.J.; Horn, E.J.; Dines, J.N.; Gittelman, R.M.; Snyder, T.M.; Emerson, R.O.; et al. Immunosequencing of the T-Cell Receptor Repertoire Reveals Signatures Specific for Diagnosis and Characterization of Early Lyme Disease. bioRxiv 2021. [Google Scholar] [CrossRef]
  215. Hefter, Y.; D’Arco, C.; Shute, T.; Dattwyler, R.; Arnaboldi, P.; Nolan, S. 1760. Interferon Gamma Release Assay for Diagnosis of Lyme Disease. Open Forum Infect Dis 2018, 5, S61. [Google Scholar] [CrossRef]
  216. Shan, J.; Jia, Y.; Teulières, L.; Patel, F.; Clokie, M.R.J. Targeting Multicopy Prophage Genes for the Increased Detection of Borrelia Burgdorferi Sensu Lato (s.l.), the Causative Agents of Lyme Disease, in Blood. Front. Microbiol. 2021, 12, 651217. [Google Scholar] [CrossRef]
  217. Zhou, Y.; Qin, S.; Sun, M.; Tang, L.; Yan, X.; Kim, T.-K.; Caballero, J.; Glusman, G.; Brunkow, M.E.; Soloski, M.J.; et al. Measurement of Organ-Specific and Acute-Phase Blood Protein Levels in Early Lyme Disease. J. Proteome Res. 2020, 19, 346–359. [Google Scholar] [CrossRef]
  218. Marques, A.; Telford, S.R., 3rd; Turk, S.-P.; Chung, E.; Williams, C.; Dardick, K.; Krause, P.J.; Brandeburg, C.; Crowder, C.D.; Carolan, H.E.; et al. Xenodiagnosis to Detect Borrelia Burgdorferi Infection: A First-in-Human Study. Clin. Infect. Dis. 2014, 58, 937–945. [Google Scholar] [CrossRef]
  219. Murfin, K.E.; Kleinbard, R.; Aydin, M.; Salazar, S.A.; Fikrig, E. Borrelia Burgdorferi Chemotaxis toward Tick Protein Salp12 Contributes to Acquisition. Ticks Tick-Borne Dis. 2019, 10, 1124–1134. [Google Scholar] [CrossRef]
  220. Shih, C.-M.; Chao, L.-L.; Yu, C.-P. Chemotactic Migration of the Lyme Disease Spirochete (Borrelia Burgdorferi) to Salivary Gland Extracts of Vector Ticks. Am. J. Trop. Med. Hyg. 2002, 66, 616–621. [Google Scholar] [CrossRef]
  221. Embers, M.E.; Barthold, S.W.; Borda, J.T.; Bowers, L.; Doyle, L.; Hodzic, E.; Jacobs, M.B.; Hasenkampf, N.R.; Martin, D.S.; Narasimhan, S.; et al. Persistence of Borrelia Burgdorferi in Rhesus Macaques Following Antibiotic Treatment of Disseminated Infection. PLoS ONE 2012, 7, e29914. [Google Scholar] [CrossRef]
  222. Embers, M.E.; Hasenkampf, N.R.; Jacobs, M.B.; Tardo, A.C.; Doyle-Meyers, L.A.; Philipp, M.T.; Hodzic, E. Variable Manifestations, Diverse Seroreactivity and Post-Treatment Persistence in Non-Human Primates Exposed to Borrelia Burgdorferi by Tick Feeding. PLoS ONE 2017, 12, e0189071. [Google Scholar] [CrossRef] [PubMed]
  223. Bockenstedt, L.K.; Mao, J.; Hodzic, E.; Barthold, S.W.; Fish, D. Detection of Attenuated, Noninfectious Spirochetes inBorrelia burgdorferi–Infected Mice after Antibiotic Treatment. J. Infect. Dis. 2002, 186, 1430–1437. [Google Scholar] [CrossRef] [PubMed]
  224. Molins, C.R.; Ashton, L.V.; Wormser, G.P.; Hess, A.M.; Delorey, M.J.; Mahapatra, S.; Schriefer, M.E.; Belisle, J.T. Development of a Metabolic Biosignature for Detection of Early Lyme Disease. Clin. Infect. Dis. 2015, 60, 1767–1775. [Google Scholar] [CrossRef]
  225. Fitzgerald, B.L.; Molins, C.R.; Islam, M.N.; Graham, B.; Hove, P.R.; Wormser, G.P.; Hu, L.; Ashton, L.V.; Belisle, J.T. Host Metabolic Response in Early Lyme Disease. J. Proteome Res. 2020, 19, 610–623. [Google Scholar] [CrossRef]
  226. Haslund-Gourley, B.S.; Grauzam, S.; Mehta, A.S.; Wigdahl, B.; Comunale, M.A. Acute lyme disease IgG N-linked glycans contrast the canonical inflammatory signature. Front. Immunol. 2022, 13, 949118. [Google Scholar] [CrossRef]
  227. Fraser, C.M.; Casjens, S.; Huang, W.M.; Sutton, G.G.; Clayton, R.; Lathigra, R.; White, O.; Ketchum, K.A.; Dodson, R.; Hickey, E.K.; et al. Genomic Sequence of a Lyme Disease Spirochaete, Borrelia Burgdorferi. Nature 1997, 390, 580–586. [Google Scholar] [CrossRef]
  228. Picardeau, M.; Lobry, J.R.; Hinnebusch, B.J. Physical Mapping of an Origin of Bidirectional Replication at the Centre of the Borrelia Burgdorferi Linear Chromosome. Mol. Microbiol. 1999, 32, 437–445. [Google Scholar] [CrossRef] [PubMed]
  229. Nguyen, K.T.; Wu, J.-C.; Boylan, J.A.; Gherardini, F.C.; Pei, D. Zinc Is the Metal Cofactor of Borrelia Burgdorferi Peptide Deformylase. Arch. Biochem. Biophys. 2007, 468, 217–225. [Google Scholar] [CrossRef] [Green Version]
  230. Ouyang, Z.; He, M.; Oman, T.; Yang, X.F.; Norgard, M.V. A Manganese Transporter, BB0219 (BmtA), Is Required for Virulence by the Lyme Disease Spirochete, Borrelia Burgdorferi. Proc. Natl. Acad. Sci. USA 2009, 106, 3449–3454. [Google Scholar] [CrossRef]
  231. Groshong, A.M.; Blevins, J.S. Insights into the Biology of Borrelia Burgdorferi Gained through the Application of Molecular Genetics. Adv. Appl. Microbiol. 2014, 86, 141–143. [Google Scholar]
  232. Posey, J.E.; Gherardini, F.C. Lack of a Role for Iron in the Lyme Disease Pathogen. Science 2000, 288, 1651–1653. [Google Scholar] [CrossRef]
  233. Troxell, B.; Xu, H.; Frank Yang, X. Borrelia Burgdorferi, a Pathogen That Lacks Iron, Encodes Manganese-Dependent Superoxide Dismutase Essential for Resistance to Streptonigrin. J. Biol. Chem. 2012, 287, 19284–19293. [Google Scholar] [CrossRef] [PubMed]
  234. Aguirre, J.D.; Clark, H.M.; McIlvin, M.; Vazquez, C.; Palmere, S.L.; Grab, D.J.; Seshu, J.; Hart, P.J.; Saito, M.; Culotta, V.C. A Manganese-Rich Environment Supports Superoxide Dismutase Activity in a Lyme Disease Pathogen, Borrelia Burgdorferi. J. Biol. Chem. 2013, 288, 8468–8478. [Google Scholar] [CrossRef] [PubMed]
  235. Troxell, B.; Yang, X.F. Metal-Dependent Gene Regulation in the Causative Agent of Lyme Disease. Front. Cell. Infect. Microbiol. 2013, 3, 79. [Google Scholar] [CrossRef] [PubMed]
  236. Casjens, S.; Eggers, C.H.; Schwartz, I. Borrelia Genomics: Chromosome, Plasmids, Bacteriophages and Genetic Variation. Borrelia: Molecular Biology, Host Interaction and Pathogenesis; Caister Academic Press: Hethersett, UK, 2010; pp. 27–53. [Google Scholar]
  237. Margos, G.; Hojgaard, A.; Lane, R.S.; Cornet, M.; Fingerle, V.; Rudenko, N.; Ogden, N.; Aanensen, D.M.; Fish, D.; Piesman, J. Multilocus Sequence Analysis of Borrelia Bissettii Strains from North America Reveals a New Borrelia Species, Borrelia Kurtenbachii. Ticks Tick Borne Dis. 2010, 1, 151–158. [Google Scholar] [CrossRef] [PubMed]
  238. Rudenko, N.; Golovchenko, M.; Grubhoffer, L.; Oliver, J.H., Jr. Borrelia carolinensis Sp. Nov., a New (14th) Member of the Borrelia Burgdorferi Sensu Lato Complex from the Southeastern Region of the United States. J. Clin. Microbiol. 2009, 47, 134–141. [Google Scholar] [CrossRef]
  239. Rudenko, N.; Golovchenko, M.; Lin, T.; Gao, L.; Grubhoffer, L.; Oliver, J.H., Jr. Delineation of a New Species of the Borrelia Burgdorferi Sensu Lato Complex, Borrelia Americana Sp. Nov. J. Clin. Microbiol. 2009, 47, 3875–3880. [Google Scholar] [CrossRef]
  240. Stanek, G.; Reiter, M. The Expanding Lyme Borrelia Complex—Clinical Significance of Genomic Species? Clin. Microbiol. Infect. 2011, 17, 487–493. [Google Scholar] [CrossRef]
  241. Lescot, M.; Audic, S.; Robert, C.; Nguyen, T.T.; Blanc, G.; Cutler, S.J.; Wincker, P.; Couloux, A.; Claverie, J.-M.; Raoult, D.; et al. The Genome of Borrelia Recurrentis, the Agent of Deadly Louse-Borne Relapsing Fever, Is a Degraded Subset of Tick-Borne Borrelia Duttonii. PLoS Genet. 2008, 4, e1000185. [Google Scholar] [CrossRef]
  242. Kuleshov, K.V.; Margos, G.; Fingerle, V.; Koetsveld, J.; Goptar, I.A.; Markelov, M.L.; Kolyasnikova, N.M.; Sarksyan, D.S.; Kirdyashkina, N.P.; Shipulin, G.A.; et al. Whole Genome Sequencing of Borrelia Miyamotoi Isolate Izh-4: Reference for a Complex Bacterial Genome. BMC Genom. 2020, 21, 16. [Google Scholar] [CrossRef] [PubMed]
  243. Carlyon, J.A.; Marconi, R.T. Cloning and Molecular Characterization of a Multicopy, Linear Plasmid-Carried, Repeat Motif-Containing Gene from Borrelia Turicatae, a Causative Agent of Relapsing Fever. J. Bacteriol. 1998, 180, 4974–4981. [Google Scholar] [CrossRef] [PubMed]
  244. Genomics of Relapsing Fever Spirochetes. In Lyme Disease and Relapsing Fever Spirochetes: Genomics, Molecular Biology, Host Interactions and Disease Pathogenesis; Caister Academic Press: Hethersett, UK, 2021; ISBN 9781913652616.
  245. Barbour, A.G. Phylogeny of a Relapsing Fever Borrelia Species Transmitted by the Hard Tick Ixodes Scapularis. Infect. Genet. Evol. 2014, 27, 551–558. [Google Scholar] [CrossRef] [PubMed]
  246. Purser, J.E.; Norris, S.J. Correlation between Plasmid Content and Infectivity in Borrelia Burgdorferi. Proc. Natl. Acad. Sci. USA 2000, 97, 13865–13870. [Google Scholar] [CrossRef] [PubMed]
  247. Schwartz, I.; Margos, G.; Casjens, S.R.; Qiu, W.-G.; Eggers, C.H. Multipartite Genome of Lyme Disease Borrelia: Structure, Variation and Prophages. Curr. Issues Mol. Biol. 2021, 42, 409–454. [Google Scholar]
  248. Casjens, S.R. The Unusual Linear Plasmids of the Lyme Disease Spirochete Borrelia Burgdorferi. Biochem. Soc. Transact. 2000, 28, A102. [Google Scholar] [CrossRef]
  249. Casjens, S.R.; Gilcrease, E.B.; Vujadinovic, M.; Mongodin, E.F.; Luft, B.J.; Schutzer, S.E.; Fraser, C.M.; Qiu, W.-G. Plasmid Diversity and Phylogenetic Consistency in the Lyme Disease Agent Borrelia Burgdorferi. BMC Genom. 2017, 18, 165. [Google Scholar] [CrossRef]
  250. Balakirev, E.S.; Ayala, F.J. Pseudogenes: Are They “Junk” or Functional DNA? Annu. Rev. Genet. 2003, 37, 123–151. [Google Scholar] [CrossRef]
  251. Casjens, S.R.; Di, L.; Akther, S.; Mongodin, E.F.; Luft, B.J.; Schutzer, S.E.; Fraser, C.M.; Qiu, W.-G. Primordial Origin and Diversification of Plasmids in Lyme Disease Agent Bacteria. BMC Genom. 2018, 19, 218. [Google Scholar] [CrossRef]
  252. Dulebohn, D.P.; Bestor, A.; Rosa, P.A. Borrelia Burgdorferi Linear Plasmid 28-3 Confers a Selective Advantage in an Experimental Mouse-Tick Infection Model. Infect. Immun. 2013, 81, 2986–2996. [Google Scholar] [CrossRef]
  253. Kobryn, K. Replication of the Borrelia Burgdorferi Genome. In Lyme Disease and Relapsing Fever Spirochetes: Genomics, Molecular Biology, Host Interactions and Disease Pathogenesis 2021; Caister Academic Press: Wymondham, UK, 2021. [Google Scholar]
  254. Kobryn, K.; Chaconas, G. ResT, a Telomere Resolvase Encoded by the Lyme Disease Spirochete. Mol. Cell 2002, 9, 195–201. [Google Scholar] [CrossRef]
  255. Iyer, R.; Kalu, O.; Purser, J.; Norris, S.; Stevenson, B.; Schwartz, I. Linear and Circular Plasmid Content in Borrelia Burgdorferi Clinical Isolates. Infect. Immun. 2003, 71, 3699–3706. [Google Scholar] [CrossRef] [PubMed]
  256. Stevenson, B.; Porcella, S.F.; Oie, K.L.; Fitzpatrick, C.A.; Raffel, S.J.; Lubke, L.; Schrumpf, M.E.; Schwan, T.G. The Relapsing Fever Spirochete Borrelia Hermsii Contains Multiple, Antigen-Encoding Circular Plasmids That Are Homologous to the cp32 Plasmids of Lyme Disease Spirochetes. Infect. Immun. 2000, 68, 3900–3908. [Google Scholar] [CrossRef] [PubMed]
  257. Innamorati, K.A.; Earl, J.P.; Aggarwal, S.D.; Ehrlich, G.D.; Hiller, N.L. The Bacterial Guide to Designing a Diversified Gene Portfolio. In The Pangenome: Diversity, Dynamics and Evolution of Genomes; Tettelin, H., Medini, D., Eds.; Springer International Publishing: Cham, The Netherlands, 2020; pp. 51–87. ISBN 9783030382810. [Google Scholar]
  258. van Vliet, A.H.M. Use of Pan-Genome Analysis for the Identification of Lineage-Specific Genes of Helicobacter Pylori. FEMS Microbiol. Lett. 2017, 364, fnw296. [Google Scholar] [CrossRef]
  259. Hogg, J.S.; Hu, F.Z.; Janto, B.; Boissy, R.; Hayes, J.; Keefe, R.; Post, J.C.; Ehrlich, G.D. Characterization and Modeling of the Haemophilus Influenzae Core and Supragenomes Based on the Complete Genomic Sequences of Rd and 12 Clinical Nontypeable Strains. Genome Biol. 2007, 8, R103. [Google Scholar] [CrossRef]
  260. Forgetta, V.; Oughton, M.T.; Marquis, P.; Brukner, I.; Blanchette, R.; Haub, K.; Magrini, V.; Mardis, E.R.; Gerding, D.N.; Loo, V.G.; et al. Fourteen-Genome Comparison Identifies DNA Markers for Severe-Disease-Associated Strains of Clostridium Difficile. J. Clin. Microbiol. 2011, 49, 2230–2238. [Google Scholar] [CrossRef]
  261. Udaondo, Z.; Duque, E.; Ramos, J.-L. The Pangenome of the Genus Clostridium. Environ. Microbiol. 2017, 19, 2588–2603. [Google Scholar] [CrossRef]
  262. Marshall, D.J.; Heisler, L.M.; Lyamichev, V.; Murvine, C.; Olive, D.M.; Ehrlich, G.D.; Neri, B.P.; de Arruda, M. Determination of Hepatitis C Virus Genotypes in the United States by Cleavase Fragment Length Polymorphism Analysis. J. Clin. Microbiol. 1997, 35, 3156–3162. [Google Scholar] [CrossRef]
  263. Shen, K.; Wang, X.; Post, J.C. Molecular and Translational Research Approaches for the Study of Bacterial Pathogenesis in Otitis Media. In Evidence-Based Otitis Media, 2nd ed.; Rosenfeld, R., Bluestone, C.D., Eds.; B.C. Decker Inc.: Hamilton, ON, Canada, 2003; pp. 91–119. [Google Scholar]
  264. Ehrlich, G.D.; Hu, F.Z.; Post, J.C. Role for Biofilms in Infectious Disease. In Microbial Biofilms; American Society of Microbiology: Washington, DC, USA, 2004; pp. 332–358. ISBN 9781555818944. [Google Scholar]
  265. Gladitz, J.; Shen, K.; Antalis, P.; Hu, F.Z.; Post, J.C.; Ehrlich, G.D. Codon Usage Comparison of Novel Genes in Clinical Isolates of Haemophilus Influenzae. Nucleic Acids Res. 2005, 33, 3644–3658. [Google Scholar] [CrossRef]
  266. Shen, K.; Antalis, P.; Gladitz, J.; Sayeed, S.; Ahmed, A.; Yu, S.; Hayes, J.; Johnson, S.; Dice, B.; Dopico, R.; et al. Identification, Distribution, and Expression of Novel Genes in 10 Clinical Isolates of Nontypeable Haemophilus Influenzae. Infect. Immun. 2005, 73, 3479–3491. [Google Scholar] [CrossRef]
  267. Shen, K.; Gladitz, J.; Antalis, P.; Dice, B.; Janto, B.; Keefe, R.; Hayes, J.; Ahmed, A.; Dopico, R.; Ehrlich, N.; et al. Characterization, Distribution, and Expression of Novel Genes among Eight Clinical Isolates of Streptococcus Pneumoniae. Infect. Immun. 2006, 74, 321–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. Livingstone, P.G.; Morphew, R.M.; Whitworth, D.E. Genome Sequencing and Pan-Genome Analysis of 23 Corallococcus spp. Strains Reveal Unexpected Diversity, With Particular Plasticity of Predatory Gene Sets. Front. Microbiol. 2018, 9, 3187. [Google Scholar] [CrossRef] [PubMed]
  269. Wu, Y.; Zaiden, N.; Cao, B. The Core- and Pan-Genomic Analyses of the Genus Comamonas: From Environmental Adaptation to Potential Virulence. Front. Microbiol. 2018, 9, 3096. [Google Scholar] [CrossRef] [PubMed]
  270. Earl, J.P.; de Vries, S.P.W.; Ahmed, A.; Powell, E.; Schultz, M.P.; Hermans, P.W.M.; Hill, D.J.; Zhou, Z.; Constantinidou, C.I.; Hu, F.Z.; et al. Comparative Genomic Analyses of the Moraxella Catarrhalis Serosensitive and Seroresistant Lineages Demonstrate Their Independent Evolution. Genome Biol. Evol. 2016, 8, 955–974. [Google Scholar] [CrossRef]
  271. Nistico, L.; Earl, J.; Hiller, L. Using the Core and Supra Genomes to Determine Diversity and Natural Proclivities among Bacterial Strains. In Application of Molecular Microbiological Methods; Skovhus, T.L., Caffrey, S., Hubert, C., Eds.; Caister Academic Press: Norfolk, UK, 2014. [Google Scholar]
  272. Kress-Bennett, J.M.; Hiller, N.L.; Eutsey, R.A.; Powell, E.; Longwell, M.J.; Hillman, T.; Blackwell, T.; Byers, B.; Mell, J.C.; Post, J.C.; et al. Identification and Characterization of Msf, a Novel Virulence Factor in Haemophilus Influenzae. PLoS ONE 2016, 11, e0149891. [Google Scholar]
  273. Margos, G.; Hepner, S.; Mang, C.; Marosevic, D.; Reynolds, S.E.; Krebs, S.; Sing, A.; Derdakova, M.; Reiter, M.A.; Fingerle, V. Lost in Plasmids: Next Generation Sequencing and the Complex Genome of the Tick-Borne Pathogen Borrelia Burgdorferi. BMC Genom. 2017, 18, 422. [Google Scholar] [CrossRef]
  274. Mongodin, E.F.; Casjens, S.R.; Bruno, J.F.; Xu, Y.; Drabek, E.F.; Riley, D.R.; Cantarel, B.L.; Pagan, P.E.; Hernandez, Y.A.; Vargas, L.C.; et al. Inter- and Intra-Specific Pan-Genomes of Borrelia Burgdorferi Sensu Lato: Genome Stability and Adaptive Radiation. BMC Genom. 2013, 14, 693. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The anatomy of the adult I. scapularis hard tick underneath a dissection microscope. The red arrows indicate the different anatomical parts of the I. scapularis.
Figure 1. The anatomy of the adult I. scapularis hard tick underneath a dissection microscope. The red arrows indicate the different anatomical parts of the I. scapularis.
Genes 13 01604 g001
Figure 2. Transmission and acquisition of Borreliellal spirochetes. The image was created in BioRender by KM Socarras 2022.
Figure 2. Transmission and acquisition of Borreliellal spirochetes. The image was created in BioRender by KM Socarras 2022.
Genes 13 01604 g002
Figure 3. Dark field microscopic images of wet mount Borrelial spirochaetes (Borrelia hermsii strain DAH). Blood from C3H-Hej mice infected with B. hermsii was diluted 4-fold with phosphate buffered saline. (A): 100×, (B,C): 400×.
Figure 3. Dark field microscopic images of wet mount Borrelial spirochaetes (Borrelia hermsii strain DAH). Blood from C3H-Hej mice infected with B. hermsii was diluted 4-fold with phosphate buffered saline. (A): 100×, (B,C): 400×.
Genes 13 01604 g003
Table 1. Leading anthropophilic ticks within the United States.
Table 1. Leading anthropophilic ticks within the United States.
Amblyomma americanumAmblyomma maculatumDermacentor andersoniiDermacentor variabilisIxodes cookeiIxodes pacificusIxodes scapularisRhicephalus sanguineusOrnithodoros hermsii
Common name 1Lone Star TickGulf Coast TickRocky Mountain Wood TickAmerican Dog TickGroundhog TickWestern Blacklegged tickBlacklegged Tick/Deer TickBrown Dog Tick
Type 3HardHardHardHardHardHardHardHardSoft
Prey 1,3rodents, domestic animalsrodents, domestic animalsrodents, domestic animalsdomestic animalsgroundhogs, skunks, rodents, racoons, foxes, weasels, domestic animalsmice, voles, weasels, deer, birds, lizardsmice, deer, birdsdomestic animalsrodents
Feeding style 33-host tick3-host tick3-host tick3-host tick3-host tick3-host3-host3-host tick
Dimorphic 3YesYesYesYesYesYesYesYesYes
Zone 1,2East and Southern USSouth-mid US, Southern Arizona East of Rocky Mountains, US, pacific coastEastern U.SWestern USEastern, Upper Midwest, and Southern USWorld-wide, * Southern border of U.SNorthwest coast, Texas, Florida
Peak activity 1,2Early Spring- Late fallEarly Spring- Late fall Spring and SummerEarly spring-Mid FallEarly spring-Mid FallEarly spring-Mid Fall Year-round
States 1,2AL, AR, CT, DE, FL, GA, IL, IN, IA, KS, KY, LA, ME, MD, MA, MS, MO, NE, NH, NJ, NY, NC, OH, OK, PA, RI, SC, TN, TX, VT, VA, WV, DCAL, AR, FL, GA, KS, LA, MS, MO, NC, OK, SC, TN, TX, VAAZ, CA, CO, ID, KS, MT
NE, NV, NM, ND, OK, OR, SD, UT, WA, WY
AL, AR, CA, CT, DE, FL, GA, IL, IN, IA, KS, KY, LA, ME, MD, MA, MI, MN, MS, MO, MT, NE
NH, NJ, NY, NC, ND, OH, OK, PA, RI, SC, SD, TN, TX, VT, VA, WV, WI, WY, DC
MEAZ, CA, NV, OR, UT, WAAL, AR, CT, DE, FL, GA, IL, IN, IA, KS, KY, LA, ME, MD, MA, MI, MN, MS, MO, NH, NJ, NY, NC, ND, OH, OK, PA, RI, SC, SD, TN, TX, VT, VA, WV, WI, DCAL, AK, AZ, AR, CA, CO, CT, DE, FL, GA, HI, ID, IL, IN, IA, KS, KY, LA, ME, MD
MA, MI, MN, MS, MO, MT, NE, NV, NH, NJ, NM, NY, NC, ND, OH, OK, OR, PA. RI, SC, SD, TN, TX, UT, VT, VA, WA, WV, WI, WY, DC
Habitat 1,2Wooded areasCoastal areasscrublands, lightly wooded areas, and open grasslandsHuman settlements Human settlements
1 Center for Disease Control and Prevention 2022. 2 National Environmental Health Association 2022. 3 Soneshine, Daniel E. (1992). Biology of Ticks Volume I. Oxford University Press. * This tick species is found within the United States and several countries within Europe.
Table 2. Leading tick-borne pathogens within the United States and their vectors.
Table 2. Leading tick-borne pathogens within the United States and their vectors.
Babesia microtiAnaplasma phagocytophilumBorrelia burgdorferiBorrelia miyamotoiBorrelia mayoniiEhrlichia chafeensisEhrlichia murisEhrlichia ewingiiFrancisella tularensisRickettsia parkeriRickettsia rickettsiiBourbon virusColtivirusHeartland virusPowassan virus
TypeParasiteBacteriaBacteriaBacteriaBacteriaBacteriaBacteriaBacteriaBacteriaBacteriaBacteriaVirusVirusVirusVirus
Disease Name 1BabesiosisAnaplasmosisLyme BorreliosisRelapsing FeverLyme borreliosisEhrlichiosisEhrlichiosisEhrlichiosisTularemiaR. parkeri spotted feverRocky Mountain Spotted FeverBourbon virus diseaseColorado Tick FeverHeartland virus diseasePowassan virus disease
Discovery 11990 [63]1932 [64]19821995 [65]2016 [66]1986 [67]2009 [68]19961912 [69]1937 [70]19062014 [71]1950 [72]2012 [73]1990 [74]
Reservoir 1Small mammals: [55] Peromyscus leucopus, Procyon lotor, Blarina brevicauda, Tamias striatusPeromyscus leucopus, Odocoileus virginianusPeromyscus leucopus, Odocoileus virginianus, Tamias striatus, Blarina brevicauda, Sorex cinereus, Sciurus carolinensis, etc. [53]Peromyscus leucopus, Apodemus spp., Microtus spp., Tamias spp., Sciuridae spp., etc. [75]Peromyscus leucopus, Tamiasciurus hudsonicus [76]Odocoileus virginianusPeromyscus leucopusOdocoileus virginianusRodentsUnknown Deer
TickVector 1I. scapularisI. scapularisI. scapularisI. scapularisI. pacificusA. americanumI. scapularisA. americanumA.Americanum, D. variabilis, D. andersoniiD. andersoniiD. variabilis, R. sanguineus, D. andersoniiA. americanumD. andersoniiA. americanumI. scapularis, I. cookei
1 Center for Disease Control and Prevention 2022. In contrast, the lone star tick, A. americanum, is found only in select regions in North America. This arachnid has a notably aggressive feeding behavior towards prey during all life stages. It targets primarily large prey such as O. virginianus, but also domestic animals and humans. While doing so, it can transmit several tick-borne pathogens such as Ehrlichia ewingii and E. chaffeensis (Table 2) [67,77]. To date, this tick has not been documented as capable of transmitting Borrelial spirochetes and as such is not considered a Lb vector [78].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Socarras, K.M.; Haslund-Gourley, B.S.; Cramer, N.A.; Comunale, M.A.; Marconi, R.T.; Ehrlich, G.D. Large-Scale Sequencing of Borreliaceae for the Construction of Pan-Genomic-Based Diagnostics. Genes 2022, 13, 1604. https://doi.org/10.3390/genes13091604

AMA Style

Socarras KM, Haslund-Gourley BS, Cramer NA, Comunale MA, Marconi RT, Ehrlich GD. Large-Scale Sequencing of Borreliaceae for the Construction of Pan-Genomic-Based Diagnostics. Genes. 2022; 13(9):1604. https://doi.org/10.3390/genes13091604

Chicago/Turabian Style

Socarras, Kayla M., Benjamin S. Haslund-Gourley, Nicholas A. Cramer, Mary Ann Comunale, Richard T. Marconi, and Garth D. Ehrlich. 2022. "Large-Scale Sequencing of Borreliaceae for the Construction of Pan-Genomic-Based Diagnostics" Genes 13, no. 9: 1604. https://doi.org/10.3390/genes13091604

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop